1
|
Nourazarian A, Aghaei-Zarch SM, Panahi Y. Delayed complications of sulfur mustard poisoning: a focus on inflammation and telomere footprint. Arch Toxicol 2025:10.1007/s00204-025-04033-z. [PMID: 40335638 DOI: 10.1007/s00204-025-04033-z] [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: 02/02/2025] [Accepted: 03/19/2025] [Indexed: 05/09/2025]
Abstract
Sulfur mustard (SM), a potent alkylating agent, has been widely used in chemical warfare, causing severe acute and long-term health complications. While its immediate toxic effects are well documented, the late-onset complications remain poorly understood. Chronic exposure to SM has been linked to persistent oxidative stress, inflammation, and genomic instability, contributing to the progression of various diseases, including pulmonary fibrosis, chronic obstructive pulmonary disease (COPD), and cancer. This review explores the emerging role of telomere biology in the delayed pathophysiology of SM exposure. Evidence suggests that telomere shortening and dysregulation of telomeric repeat-containing RNA (TERRA) may serve as key molecular indicators of SM-induced aging and cellular dysfunction. Furthermore, inflammatory pathways, particularly NF-κB and TGF-β signaling, appear to be closely associated with telomere attrition, perpetuating chronic inflammation and fibrosis. By integrating oxidative stress, inflammation, and telomere dynamics, we propose a novel model linking telomere biology to SM-induced late complications. Understanding these mechanisms could pave the way for targeted therapeutic strategies, including antioxidant and epigenetic interventions, to mitigate long-term effects. Future research should focus on validating telomere-based biomarkers for early detection and exploring novel interventions to alleviate SM-induced chronic health conditions.
Collapse
Affiliation(s)
- Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Seyed Mohsen Aghaei-Zarch
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yasin Panahi
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, 58147-43343, Iran.
| |
Collapse
|
2
|
Perera K, Ghumman M, Sorkhdini P, Norbrun C, Negash S, Zhou Y, Menon JU. Citrus pectin-coated inhalable PLGA nanoparticles for treatment of pulmonary fibrosis. J Mater Chem B 2025; 13:3325-3339. [PMID: 39918485 PMCID: PMC11804936 DOI: 10.1039/d4tb01682c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/26/2025] [Indexed: 02/09/2025]
Abstract
Pulmonary fibrosis (PF) is a chronic interstitial disorder of the respiratory system that can be debilitating as it progresses and has experienced a slow rise in incidence in past years. Treatment is complicated by the complex aetiology of the disease and the off-target effects of the two FDA-approved therapeutics available on the market: pirfenidone and nintedanib. In this work, we propose a multipurpose nanoparticle system consisting of poly(lactic-co-glycolic) acid polymer (PLGA) and a coating of citrus pectin (CP) for galectin-3 targeting and anti-fibrotic therapy. Pectin from citrus peels has been observed to have anti-fibrotic activity in a range of fibrotic tissues, causing a decrease in the expression and activity of galectin-3: a key, upregulated marker of fibrosis. We show that the CP-PLGA nanoparticles (NPs) have an average diameter of 340.5 ± 10.6 nm, compatible with inhalation and retention in the deep lung, and that CP constitutes, on average, 40.3% of the final CP-PLGA formulation. The NPs are well-tolerated by MRC-5 lung fibroblasts up to 2 mg mL-1. We demonstrate the NPs' ability to target transforming growth factor β (TGFβ)-treated fibrotic MRC-5 cells in a specific, dose-dependent manner, saturating at approx. 250 μg mL-1in vitro, and that our NPs have potent anti-fibrotic activity in vivo in particular, reversing bleomycin-induced fibrosis in mouse lungs, accompanied by marked reduction in profibrotic markers including collagen 1, fibronectin, α-smooth muscle actin, β-catenin and galectin-3. In all, we present an inherently therapeutic inhalable nanocarrier for galectin-3 targeting and anti-fibrotic therapy. We envision this carrier to be doubly effective against fibrotic lung tissue when combined with an encapsulated anti-fibrotic drug, improving overall/total therapeutic efficacy and patient compliance via the reduction of off-target effects and additive therapeutic effects.
Collapse
Affiliation(s)
- Kalindu Perera
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Moez Ghumman
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Parand Sorkhdini
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912, USA
| | - Carmelissa Norbrun
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912, USA
| | - Seraphina Negash
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA
| | - Yang Zhou
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912, USA
| | - Jyothi U Menon
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
- Department of Chemical Engineering, College of Engineering, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
3
|
Saygun I, Slezovic MÖ, Özkan CK, Bengi VU, Elçi P, Serdar M, Kantarci A. Anti-proliferative impact of resveratrol on gingival fibroblasts from juvenile hyaline fibromatosis. Clin Oral Investig 2024; 28:448. [PMID: 39060456 PMCID: PMC11281951 DOI: 10.1007/s00784-024-05771-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/11/2024] [Indexed: 07/28/2024]
Abstract
AIM Resveratrol is a natural polyphenolic compound with biological activities such as anti-inflammation and antioxidation. Its anti-fibrotic effect has been experimentally demonstrated in the pancreas and liver. This study aims to determine the anti-proliferative effect of resveratrol on fibroblasts obtained from hyperplastic gingival tissues from a patient diagnosed with Juvenile Hyaline Fibromatosis (JHF). MATERIALS AND METHODS Primary gingival fibroblast cell lines were obtained from gingival growth tissues by the gingivectomy of a patient with JHF. Gingival fibroblasts were treated with or without 3 different doses of resveratrol (50, 100, 200 µM). Cytotoxicity and cell proliferation were evaluated after 24, 48, and 72 h. Collagen, TGF, and CTGF were analyzed by ELISA in the 48-hour supernatants. RESULTS All three doses of resveratrol suppressed the proliferation of JHF gingival fibroblasts at 24 and 48 h without showing any cytotoxic effect compared to the control group (p < 0.0001). At 72 h, 100 and 200 µM resveratrol showed significantly less proliferation (p < 0.0001), less collagen, CTGF, and TGF- β (p < 0.001) than the control group. CONCLUSION Resveratrol had a profound anti-proliferative effect on gingival fibroblasts obtained from gingival enlargements with JHF, suggesting that it can be used as a therapeutic to prevent excessive cell growth by suppressing collagen, CTGF, and TGF- β synthesis in the pathogenesis of hyperplasia.
Collapse
Affiliation(s)
- Işıl Saygun
- Department of Periodontology, Gulhane Faculty of Dental Medicine, University of Health Sciences, Ankara, Turkey.
| | - Melis Özgül Slezovic
- Department of Periodontology, Gulhane Faculty of Dental Medicine, University of Health Sciences, Ankara, Turkey.
| | - Cansel Köse Özkan
- Gulhane Faculty of Pharmacy, University of Health Sciences, Ankara, Turkey
| | - Vahdi Umut Bengi
- Department of Periodontology, Gulhane Faculty of Dental Medicine, University of Health Sciences, Ankara, Turkey
| | - Pınar Elçi
- Gulhane Health Sciences Institute, Stem Cell Lab, University of Health Sciences, Ankara, Turkey
| | - Muhittin Serdar
- Department of Medical Biochemistry, Acıbadem Mehmet Ali Aydınlar University, Ankara, Turkey
| | | |
Collapse
|
4
|
Luo J, Li P, Dong M, Zhang Y, Lu S, Chen M, Zhou H, Lin N, Jiang H, Wang Y. SLC15A3 plays a crucial role in pulmonary fibrosis by regulating macrophage oxidative stress. Cell Death Differ 2024; 31:417-430. [PMID: 38374230 PMCID: PMC11043330 DOI: 10.1038/s41418-024-01266-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/21/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal and irreversible disease with few effective treatments. Alveolar macrophages (AMs) are involved in the development of IPF from the initial stages due to direct exposure to air and respond to external oxidative damage (a major inducement of pulmonary fibrosis). Oxidative stress in AMs plays an indispensable role in promoting fibrosis development. The oligopeptide histidine transporter SLC15A3, mainly expressed on the lysosomal membrane of macrophages and highly expressed in the lung, has proved to be involved in innate immune and antiviral signaling pathways. In this study, we demonstrated that during bleomycin (BLM)- or radiation-induced pulmonary fibrosis, the recruitment of macrophages induced an increase of SLC15A3 in the lung, and the deficiency of SLC15A3 protected mice from pulmonary fibrosis and maintained the homeostasis of the pulmonary microenvironment. Mechanistically, deficiency of SLC15A3 resisted oxidative stress in macrophages, and SLC15A3 interacted with the scaffold protein p62 to regulate its expression and phosphorylation activation, thereby regulating p62-nuclear factor erythroid 2-related factor 2 (NRF2) antioxidant stress pathway protein, which is related to the production of reactive oxygen species (ROS). Overall, our data provided a novel mechanism for targeting SLC15A3 to regulate oxidative stress in macrophages, supporting the therapeutic potential of inhibiting or silencing SLC15A3 for the precautions and treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Jun Luo
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ping Li
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China
| | - Minlei Dong
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yingqiong Zhang
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Shuanghui Lu
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Mingyang Chen
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hui Zhou
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Nengming Lin
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China
| | - Huidi Jiang
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
| | - Yuqing Wang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, China.
| |
Collapse
|
5
|
Ozawa S, Mukudai S, Kaneko M, Kinoshita S, Hashimoto K, Sugiyama Y, Hashimoto S, Akaki J, Hirano S. Anti-inflammatory and Antioxidant Effects of Japanese Herbal Medicine Kyoseihatekigan on Vocal Fold Wound Healing. J Voice 2024; 38:503-509. [PMID: 34836738 DOI: 10.1016/j.jvoice.2021.10.003] [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: 04/30/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVES The Japanese herbal medicine kyoseihatekigan (KHG) has been used to alleviate the symptoms of croaky voice and globus hystericus, and each of its components has anti-inflammatory and antioxidant effects. However, the mechanisms underlying these beneficial actions of KHG on the vocal folds remain largely unknown. We examined the effects of KHG on rat vocal fold wound healing and assessed its anti-inflammatory and antioxidant properties. STUDY DESIGN Animal model. METHODS The vocal folds of Sprague-Dawley rats were unilaterally injured under endoscopy. Rats were divided into three groups based on KHG dosing from pre injury day 4 to post injury day 3: 0 mg/kg/day (sham group), 500 mg/kg/day (1% KHG group) and 1000 mg/kg/day (2% KHG group). Histologic changes were examined to assess the degree of inflammation and oxidative stress at day 3, and fibrosis at day 56. In addition, gene expression related to pro-inflammatory cytokines and transforming growth factor-beta1 (TGF-β1) signaling was examined by quantitative real-time polymerase chain reaction (qPCR). RESULTS Histologic analysis showed that the 1% and 2% KHG treatments significantly decreased cell infiltration and the 4-hydroxy-2-nonenalx-immunopositive area, and increased hyaluronic acid at day 3. Both KHG treatments significantly decreased fibrosis at day 56. qPCR revealed that mRNA of interleukin-1β and cyclooxygenase-2 were significantly suppressed at day 1 and TGF-β1 mRNA was significantly downregulated at day 5 in both KHG groups. CONCLUSIONS The current findings suggest that KHG has anti-inflammatory and antioxidant effects in the early phase of vocal fold wound healing, which can lead to better wound healing with less scar formation.
Collapse
Affiliation(s)
- Satomi Ozawa
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeyuki Mukudai
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Mami Kaneko
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shota Kinoshita
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Keiko Hashimoto
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoichiro Sugiyama
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Junji Akaki
- Kobayashi Pharmaceutical Co., Ltd., Osaka, Japan
| | - Shigeru Hirano
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
6
|
Cui Y, Yang Z, Lv Z, Lei J. Disruption of extracellular redox balance drives persistent lung fibrosis and impairs fibrosis resolution. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166842. [PMID: 37558008 DOI: 10.1016/j.bbadis.2023.166842] [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: 03/21/2023] [Revised: 07/13/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023]
Abstract
Lung fibrosis is a devastating outcome of various diffuse parenchymal lung diseases. Despite rigorous research efforts, the mechanisms that propagate its progressive and nonresolving nature remain enigmatic. Oxidative stress has been implicated in the pathogenesis of lung fibrosis. However, the role of extracellular redox state in disease progression and resolution remains largely unexplored. Here, we show that compartmentalized control over extracellular reactive oxygen species (ROS) by aerosolized delivery of recombinant extracellular superoxide dismutase (ECSOD) suppresses an established bleomycin-induced fibrotic process in mice. Further analysis of publicly available microarray, RNA-seq and single-cell RNAseq datasets reveals a significant decrease in ECSOD expression in fibrotic lung tissues that can be spontaneously restored during fibrosis resolution. Therefore, we investigate the effect of siRNA-mediated ECSOD depletion during the established fibrotic phase on the self-limiting nature of the bleomycin mouse model. Our results demonstrate that in vivo knockdown of ECSOD in mouse fibrotic lungs impairs fibrosis resolution. Mechanistically, we demonstrate that transforming growth factor (TGF)-β1 downregulates endogenous ECSOD expression, leading to the accumulation of extracellular superoxide via Smad-mediated signaling and the activation of additional stores of latent TGF-β1. In addition, depletion of endogenous ECSOD during the fibrotic phase in the bleomycin model induces an apoptosis-resistant phenotype in lung fibroblasts through unrestricted Akt signaling. Taken together, our data strongly support the critical role of extracellular redox state in fibrosis persistence and resolution. Based on these findings, we propose that compartment-specific control over extracellular ROS may be a potential therapeutic strategy for managing fibrotic lung disorders.
Collapse
Affiliation(s)
- Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, People's Republic of China.
| | - Zeran Yang
- Interventional Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, People's Republic of China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, People's Republic of China
| | - Jianfeng Lei
- Medical Imaging Laboratory, Research Core Facilities, Capital Medical University, Beijing 100069, People's Republic of China
| |
Collapse
|
7
|
Liu Q, Ren Y, Jia H, Yuan H, Tong Y, Kotha S, Mao X, Huang Y, Chen C, Zheng Z, Wang L, He W. Vanadium Carbide Nanosheets with Broad-Spectrum Antioxidant Activity for Pulmonary Fibrosis Therapy. ACS NANO 2023; 17:22527-22538. [PMID: 37933888 DOI: 10.1021/acsnano.3c06105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Idiopathic pulmonary fibrosis is a chronic and highly lethal lung disease that largely results from oxidative stress; however, effective antioxidant therapy by targeting oxidative stress pathogenesis is still lacking. The big challenge is to develop an ideal antioxidant material with superior antifibrotic effects. Herein, we report that V4C3 nanosheets (NSs) can serve as a potential antioxidant for treatment of pulmonary fibrosis by scavenging reactive oxygen and nitrogen species. Interestingly, subtle autoxidation can adjust the valence composition of V4C3 NSs and significantly improve their antioxidant behavior. Valence engineering triggers multiple antioxidant mechanisms including electron transfer, H atom transfer, and enzyme-like catalysis, thus endowing V4C3 NSs with broad-spectrum, high-efficiency, and persistent antioxidant capacity. Benefiting from antioxidant properties and good biocompatibility, V4C3 NSs can significantly prevent myofibroblast proliferation and extracellular matrix abnormality, thus alleviating the progression of bleomycin-induced pulmonary fibrosis in vivo by scavenging ROS, anti-inflammation, and rebuilding antioxidant defenses. This study not only provides an important strategy for designing excellent antioxidant nanomaterials, but also proposes a proof-of-concept demonstration for the treatment of pulmonary fibrosis and other oxidative stress-related diseases.
Collapse
Affiliation(s)
- Quan Liu
- Key Laboratory of Micro-Nano Materials for Energy Storage and Conversion of Henan Province, Institute of Surface Micro and Nano Materials, College of Chemical and Materials Engineering, Xuchang University, Xuchang, Henan 461000, P. R. China
- School of Materials, North China University of Water Resources and Electric Power, Zhengzhou, Henan 450045, P. R. China
| | - Yaping Ren
- Key Laboratory of Micro-Nano Materials for Energy Storage and Conversion of Henan Province, Institute of Surface Micro and Nano Materials, College of Chemical and Materials Engineering, Xuchang University, Xuchang, Henan 461000, P. R. China
- Laboratory for NanoMedical Photonics, School of Basic Medical Science, Henan University, Zhengzhou 450046, P. R. China
| | - Huimin Jia
- Key Laboratory of Micro-Nano Materials for Energy Storage and Conversion of Henan Province, Institute of Surface Micro and Nano Materials, College of Chemical and Materials Engineering, Xuchang University, Xuchang, Henan 461000, P. R. China
| | - Hao Yuan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS-HKU Joint Laboratory of Metallomics on Health and Environment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yuping Tong
- School of Materials, North China University of Water Resources and Electric Power, Zhengzhou, Henan 450045, P. R. China
| | - Sumasri Kotha
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Xiaobo Mao
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Yongwei Huang
- Laboratory for NanoMedical Photonics, School of Basic Medical Science, Henan University, Zhengzhou 450046, P. R. China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Zhi Zheng
- Key Laboratory of Micro-Nano Materials for Energy Storage and Conversion of Henan Province, Institute of Surface Micro and Nano Materials, College of Chemical and Materials Engineering, Xuchang University, Xuchang, Henan 461000, P. R. China
| | - Liming Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS-HKU Joint Laboratory of Metallomics on Health and Environment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Weiwei He
- Key Laboratory of Micro-Nano Materials for Energy Storage and Conversion of Henan Province, Institute of Surface Micro and Nano Materials, College of Chemical and Materials Engineering, Xuchang University, Xuchang, Henan 461000, P. R. China
| |
Collapse
|
8
|
Yang F, Wendusubilige, Kong J, Zong Y, Wang M, Jing C, Ma Z, Li W, Cao R, Jing S, Gao J, Li W, Wang J. Identifying oxidative stress-related biomarkers in idiopathic pulmonary fibrosis in the context of predictive, preventive, and personalized medicine using integrative omics approaches and machine-learning strategies. EPMA J 2023; 14:417-442. [PMID: 37605652 PMCID: PMC10439879 DOI: 10.1007/s13167-023-00334-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/09/2023] [Indexed: 08/23/2023]
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a rare interstitial lung disease with a poor prognosis that currently lacks effective treatment methods. Preventing the acute exacerbation of IPF, identifying the molecular subtypes of patients, providing personalized treatment, and developing individualized drugs are guidelines for predictive, preventive, and personalized medicine (PPPM / 3PM) to promote the development of IPF. Oxidative stress (OS) is an important pathological process of IPF. However, the relationship between the expression levels of oxidative stress-related genes (OSRGs) and clinical indices in patients with IPF is unclear; therefore, it is still a challenge to identify potential beneficiaries of antioxidant therapy. Because PPPM aims to recognize and manage diseases by integrating multiple methods, patient stratification and analysis based on OSRGs and identifying biomarkers can help achieve the above goals. Methods Transcriptome data from 250 IPF patients were divided into training and validation sets. Core OSRGs were identified in the training set and subsequently clustered to identify oxidative stress-related subtypes. The oxidative stress scores, clinical characteristics, and expression levels of senescence-associated secretory phenotypes (SASPs) of different subtypes were compared to identify patients who were sensitive to antioxidant therapy to conduct differential gene functional enrichment analysis and predict potential therapeutic drugs. Diagnostic markers between subtypes were obtained by integrating multiple machine learning methods, their expression levels were tested in rat models with different degrees of pulmonary fibrosis and validation sets, and nomogram models were constructed. CIBERSORT, single-cell RNA sequencing, and immunofluorescence staining were used to explore the effects of OSRGs on the immune microenvironment. Results Core OSRGs classified IPF into two subtypes. Patients classified into subtypes with low oxidative stress levels had better clinical scores, less severe fibrosis, and lower expression of SASP-related molecules. A reliable nomogram model based on five diagnostic markers was constructed, and these markers' expression stability was verified in animal experiments. The number of neutrophils in the immune microenvironment was significantly different between the two subtypes and was closely related to the degree of fibrosis. Conclusion Within the framework of PPPM, this work comprehensively explored the role of OSRGs and their mediated cellular senescence and immune processes in the progress of IPF and assessed their capabilities aspredictors of high oxidative stress and disease progression,targets of the vicious loop between regulated pulmonary fibrosis and OS for targeted secondary and tertiary prevention, andreferences for personalized antioxidant and antifibrotic therapies. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-023-00334-4.
Collapse
Affiliation(s)
- Fan Yang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wendusubilige
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Institute of Ethnic Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jingwei Kong
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuhan Zong
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Manting Wang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chuanqing Jing
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhaotian Ma
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Institute of Ethnic Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wanyang Li
- Department of Clinical Nutrition, Chinese Academy of Medical Sciences - Peking Union Medical College, Peking Union Medical College Hospital (Dongdan campus), Beijing, China
| | - Renshuang Cao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuwen Jing
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Gao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenxin Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ji Wang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
9
|
Makena P, Kikalova T, Prasad GL, Baxter SA. Oxidative Stress and Lung Fibrosis: Towards an Adverse Outcome Pathway. Int J Mol Sci 2023; 24:12490. [PMID: 37569865 PMCID: PMC10419527 DOI: 10.3390/ijms241512490] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Lung fibrosis is a progressive fatal disease in which deregulated wound healing of lung epithelial cells drives progressive fibrotic changes. Persistent lung injury due to oxidative stress and chronic inflammation are central features of lung fibrosis. Chronic cigarette smoking causes oxidative stress and is a major risk factor for lung fibrosis. The objective of this manuscript is to develop an adverse outcome pathway (AOP) that serves as a framework for investigation of the mechanisms of lung fibrosis due to lung injury caused by inhaled toxicants, including cigarette smoke. Based on the weight of evidence, oxidative stress is proposed as a molecular initiating event (MIE) which leads to increased secretion of proinflammatory and profibrotic mediators (key event 1 (KE1)). At the cellular level, these proinflammatory signals induce the recruitment of inflammatory cells (KE2), which in turn, increase fibroblast proliferation and myofibroblast differentiation (KE3). At the tissue level, an increase in extracellular matrix deposition (KE4) subsequently culminates in lung fibrosis, the adverse outcome. We have also defined a new KE relationship between the MIE and KE3. This AOP provides a mechanistic platform to understand and evaluate how persistent oxidative stress from lung injury may develop into lung fibrosis.
Collapse
Affiliation(s)
- Patrudu Makena
- RAI Services Company, P.O. Box 1487, Winston-Salem, NC 27102, USA;
| | - Tatiana Kikalova
- Clarivate Analytics, 1500 Spring Garden, Philadelphia, PA 19130, USA
| | - Gaddamanugu L. Prasad
- Former Employee of RAI Services Company, Winston-Salem, NC 27101, USA
- Prasad Scientific Consulting LLC, 490 Friendship Place Ct, Lewisville, NC 27023, USA
| | - Sarah A. Baxter
- RAI Services Company, P.O. Box 1487, Winston-Salem, NC 27102, USA;
| |
Collapse
|
10
|
Zhang Z, Huang W, Ren F, Luo L, Zhou J, Tang L. Measurement of superoxide dismutase: clinical usefulness for patients with anti-neutrophil cytoplasmic antibody-associated vasculitis. Adv Rheumatol 2023; 63:28. [PMID: 37381048 DOI: 10.1186/s42358-023-00312-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/15/2023] [Indexed: 06/30/2023] Open
Abstract
OBJECTIVE To investigate the clinical usefulness of serum superoxide dismutase (SOD) measurement in patients with anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). METHODS In this single-center retrospective study, demographic data, serum SOD levels, erythrocyte sedimentation rate (ESR), C reactive protein (CRP), the Birmingham Vasculitis Activity Score (BVAS), ANCA, organ involvement, and outcomes were analyzed for 152 AAV patients hospitalized in the Second Affiliated Hospital of Chongqing Medical University. Meanwhile, the serum SOD levels of 150 healthy people were collected as the control group. RESULTS Compared to the healthy control group, serum SOD levels of the AAV group were significantly lower (P < 0.001). SOD levels of AAV patients were negatively correlated to ESR, CRP, and BVAS (ESR rho = - 0.367, P < 0.001; CRP rho = - 0.590, P < 0.001; BVAS rho = - 0.488, P < 0.001). SOD levels for the MPO-ANCA group were significantly lower than the PR3-ANCA group (P = 0.045). SOD levels for the pulmonary involvement group and the renal involvement group were significantly lower than those for the non-pulmonary involvement group and the non-renal involvement group (P = 0.006; P < 0.001, respectively). SOD levels in the death group were significantly lower than the survival group (P = 0.001). CONCLUSIONS In AAV patients, low SOD levels might indicate disease associated oxidative stress. SOD levels in AAV patients were decreased with inflammation, suggesting that SOD levels could potentially be a surrogate marker for disease activity. SOD levels in AAV patients were closely related to ANCA serology, pulmonary involvement, and renal involvement, with low SOD levels an important indicator of a poor prognosis for AAV patients.
Collapse
Affiliation(s)
- Zhihuan Zhang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Wenhan Huang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Feifeng Ren
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Lei Luo
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Jun Zhou
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Lin Tang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
11
|
Chen D, Ai X, Li Y, Li Y, Ao Y, Rong J, Li G. Protective effects of Cu/Zn-SOD and Mn-SOD on UVC radiation-induced damage in NIH/3T3 cells and murine skin. Acta Histochem 2023; 125:152030. [PMID: 37099996 DOI: 10.1016/j.acthis.2023.152030] [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: 03/07/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/28/2023]
Abstract
Superoxide dismutase (SOD) is an antioxidant enzyme with multiple metal cofactors that can specifically clear reactive oxygen species (ROS), which plays an important role in a variety of ultraviolet-induced lesions. Therefore, SOD has the anti-ultraviolet radiation effect. The objective of this study was to compare the differences in the anti-ultraviolet radiation effect of SOD with distinct metal cofactors: Cu/Zn-SOD and Mn-SOD. SOD was first purified using hydrophobic interaction chromatography and ion-exchange chromatography. Second, the Methylthiazolyldiphenyl-tetrazolium bromide method and cell senescence kits were used to study the protective effect of SOD on ultraviolet-induced cell damage. Finally, the protective effect of SOD on ultraviolet -induced skin damage was histopathologically evaluated, and the expression levels of malondialdehyde (MDA) and matrix metalloproteinases (MMPs) in tissues were detected. The results showed that Cu/Zn-SOD was superior to Mn-SOD in promoting cell proliferation, alleviating cell damage, protecting skin structure, and regulating the expression levels of MDA and MMPs, and it has no side effects. In conclusion, Cu/Zn-SOD had a better anti-ultraviolet radiation effect than Mn-SOD, and it can be used in anti-aging and anti-ultraviolet skin-care products.
Collapse
Affiliation(s)
- Disi Chen
- College of Life Science, Yangtze University, Jingzhou 434000, PR China
| | - Xiaoyang Ai
- College of Life Science, Yangtze University, Jingzhou 434000, PR China
| | - Yang Li
- College of Life Science, Yangtze University, Jingzhou 434000, PR China
| | - Yue Li
- College of Life Science, Yangtze University, Jingzhou 434000, PR China
| | - Yunfan Ao
- College of Life Science, Yangtze University, Jingzhou 434000, PR China
| | - Jun Rong
- College of Life Science, Yangtze University, Jingzhou 434000, PR China; Jingzhou Changxin Biotechnology Co., Ltd., Jingzhou 434000, PR China.
| | - Guopan Li
- College of Life Science, Yangtze University, Jingzhou 434000, PR China; Jingzhou Changxin Biotechnology Co., Ltd., Jingzhou 434000, PR China.
| |
Collapse
|
12
|
Li Y, Lian Z, Li Q, Ding W, Wang W, Zhang L, Muhataer X, Zhou Y, Yang X, Wu C. Molecular mechanism by which the Notch signaling pathway regulates autophagy in a rat model of pulmonary fibrosis in pigeon breeder's lung. Open Med (Wars) 2023; 18:20230629. [PMID: 36785767 PMCID: PMC9921914 DOI: 10.1515/med-2023-0629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/17/2022] [Accepted: 12/09/2022] [Indexed: 02/11/2023] Open
Abstract
This study investigated the molecular mechanisms underlying the involvement of the Notch signaling pathway and autophagy in the development of pulmonary fibrosis in pigeon breeder's lung (PBL). Rats were divided into control (Ctrl), PBL model (M), M + D (Notch signaling inhibition), M + W (autophagy inhibition), and M + R (autophagy induction) groups. Lyophilized protein powder from pigeon shedding materials was used as an allergen to construct a fibrotic PBL rat model. The mechanism by which Notch signaling regulated autophagy in the pulmonary fibrosis of PBL was investigated by inhibiting the Notch pathway and interfering with autophagy. Pulmonary interstitial fibrosis was significantly greater in the M group and the M + W group than in the M + D and M + R groups. The expression of α-smooth muscle actin was significantly higher in the M, M + D, and M + W groups than in the Ctrl group (P < 0.05). The expression of the cell autophagy markers Beclin1 and LC3 was lower in the M, M + D, and M + W groups than in the Ctrl group (P < 0.05), whereas Beclin1 and LC3 expressions were higher in the M + D and M + R groups than in the M group. The levels of reactive oxygen species in serum and lung tissues were higher in the M, M + D, M + W, and M + R groups than in the Ctrl group (P < 0.05). The Notch signaling pathway is involved in the pathological process of pulmonary fibrosis in the rat model of PBL by regulating autophagy.
Collapse
Affiliation(s)
- Yafang Li
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, 830001 Urumqi, China,Xinjiang Clinical Research Center for Interstitial Lung Diseases, 830001 Urumqi, China
| | - Zhichuang Lian
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, 830001 Urumqi, China
| | - Qifeng Li
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, 830001 Urumqi, China
| | - Wei Ding
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, 830001 Urumqi, China
| | - Wenyi Wang
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, 830001 Urumqi, China
| | - Ling Zhang
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, 830001 Urumqi, China
| | - Xirennayi Muhataer
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, 830001 Urumqi, China
| | - Yuan Zhou
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, 830001 Urumqi, China
| | - Xiaohong Yang
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, 830001 Urumqi, China,Xinjiang Clinical Research Center for Interstitial Lung Diseases, 830001 Urumqi, China
| | - Chao Wu
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, 830001 Urumqi, China,Xinjiang Clinical Research Center for Interstitial Lung Diseases, 830001 Urumqi, China
| |
Collapse
|
13
|
Ning W, Xu X, Zhou S, Wu X, Wu H, Zhang Y, Han J, Wang J. Effect of high glucose supplementation on pulmonary fibrosis involving reactive oxygen species and TGF-β. Front Nutr 2022; 9:998662. [PMID: 36304232 PMCID: PMC9593073 DOI: 10.3389/fnut.2022.998662] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/21/2022] [Indexed: 11/24/2022] Open
Abstract
This study explored the profibrotic impact of high glucose in the lung and potential mechanisms using latent TGF-β1-induced human epithelial cell pulmonary fibrosis and bleomycin (BLM)-induced pulmonary fibrosis models. Results demonstrated that high glucose administration induced epithelial–mesenchymal transition (EMT) in human epithelial cells in a dose-dependent manner via activating latent TGF-β1, followed by increased expression of mesenchymal-related proteins and decreased expression of epithelial marker protein E-cadherin. Further mechanism analysis showed that administration of high glucose dose-dependently promoted total and mitochondrial reactive oxygen species (ROS) accumulation in human epithelial cells, which promoted latent TGF-β1 activation. However, N-acetyl-L-cysteine, a ROS eliminator, inhibited such effects. An in vivo feed study found that mice given a high-glucose diet had more seriously pathological characteristics of pulmonary fibrosis in BLM-treated mice, including increasing infiltrated inflammatory cells, collagen I deposition, and the expression of mesenchymal-related proteins while decreasing the expression of the epithelial marker E-cadherin. In addition, high glucose intake further increased TGF-β1 concentration and upregulated p-Smad2/3 and snail in lung tissues from BLM-treated mice when compared to BLM-treated mice. Finally, supplementation with high glucose further increased the production of lipid peroxidation metabolite malondialdehyde and decreased superoxide dismutase activity in BLM-treated mice. Collectively, these findings illustrate that high glucose supplementation activates a form of latent TGF-β1 by promoting ROS accumulation and ultimately exacerbates the development of pulmonary fibrosis.
Collapse
|
14
|
Wang B, Yu L, Liu W, Yang M, Fan L, Zhou M, Ma J, Wang X, Nie X, Cheng M, Qiu W, Ye Z, Song J, Chen W. Cross-sectional and longitudinal associations of acrolein exposure with pulmonary function alteration: Assessing the potential roles of oxidative DNA damage, inflammation, and pulmonary epithelium injury in a general adult population. ENVIRONMENT INTERNATIONAL 2022; 167:107401. [PMID: 35850081 DOI: 10.1016/j.envint.2022.107401] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/13/2022] [Accepted: 07/06/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Acrolein is a significant high priority hazardous air pollutant with pulmonary toxicity and the leading cause of most noncancer adverse respiratory effects among air toxics that draws great attention. Whether and how acrolein exposure impacts pulmonary function remain inconclusive. OBJECTIVES To assess the association of acrolein exposure with pulmonary function and the underlying roles of oxidative DNA damage, inflammation, and pulmonary epithelium integrity. METHODS Among 3,279 Chinese adults from the Wuhan-Zhuhai cohort, associations of urinary acrolein metabolites (N-Acetyl-S-(2-carboxyethyl)-L-cysteine, CEMA; N-Acetyl-S-(3-hydroxypropyl)-L-cysteine, 3HPMA) as credible biomarkers of acrolein exposure with pulmonary function were analyzed by linear mixed models. Joint effects of biomarkers of oxidative DNA damage (8-hydroxy-deoxyguanosine), inflammation (C-reactive protein, CRP), and pulmonary epithelium integrity (Club cell secretory protein, CC16) with acrolein metabolites on pulmonary function and the mediating roles of these biomarkers were assessed. Besides, a subgroup (N = 138) was randomly recruited from the cohort to assess the stabilities of acrolein metabolites and their longitudinal associations with pulmonary function change in three years. RESULTS Significant inverse dose-response relationships between acrolein metabolites and pulmonary function were found. Each 10-fold increment in CEMA, 3HPMA, or ΣUACLM (CEMA + 3HPMA) was cross-sectionally related to a 68.56-, 40.98-, or 46.02-ml reduction in FVC and a 61.54-, 43.10-, or 50.14-ml reduction in FEV1, respectively (P < 0.05). Furthermore, acrolein metabolites with fair to excellent stabilities were found to be longitudinally associated with pulmonary function decline in three years. Joint effects of acrolein metabolites with 8-hydroxy-deoxyguanosine, CRP, and CC16 on pulmonary function were identified. CRP significantly mediated 5.97% and 5.51% of CEMA-associated FVC and FEV1 reductions, respectively. 8-hydroxy-deoxyguanosine significantly mediated 6.78%, 6.88%, and 7.61% of CEMA-, 3HPMA-, and ΣUACLM-associated FVC reductions, respectively. CONCLUSIONS Acrolein exposure of general adults was cross-sectionally and longitudinally related to pulmonary function decline, which was aggravated and/or partly mediated by oxidative DNA damage, inflammation, and pulmonary epithelium injury.
Collapse
Affiliation(s)
- Bin Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Linling Yu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wei Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Meng Yang
- Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430019, China
| | - Lieyang Fan
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Min Zhou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jixuan Ma
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xing Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiuque Nie
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Man Cheng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Weihong Qiu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zi Ye
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jiahao Song
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Weihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
15
|
Jeyanathan V, Afkhami S, D’Agostino MR, Zganiacz A, Feng X, Miller MS, Jeyanathan M, Thompson MR, Xing Z. Differential Biodistribution of Adenoviral-Vectored Vaccine Following Intranasal and Endotracheal Deliveries Leads to Different Immune Outcomes. Front Immunol 2022; 13:860399. [PMID: 35757753 PMCID: PMC9231681 DOI: 10.3389/fimmu.2022.860399] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 05/11/2022] [Indexed: 11/22/2022] Open
Abstract
Infectious diseases of the respiratory tract are one of the top causes of global morbidity and mortality with lower respiratory tract infections being the fourth leading cause of death. The respiratory mucosal (RM) route of vaccine delivery represents a promising strategy against respiratory infections. Although both intranasal and inhaled aerosol methods have been established for human application, there is a considerable knowledge gap in the relationship of vaccine biodistribution to immune efficacy in the lung. Here, by using a murine model and an adenovirus-vectored model vaccine, we have compared the intranasal and endotracheal delivery methods in their biodistribution, immunogenicity and protective efficacy. We find that compared to intranasal delivery, the deepened and widened biodistribution in the lung following endotracheal delivery is associated with much improved vaccine-mediated immunogenicity and protection against the target pathogen. Our findings thus support further development of inhaled aerosol delivery of vaccines over intranasal delivery for human application.
Collapse
Affiliation(s)
- Vidthiya Jeyanathan
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Michael R. D’Agostino
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Anna Zganiacz
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Xueya Feng
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Matthew S. Miller
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Mangalakumari Jeyanathan
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Michael R. Thompson
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Zhou Xing
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, Hamilton, ON, Canada,Department of Medicine, McMaster University, Hamilton, ON, Canada,*Correspondence: Zhou Xing,
| |
Collapse
|
16
|
Assayag M, Goldstein S, Samuni A, Kaufman A, Berkman N. The nitroxide/antioxidant 3-carbamoyl proxyl attenuates disease severity in murine models of severe asthma. Free Radic Biol Med 2021; 177:181-188. [PMID: 34678420 DOI: 10.1016/j.freeradbiomed.2021.10.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 11/19/2022]
Abstract
Asthma is characterized by airway inflammation, hyper-responsiveness, symptoms of dyspnea, wheezing and coughing. In most patients, asthma is well controlled using inhaled corticosteroids and bronchodilators. A minority of patients with asthma develop severe disease, which is frequently only partially responsive or even resistant to treatment with corticosteroids. Severe refractory asthma is associated with structural changes in the airways, termed "airway remodeling", and/or with neutrophilic rather than eosinophilic airway inflammation. While oxidative stress plays an important role in the pathophysiology of asthma, cyclic nitroxide stable radicals, which are unique and efficient catalytic antioxidants, effectively protect against oxidative injury. We have demonstrated that the nitroxide 3-carbamoyl proxyl (3-CP) attenuates airway inflammation and hyperresponsiveness in allergic asthma as well as bleomycin-induced fibrosis both using murine models, most probably through modulation of oxidative stress. The present study evaluates the effect of 3-CP on airway inflammation and remodeling using two murine models of severe asthma where mice are sensitized and challenged either by ovalbumin (OVA) or by house dust mite (HDM). 3-CP was orally administered during the entire period of the experiment or during the challenge period alone where its effect was compared to that of dexamethasone. The induced increase by OVA and by HDM of BALf cell counts, airway hyperresponsiveness, fibrosis, transforming growth factor-beta (TGF-β) levels in BALf and protein nitration levels of the lung tissue was significantly reduced by 3-CP. The effect of 3-CP, using two different murine models of severe asthma, is associated at least partially with attenuation of oxidative stress and with TGF-β expression in the lungs. The results of this study suggest a potential use of 3-CP as a novel therapeutic agent in different forms of severe asthma.
Collapse
Affiliation(s)
- Miri Assayag
- Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, 91120, Israel
| | - Sara Goldstein
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel.
| | - Amram Samuni
- Institute of Medical Research, Israel-Canada Medical School, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Alexander Kaufman
- Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, 91120, Israel
| | - Neville Berkman
- Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, 91120, Israel
| |
Collapse
|
17
|
Yu X, Su Q, Geng J, Liu H, Liu Y, Liu J, Shi Y, Zou Y. Ginkgo biloba leaf extract prevents diabetic nephropathy through the suppression of tissue transglutaminase. Exp Ther Med 2021; 21:333. [PMID: 33732306 PMCID: PMC7903480 DOI: 10.3892/etm.2021.9764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/18/2021] [Indexed: 12/28/2022] Open
Abstract
The present study aimed to investigate the preventive effects of Ginkgo biloba leaf extract (GBE) against extracellular matrix (ECM) accumulation in a streptozotocin (STZ)-induced rat model of diabetic nephropathy (DN), and to determine its underlying molecular mechanism. In vivo, a rat model of DN was established by intraperitoneal injection of STZ, and the rats were subsequently administered GBE. The results demonstrated that GBE significantly decreased blood glucose, the urine protein excretion rate and ECM accumulation in DN rats. In addition, the development of DN significantly induced tissue transglutaminase (tTG) protein expression, which was detected by immunohistochemistry, western blotting and PCR analyses, while GBE administration decreased tTG expression in the diabetic kidney. In vitro, rat glomerular mesangial cells (HBZY-1 cells) cultured with high glucose were also treated with GBE. The concentrations of tTG, fibronectin, type IV collagen, transforming growth factor (TGF)-β and connective tissue growth factor (CTGF) were detected via ELISA. The results demonstrated that GBE notably decreased the concentration of these proteins, and tTG expression was positively associated with TGF-β. GBE also suppressed tTG expression of high glucose-treated HBZY-1 cells in a concentration-dependent manner. Furthermore, tTG protein expression was detected in high glucose-treated HBZY-1 cells transfected with small interfering RNA (siRNA) oligonucleotides against TGF-β and CTGF to investigate a possible mechanism of GBE-mediated inhibition of tTG. The results demonstrated that the tTG levels remained unchanged in CTGF siRNA-transfected cells, but were decreased in the GBE + CTGF siRNA group compared with the control siRNA group, suggesting that tTG may not be regulated by CTGF, and the inhibitory effect of GBE on tTG may not be associated with the direct inhibition of CTGF. However, tTG expression was decreased following the transfection with TGF-β siRNA, in which levels of tTG were similar compared with both the GBE group and GBE + TGF-β siRNA group, indicating that tTG may be regulated by TGF-β, and that the GBE-induced repression of tTG expression may be associated with the downregulation of TGF-β. Taken together, the results of the present study suggest that GBE prevented ECM accumulation by suppressing tTG expression in DN, which was predominantly mediated by TGF-β.
Collapse
Affiliation(s)
- Xiaoyan Yu
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Qing Su
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jianan Geng
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hui Liu
- Department of Anatomy, College of Basic Medical Science, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yumeng Liu
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jinming Liu
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yan Shi
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yinggang Zou
- Department of Obstetrics and Gynecology, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
18
|
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
|
19
|
Pan L, Lu Y, Li Z, Tan Y, Yang H, Ruan P, Li R. Ginkgo biloba Extract EGb761 Attenuates Bleomycin-Induced Experimental Pulmonary Fibrosis in Mice by Regulating the Balance of M1/M2 Macrophages and Nuclear Factor Kappa B (NF-κB)-Mediated Cellular Apoptosis. Med Sci Monit 2020; 26:e922634. [PMID: 32799214 PMCID: PMC7448693 DOI: 10.12659/msm.922634] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background The aim of this study was to show whether the standardized Ginkgo biloba extract EGb761, a traditional Chinese medicine, has a therapeutic effect on pulmonary fibrosis (PF). Material/Methods Bleomycin (BLM) was used for establishing the PF mouse model. The mice were treated with a gradient of EGb761 for 28 days to determine an appropriate drug dose. On day 28, the effect of EGb761 on lung injury and inflammation was confirmed by hematoxylin and eosin and Masson staining and evaluated by pulmonary alveolitis and Ashcroft score. The balance of M1/M2 macrophages was evaluated with the respective markers inducible nitric oxide synthase and and interleukin-10 by real-time polymerase chain reaction. Furthermore, the expressions of fibrosis-associated protein α-smooth muscle actin (SMA), related inflammatory protein transforming growth factor (TGF)-β1, the apoptosis-related proteins B-cell lymphoma-associated X protein (Bax), B-cell lymphoma (Bcl)-2, caspase-3, caspase-9, and phosphorylated nuclear factor (NF)-κB (p65) were assessed by western blot. Results On day 28, PF was induced by treating with BLM, whereas EGb761 suppressed the PF of lung tissue. The BLM-induced imbalance of M1/M2 macrophages was reduced by EGb761. Furthermore, the increasing amounts of α-SMA and TGF-β1 induced by BLM were suppressed by EGb761. In addition, the protein or messenger ribonucleic acid expression levels of phosphorylated NF-κB (p65), caspase-3, and caspase-9 were upregulated, whereas Bax and Bcl-2 were downregulated. Treatment with EGb761 restored the levels of these proteins except for caspase-9. Conclusions This study illustrated the protective effect of EGb761 on BLM-induced PF by regulating the balance of M1/M2 macrophages and NF-κB (p65)-mediated apoptosis. The results demonstrated the potential clinical therapeutic effect of EGb761, providing a novel possibility for curing PF.
Collapse
Affiliation(s)
- Ling Pan
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| | - Yuehong Lu
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| | - Zhanhua Li
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| | - Yuping Tan
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| | - Hongmei Yang
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| | - Ping Ruan
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| | - Ruixiang Li
- Respiratory Medicine Department, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medicine University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
20
|
Song MK, Kim DI, Lee K. Time-course transcriptomic alterations reflect the pathophysiology of polyhexamethylene guanidine phosphate-induced lung injury in rats. Inhal Toxicol 2020; 31:457-467. [PMID: 31971030 DOI: 10.1080/08958378.2019.1707912] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Objective: Humidifier-disinfectant-induced lung injury is a new syndrome associated with a high mortality rate and characterized by severe hypersensitivity pneumonitis, acute interstitial pneumonia, or acute respiratory distress syndrome. Polyhexamethylene guanidine phosphate (PHMG-P), a guanidine-based antimicrobial agent, is a major component associated with severe lung injury. In-depth studies are needed to determine how PHMG-P affects pathogenesis at the molecular level. Therefore, in this study, we analyzed short-term (4 weeks) and long-term (10 weeks) PHMG-P-exposure-specific gene-expression patterns in rats to improve our understanding of time-dependent changes in fibrosis.Materials and methods: Gene-expression profiles were analyzed in rat lung tissues using DNA microarrays and bioinformatics tools.Results: Clustering analysis of gene-expression data showed different gene-alteration patterns in the short- and long-term exposure groups and higher sensitivity to gene-expression changes in the long-term exposure group than in the short-term exposure group. Supervised analysis revealed 34 short-term and 335 long-term exposure-specific genes, and functional analysis revealed that short-term exposure-specific genes were involved in PHMG-P-induced initial inflammatory responses, whereas long-term exposure-specific genes were involved in PHMG-P-related induction of chronic lung fibrosis.Conclusion: The results of transcriptomic analysis were consistent with lung histopathology results. These findings indicated that exposure-time-specific changes in gene expression closely reflected time-dependent pathological changes in PHMG-P-induced lung injury.
Collapse
Affiliation(s)
- Mi-Kyung Song
- National Center for Efficacy Evaluation for Respiratory Disease Product, Korea Institute of Toxicology, Jeongeup, Republic of Korea.,Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, Republic of Korea
| | - Dong Im Kim
- National Center for Efficacy Evaluation for Respiratory Disease Product, Korea Institute of Toxicology, Jeongeup, Republic of Korea
| | - Kyuhong Lee
- National Center for Efficacy Evaluation for Respiratory Disease Product, Korea Institute of Toxicology, Jeongeup, Republic of Korea.,Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
21
|
Xian Z, Choi YH, Zheng M, Jiang J, Zhao Y, Wang C, Li J, Li Y, Li L, Piao H, Yan G. Imperatorin alleviates ROS-mediated airway remodeling by targeting the Nrf2/HO-1 signaling pathway. Biosci Biotechnol Biochem 2020; 84:898-910. [PMID: 31900049 DOI: 10.1080/09168451.2019.1710107] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this study, we investigated the role and mechanism of imperatorin (IMP) in chronic inflammation and airway remodeling. The levels of TNF-α, IL-1β, IL-6, IL-8, VEGF, α-SMA, and ROS were detected by ELISA, immunohistochemistry (IHC), immunofluorescence, and Western blot. In addition, we evaluated the effect of IMP on MAPK, PI3K/Akt, NF-κB, and Nrf2/HO-1 signaling pathways. IMP treatment obviously attenuated the production of inflammatory cytokines and inflammatory cells in bronchoalveolar lavage fluid of OVA-induced airway remodeling model. Meanwhile, it significantly inhibited inflammatory cell infiltration, goblet cell hyperplasia, collagen deposition, VEGF production, α-SMA, and ROS expression. Our study has shown that IMP could regulate the signaling pathways including MAPK, PI3K/Akt, NF-κB, and Nrf2/HO-1 to release the inflammatory responses. IMP might attenuate airway remodeling by the down-regulation of Nrf2/HO-1/ROS/PI3K/Akt, Nrf2/HO-1/ROS/MAPK, and Nrf2/HO-1/ROS/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Zhemin Xian
- Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji, P.R. China
| | - Yun Ho Choi
- Department of Anatomy, Medical School, Institute for Medical Sciences, Chonbuk National University, Jeonju, Republic of Korea
| | - Mingyu Zheng
- College of Pharmacy, Yanbian University, Yanji, P.R. China
| | - Jingzhi Jiang
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P.R. China
| | - Yuzhe Zhao
- Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji, P.R. China
| | - Chongyang Wang
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P.R. China
| | - Junfeng Li
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P.R. China
| | - Yan Li
- Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji, P.R. China
| | - Liangchang Li
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P.R. China
| | - Hongmei Piao
- Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji, P.R. China
| | - Guanghai Yan
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P.R. China
| |
Collapse
|
22
|
Zhang P, Liu J, Zong R. Triptolide protects against TGF-β1-induced pulmonary fibrosis by regulating FAK/calpain signaling. Exp Ther Med 2019; 18:4781-4789. [PMID: 31777561 PMCID: PMC6862707 DOI: 10.3892/etm.2019.8127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 09/04/2019] [Indexed: 11/17/2022] Open
Abstract
The present study aimed to investigate the mechanism of anti-proliferative, anti-inflammatory and anti-fibrotic effects of triptolide (TPL) on activated lung fibroblasts by regulating the focal adhesion kinase (FAK) and calpain signaling pathways. The HFL-1 human foetal lung fibroblast cell line was cultured in vitro and treated with 50 ng/ml transforming growth factor (TGF)-β1 for 48 h to establish the model of pulmonary fibrosis. Subsequently, the cells were divided into five groups, including a control, model, TPL, FAK inhibitor and calpeptin group. Subsequently, the proliferation of lung fibroblasts was detected using the Cell Counting Kit-8 assay. The concentration of interleukin (IL)-6 in the cell culture supernatant was examined by ELISA and the mRNA expression levels of collagen type I (ColI)α and ColIII in lung fibroblasts were quantified by reverse transcription-quantitative PCR. The protein levels of FAK, phosphorylated (p)-FAK, calpain 1 and calpain 2 were detected by western blot analysis. TGF-β1 induced the proliferation of lung fibroblasts, whereas TPL inhibited this proliferation in a dose-dependent manner. TPL also decreased the TGF-β1-induced production of IL-6 and reduced the upregulation of ColIα, ColIII, FAK, p-FAK, and inhibited the decrease of calpain 1 and calpain 2 induced by TGF-β1. In addition, the FAK inhibitor acted synergistically with TPL to decrease TGF-β1-induced production of IL-6 and attenuate TGF-β1-induced synthesis of ColIα and ColIII, while calpeptin had an antagonistic effect on the function of TPL. Furthermore, treatment with the FAK inhibitor and TPL markedly decreased the protein levels of FAK and p-FAK, and increased the protein expression of calpain 1 and calpain 2 in lung fibroblasts stimulated by TGF-β1 to a greater extent than TPL alone, while calpeptin had an antagonistic effect on the action of TPL. In conclusion, the present study indicated that TPL protected against TGF-β1-induced proliferation, inflammation and fibrosis by regulating the FAK and calpain signaling pathways.
Collapse
Affiliation(s)
- Pingheng Zhang
- Traditional Chinese Medicine Department, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Jian Liu
- Rheumatism Department, Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Ruikai Zong
- Rheumatism Department, Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| |
Collapse
|
23
|
Elrashidy RA, Liu G. Long-term diabetes causes molecular alterations related to fibrosis and apoptosis in rat urinary bladder. Exp Mol Pathol 2019; 111:104304. [PMID: 31479659 DOI: 10.1016/j.yexmp.2019.104304] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/23/2019] [Accepted: 08/30/2019] [Indexed: 02/05/2023]
Abstract
Diabetes induces time-dependent alterations in urinary bladders. Long-term diabetes causes an underactive bladder. However, the fundamental mechanisms are still elusive. This study aimed to examine the histological changes and the potential molecular pathways affected by long-term diabetes in the rat bladder. Diabetes was induced in 8-week-old male Lewis rats by streptozotocin, while age-matched control rats received citrate buffer only. Forty-four weeks after diabetes induction, bladders were harvested for histological and molecular analyses. The expressions of proteins related to fibrosis, apoptosis and oxidative stress as well as the cellular signaling pathway in the bladder were examined by immunoblotting. Histological examinations illustrated diabetes caused detrusor hypertrophy and fibrotic changes in the bladder. Immunoblotting analysis demonstrated higher collagen I but lower elastin expression in the bladder in diabetic rats. These were accompanied by an increase in the expression of transforming growth factor-beta1, along with the downregulation of matrix metalloptoteinase-1, and upregulation of tissue inhibitor of metalloproteinase-1. Diabetic rats showed an increase in nitrotyrosine, but decrease in nuclear factor erythroid-related factor 2 (Nrf2) levels in the bladder. Enhanced apoptotic signaling was observed, characterized by increased expression of Bcl-2-associated X protein (Bax), decreased expression of Bcl-2, in the diabetic bladder. The nerve growth factor level was decreased in the diabetic bladder. A significant suppression in the protein expressions of phosphorylated extracellular signal-regulated kinases 1/2 was found in diabetic bladders. This study demonstrated that long-term diabetes caused molecular changes that could promote fibrosis and apoptosis in the bladder. Oxidative stress may be involved in this context.
Collapse
Affiliation(s)
- Rania A Elrashidy
- Department of Surgery, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, USA; Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Guiming Liu
- Department of Surgery, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
24
|
Zhou Y, Chai X. Protective effect of bicyclol against pulmonary fibrosis via regulation of microRNA-455-3p in rats. J Cell Biochem 2019; 121:651-660. [PMID: 31407409 DOI: 10.1002/jcb.29310] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 07/15/2019] [Indexed: 12/20/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF), a chronic, progressive and irreversible disease, needs long-term treatment. Bicyclol was found to play a great role in pulmonary fibrosis, and the present study is to explore how bicyclol affects IPF with the involvement of microRNA-455-3p (miR-455-3p) and Bax. Bleomycin (BLM) was used to induce the IPF model in Sprague-Dawley rats to detect the expression of miR-455-3p, Bax, and B-cell lymphoma factor 2 (Bcl-2). Moreover, to further investigate the mechanisms of bicyclol, the BLM-induced fibrotic cell model was used after the lung epithelial cells HPAEpiC received miR-455-3p knockout treatment. The rats were then treated with vehicle and bicyclol, respectively. The apoptosis of fibrotic cells and Bax/Bcl-2 were identified. Inhibition function of bicyclol was optimal at a dose of 150 mg/kg. Bicyclol inhibited cell apoptosis and reduced Bax/Bcl-2 expression in rats. miR-455-3p could potentially bind to Bax gene. Bicyclol reduced the levels of methylenedioxyamphetamine, superoxide dismutase, and glutathione in rat lung tissue, inhibited the apoptosis of rats with IPF and upregulated miR-455-3p expression. In vitro studies showed that bicyclol significantly promoted miR-455-3p expression in HPAEpiC fibrosis. Bicyclol inhibited fibrosis-induced apoptosis of HPAEpiC in alveolar epithelial cells through promoting miR-455-3p, which inhibited Bax expression in IPF. Bicyclol may suppress the apoptosis of alveolar epithelial cells by upregulating miR-455-3p. This study laid a theoretical foundation for further understanding of IPF and searching for new molecular therapeutic targets.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Pneumology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xiujuan Chai
- Department of Pneumology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Wang D, Yan Z, Bu L, An C, Deng B, Zhang J, Rao J, Cheng L, Zhang J, Zhang B, Xie J. Protective effect of peptide DR8 on bleomycin-induced pulmonary fibrosis by regulating the TGF-β/MAPK signaling pathway and oxidative stress. Toxicol Appl Pharmacol 2019; 382:114703. [PMID: 31398421 DOI: 10.1016/j.taap.2019.114703] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/02/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023]
Abstract
Pulmonary fibrosis (PF) is a fatal and irreversible lung disease that eventually causes respiratory failure, lung dysfunction and death. The peptide DHNNPQIR-NH2 (DR8) has been reported to possess potent antioxidant activity, and an imbalance of oxidation/antioxidation is a crucial mechanism that causes PF. Here, we studied the ability of DR8 to improve PF and further explored the pathway in which DR8 plays a critical role. We found that after prophylactic or therapeutic treatment with DR8, fibrosis-associated indices, including marker proteins, proinflammatory cytokines and profibrogenic cytokines, were significantly downregulated. Importantly, DR8 could reduce bleomycin-induced pathological changes and collagen deposition, especially collagen I content. Furthermore, DR8 prominently upregulated nonenzymatic antioxidants and enzymatic antioxidants. Consistent with the in vivo results, we observed that DR8 significantly inhibited the proliferation and reactive oxygen species (ROS) generation of A549 cells and NIH3T3 cells stimulated with transforming growth factor-β1 (TGF-β1), as well as decreased NADPH oxidase 4 (NOX4) levels under the same conditions. Moreover, DR8 reversed the TGF-β1-induced upregulation of phosphorylated ERK1/2 and p38 MAPK in cells and the bleomycin-induced upregulation of these indices in mice. Our results indicate that DR8 could prevent and treat PF by reducing oxidative damage and suppressing the TGF-β/MAPK pathway. Because of the high efficiency and low toxicity of DR8, we consider that DR8 could be a candidate drug for PF, and our studies establish a foundation for the development of a lead compound to be used as a therapy for fibrosis-related diseases.
Collapse
Affiliation(s)
- Dan Wang
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Zhibin Yan
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Lili Bu
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Chunmei An
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Bochuan Deng
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Jianfeng Zhang
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Jing Rao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Lu Cheng
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Jingying Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Bangzhi Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China.
| | - Junqiu Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
26
|
Sun Q, Hu J, Yu P, Zhu Z, Yu R, Ge C, Li C, Wu G, Lin B, Liu G, Liu M, Bian H, Xu H, Jia S. Peptide PD29 treats bleomycin-induced pulmonary fibrosis by inhibiting the TGF-β/smad signaling pathway. Exp Lung Res 2019; 45:123-134. [PMID: 31210057 DOI: 10.1080/01902148.2019.1614696] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Pulmonary fibrosis (PF) is an end-stage change in lung disease characterized by fibroblast proliferation, massive extracellular matrix (ECM) aggregation with inflammatory damage, and severe structural deterioration. PD29 is a 29-amino acid peptide which has the potential to alleviate PF pathogenesis via three mechanisms: anti-angiogenesis, inhibition of matrix metalloproteinase activities, and inhibition of integrins. In this study, fibrotic lung injuries were induced in SD rats by a single intratracheal instillation of 5 mg/kg bleomycin (BLM). Then, these rats were administered 7.5, 5, or 2.5 mg/kg PD29 daily for 30 days. BLM induced-syndromes including structure distortion, excessive deposition of ECM, excessive inflammatory infiltration, and pro-inflammatory cytokine release were used to evaluate the protective effect of PD-29. Oxidative stress damage in lung tissues was attenuated by PD29 in a dose-dependent manner. The expression of TGF-β1 and the phosphorylation of Smad-2/-3-its downstream targets-were enhanced by BLM and weakened by PD29. In vitro, PD29 inhibited TGF-β1-induced epithelial-mesenchymal transition (EMT) and transformation in A549 cells and mouse primary fibroblasts into myofibroblasts. In summary, PD29 reversed EMT and transformation of fibroblasts into myofibroblasts in vitro and prevented PF in vivo possibly by suppressing the TGF-β1/Smad pathway.
Collapse
Affiliation(s)
- Qingbo Sun
- a Jiangsu Provincial Research Center for Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China
| | - Jialiang Hu
- a Jiangsu Provincial Research Center for Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China
| | - Pengcheng Yu
- a Jiangsu Provincial Research Center for Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China
| | - Zhaohao Zhu
- a Jiangsu Provincial Research Center for Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China
| | - Ruihe Yu
- a Jiangsu Provincial Research Center for Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China
| | - Chuang Ge
- a Jiangsu Provincial Research Center for Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China
| | - Chencheng Li
- a Jiangsu Provincial Research Center for Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China
| | - Guiyue Wu
- a Jiangsu Provincial Research Center for Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China
| | - Bingjing Lin
- a Jiangsu Provincial Research Center for Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China
| | - Guangpan Liu
- a Jiangsu Provincial Research Center for Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China
| | - Meng Liu
- a Jiangsu Provincial Research Center for Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China
| | - Huan Bian
- a Jiangsu Provincial Research Center for Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China
| | - Hanmei Xu
- a Jiangsu Provincial Research Center for Synthetic Peptide Drug Discovery and Evaluation, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China.,b State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University , Nanjing , Jiangsu , People's Republic of China
| | - Shaochang Jia
- c Department of Bio-Treatment Jinling Hospital , Nanjing , Jiangsu , People's Republic of China
| |
Collapse
|
27
|
Jia L, Sun P, Gao H, Shen J, Gao Y, Meng C, Fu S, Yao H, Zhang G. Mangiferin attenuates bleomycin-induced pulmonary fibrosis in mice through inhibiting TLR4/p65 and TGF-β1/Smad2/3 pathway. J Pharm Pharmacol 2019; 71:1017-1028. [PMID: 30847938 DOI: 10.1111/jphp.13077] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/12/2019] [Indexed: 12/30/2022]
Abstract
Abstract
Objectives
Investigating the antipulmonary fibrosis effect of mangiferin from Mangifera indica and the possible molecular mechanism.
Methods
In vivo, bleomycin (BLM)-induced pulmonary fibrosis experimental model was used for evaluating antipulmonary fibrosis effect of mangiferin. Histopathologic examination and collagen deposition were investigated by HE and Masson staining as well as detecting the content of hydroxyproline. The expression of transforming growth factor-β1 (TGF-β1), α-smooth muscle actin (α-SMA), TLR4 and p-P65 in lung tissue was analysed through immunofluorescence. Leucocytes and inflammatory cytokines including IL-1β, IL-6, TNF-α and MCP-1 in bronchoalveolar lavage fluid were detected by cell counting and enzyme-linked immunosorbent assay. In vitro, TGF-β1-induced A549 epithelial–mesenchymal transition (EMT) cell model was used for investigating the possible molecular mechanism. Reactive oxygen species (ROS) generation was detected by DCFH-DA assay. Expression of all proteins was examined by Western blot.
Key findings
Oral administration of mangiferin could attenuate the severity of BLM-induced pulmonary fibrosis through increasing the survival rate, improving histopathological lesion and body weight loss as well as decreasing pulmonary index visibly. Pulmonary hydroxyproline content, TGF-β1, and α-SMA levels were reduced significantly. The molecular mechanism of mangiferin for inhibiting pulmonary fibrosis is that it could obviously inhibit the occurrence of inflammation and the secretion of inflammatory cytokine through inhibiting activation of TLR4 and phosphorylation of p65. Meanwhile, EMT process was suppressed obviously by mangiferin through blocking the phosphorylation of Smad2/3 and reducing MMP-9 expression. Besides, mangiferin could significantly inhibit the process of oxidant stress through downregulating the intracellular ROS generation.
Conclusions
Mangiferin attenuates BLM-induced pulmonary fibrosis in mice through inhibiting TLR4/p65 and TGF-β1/Smad2/3 pathway.
Collapse
Affiliation(s)
- Li Jia
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Ping Sun
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Hui Gao
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Jie Shen
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Yuan Gao
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Cheng Meng
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Shidong Fu
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Huijuan Yao
- Yanan's People Hospital, Yanan, Shanxi, China
| | - Gong Zhang
- Yanan University Affiliated Hospital, Yanan, Shanxi, China
| |
Collapse
|
28
|
Meng Y, Pan M, Zheng B, Chen Y, Li W, Yang Q, Zheng Z, Sun N, Zhang Y, Li X. Autophagy Attenuates Angiotensin II-Induced Pulmonary Fibrosis by Inhibiting Redox Imbalance-Mediated NOD-Like Receptor Family Pyrin Domain Containing 3 Inflammasome Activation. Antioxid Redox Signal 2019; 30:520-541. [PMID: 29486589 DOI: 10.1089/ars.2017.7261] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
AIMS The NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, which is activated by reactive oxygen species (ROS) and repressed by autophagy, has been identified as a novel agent of pulmonary fibrosis. Angiotensin II (AngII), the bioactive pro-oxidant in the renin-angiotensin system, aggravates lung fibrosis. However, the effect of AngII on NLRP3 inflammasome and autophagy in lung fibrosis remains unknown. This study investigates the potential link between AngII-induced autophagy in the regulation of NLRP3 inflammasome/IL-1β axis in lung fibrosis. RESULTS In vivo, autophagy and the NLRP3 inflammasome were activated in fibrotic patients and positively correlated with oxidation. Treatment with rapamycin promoted autophagy but inhibited oxidation, NLRP3 inflammasome, and lung fibrosis after bleomycin (BLM) infusion. The autophagy inhibitor 3-methyladenine reduced BLM-induced lung fibrosis and concurrently facilitated NLRP3 inflammasome activation and oxidation in fibroblasts. In vitro, AngII promoted intercellular ROS, hydrogen peroxide, and NADPH oxidase 4 (NOX4) protein levels and reduced the glutathione concentration, thereby leading to NLRP3 inflammasome activation and consequent collagen synthesis. AngII induced autophagy, while VAS2870, NOX4, small-interfering RNA (siRNA), and compound C eliminated AngII-induced LC3B augmentation. Moreover, blocking autophagy with bafilomycin A1 or LC3B siRNA resulted in oxidant accumulation, NLRP3 inflammasome hyperactivation, and collagen deposition. Finally, AngII induced P62/SQSTM1, targeting ubiquitinated apoptosis-associated speck-like protein containing a CARD for degradation, thereby contributing to NLRP3 inflammasome inactivation. Innovation and Conclusion: Autophagy attenuates pulmonary fibrosis by regulating NLRP3 inflammasome activation induced by AngII-mediated ROS via redox balance modulation.
Collapse
Affiliation(s)
- Ying Meng
- 1 Department of Respiratory and Critical Care Medicine and Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Miaoxia Pan
- 1 Department of Respiratory and Critical Care Medicine and Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Bojun Zheng
- 1 Department of Respiratory and Critical Care Medicine and Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Yan Chen
- 2 Department of General Medicine, Panyu Central Hospital , Guangzhou, China
| | - Wei Li
- 3 Department of Intensive Care Unit, Foshan First People's Hospital, Sun Yat-Sen University , Foshan, China
| | - Qianjie Yang
- 1 Department of Respiratory and Critical Care Medicine and Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Zemao Zheng
- 1 Department of Respiratory and Critical Care Medicine and Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Nana Sun
- 1 Department of Respiratory and Critical Care Medicine and Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Yue Zhang
- 1 Department of Respiratory and Critical Care Medicine and Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Xu Li
- 5 Department of Emergency Medicine, Nanfang Hospital, Southern Medical University , Guangzhou, China
- 4 State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University , Guangzhou, China
| |
Collapse
|
29
|
Nokes B, Afshar K. Adjunctive therapies in idiopathic pulmonary fibrosis-where do we stand? J Thorac Dis 2019; 11:357-360. [PMID: 30962973 DOI: 10.21037/jtd.2018.11.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Brandon Nokes
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego. San Diego, CA, USA
| | - Kamyar Afshar
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego. San Diego, CA, USA
| |
Collapse
|
30
|
Eberhardt W, Nasrullah U, Pfeilschifter J. Activation of renal profibrotic TGFβ controlled signaling cascades by calcineurin and mTOR inhibitors. Cell Signal 2018; 52:1-11. [PMID: 30145216 DOI: 10.1016/j.cellsig.2018.08.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/11/2022]
Abstract
The calcineurin inhibitors (CNI) cyclosporine A (CsA) and tacrolimus represent potent immunosuppressive agents frequently used for solid organ transplantation and treatment of autoimmune disorders. Despite of their immense therapeutic benefits, residual fibrosis mainly in the kidney represents a common side effect of long-term therapy with CNI. Regardless of the immunosuppressive action, an increasing body of evidence implicates that a drug-induced increase in TGFβ and subsequent activation of TGFβ-initiated signaling pathways is closely associated with the development and progression of CNI-induced nephropathy. Mechanistically, an increase in reactive oxygen species (ROS) generation due to drug-induced changes in the intracellular redox homeostasis functions as an important trigger of the profibrotic signaling cascades activated under therapy with CNI. Although, inhibitors of the mechanistic target of rapamycin (mTOR) kinase have firmly been established as alternative compounds with a lower nephrotoxic potential, an activation of fibrogenic signaling cascades has been reported for these drugs as well. This review will comprehensively summarize recent advances in the understanding of profibrotic signaling events modulated by these widely used compounds with a specific focus put on mechanisms occurring independent of their respective immunosuppressive action. Herein, the impact of redox modulation, the activation of canonical TGFβ and non-Smad pathways and modulation of autophagy by both classes of immunosuppressive drugs will be highlighted and discussed in a broader perspective. The comprehensive knowledge of profibrotic signaling events specifically accompanying the immunomodulatory activity of these widely used drugs is needed for a reliable benefit-risk assessment under therapeutic regimens.
Collapse
Affiliation(s)
- Wolfgang Eberhardt
- Pharmazentrum frankfurt/ZAFES, Universitätsklinikum und Goethe-Universität, Frankfurt am Main, Germany.
| | - Usman Nasrullah
- Pharmazentrum frankfurt/ZAFES, Universitätsklinikum und Goethe-Universität, Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Pharmazentrum frankfurt/ZAFES, Universitätsklinikum und Goethe-Universität, Frankfurt am Main, Germany
| |
Collapse
|
31
|
Wieczfinska J, Kowalczyk T, Sitarek P, Skała E, Pawliczak R. Analysis of Short-Term Smoking Effects in PBMC of Healthy Subjects-Preliminary Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E1021. [PMID: 29783661 PMCID: PMC5982060 DOI: 10.3390/ijerph15051021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/20/2022]
Abstract
Early structural changes exist in the small airways before the establishment of Chronic Obstructive Pulmonary Disease (COPD). These changes are believed to be induced by oxidation. The aim of this study was to analyze the influence of short-term smoking on the expression of the genes contributing to airway remodeling and their relationship with the oxidative status of human blood cells. Blood mononuclear cells were isolated from 16 healthy volunteers and treated with cigarette smoke ingredients (CSI): nicotine, 1-Nitrosodimethylamine, N-Nitrosopyrrolidyne, vinyl chloride, acetone, and acrolein. The expression of TGF-β1, TIMP-1, SOD1, and arginase I was determined by qPCR. Additionally, thiol groups and TBARs were assessed. CSI induced TGF and TIMP-1 expression in peripheral blood mononuclear cells (PBMC), and apocynin alleviated this effect. The changes were more noticeable in the smoking group (p < 0.05). TBARs concentrations were higher in smokers, and in this group, apocynin acted more effectively. SOD1 correlated with arginase expression in smokers (p < 0.05). MMP-9 showed a significant correlation with SOD1 in both groups, but only on the protein level. Blood cells appear to mirror the general changes caused by cigarette smoke ingredients, which seem to be connected with the oxidative status of the cell. Our findings indicate that a short period of smoking influences the gene expression and oxidative balance of blood cells, which might result in the development of serious disorders such as COPD.
Collapse
Affiliation(s)
- Joanna Wieczfinska
- Department of Immunopathology, Medical University of Lodz, Lodz 90-752, Poland.
| | - Tomasz Kowalczyk
- Department of Genetics and Plant Molecular Biology and Biotechnology, The University of Lodz, Lodz 90-237, Poland.
| | - Przemyslaw Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Lodz 90-151, Poland.
| | - Ewa Skała
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Lodz 90-151, Poland.
| | - Rafal Pawliczak
- Department of Immunopathology, Medical University of Lodz, Lodz 90-752, Poland.
| |
Collapse
|
32
|
Yang L, Lin Z, Wang Y, Li C, Xu W, Li Q, Yao W, Song Z, Liu G. Nickle(II) ions exacerbate bleomycin-induced pulmonary inflammation and fibrosis by activating the ROS/Akt signaling pathway. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:4406-4418. [PMID: 29185215 DOI: 10.1007/s11356-017-0525-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 10/19/2017] [Indexed: 06/07/2023]
Abstract
Nickle (Ni) is a heavy metal found in particulate matter. We previously reported that Ni ions are strongly associated with high apoptosis rates and high expression of IL-1β in human bronchial epithelial cells following exposure to PM2.5; however, the effects of Ni ions on pulmonary fibrosis have not been fully elucidated. In the current study, we evaluated whether Ni ions can exacerbate bleomycin (BLM)-induced pulmonary fibrosis in a mouse model and illustrated the potential mechanism. Ni ions inhibited cell proliferation and induced apoptosis in A549 and MRC-5 cells. BLM-induced lung injury and fibrosis in mice were significantly enhanced by nickel treatment, and these findings were also supported by inflammatory cell accumulation in bronchoalveolar lavage fluid and elevated levels of pro-inflammatory cytokines in lung tissues. Ni ions also increased extracellular matrix protein levels, including those of type I collagen and MMP9 in mouse lung tissues and cell lines. Moreover, Ni ions promoted the phosphorylation of AKT in this mouse model. The effect of increased collagen levels and MMP9 expression was inhibited by blocking the AKT phosphorylation. Together, these findings suggest AKT activation as a critical contributor to this Ni-exacerbated pulmonary fibrotic process.
Collapse
Affiliation(s)
- Lawei Yang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Ziying Lin
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yahong Wang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Chunyan Li
- Department of Respiratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Wenya Xu
- Department of Respiratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Qinglan Li
- Department of Respiratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Weimin Yao
- Department of Respiratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Zeqing Song
- Department of Respiratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Gang Liu
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
- Department of Respiratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
33
|
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: 44] [Impact Index Per Article: 5.5] [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
|
34
|
Pulmonary Hypertension Associated with Idiopathic Pulmonary Fibrosis: Current and Future Perspectives. Can Respir J 2017; 2017:1430350. [PMID: 28286407 PMCID: PMC5327768 DOI: 10.1155/2017/1430350] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/19/2017] [Indexed: 12/12/2022] Open
Abstract
Pulmonary hypertension (PH) is commonly present in patients with chronic lung diseases such as Chronic Obstructive Pulmonary Disease (COPD) or Idiopathic Pulmonary Fibrosis (IPF) where it is classified as Group III PH by the World Health Organization (WHO). PH has been identified to be present in as much as 40% of patients with COPD or IPF and it is considered as one of the principal predictors of mortality in patients with COPD or IPF. However, despite the prevalence and fatal consequences of PH in the setting of chronic lung diseases, there are limited therapies available for patients with Group III PH, with lung transplantation remaining as the most viable option. This highlights our need to enhance our understanding of the molecular mechanisms that lead to the development of Group III PH. In this review we have chosen to focus on the current understating of PH in IPF, we will revisit the main mediators that have been shown to play a role in the development of the disease. We will also discuss the experimental models available to study PH associated with lung fibrosis and address the role of the right ventricle in IPF. Finally we will summarize the current available treatment options for Group III PH outside of lung transplantation.
Collapse
|
35
|
Satpute RM, Pawar PP, Puttewar S, Sawale SD, Ambhore PD. Effect of resveratrol and tetracycline on the subacute paraquat toxicity in mice. Hum Exp Toxicol 2017; 36:1303-1314. [PMID: 28090784 DOI: 10.1177/0960327116688070] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Paraquat (PQ) is a nonselective bipyridyl herbicide widely used in agriculture to control weeds, but its accidental, occupational, or intentional exposure in humans is known to cause pneumo- and neurotoxicity which may proves fatal. Oxidative stress is reported as an underlined mechanism of PQ-induced toxicity in alveolar cells, neurons, and astroglia. PQ generates superoxides both through electron transport reaction (ETC) with nicotinamide adenine dinucleotide-dependent oxidoreductase and by the redox cycling via reaction with molecular oxygen. In lungs, it causes edema and inflammation resulting in neutrophils infiltration and subsequent activation of pro-inflammatory cytokines. In the present study, toxicity of subacute oral PQ exposure and effect of resveratrol (Res) and/or tetracycline (TC) on oxidative stress and inflammatory markers in lungs, brain, and liver was studied. Levels of glutathione and malondialdehyde and activities of myeloperoxidase, glutathione peroxidase, and catalase were measured in lungs, brain, and liver. PQ interferes in the function of mitochondrial ETC complexes causing decreased adenosine triphosphate levels, and hence the activities of complexes I and IV were studied in brain tissues. Res, a natural antioxidant, and TC, an antibiotic with its antimicrobial and anti-inflammatory properties, offered significant protection from severe oxidative stress and inflammation and ameliorated the general well-being of mice against the toxic outcome of PQ.
Collapse
Affiliation(s)
- R M Satpute
- 1 Toxicology Laboratory, Defence Research and Development Establishment, Nagpur, Maharashtra, India
| | - P P Pawar
- 2 Department of Biotechnology, S.F.S. College, Seminari Hills, Nagpur, India
| | - S Puttewar
- 2 Department of Biotechnology, S.F.S. College, Seminari Hills, Nagpur, India
| | - S D Sawale
- 3 Department of Biotechnology, G.H. Raisoni Institute of Information Technology, Nagpur, India
| | - P D Ambhore
- 4 Department of Biotechnology, Rajarshi Shahu Mahavidyalaya, Latur, India
| |
Collapse
|
36
|
Zhao J, Liu J, Lee JF, Zhang W, Kandouz M, VanHecke GC, Chen S, Ahn YH, Lonardo F, Lee MJ. TGF-β/SMAD3 Pathway Stimulates Sphingosine-1 Phosphate Receptor 3 Expression: IMPLICATION OF SPHINGOSINE-1 PHOSPHATE RECEPTOR 3 IN LUNG ADENOCARCINOMA PROGRESSION. J Biol Chem 2016; 291:27343-27353. [PMID: 27856637 DOI: 10.1074/jbc.m116.740084] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 11/04/2016] [Indexed: 12/20/2022] Open
Abstract
Previously, we showed that levels of sphingosine-1 phosphate receptor 3 (S1PR3) are increased in a panel of cultured human lung adenocarcinoma cell lines, and that S1PR3-mediated signaling pathways regulate proliferation, soft agar growth, and invasion of human lung adenocarcinoma cells in vitro In the present study, we examine S1PR3 levels in human lung adenocarcinoma specimens. cDNA array and tumor microarray analysis shows that mRNA and protein levels of S1PR3 are significantly increased in human lung adenocarcinomas when compared with normal lung epithelial cells. Promoter analysis shows 16 candidate SMAD3 binding sites in the promoter region of S1PR3. ChIP indicates that TGF-β treatment stimulates the binding of SMAD3 to the promoter region of S1PR3. Luciferase reporter assay demonstrates that SMAD3 transactivates S1PR3 promoter. TGF-β stimulation or ectopic expression of TGF-β up-regulates S1PR3 levels in vitro and ex vivo Pharmacologic inhibition of TGF-β receptor or SMAD3 abrogates the TGF-β-stimulated S1PR3 up-regulation. Moreover, S1PR3 knockdown dramatically inhibits tumor growth and lung metastasis, whereas ectopic expression of S1PR3 promotes the growth of human lung adenocarcinoma cells in animals. Pharmacological inhibition of S1PR3 profoundly inhibits the growth of lung carcinoma in mice. Our studies suggest that levels of S1PR3 are up-regulated in human lung adenocarcinomas, at least in part due to the TGF-β/SMAD3 signaling axis. Furthermore, S1PR3 activity promotes the progression of human lung adenocarcinomas. Therefore, S1PR3 may represent a novel therapeutic target for the treatment of deadly lung adenocarcinomas.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shiyou Chen
- the Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia 30602
| | | | - Fulvio Lonardo
- From the Departments of Pathology and.,Karmanos Cancer Institute, and
| | - Menq-Jer Lee
- From the Departments of Pathology and .,Karmanos Cancer Institute, and.,Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, Michigan 48201 and
| |
Collapse
|
37
|
Qin T, Yin S, Yang J, Zhang Q, Liu Y, Huang F, Cao W. Sinomenine attenuates renal fibrosis through Nrf2-mediated inhibition of oxidative stress and TGFβ signaling. Toxicol Appl Pharmacol 2016; 304:1-8. [PMID: 27211841 DOI: 10.1016/j.taap.2016.05.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/05/2016] [Accepted: 05/16/2016] [Indexed: 02/06/2023]
Abstract
Renal fibrosis is the common feature of chronic kidney disease and mainly mediated by TGFβ-associated pro-fibrogenic signaling, which causes excessive extracellular matrix accumulation and successive loss of kidney functions. Sinomenine (SIN), an alkaloid derived from medicinal herb extensively used in treatment of rheumatoid arthritis and various inflammatory disorders, displays renal protective properties in experimental animals; however its pharmacological potency against renal fibrosis is not explored. In this study we report that SIN possesses strong anti-renal fibrosis functions in kidney cell and in mouse fibrotic kidney. SIN beneficially modulated the pro-fibrogenic protein expression in TGFβ-treated kidney cells and attenuated the renal fibrotic pathogenesis incurred by unilateral ureteral obstruction (UUO), which correlated with its activation of Nrf2 signaling - the key defender against oxidative stress with anti-fibrotic potentials. Further investigation on its regulation of Nrf2 downstream events revealed that SIN significantly balanced oxidative stress via improving the expression and activity of anti-oxidant and detoxifying enzymes, and interrupted the pro-fibrogenic signaling of TGFβ/Smad and Wnt/β-catenin. Even more impressively SIN achieved its anti-fibrotic activities in an Nrf2-dependent manner, suggesting that SIN regulation of Nrf2-associated anti-fibrotic activities constitutes a critical component of SIN's renoprotective functions. Collectively our studies have demonstrated a novel anti-fibrotic property of SIN and its upstream events and provided a molecular basis for SIN's potential applications in treatment of renal fibrosis-associated kidney disorders.
Collapse
Affiliation(s)
- Tian Qin
- School of Life Science & Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Shasha Yin
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University School of Medicine, Nanjing 210093, China
| | - Jun Yang
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University School of Medicine, Nanjing 210093, China
| | - Qin Zhang
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University School of Medicine, Nanjing 210093, China
| | - Yangyang Liu
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University School of Medicine, Nanjing 210093, China
| | - Fengjie Huang
- School of Life Science & Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Wangsen Cao
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University School of Medicine, Nanjing 210093, China.
| |
Collapse
|
38
|
Becerik S, Celec P, Gürkan A, Öztürk VÖ, Kamodyova N, Atilla G, Emingil G. Gingival Crevicular Fluid and Plasma Levels of Transglutaminase-2 and Oxidative Stress Markers in Cyclosporin A-Induced Gingival Overgrowth. J Periodontol 2016; 87:1508-1516. [PMID: 27468796 DOI: 10.1902/jop.2016.160031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Transglutaminase (TGM)-2 has been shown to contribute to fibrosis by extracellular matrix accumulation in some organs and is activated by intracellular reactive oxygen species. The aim of this study is to investigate levels of gingival crevicular fluid (GCF) and plasma TGM-2 and oxidative stress markers (OSMs) in cyclosporin A (CsA)-induced gingival overgrowth (GO). METHODS The study enrolled 20 healthy (H) individuals; 20 patients with gingivitis (G); 20 CsA-medicated patients with GO (CsA GO+); and 20 CsA-medicated patients without GO (CsA GO-). GCF and plasma levels of TGM-2 were analyzed by enzyme-linked immunosorbent assay. Spectrofluorometry was used to analyze thiobarbituric acid reactive substance (TBARS); ferric-reducing antioxidant power (FRAP); total oxidant status (TOS); and total antioxidant capacity (TAC). RESULTS GCF TGM-2 level was elevated in CsA GO+ compared with G (P = 0.048) and H (P = 0.001) groups. GCF TBARS level was elevated in CsA GO+ compared with other groups (CsA GO- group: P = 0.003; G group: P <0.001; and H group: P <0.001) and was higher in CsA GO- than in H (P = 0.048). GCF FRAP level was lower in CsA GO- than in H (P = 0.04). Both CsA GO+ and CsA GO- groups had lower GCF TOS levels than H (P <0.001 and P = 0.002) and G (P = 0.003 and P = 0.04). GCF TAC was higher in CsA GO+ than in H (P = 0.02). Plasma TGM-2 level was elevated in CsA GO+ compared with G (P = 0.048) and H (P = 0.002). Plasma FRAP level was higher in H and CsA GO- than in CsA GO+ (P = 0.008 and P = 0.02). CONCLUSIONS CsA use significantly alters GCF and plasma levels of TGM-2 and OSMs. TGM-2 may contribute to CsA-induced GO in CsA-treated patients by changing GCF and plasma levels of OSMs. Further studies are needed to prove causality and its direction.
Collapse
Affiliation(s)
- Sema Becerik
- Department of Periodontology, School of Dentistry, Ege University, İzmir, Turkey
| | - Peter Celec
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| | - Ali Gürkan
- Department of Periodontology, School of Dentistry, Ege University, İzmir, Turkey
| | - Veli Özgen Öztürk
- Department of Periodontology, School of Dentistry, Adnan Menderes University, Aydın, Turkey
| | - Natalia Kamodyova
- Faculty of Medicine, Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| | - Gül Atilla
- Department of Periodontology, School of Dentistry, Ege University, İzmir, Turkey
| | - Gülnur Emingil
- Department of Periodontology, School of Dentistry, Ege University, İzmir, Turkey
| |
Collapse
|
39
|
Du J, Li X, Lin C, He X. Protective Effects of Arachidonic Acid Against Paraquat-Induced Pulmonary Injury. Inflammation 2016; 38:1458-63. [PMID: 25633425 DOI: 10.1007/s10753-015-0120-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
In this study, we aimed to study the effects of arachidonic acid (AA) on acute lung injury (ALI) caused by paraquat (PQ) in mice. Male Kunming mice were randomly divided into three groups: control group, PQ group, and PQ + AA group (n = 24). The mice in the PQ and PQ + AA groups received a single oral dose of 20 mg/kg bodyweight PQ, and the mice of the PQ + AA group were challenged by 500 mg/kg bodyweight AA posttreatment 2 h after PQ administration. The results indicated that the administration of AA significantly increased the activity of superoxide dismutase (SOD), decreased the activity of myeloperoxidase (MPO), the content of malondialdehyde (MDA), and the level of lactate dehydrogenase (LDH). Pathological examination also revealed that AA effectively alleviated PQ-induced histological damage. Furthermore, AA significantly reduced PQ-induced upregulations of inflammatory mediators such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and IL-8. These results demonstrated that AA had effective protection against PQ-induced ALI.
Collapse
Affiliation(s)
- Jianxin Du
- Pulmonary Department, Affiliated Hospital of QingDao University, Qingdao, 266003, People's Republic China
| | | | | | | |
Collapse
|
40
|
Trajano LASN, Trajano ETL, Lanzetti M, Mendonça MSA, Guilherme RF, Figueiredo RT, Benjamim CF, Valenca SS, Costa AMA, Porto LC. Elastase modifies bleomycin-induced pulmonary fibrosis in mice. Acta Histochem 2016; 118:203-12. [PMID: 26852294 DOI: 10.1016/j.acthis.2015.12.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 11/17/2015] [Accepted: 12/23/2015] [Indexed: 01/24/2023]
Abstract
Pulmonary fibrosis (PF) is characterized by excessive accumulation of collagen in the lungs. Emphysema is characterized by loss of the extracellular matrix (ECM) and alveolar enlargement. We studied the co-participation of elastase-induced mild emphysema in bleomycin-induced PF in mice by analyzing oxidative stress, inflammation and lung histology. C57BL/6 mice were divided into four groups: control; bleomycin (0.1U/mouse); elastase (using porcine pancreatic elastase (PPE)+bleomycin (3U/mouse 14 days before 0.1U/mouse of bleomycin; PPE+B); elastase (3U/mouse). Mice were humanely sacrificed 7, 14 and 21 days after treatment with bleomycin or vehicle. PF was observed 14 days and 21 days after bleomycin treatment but was observed after 14 days only in the PPE+B group. In the PPE+B group at 21 days, we observed many alveoli and alveolar septa with few PF areas. We also observed marked and progressive increases of collagens 7, 14 and 21 days after bleomycin treatment whereas, in the PPE+B group, collagen deposition was observed only at 14 days. There was a reduction in activities of the antioxidant enzymes superoxide dismutase (p<0.05), catalase (p<0.01) and glutathione peroxidase (p<0.01) parallel with an increase in nitrite (p<0.01) 21 days after bleomycin treatment compared with the control group. These endpoints were also reduced (p<0.05, p<0.05 and p<0.01, respectively) and increased (p<0.01) in the PPE+B group at 21 days compared with the control group. Interleukin (IL)-1β expression was upregulated (p<0.01) whereas IL-6 was downregulated (p<0.05) in the PPE+B group at 21 days compared with the control group. PF and emphysema did not coexist in our model of lung disease and despite increased levels of oxidative stress and inflammatory markers after combined stimulus (elastase and bleomycin) overall histology was improved to that of the nearest control group.
Collapse
|
41
|
Khalighi Z, Rahmani A, Cheraghi J, Ahmadi MRH, Soleimannejad K, Asadollahi R, Asadollahi K. Perfluorocarbon attenuates inflammatory cytokines, oxidative stress and histopathologic changes in paraquat-induced acute lung injury in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 42:9-15. [PMID: 26766533 DOI: 10.1016/j.etap.2015.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 12/04/2015] [Accepted: 12/13/2015] [Indexed: 06/05/2023]
Abstract
The effects of perfluorocarbon (PFC) on paraquat (PQ) induced acute lung injury (ALI) was evaluated among rats. Twenty four Wistar rats were divided into 4 groups: control group injected by saline physiologic 0.9%, PFC group injected by Perfluorocarbon, PQ group injected by PQ and PQ+PFC group injected by PFC one hour after receiving paraquat. Bronchoalveular fluid content, inflammatory cytokines, oxidative and histopathologic changes were measured after 72 h. The levels of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and transforming growth factor-β1(TGF-β1) in the PQ group were increased compared to either control or PFC groups, but their levels decreased in PQ+PFC group significantly (p<0.05). Also, histopathologic evaluation revealed an increase in malondialdehyde (MDA) and hydroxyproline (HP) in the PQ group but a decrease in PQ+PFC group significantly (p<0.01). PFC emulsion by its anti-inflammatory, anti-oxidative and anti-fibrotic properties can reduce the inflammatory and fibrotic alterations, pulmonary oedema, and pulmonary histopathologic changes created by PQ.
Collapse
Affiliation(s)
- Zahra Khalighi
- Department of Internal Medicine, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Asghar Rahmani
- Student Researches Committee, Ilam University of Medical Sciences, Ilam, Iran
| | - Javad Cheraghi
- Department of Physiology, Faculty of Veterinary, Ilam University, Ilam, Iran
| | | | - Koroush Soleimannejad
- Department of Cardiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Ruhangiz Asadollahi
- Department of Pathology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Khairollah Asadollahi
- Department of Epidemiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran.
| |
Collapse
|
42
|
Correlation of TGF-β1 and oxidative stress in the blood of patients with melanoma: a clue to understanding melanoma progression? Tumour Biol 2016; 37:10753-61. [PMID: 26873487 DOI: 10.1007/s13277-016-4967-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/02/2016] [Indexed: 12/15/2022] Open
Abstract
TGF-β1 and oxidative stress are involved in cancer progression, but in melanoma, their role is still controversial. Our aim was to correlate plasma TGF-β1 levels and systemic oxidative stress biomarkers in patients with melanoma, with or without disease metastasis, to understand their participation in melanoma progression. Thirty patients were recruited for melanoma surveillance, together with 30 healthy volunteers. Patients were divided into two groups: Non-metastasis, comprising patients with tumor removal and no metastatic episode for 3 years; and Metastasis, comprising patients with a metastatic episode. The plasmatic cytokines TGF-β1, IL-1 β, and TNF-α were analyzed by ELISA. For oxidative stress, the following assays were performed: malondialdehyde (MDA), advanced oxidation protein products (AOPP) levels, total radical-trapping antioxidant parameter (TRAP) and thiol in plasma, and lipid peroxidation, SOD and catalase activity and GSH in erythrocytes. Patients with a metastatic episode had less circulating TGF-β1 and increased TRAP, thiol, AOPP and lipid peroxidation levels. MDA was increased in both melanoma groups, while catalase, GSH, and IL-1β was decreased in Non-metastasis patients. Significant negative correlations were observed between TGF-β1 levels and systemic MDA, and TGF-β1 levels and systemic AOPP, while a positive correlation was observed between TGF-β1 levels and erythrocyte GSH. Lower levels of TGF-β1 were related to increased oxidative stress in Metastasis patients, reinforcing new evidence that in melanoma TGF-β1 acts as a tumor suppressor, inhibiting tumor relapse. These findings provide new knowledge concerning this cancer pathophysiology, extending the possibilities of investigating new therapies based on this evidence.
Collapse
|
43
|
Shao X, Li M, Luo C, Wang YY, Lu YY, Feng S, Li H, Lang XB, Wang YC, Lin C, Shen XJ, Zhou Q, Jiang H, Chen JH. Effects of rapamycin against paraquat-induced pulmonary fibrosis in mice. J Zhejiang Univ Sci B 2015; 16:52-61. [PMID: 25559956 DOI: 10.1631/jzus.b1400229] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND AND AIMS Ingestion of paraquat (PQ), a widely used herbicide, can cause severe toxicity in humans, leading to a poor survival rate and prognosis. One of the main causes of death by PQ is PQ-induced pulmonary fibrosis, for which there are no effective therapies. The aim of this study was to evaluate the effects of rapamycin (RAPA) on inhibiting PQ-induced pulmonary fibrosis in mice and to explore its possible mechanisms. METHODS Male C57BL/6J mice were exposed to either saline (control group) or PQ (10 mg/kg body weight, intraperitoneally; test group). The test group was divided into four subgroups: a PQ group (PQ-exposed, non-treated), a PQ+RAPA group (PQ-exposed, treated with RAPA at 1 mg/kg intragastrically), a PQ+MP group (PQ-exposed, treated with methylprednisolone (MP) at 30 mg/kg intraperitoneally), and a PQ+MP+RAPA group (PQ-exposed, treated with MP at 30 mg/kg intraperitoneally and with RAPA at 1 mg/kg intragastrically). The survival rate and body weight of all the mice were recorded every day. Three mice in each group were sacrificed at 14 d and the rest at 28 d after intoxication. Lung tissues were excised and stained with hematoxylin-eosin (H&E) and Masson's trichrome stain for histopathological analysis. The hydroxyproline (HYP) content in lung tissues was detected using an enzyme-linked immunosorbent assay (ELISA) kit. The expression of transforming growth factor-β1 (TGF-β1) and α-smooth muscle actin (α-SMA) in lung tissues was detected by immunohistochemical staining and Western blotting. RESULTS A mice model of PQ-induced pulmonary fibrosis was established. Histological examination of lung tissues showed that RAPA treatment moderated the pathological changes of pulmonary fibrosis, including alveolar collapse and interstitial collagen deposition. HYP content in lung tissues increased soon after PQ intoxication but had decreased significantly by the 28th day after RAPA treatment. Immunohistochemical staining and Western blotting showed that RAPA treatment significantly down-regulated the enhanced levels of TGF-β1 and α-SMA in lung tissues caused by PQ exposure. However, RAPA treatment alone could not significantly ameliorate the lower survival rate and weight loss of treated mice. MP treatment enhanced the survival rate, but had no significant effects on attenuating PQ-induced pulmonary fibrosis or reducing the expression of TGF-β1 and α-SMA. CONCLUSIONS This study demonstrates that RAPA treatment effectively suppresses PQ-induced alveolar collapse and collagen deposition in lung tissues through reducing the expression of TGF-β1 and α-SMA. Thus, RAPA has potential value in the treatment of PQ-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Xue Shao
- Kidney Disease Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
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: 488] [Impact Index Per Article: 48.8] [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
|
45
|
Huang H, Dai HP, Kang J, Chen BY, Sun TY, Xu ZJ. Double-Blind Randomized Trial of Pirfenidone in Chinese Idiopathic Pulmonary Fibrosis Patients. Medicine (Baltimore) 2015; 94:e1600. [PMID: 26496265 PMCID: PMC4620844 DOI: 10.1097/md.0000000000001600] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) lacks effective treatment. Pirfenidone has been used to treat IPF patients. N-acetylcysteine (NAC) exerts antioxidant and antifibrotic effects on IPF cases.This study is a double-blind, modified placebo-controlled, randomized phase II trial of pirfenidone in Chinese IPF patients. We randomly assigned the enrolled Chinese IPF patients with mild to moderate impairment of pulmonary function to receive either oral pirfenidone (1800 mg per day) and NAC (1800 mg per day) or placebo and NAC (1800 mg per day) for 48 weeks. The primary endpoints were the changes in forced vital capacity (FVC) and walking distance and the lowest SPO2 during the 6-minute walk test (6MWT) at week 48. The key secondary endpoint was the progression-free survival time. This study is registered in ClinicalTrials.gov as number NCT01504334.Eighty-six patients were screened, and 76 cases were enrolled (pirfenidone + NAC: 38; placebo + NAC: 38). The effect of pirfenidone treatment was significant at the 24th week, but this effect did not persist to the 48th week. At the 24th week, the mean decline in both FVC and ΔSPO2 (%) during the 6MWT in the pirfenidone group was lower than that in the control group (-0.08 ± 0.20 L vs -0.22 ± 0.29 L, P = 0.02 and -3.44% ± 4.51% vs -6.29% ± 6.06%, P = 0.03, respectively). However, there was no significant difference between these 2 groups at the 48th week (-0.15 ± 0.25 L vs -0.25 ± 0.28 L, P = 0.11 and -4.25% ± 7.27% vs -5.31% ± 5.49%, P = 0.51, respectively). The pirfenidone treatment group did not achieve the maximal distance difference on the 6MWT at either the 24th or the 48th week. But pirfenidone treatment prolonged the progression-free survival time in the IPF patients (hazard ratio = 1.88, 95% confidence interval: 1.092-3.242, P = 0.02). In the pirfenidone group, the adverse event (AE) rate (52.63%) was higher than that in the control group (26.3%, P = 0.03). Rash was more common in the pirfenidone group (39.5% vs 13.2%, P = 0.02).Compared with placebo combined with high-dose NAC, pirfenidone combined with high-dose NAC prolonged the progression-free survival of Chinese IPF patients with mild to moderate impairment of pulmonary function. (ClinicalTrials.gov number, NCT01504334).
Collapse
Affiliation(s)
- Hui Huang
- From the Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (HH, ZJX); Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University; Beijing Key Laboratory of Respiratory and Pulmonary Circulation, Beijing Institute of Respiratory Medicine, Beijing (HPD); Department of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang (JK); Department of Respiratory Diseases, Tianjin Medical University General Hospital, Tianjin (BYC); and Department of Respiratory and Critical Care Medicine, Beijing Hospital, Beijing, China (TYS)
| | | | | | | | | | | |
Collapse
|
46
|
Lee SS, Chen YJ, Tsai CH, Huang FM, Chang YC. Elevated transglutaminase-2 expression mediates fibrosis in areca quid chewing-associated oral submucocal fibrosis via reactive oxygen species generation. Clin Oral Investig 2015; 20:1029-34. [DOI: 10.1007/s00784-015-1579-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 08/26/2015] [Indexed: 12/24/2022]
|
47
|
Cui Y, Wilder J, Rietz C, Gigliotti A, Tang X, Shi Y, Guilmette R, Wang H, George G, Nilo de Magaldi E, Chu SG, Doyle-Eisele M, McDonald JD, Rosas IO, El-Chemaly S. Radiation-induced impairment in lung lymphatic vasculature. Lymphat Res Biol 2015; 12:238-50. [PMID: 25412238 DOI: 10.1089/lrb.2014.0012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The lymphatic vasculature has been shown to play important roles in lung injury and repair, particularly in lung fibrosis. The effects of ionizing radiation on lung lymphatic vasculature have not been previously reported. METHODS AND RESULTS C57Bl/6 mice were immobilized in a lead shield exposing only the thoracic cavity, and were irradiated with a single dose of 14 Gy. Animals were sacrificed and lungs collected at different time points (1, 4, 8, and 16 weeks) following radiation. To identify lymphatic vessels in lung tissue sections, we used antibodies that are specific for lymphatic vessel endothelial receptor 1 (LYVE-1), a marker of lymphatic endothelial cells (LEC). To evaluate LEC cell death and oxidative damage, lung tissue sections were stained for LYVE-1 and with TUNEL staining, or 8-oxo-dG respectively. Images were imported into ImageJ v1.36b and analyzed. Compared to a non-irradiated control group, we observed a durable and progressive decrease in the density, perimeter, and area of lymphatic vessels over the study period. The decline in the density of lymphatic vessels was observed in both subpleural and interstitial lymphatics. Histopathologically discernible pulmonary fibrosis was not apparent until 16 weeks after irradiation. Furthermore, there was significantly increased LEC apoptosis and oxidative damage at one week post-irradiation that persisted at 16 weeks. CONCLUSIONS There is impairment of lymphatic vasculature after a single dose of ionizing radiation that precedes architectural distortion and fibrosis, suggesting important roles for the lymphatic circulation in the pathogenesis of the radiation-induced lung injury.
Collapse
Affiliation(s)
- Ye Cui
- 1 Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital , Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Total Glucosides of Danggui Buxue Tang Attenuate BLM-Induced Pulmonary Fibrosis via Regulating Oxidative Stress by Inhibiting NOX4. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:645814. [PMID: 26347805 PMCID: PMC4548145 DOI: 10.1155/2015/645814] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 06/22/2015] [Accepted: 07/14/2015] [Indexed: 11/17/2022]
Abstract
Pulmonary fibrosis (PF) is a serious chronic lung disease with unknown pathogenesis. Researches have confirmed that oxidative stress which is regulated by NADPH oxidase-4 (NOX4), a main source of reactive oxygen species (ROS), is an important molecular mechanism underlying PF. Previous studies showed that total glucosides of Danggui Buxue Tang (DBTG), an extract from a classical traditional Chinese herbal formula, Danggui Buxue Tang (DBT), attenuated bleomycin-induced PF in rats. However, the mechanisms of DBTG are still not clear. We hypothesize that DBTG attenuates PF through regulating the level of oxidative stress by inhibiting NOX4. And we found that fibrosis indexes hydroxyproline (HYP) and type I collagen (Col-I) were lower in DBTG groups compared with the model group. In addition, the expression of transforming growth factor-β1 (TGF-β1) and expression of alpha smooth muscle actin (α-SMA) were also much more decreased than the model group. For oxidative stress indicators, DBTG blunted the decrease of superoxide dismutase (SOD) activity, total antioxidant capacity (T-AOC), and the increase in malondialdehyde (MDA), 8-iso-prostaglandin in lung homogenates. Treatment with DBTG restrained the expression of NOX4 compared to the model group. Present study confirms that DBTG inhibits BLM-induced PF by modulating the level of oxidative stress via suppressing NOX4.
Collapse
|
49
|
Wieczfinska J, Sokolowska M, Pawliczak R. NOX Modifiers-Just a Step Away from Application in the Therapy of Airway Inflammation? Antioxid Redox Signal 2015; 23:428-45. [PMID: 24383678 PMCID: PMC4543397 DOI: 10.1089/ars.2013.5783] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE NADPH oxidase (NOX) enzymes, which are widely expressed in different airway cell types, not only contribute to the maintenance of physiological processes in the airways but also participate in the pathogenesis of many acute and chronic diseases. Therefore, the understanding of NOX isoform regulation, expression, and the manner of their potent inhibition might lead to effective therapeutic approaches. RECENT ADVANCES The study of the role of NADPH oxidases family in airway physiology and pathophysiology should be considered as a work in progress. While key questions still remain unresolved, there is significant progress in terms of our understanding of NOX importance in airway diseases as well as a more efficient way of using NOX modifiers in human settings. CRITICAL ISSUES Agents that modify the activity of NADPH enzyme components would be considered useful tools in the treatment of various airway diseases. Nevertheless, profound knowledge of airway pathology, as well as the mechanisms of NOX regulation is needed to develop potent but safe NOX modifiers. FUTURE DIRECTIONS Many compounds seem to be promising candidates for development into useful therapeutic agents, but their clinical potential is yet to be demonstrated. Further analysis of basic mechanisms in human settings, high-throughput compound scanning, clinical trials with new and existing molecules, and the development of new drug delivery approaches are the main directions of future studies on NOX modifiers. In this article, we discuss the current knowledge with regard to NOX isoform expression and regulation in airway inflammatory diseases as well as the aptitudes and therapeutic potential of NOX modifiers.
Collapse
Affiliation(s)
- Joanna Wieczfinska
- 1 Department of Immunopathology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz , Lodz, Poland
| | - Milena Sokolowska
- 2 Critical Care Medicine Department, Clinical Center, National Institutes of Health , Bethesda, Maryland
| | - Rafal Pawliczak
- 1 Department of Immunopathology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz , Lodz, Poland
| |
Collapse
|
50
|
Liu YN, Zha WJ, Ma Y, Chen FF, Zhu W, Ge A, Zeng XN, Huang M. Galangin attenuates airway remodelling by inhibiting TGF-β1-mediated ROS generation and MAPK/Akt phosphorylation in asthma. Sci Rep 2015; 5:11758. [PMID: 26156213 PMCID: PMC4496730 DOI: 10.1038/srep11758] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 06/05/2015] [Indexed: 02/07/2023] Open
Abstract
Galangin, a natural flavonol, has attracted much attention for its potential anti-inflammatory properties. However, its role in the regulation of airway remodelling in asthma has not been explored. The present study aimed to elucidate the effects of galangin on chronic inflammation and airway remodelling and to investigate the underlying mechanisms both in vivo and in vitro. Ovalbumin (OVA)-sensitised mice were administered with galangin 30 min before challenge. Our results showed that severe inflammatory responses and airway remodelling occurred in OVA-induced mice. Treatment with galangin markedly attenuated the leakage of inflammatory cells into bronchoalveolar lavage fluid (BALF) and decreased the level of OVA-specific IgE in serum. Galangin significantly inhibited goblet cell hyperplasia, collagen deposition and α-SMA expression. Lowered level of TGF-β1 and suppressed expression of VEGF and MMP-9 were observed in BALF or lung tissue, implying that galangin has an optimal anti-remodelling effect in vivo. Consistently, the TGF-β1-induced proliferation of airway smooth muscle cells was reduced by galangin in vitro, which might be due to the alleviation of ROS levels and inhibition of MAPK pathway. Taken together, the present findings highlight a novel role for galangin as a promising anti-remodelling agent in asthma, which likely involves the TGF-β1-ROS-MAPK pathway.
Collapse
Affiliation(s)
- Ya-Nan Liu
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wang-Jian Zha
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuan Ma
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fei-Fei Chen
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wen Zhu
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ai Ge
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao-Ning Zeng
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mao Huang
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|