1
|
Guo Y, Jin Q, Kang Y, Jin W, Liu Y, Chen Q, Liu J, Wang YG. Integrating machine learning and neural networks for new diagnostic approaches to idiopathic pulmonary fibrosis and immune infiltration research. PLoS One 2025; 20:e0320242. [PMID: 40273141 PMCID: PMC12021136 DOI: 10.1371/journal.pone.0320242] [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: 08/16/2024] [Accepted: 02/15/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease with a fatal outcome, known for its rapid progression and unpredictable clinical course. However, the tools available for diagnosing and treating IPF are quite limited. This study aims to identify and screen potential biomarkers for IPF diagnosis, thereby providing new diagnostic approaches. METHODS We choosed datasets from the Gene Expression Omnibus (GEO) database, including samples from both IPF patients and healthy controls. For the training set, we combined two gene array datasets (GSE24206 and GSE10667) and utilized GSE32537 as the test set. We identified differentially expressed genes (DEGs) between IPF and normal tissues and determined IPF-related modules using Weighted Gene Co-expression Network Analysis (WGCNA). Subsequently, we employed two machine learning strategies to screen potential diagnostic biomarkers. Candidate biomarkers were quantitatively evaluated using Receiver Operating Characteristic (ROC) curves to identify key diagnostic genes, followed by the construction of a nomogram. Further validation of the expression of these genes through transcriptomic sequencing data from IPF and normal group animal models. Next, we conducted immune infiltration analysis, single-gene Gene Set Enrichment Analysis (GSEA), and targeted drug prediction. Finally, we created an artificial neural network model specifically for IPF. RESULTS We identified ASPN, COMP, and GPX8 as candidate biomarker genes for IPF, all of which exhibited Area Under the Curve (AUC) above 0.90. These genes were validated by RT-qPCR. Immune infiltration analysis revealed that specific immune cell types are closely related to IPF, suggesting that these immune cells may play a significant role in the pathogenesis of IPF. CONCLUSION ASPN, COMP, and GPX8 have been identified as potential diagnostic genes for IPF, and the most relevant immune cell types have been determined. Our research results propose potential biomarkers for diagnosing IPF and present new pathways for investigating its pathogenesis and devising novel therapeutic approaches.
Collapse
Affiliation(s)
- Yali Guo
- Department of Respiratory Medicine, Beijing Hospital of Traditional Chinese Medicine, Affiliated to Capital Medical University, Beijing, China
| | - Qian Jin
- Department of Respiratory Medicine, Beijing Hospital of Traditional Chinese Medicine, Affiliated to Capital Medical University, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China.
| | - Yi Kang
- Beijing University of Chinese Medicine, Beijing, China.
| | - Wenwen Jin
- Department of Respiratory Medicine, Beijing Hospital of Traditional Chinese Medicine, Affiliated to Capital Medical University, Beijing, China
| | - Ying Liu
- Beijing University of Chinese Medicine, Beijing, China.
| | - Qian Chen
- Department of Respiratory Medicine, Beijing Hospital of Traditional Chinese Medicine, Affiliated to Capital Medical University, Beijing, China
| | - Jian Liu
- Department of Respiratory Medicine, Beijing Hospital of Traditional Chinese Medicine, Affiliated to Capital Medical University, Beijing, China
| | - Yu guang Wang
- Department of Respiratory Medicine, Beijing Hospital of Traditional Chinese Medicine, Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
2
|
高 志, 吴 傲, 胡 仲, 孙 培. [Bioinformatics analysis of oxidative stress and immune infiltration in rheumatoid arthritis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:862-870. [PMID: 40294937 PMCID: PMC12037277 DOI: 10.12122/j.issn.1673-4254.2025.04.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Indexed: 04/30/2025]
Abstract
OBJECTIVES To explore the role of oxidative stress and immune infiltration in rheumatoid arthritis (RA). METHODS RA datasets GSE55235 (10 RA vs 10 normal samples) and GSE55457 (13 RA vs 10 normal samples) from the GEO database were merged as the test set to identify the differentially expressed genes (DEGs) in RA using R. The DEGs were intersected with oxidative stress-related genes to obtain oxidative stress-associated DEGs. KEGG and GO enrichment analyses of the DEGs were performed, and the RA-related pathways and biological processes were analyzed using GSEA. A protein-protein interaction (PPI) network was constructed using STRING and Cytoscape, and the top 10 key genes were obtained using the Degree algorithm. The validation dataset GSE1919 from GEO database was used for ROC analysis of the key genes to obtain the core genes, and their correlations with infiltrating immune cells were analyzed using CIBERSORT. The results were verified by RT-qPCR for detecting expression levels of the core genes in RA and normal joint samples. RESULTS We identified 89 oxidative stress-associated DEGs. Enrichment analysis suggested that these DEGs were involved in the biological processes including oxidative stress, chemical stress response, reactive oxygen species response, and lipopolysaccharide response. ROC analysis showed that the 5 core genes (STAT1, MMP9, MYC, CCL5, and JUN) all had AUC values >0.7, indicating their high diagnostic sensitivity and specificity for RA. These genes were closely correlated with immune cells, particularly T cells. RT-qPCR confirmed significant differential expressions of the core genes between RA and normal samples. CONCLUSIONS Oxidative stress and diverse immune responses are features of RA, and the immune responses contribute to activation of oxidative stress. The identified core genes can potential serve as new diagnostic markers for RA.
Collapse
|
3
|
Alzahrani KJ, El Safadi M, Alzahrani FM, Akbar A, Alsiwiehri NO. Bromoxynil induced hepatic toxicity via dysregulating TLR4/MyD88, JAK1/STAT3 and NF-κB signaling pathways: A dose-dependent investigation. Tissue Cell 2025; 93:102735. [PMID: 39827709 DOI: 10.1016/j.tice.2025.102735] [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: 10/13/2024] [Revised: 12/29/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Bromoxynil (BML) is a toxic herbicide that is reported to cause various organ toxicities. However, there is not a single investigation conducted to elucidate the adverse impacts of BML on hepatic tissues at different dose concentrations. Therefore, the current investigation was planned to assess the deleterious effects of BML on liver against different dose concentrations. Thirty-six albino rats (Sprague Dawley) were divided into four groups including the control, BML (10 mg/kg), BML (20 mg/kg) and BML (40 mg/kg). Gene expressions were assessed by qRT-PCR while other biochemical parameters were evaluated through ELISA as well as standard assays. The histological procedure was conducted as per the standard protocols of histomorphology. It is revealed that BML intoxication at all tested doses showed notable elevation in the gene expression of tumor necrosis factor-alpha (TNF-α), toll-like receptors-4 (TLR-4), interleukin-1beta (IL-1β), myeloid differentiation primary response protein-88 (MyD88), interleukin-6 (IL-6), tumor necrosis factor receptor-associated factor-6 (TRAF-6), cyclooxygenase-2 (COX-2), nuclear factor kappa-B (NF-κB), Janus kinase 1 (JAK1) and signal transducer and activator of transcription 3 (STAT3) while reducing the gene expression of inhibitor of kappa-B (I-κB). Moreover, BML exposure (10 mg/kg, 20 mg/kg, 40 mg/kg) reduced the activities of catalase (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD), glutathione (GSH), glutathione S- transferase (GST), heme-oxygenase-1 (HO-1) and glutathione reductase (GSR) while upregulating the levels of reactive oxygen species (ROS) and malondialdehyde (MDA). However, an elevation was observed in the levels of alanine transaminase (ALT), gamma-glutamyl transpeptidase (GGT), aspartate aminotransferase (AST), and alkaline phosphatase (ALP) while a reduction in the levels of total proteins and albumin was observed after high dose (20 mg/kg, 40 mg/kg) of BML. There was insignificant elevation among the values of these biomarkers at 10 mg/kg administration of BML. Besides, BML exposure at 10 mg/kg, 20 mg/kg and 40 mg/kg escalated the levels of Bcl-2-associated X protein (Bax), cysteine-aspartic acid protease-9 (Caspase-9) and cysteine-aspartic acid protease-3 (Caspase-3) while reducing the levels of B-cell lymphoma 2 (Bcl-2) in hepatic tissues. Similarly, BML at all tested concentrations showed adverse impacts on hepatic histology. These findings validated the deleterious impacts of BML on hepatic tissues owing to its pro-oxidative, pro-inflammatory and pro-apoptotic potential.
Collapse
Affiliation(s)
- Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Mahmoud El Safadi
- Department of Chemistry, College of Science, United Arab Emirates University, P.O. Box 15551, Al Ain, Abu Dhabi, United Arab Emirates
| | - Fuad M Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ali Akbar
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan.
| | - Naif O Alsiwiehri
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
4
|
Dong Z, Wang X, Wang P, Bai M, Wang T, Chu Y, Qin Y. Idiopathic Pulmonary Fibrosis Caused by Damaged Mitochondria and Imbalanced Protein Homeostasis in Alveolar Epithelial Type II Cell. Adv Biol (Weinh) 2025; 9:e2400297. [PMID: 39390651 PMCID: PMC12001015 DOI: 10.1002/adbi.202400297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/21/2024] [Indexed: 10/12/2024]
Abstract
Alveolar epithelial Type II (ATII) cells are closely associated with early events of Idiopathic pulmonary fibrosis (IPF). Proteostasis dysfunction, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction are known causes of decreased proliferation of alveolar epithelial cells and the secretion of pro-fibrotic mediators. Here, a large body of evidence is systematized and a cascade relationship between protein homeostasis, endoplasmic reticulum stress, mitochondrial dysfunction, and fibrotropic cytokines is proposed, providing a theoretical basis for ATII cells dysfunction as a possible pathophysiological initiating event for idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Zhaoxiong Dong
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- Institute of BiophysicsChinese Academy of Sciences 15 Datun RoadChaoyang DistrictBeijing100101China
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Xiaolong Wang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
| | - Peiwen Wang
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Mingjian Bai
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- School of Chemistry and Biological EngineeringUniversity of Science and Technology BeijingBeijing100101China
| | - Tianyu Wang
- School of Chemistry and Biological EngineeringUniversity of Science and Technology BeijingBeijing100101China
| | - Yanhui Chu
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Yan Qin
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- Institute of BiophysicsChinese Academy of Sciences 15 Datun RoadChaoyang DistrictBeijing100101China
| |
Collapse
|
5
|
Milivojac T, Grabež M, Amidžić L, Prtina A, Krivokuća A, Malicevic U, Barudžija M, Matičić M, Uletilović S, Mandić-Kovačević N, Cvjetković T, Stojiljković MP, Gajić Bojić M, Mikov M, Gajanin R, Bolevich S, Petrović A, Škrbić R. Ursodeoxycholic and chenodeoxycholic bile acids alleviate endotoxininduced acute lung injury in rats by modulating aquaporin expression and pathways associated with apoptosis and inflammation. Front Pharmacol 2025; 16:1484292. [PMID: 40115259 PMCID: PMC11922783 DOI: 10.3389/fphar.2025.1484292] [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: 08/21/2024] [Accepted: 02/06/2025] [Indexed: 03/23/2025] Open
Abstract
Introduction This study aimed to investigate the anti-inflammatory, antioxidant, and anti-apoptotic properties of ursodeoxycholic (UDCA) and chenodeoxycholic (CDCA) bile acids in a rat model of endotoxin (lipopolysaccharide, LPS)-induced acute lung injury (ALI). Methods The study included six groups of Wistar rats exposed to different pretreatments. The control and endotoxin groups were pretreated with propylene glycol, a solvent for bile acids, while the other groups received UDCA or CDCA for 10 days. On the 10th day, an endotoxin injection was given to evaluate the impact of these pretreatments. Lung tissue sections were analyzed by immunohistochemistry, targeting the pro-inflammatory marker nuclear factor kappa B (NF-κB), the anti-apoptotic marker B-cell lymphoma 2 (BCL-2), pro-apoptotic markers BCL-2-associated X protein (BAX) and caspase 3, as well as the aquaporins 1 and 5 (AQP1 and AQP5). Oxidative stress was assessed in bronchoalveolar lavage fluid (BALF). Results and discussion This study demonstrates that UDCA and CDCA can mitigate endotoxin-induced lung injury in rats. These effects are achieved through modulation of AQP1 and AQP5 expression, reduction of oxidative stress, regulation of apoptotic pathways (BAX, caspase 3, BCL-2), and attenuation of pro-inflammatory activity of NF-κB. Although the results indicate a significant association between the expression of these proteins and histopathological changes, the potential influence of additional factors cannot be excluded. These findings suggest that UDCA and CDCA provide lung protection by acting through complex mechanisms involving inflammatory, oxidative, and apoptotic pathways.
Collapse
Affiliation(s)
- Tatjana Milivojac
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Milkica Grabež
- Department of Hygiene, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Ljiljana Amidžić
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Alma Prtina
- Department of Pathophysiology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Aleksandra Krivokuća
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pathophysiology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Ugljesa Malicevic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pathophysiology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Maja Barudžija
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Histology and Embryology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Milka Matičić
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Snežana Uletilović
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Medical Biochemistry and Chemistry, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Nebojša Mandić-Kovačević
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pharmacy, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Tanja Cvjetković
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Medical Biochemistry and Chemistry, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Miloš P Stojiljković
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Milica Gajić Bojić
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Momir Mikov
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Radoslav Gajanin
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Sergey Bolevich
- Department of Pathologic Physiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Aleksandar Petrović
- Department of Histology and Embryology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Ranko Škrbić
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pathologic Physiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
6
|
Zhao W, Bai B, Li H, Feng Y, Sun J, Fang Y, Zheng P, Zhang G. The role of oxidative stress-related genes in idiopathic pulmonary fibrosis. Sci Rep 2025; 15:5954. [PMID: 39966531 PMCID: PMC11836339 DOI: 10.1038/s41598-025-89770-y] [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: 07/08/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an age-related interstitial lung disease of unknown cause. Oxidative stress, an imbalance between oxidants and antioxidants, is implicated in IPF pathogenesis and prognosis but needs further study. We used transcriptome sequencing data (GSE70866) and oxidative stress-related genes from GeneCards. A prognostic risk model for IPF patients was constructed using LASSO. Functional and pathway differences were analyzed between risk score groups, along with comparisons of immune cells and functions. An IPF rat model with vitamin D3 (VD3) intervention was also established. Finally, we used IL-4 to induce M2 macrophages to explore the mechanism of action of CCL2. We identified 483 DEGs and 50 oxidative stress-related DEGs (OSDEGs). Single-factor COX regression identified 34 prognostic OSDEGs, and LASSO identified an 8-gene signature for the risk model. The high-risk group had more CD8 + T cells, macrophages, APC costimulation, and cytokine-cytokine receptor activity. CCL2 was significantly correlated with macrophages in IPF. VD3 inhibited the TGF-β signaling pathway and reduced macrophage M2 infiltration in the rat model. In the IL-4 induced M2 macrophage model, we found that M2 macrophages produced more CCL2, and the production of CCL2 was significantly reduced after VD3 intervention. We established prognostic markers of eight oxidative stress-related genes. The risk score effectively predicts adverse outcomes in IPF. VD3 may alleviate IPF by reducing macrophage infiltration and inhibiting the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Wenfei Zhao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, People's Republic of China
| | - Bing Bai
- Fuhua Street Branch of the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 453000, Henan, People's Republic of China
- Henan Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450002, People's Republic of China
| | - Hongyun Li
- Department of Respiratory and Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 453000, Henan, People's Republic of China
| | - Yonghai Feng
- Department of Respiratory and Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 453000, Henan, People's Republic of China
| | - Jun Sun
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yang Fang
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Pengyuan Zheng
- Henan Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450002, People's Republic of China.
| | - Guojun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, People's Republic of China.
| |
Collapse
|
7
|
Wang Y, Ma Z, Peng W, Yu Q, Liang W, Cao L, Wang Z. 3,5,6,7,8,3',4'- Heptamethoxyflavonoid inhibits TGF-β1-induced epithelial-mesenchymal transition by regulating oxidative stress and autophagy through MEK/ERK/PI3K/AKT/mTOR signaling pathway. Sci Rep 2025; 15:4567. [PMID: 39915543 PMCID: PMC11802913 DOI: 10.1038/s41598-025-88869-6] [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: 06/12/2024] [Accepted: 01/31/2025] [Indexed: 02/09/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a crucial pathological process in the pathogenesis of fibrosis. 3,5,6,7,8,3',4'-hepmethoxyflavone (HMF), the main active ingredient extracted from the Chinese herb Breynia fruticosa (L.) Hook. f., has been shown to have beneficial effects on regulating apoptosis and inhibiting collagen deposition. However, it remains unclear whether and how HMF alleviates transforming growth factor-β1 (TGF-β1)-induced EMT. The objective of this study was to investigate the impact of HMF on TGF-β1-induced EMT in human alveolar Type II epithelial cells (A549) and its underlying mechanism. In vitro culture of TGF-β1-induced EMT in A549 cells revealed that HMF reduced cell viability and migration, inhibited collagen deposition, decreased expression levels of mesenchymal cell markers and fibrosis markers α-SMA, MMP2, TIMP1, β-catenin, and Snail. Meanwhile, the expression level of E-cadherin increased as an epithelial cell marker. Additionally, we discussed the effects of HMF on oxidative stress and autophagy. Various experiments confirmed that HMF regulated the expression levels of Nrf2, keap-1, HO-1, ROS, MDA, SOD, GSH, and played a role in reducing oxidative stress. At the same time, HMF significantly activated autophagy by increasing expressions of Beclin-1 and LC3B as well as enhancing autophagosome content. The addition 3-MA, an autophagy inhibitor attenuated these beneficial effects. Furthermore, HMF significantly inhibited phosphorylation levels of MEK, ERK, PI3K, AKT, and mTOR through various pathways. In conclusion, HMF effectively inhibits TGF-β1-induced EMT in A549 cells by targeting the MEK/ERK/PI3K/AKT/mTOR signaling pathway. Moreover, it exhibits a close correlation with the suppression of oxidative stress and induction of autophagy.
Collapse
Affiliation(s)
- Yiting Wang
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Zhiheng Ma
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Weiwen Peng
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Qinglian Yu
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Wenjie Liang
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Liu Cao
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Zhuqiang Wang
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China.
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China.
| |
Collapse
|
8
|
Nandha SR, Checker R, Patwardhan RS, Sharma D, Sandur SK. Anti-oxidants as therapeutic agents for oxidative stress associated pathologies: future challenges and opportunities. Free Radic Res 2025; 59:61-85. [PMID: 39764687 DOI: 10.1080/10715762.2025.2450504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/13/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Free radicals have been implicated in the pathogenesis of cancer along with cardiovascular, neurodegenerative, pulmonary and inflammatory disorders. Further, the relationship between oxidative stress and disease is distinctively established. Clinical trials using anti-oxidants for the prevention of disease progression have indicated some beneficial effects. However, these trials failed to establish anti-oxidants as therapeutic agents due to lack of efficacy. This is attributed to the fact that living systems are under dynamic redox control wherein their redox behavior is compartmentalized and simple aggregation of redox couples, distributed throughout the system, is of miniscule importance while determining their overall redox state. Further, free radical metabolism is intriguingly complex as they play plural roles segregated in a spatio-temporal manner. Depending on quality, quantity and site of generation, free radicals exhibit beneficial or harmful effects. Use of nonspecific, non-targeted, general ROS scavengers lead to systemic elimination of all types of ROS and interferes in cellular signaling. Failure of anti-oxidants to act as therapeutic agents lies in this oversimplification of extremely dynamic cellular redox environment as a static and non-compartmentalized redox state. Rather than generalizing the term "oxidative stress" if we can identify the "type of oxidative stress" in different types of diseases, a targeted and more specific anti-oxidant therapy may be developed. In this review, we discuss the concept of redox dynamics, role and type of oxidative stress in disease conditions, and current status of anti-oxidants as therapeutic agents. Further, we probe the possibility of developing novel, targeted and efficacious anti-oxidants with drug-like properties.
Collapse
Affiliation(s)
- Shivani R Nandha
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Rahul Checker
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Raghavendra S Patwardhan
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Santosh K Sandur
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
9
|
Yu JW, Lu WH. Melittin alleviates bleomycin-induced pulmonary fibrosis in vivo through regulating TGF-β1/Smad2/3 and AMPK/SIRT1/PGC-1α signaling pathways. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2025; 28:426-433. [PMID: 39968084 PMCID: PMC11831745 DOI: 10.22038/ijbms.2024.81986.17740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 09/28/2024] [Indexed: 02/20/2025]
Abstract
Objectives The present study investigated the protective effect of melittin (MEL) against bleomycin (BLM)- induced pulmonary fibrosis (PF) in mice and the mechanism underlying this effect. Materials and Methods A mouse model of PF was established by intratracheal injection of 3.5 mg/kg BLM. Twenty-four hours after the model was established, the mice in the treatment groups were intraperitoneally injected with MEL, and specimens were collected 28 days later. The body weight, survival rate, and pulmonary index (PI) of the mice were determined. Haematoxylin and eosin (HE) staining, Masson's trichrome staining, immunohistochemical staining, kit assays, and Western blot (WB) analysis were performed. Results Our study indicated that MEL significantly increased the body weight and survival rate, reduced PI, and improved lung histopathology in mice. In addition, MEL inhibited epithelial-mesenchymal transition (EMT) and extracellular matrix (ECM) deposition. Attenuated mitochondrial damage and reduced oxidative stress (OS) were also observed in MEL-treated mice. We further showed that MEL inhibited the TGF-β1/Smad2/3 pathway and activated the AMPK/SIRT1/PGC-1α pathway. Conclusion MEL is a promising future therapeutic agent for PF. Its multifaceted and complex mechanism of action inhibits both EMT and ECM production by modulating the TGF-β1/Smad2/3 pathway. It also improves mitochondrial function and reduces OS at least partially through the activation of the AMPK/SIRT1/PGC-1α signaling pathway.
Collapse
Affiliation(s)
- Jia-Wang Yu
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, 230032, China
- EICU, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, China
| | - Wei-Hua Lu
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, 230032, China
- ICU, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, China
| |
Collapse
|
10
|
Krauss E, Tello S, Naumann J, Wobisch S, Ruppert C, Kuhn S, Mahavadi P, Majeed RW, Bonniaud P, Molina-Molina M, Wells A, Hirani N, Vancheri C, Walsh S, Griese M, Crestani B, Guenther A. Protocol and research program of the European registry and biobank for interstitial lung diseases (eurILDreg). BMC Pulm Med 2024; 24:572. [PMID: 39558302 PMCID: PMC11575435 DOI: 10.1186/s12890-024-03389-9] [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: 08/14/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND AND AIMS Interstitial lung diseases (ILDs), encompassing both pediatric and adult cases, present a diverse spectrum of chronic conditions with variable prognosis. Despite limited therapeutic options beyond antifibrotic drugs and immunosuppressants, accurate diagnosis is challenging, often necessitating invasive procedures that may not be feasible for certain patients. Drawn against this background, experts across pediatric and adult ILD fields have joined forces in the RARE-ILD initiative to pioneer novel non-invasive diagnostic algorithms and biomarkers. Collaborating with the RARE-ILD consortium, the eurILDreg aims to comprehensively describe different ILDs, analyze genetically defined forms across age groups, create innovative diagnostic and therapeutic biomarkers, and employ artificial intelligence for data analysis. METHODS The foundation of eurILDreg is built on a comprehensive parameter list developed and adopted by clinical experts, encompassing over 1,800 distinct parameters related to patient history, clinical examinations, diagnosis, lung function and biospecimen collection. This robust dataset is further enriched with daily assessments captured through the patientMpower app, including handheld spirometry and exercise tests, conducted on approximately 350 patients over the course of a year. This approach involves app-based daily assessments of quality of life, symptom tracking, handheld spirometry, saturation measurement, and the 1-min sit-to-stand test (1-STST). Additionally, pediatric data from the ChILD-EU registry will be integrated into the RARE-ILD Data Warehouse, with the ultimate goal of including a total of 4.000 ILD patients and over 100.000 biospecimen. DISCUSSION The collaborative efforts within the consortium are poised to streamline research endeavors significantly, promising to advance patient-centered care, foster innovation, and shape the future landscape of interstitial lung disease research and healthcare practices. TRIAL REGISTRATION EurILDreg is registered in the German Clinical Trials Register (DRKS 00028968, 26.07.2022), and eurIPFreg is registered in ClinicalTrials.gov (NCT02951416).
Collapse
Affiliation(s)
- Ekaterina Krauss
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
- Center for Interstitial and Rare Lung Diseases, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Silke Tello
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
- Center for Interstitial and Rare Lung Diseases, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Jennifer Naumann
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
| | - Sandra Wobisch
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
| | - Clemens Ruppert
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
| | - Stefan Kuhn
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
| | - Poornima Mahavadi
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
| | - Raphael W Majeed
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
- Cardio-Pulmonary Institute (CPI), Klinikstr. 33, 35392, Giessen, Germany
| | - Philippe Bonniaud
- Service de Pneumologie Et Soins Intensifs Respiratoire, Centre de Référence Constitutif Des Maladies Pulmonaires Rares de L'Adultes de Dijon, Centre Hospitalier Universitaire de Dijon-Bourgogne, INSERM U1231, Equipe HSP-Pathies, Faculty of Medicine and Pharmacy, Université de Bourgogne, Dijon, France
| | - Maria Molina-Molina
- ILD Unit, Respiratory Department, University Hospital of Bellvitge (HUB), Biomedical Research Institute of Bellvitge (IDIBELL), Barcelona, Spain
| | | | - Nik Hirani
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Carlo Vancheri
- Department of Clinical and Experimental Medicine, Regional Referral Center for Rare Lung Diseases, University Hospital Policlinico, University of Catania, Catania, Italy
| | - Simon Walsh
- King's College Hospital Foundation Trust, Denmark Hill, London, UK
| | - Matthias Griese
- ChILD-EU, Hauner Children's Hospital, University of Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Bruno Crestani
- Institute National de La Sainté Et de La Recherche Médicale, Hopital Bichat, Service de Pneumologie, Paris, France
| | - Andreas Guenther
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany.
- Center for Interstitial and Rare Lung Diseases, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany.
- Cardio-Pulmonary Institute (CPI), Klinikstr. 33, 35392, Giessen, Germany.
- Agaplesion Lung Clinic "Evangelisches Krankenhaus Mittelhessen", Paul-Zipp Str. 171, 35398, Giessen, Germany.
- Institute for Lung Health (ILH), Giessen, Germany.
- eurILDreg Investigators, European ILD Registry (eurILDreg), Klinikstrasse 36, Giessen, 35392, Germany.
| |
Collapse
|
11
|
Baraka SM, Hussien YA, Ahmed-Farid OA, Hassan A, Saleh DO. Acrylamide-induced hypothalamic-pituitary-gonadal axis disruption in rats: Androgenic protective roles of apigenin by restoring testicular steroidogenesis through upregulation of 17β-HSD, CYP11A1 and CYP17A1. Food Chem Toxicol 2024; 194:115078. [PMID: 39515511 DOI: 10.1016/j.fct.2024.115078] [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: 09/26/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Acrylamide (ARL) exposure induces significant toxicity to the hypothalamic-pituitary-gonadal (HPG) axis, leading to detrimental effects on behavior, neuroendocrine functions, steroidogensis, oxidative stress, inflammation, hormonal balance, sperm quality, and histopathological integrity in rats. This study investigates the protective role of oral apigenin (API; 10 or 20 mg/kg/day for 28 days) against ARL-induced toxicity in the HPG axis of male Wistar rats. Behavioral assessments revealed that ARL exposure impaired motor coordination and balance, as evidenced by increased landing foot splay distance and gait score. ARL-induced toxicity elevated brain Tau protein levels and disrupted hypothalamic GnRH levels, both mitigated by API. ARL triggered oxidative/nitrosative stress, reducing GSH contents and increasing MDA and NO levels in brain and testicular tissues, which were reversed by API. Hormonal imbalance, marked by decreased serum testosterone, FSH, and LH levels, was corrected by API. API enhanced semen quality parameters, with elevation in sperm count concentration and the percentages of both progressive motility and individual motility. It also normalized testicular PS and PC content, enhanced testicular cellular energy and restored seminal amino acid. The repression of testicular steroidogenesis-related enzymes CYP11A1, CYP17A1, and 17β-HSD following ARL exposure was alleviated by API administration. API also mitigated the inflammatory effects of ARL by reducing the expression of p-NF-κB p65 and TNF-α in testicular tissue. Histopathological examinations showed that API reduced neuronal and testicular degeneration, improving spermatogenesis. These findings suggest that API confers significant protective effects against ARL-induced HPG axis toxicity by restoring testicular steroidogenesis through the upregulation of 17β-HSD, CYP11A1, and CYP17A1, potentially due to its antioxidant, anti-inflammatory, and neuroprotective properties.
Collapse
Affiliation(s)
- Sara M Baraka
- Chemistry of Natural Compounds Department, National Research Centre, Giza, 12622, Egypt
| | - Yosra A Hussien
- Pharmacology Department, National Research Centre, Giza, 12622, Egypt
| | - Omar A Ahmed-Farid
- Physiology Department, National Organization for Drug Control and Research, Giza, Egypt
| | - Azza Hassan
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Dalia O Saleh
- Pharmacology Department, National Research Centre, Giza, 12622, Egypt.
| |
Collapse
|
12
|
Bahri S, Bouazizi S, Nahdi A, Mlika M, Hamdi M, Jameleddine S. Insights on the Tunisian Prickly Pear Molasses as a Potential Antifibrotic and Antioxidant Candidate against Lung Fibrosis. Chem Biodivers 2024; 21:e202401030. [PMID: 39073317 DOI: 10.1002/cbdv.202401030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 07/30/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive interstitial disease leading to pulmonary damage and respiratory failure. We aimed to investigate the effect of prickly pear molasses (PPM) on an experimental model of pulmonary fibrosis induced by bleomycin (BLM) in Wistar rat. Animals were divided into 5 groups: the control group (G1), the BLM group (G2) and three groups (G3, G4, G5) receiving a single intra-tracheal injection of BLM (4 mg/kg) and PPM (at 2, 4.5 and10 %) that was introduced into the diet one week before BLM injection and continued for 3 weeks. Our phytochemical results revealed significant polyphenol and flavonoid content. LCMS analysis revealed the presence of Sinapinic acid, t-ferulic acid, t-cinnamic acid, Caffeic acid, gallic acid and vallinic acid among others. Our histological study revealed significant decrease in collagen deposition in the groups of rats treated with 4.5 % and 10 % molasses compared to BLM group. Oxidative stress in pulmonary tissues was investigated using catalase (CAT), superoxide dismutase (SOD), and malondialdehyde (MDA) assays. Treatment with PPM normalized the disturbance in the level of these oxidative markers in G3,G4, G5 compared to G2. In conclusion, PPM exhibit antifibrotic and antioxidant activities in BLM model of lung fibrosis.
Collapse
Affiliation(s)
- Sana Bahri
- Laboratory of Physiology, Faculty of Medicineof Tunis, University of Tunis El Manar, Tunis, La Rabta, 1007, Tunisia
- Laboratory of Physiopathology, Food and Biomolecules (LR-17-ES-03), Technology Center of Sidi Thabet, University of Manouba, Tunis, Tunisa
| | - Souhir Bouazizi
- Laboratory of Microbial Ecology and Technology, Department of Biological and Chemical Engineering, National Institute of Applied Sciences and Technology (INSAT), Centre Urbain Nord, Boulevard de la Terre2, B.P.676, 1080, Tunis, Tunisia
| | - Afef Nahdi
- Research Unit n 17/ES/13, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Mona Mlika
- Laboratory of Anatomy and Pathology, Abderhaman Mami Hospital, 2080, Ariana, Tunisia
| | - Mokhtar Hamdi
- Laboratory of Microbial Ecology and Technology, Department of Biological and Chemical Engineering, National Institute of Applied Sciences and Technology (INSAT), Centre Urbain Nord, Boulevard de la Terre2, B.P.676, 1080, Tunis, Tunisia
| | - Saloua Jameleddine
- Laboratory of Physiology, Faculty of Medicineof Tunis, University of Tunis El Manar, Tunis, La Rabta, 1007, Tunisia
- Laboratory of Physiopathology, Food and Biomolecules (LR-17-ES-03), Technology Center of Sidi Thabet, University of Manouba, Tunis, Tunisa
| |
Collapse
|
13
|
Yue Z, Jiang Z, Qian L, Li L, Qi X, Hu K. Associations of dietary sources of antioxidant intake and NAFLD: NHANES 2017-2020 and Mendelian randomization. Front Nutr 2024; 11:1447524. [PMID: 39555193 PMCID: PMC11565937 DOI: 10.3389/fnut.2024.1447524] [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: 06/11/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
Purpose To determine the association between dietary antioxidant sources and non-alcoholic fatty liver disease (NAFLD). Methods In this observational study, we utilized NHANES 2017-2020 data to identify the factors associated with NAFLD in dietary antioxidant sources via weighted multivariate logistic regression models. Then, Mendelian randomization (MR) was applied to investigate the effect of dietary antioxidant sources on NAFLD at the genetic level. Results Of the six dietary sources of antioxidants, only vitamin E (Vit E) was significantly associated with NAFLD (OR = 0.98; 95% CI: 0.97-0.99; p = 0.001). Upon adjusting for all covariates, it was determined that the highest quartile of dietary Vit E intake was associated with a decreased NAFLD occurrence compared with the lowest quartile of dietary Vit E intake (p < 0.001). The results of IVW-MR analysis revealed an association between Vit E and NAFLD (OR = 0.028; p = 0.039). Conclusion Our research indicates a negative and linear relationship between daily vitamin E intake and NAFLD.
Collapse
Affiliation(s)
- Zilong Yue
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
- General Surgery Department, Guoyang Branch of Anhui Provincial Hospital, Bozhou, Anhui, China
| | - Ziming Jiang
- Department of Urology, Shanghai 10th People’s Hospital Affiliated to Tongji University, Shanghai, China
| | - Long Qian
- General Surgery Department, Wuhu Hospital of Traditional Chinese Medicine, Wuhu, Anhui, China
| | - Lele Li
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Xianliang Qi
- General Surgery Department, Guoyang Branch of Anhui Provincial Hospital, Bozhou, Anhui, China
| | - Kaifeng Hu
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| |
Collapse
|
14
|
Xie Y, Shi S, Lv W, Wang X, Yue L, Deng C, Wang D, Han J, Ye T, Lin Y. Tetrahedral Framework Nucleic Acids Delivery of Pirfenidone for Anti-Inflammatory and Antioxidative Effects to Treat Idiopathic Pulmonary Fibrosis. ACS NANO 2024; 18:26704-26721. [PMID: 39276332 DOI: 10.1021/acsnano.4c06598] [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: 09/17/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and irreversible lung disease, and developing an effective treatment remains a challenge. The limited therapeutic options are primarily delivered by the oral route, among which pirfenidone (PFD) improves pulmonary dysfunction and patient quality of life. However, its high dose and severe side effects (dyspepsia and systemic photosensitivity) limit its clinical value. Intratracheal aerosolization is an excellent alternative method for treating lung diseases because it increases the concentration of the drug needed to reach the focal site. Tetrahedral framework nucleic acid (tFNA) is a drug delivery system with exceptional delivery capabilities. Therefore, we synthesized a PFD-tFNA (Pt) complex using tFNA as the delivery vehicle and achieved quantitative nebulized drug delivery to the lungs via micronebulizer for lung fibrosis treatment. In vivo, Pt exhibited excellent immunomodulatory capacity and antioxidant effects. Furthermore, Pt reduced mortality, gradually restored body weight and improved lung tissue structure. Similarly, Pt also exhibited superior fibrosis inhibition in an in vitro fibrosis model, as shown by the suppression of excessive fibroblast activation and epithelial-mesenchymal transition (EMT) in epithelial cells exposed to TGF-β1. Conclusively, Pt, a complex with tFNA as a transport system, could enrich the therapeutic regimen for IPF via intratracheal aerosolization inhalation.
Collapse
Affiliation(s)
- Yuting Xie
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu 610041, China
| | - Weitong Lv
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xinyu Wang
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Yue
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Conghui Deng
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Doudou Wang
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Han
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People's Hospital, Guiyang 550000, China
- National Health Commission Key Laboratory for Diagnosis and Treatment of Pulmonary Immune Diseases, Guiyang 550000, China
| | - Tinghong Ye
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu 610041, China
| |
Collapse
|
15
|
Zou Z, Li Y, Liu J, Huang B. Identification and Validation of Oxidative Stress-Related Biomarkers for Bronchopulmonary Dysplasia. Mol Biotechnol 2024:10.1007/s12033-024-01281-9. [PMID: 39292413 DOI: 10.1007/s12033-024-01281-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024]
Abstract
The objective of this study was to identify and characterize oxidative stress (OS)-related biomarkers in bronchopulmonary dysplasia (BPD) through a combination of bioinformatics analyses and wet experiments. The study utilized the Gene Expression Omnibus database to obtain the mRNA expression profile dataset GSE32472. Differential expression analysis and functional enrichment analysis were employed to investigate the role of OS-related genes in BPD. Gene Ontology Function Enrichment Analysis and Gene Set Enrichment Analysis were conducted to understand the mechanisms behind the signature. Protein-protein interaction analysis to identify hub genes in BPD, and predictions were made for microRNAs (miRNAs), transcription factors (TFs), and potential medications targeting these genes. CIBERSORT was utilized to investigate the correlation between hub genes and the infiltration of immune cells. Hub genes were ultimately determined and confirmed using expression analysis, correlation analysis, receiver operating characteristic (ROC) analysis, and quantitative real-time PCR (qRT-PCR). A novel OS-related gene signature (ARG1, CSF3R, IL1R1, IL1R2, MMP9, RETN, S100A12, and SOCS3) was constructed for the prediction of BPD. We identified 18 miRNAs, 14 TFs, and 30 potential medications targeting these genes. ROC analysis further validated that these genes could diagnose BPD with high specificity and sensitivity. The qRT-PCR revealed that IL1R1 and ARG1 were highly expressed in the lung tissue of the model group, while the expressions of RETN, SOCS3, IL1R2, and MMP9 were decreased. This study demonstrated that ARG1, CSF3R, IL1R1, IL1R2, MMP9, RETN, S100A12, and SOCS3 may serve as potential diagnostic biomarkers in BPD. Furthermore, a significant association between IL1R1 and the pathogenesis of BPD is observed.
Collapse
Affiliation(s)
- Zhenzhuang Zou
- Department of Pediatrics, The Fifth Affiliated Hospital of Zunyi Medical University, No.1439 Zhufeng Avenue, Doumen District, Zhuhai, 519100, Guangdong, People's Republic of China
- Department of Pediatrics, Women and Children, Health Institute of Futian Shenzhen, Shenzhen, 518000, China
| | - Yunrong Li
- Department of PICU, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Jiaying Liu
- Department of Pediatrics, The Fifth Affiliated Hospital of Zunyi Medical University, No.1439 Zhufeng Avenue, Doumen District, Zhuhai, 519100, Guangdong, People's Republic of China
| | - Bo Huang
- Department of Pediatrics, The Fifth Affiliated Hospital of Zunyi Medical University, No.1439 Zhufeng Avenue, Doumen District, Zhuhai, 519100, Guangdong, People's Republic of China.
| |
Collapse
|
16
|
Li Q, Yang S, Ma Y, Huang H, Zhi L, Wang S, Lu L. Hypothyroidism and dermato/polymyositis: a two-sample Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1361581. [PMID: 39296720 PMCID: PMC11408279 DOI: 10.3389/fendo.2024.1361581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/21/2024] [Indexed: 09/21/2024] Open
Abstract
Objective Observational studies have revealed a higher probability of hypothyroidism in patients with dermatomyositis (DM) or polymyositis (PM), but there is no consensus on whether hypothyroidism causally influences DM or PM. In the present study, we assessed the causal association between hypothyroidism and the risk of dermatomyositis or polymyositis using two-sample Mendelian randomization (TSMR). Methods The genome-wide association data of hypothyroidism and dermatomyositis/polymyositis were obtained from the IEU Open GWAS project. Then, TSMR was used to determine whether hypothyroidism is causally associated with DM or PM. Single-nucleotide polymorphisms (SNPs) significantly associated with hypothyroidism were identified and used as instrumental variables (IVs), and the causal relationship between hypothyroidism and DM/PM was examined using TSMR. MR pleiotropy and Cochran's Q test were used to confirm the heterogeneity and pleiotropy of identified IVs, then four different models, including the inverse variance weighted model (IVW), MR-Egger, weighted median and weighted model were applied in this MR analysis. Results Sixty-eight SNPs for DM and 68 SNPs for PM were selected as the IVs (P<5×10-8; linkage disequilibrium R2 <0.001) to assess the causal association between hypothyroidism and DM/PM selected from GWASs on hypothyroidism. The results revealed a positive causal effect of hypothyroidism on both DM and PM (DM: OR 2.563, 95% CI [1.348, 4.874], P = 0.00156; PM: OR1.709, 95% CI [1.157, 2.525], P =0.007). Moreover, there was no heterogeneity or pleiotropy in the results. Conclusion In conclusion, the MR analysis results provided strong evidence to indicate that hypothyroidism might be causally associated with DM and PM. These findings may have important implications for the pathogenesis and possible future therapies of DM/PM.
Collapse
Affiliation(s)
- Qianqian Li
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaoying Yang
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Ma
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huijing Huang
- Department of Rheumatology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Langxian Zhi
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Suli Wang
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liangjing Lu
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Saha P, Talwar P. Idiopathic pulmonary fibrosis (IPF): disease pathophysiology, targets, and potential therapeutic interventions. Mol Cell Biochem 2024; 479:2181-2194. [PMID: 37707699 DOI: 10.1007/s11010-023-04845-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/26/2023] [Indexed: 09/15/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, degenerative pulmonary condition. Transforming growth factor (TGF)-β, platelet-derived growth factor (PDGF), and tumor necrosis factor-α (TNF-α) are the major modulators of IPF that mediate myofibroblast differentiation and promote fibrotic remodeling of the lung. Cigarette smoke, asbestos fiber, drugs, and radiation are known to favor fibrotic remodeling of the lungs. Oxidative stress in the endoplasmic reticulum (ER) also leads to protein misfolding and promotes ER stress, which is predominant in IPF. This phenomenon further results in excess reactive oxygen species (ROS) aggregation, increasing oxidative stress. During protein folding in the ER, thiol groups on the cysteine residue are oxidized and disulfide bonds are formed, which leads to the production of hydrogen peroxide (H2O2) as a by-product. With the accumulation of misfolded proteins in the ER, multiple signaling cascades are initiated by the cell, collectively termed as the unfolded protein response (UPR). UPR also induces ROS production within the ER and mitochondria and promotes both pro-apoptotic and pro-survival pathways. The prevalence of post-COVID-19 pulmonary fibrosis (PCPF) is 44.9%, along with an alarming increase in "Coronavirus Disease 2019" (COVID-19) comorbidities. Fibrotic airway remodeling and declined lung function are the common endpoints of SARS-CoV-2 infection and IPF. Flavonoids are available in our dietary supplements and exhibit medicinal properties. Apigenin is a flavonoid found in plants, including chamomile, thyme, parsley, garlic, guava, and broccoli, and regulates several cellular functions, such as oxidative stress, ER stress, and fibrotic responses. In this study, we focus on the IPF and COVID-19 pathogenesis and the potential role of Apigenin in addressing disease progression.
Collapse
Affiliation(s)
- Pritha Saha
- Apoptosis and Cell Survival Research Laboratory, 412G Pearl Research Park, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Priti Talwar
- Apoptosis and Cell Survival Research Laboratory, 412G Pearl Research Park, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
- Apoptosis and Cell Survival Research Laboratory, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
18
|
Li C, An Q, Jin Y, Jiang Z, Li M, Wu X, Dang H. Identification of oxidative stress-related diagnostic markers and immune infiltration features for idiopathic pulmonary fibrosis by bibliometrics and bioinformatics. Front Med (Lausanne) 2024; 11:1356825. [PMID: 39165378 PMCID: PMC11333355 DOI: 10.3389/fmed.2024.1356825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 07/29/2024] [Indexed: 08/22/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) garners considerable attention due to its high fatality rate and profound impact on quality of life. Our study conducts a comprehensive literature review on IPF using bibliometric analysis to explore existing hot research topics, and identifies novel diagnostic and therapeutic targets for IPF using bioinformatics analysis. Publications related to IPF from 2013 to 2023 were searched on the Web of Science Core Collection (WoSCC) database. Data analysis and visualization were conducted using CiteSpace and VOSviewer software primarily. The gene expression profiles GSE24206 and GSE53845 were employed as the training dataset. The GSE110147 dataset was employed as the validation dataset. We identified differentially expressed genes (DEGs) and differentially expressed genes related to oxidative stress (DEOSGs) between IPF and normal samples. Then, we conducted Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The hub genes were screened by protein-protein interaction (PPI) networks and machine learning algorithms. The CIBERSORT was used to analyze the immune infiltration of 22 kinds of immune cells. Finally, we conducted the expression and validation of hub genes. The diagnostic efficacy of hub genes was evaluated by employing Receiver Operating Characteristic (ROC) curves and the associations between hub genes and immune cells were analyzed. A total of 6,500 articles were identified, and the annual number of articles exhibited an upward trend. The United States emerged as the leading contributor in terms of publication count, institutional affiliations, highly cited articles, and prolific authorship. According to co-occurrence analysis, oxidative stress and inflammation are hot topics in IPF research. A total of 1,140 DEGs were identified, and 72 genes were classified as DEOSGs. By employing PPI network analysis and machine learning algorithms, PON2 and TLR4 were identified as hub genes. A total of 10 immune cells exhibited significant differences between IPF and normal samples. PON2 and TLR4, as oxidative stress-related genes, not only exhibit high diagnostic efficacy but also show close associations with immune cells. In summary, our study highlights oxidative stress and inflammation are hot topics in IPF research. Oxidative stress and immune cells play a vital role in the pathogenesis of IPF. Our findings suggest the potential of PON2 and TLR4 as novel diagnostic and therapeutic targets for IPF.
Collapse
Affiliation(s)
- Chang Li
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Graduate School, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Qing An
- Graduate School, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yi Jin
- Graduate School, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Zefei Jiang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meihe Li
- Department of Renal Transplantation, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoling Wu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Huimin Dang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
19
|
Lian Z, Kuerban R, Niu Z, Aisaiti P, Wu C, Yang X. Notch Signaling Is Associated with Pulmonary Fibrosis in Patients with Pigeon Breeder's Lung by Regulating Oxidative Stress. Emerg Med Int 2024; 2024:7610032. [PMID: 39139588 PMCID: PMC11321885 DOI: 10.1155/2024/7610032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/03/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024] Open
Abstract
This study explored the molecular mechanism underlying the association of Notch signaling and oxidative stress with the occurrence of pulmonary fibrosis in patients with pigeon breeder's lung (PBL). Rat models of fibrotic PBL were constructed with freeze-dried protein powder, and the animals were divided into the control (intratracheal instillation of normal saline; n = 9), M (PBL model; intratracheal instillation of freeze-dried protein powder; n = 9), and M + D (PBL+ the Notch inhibitor DAPT; n = 9) groups. Immunohistochemistry was employed to observe the protein levels of pathway factors and α-SMA, and the levels of ROS, GSH-PX, SOD, and MDA were observed using ELISA. To verify the results of the animal experiment, cytological models were constructed. The M group and the M + D group had significantly increased α-SMA levels (P < 0.05). Although both groups had significantly higher key protein levels in the Notch channel, the M + D group had significantly lower levels relative to the M group (P < 0.05). Oxidative stress products were examined, and the levels of MDA and ROS were significantly increased, while those of GSH-PX and SOD were significantly decreased in the M and M + D groups as compared to the control, but the M group and the M + D group significantly differed (P < 0.05). These findings were further validated by the cytological experiment. Notch signaling is associated with pulmonary fibrosis in PBL by regulating cellular oxidative stress, and inhibiting this pathway can slow down pulmonary fibrosis progression.
Collapse
Affiliation(s)
- Zhichuang Lian
- Graduate SchoolXinjiang Medical University, Urumqi 830001, China
| | - Remila Kuerban
- Department of Respiratory and Critical Care MedicinePeople's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, China
| | - Zongxin Niu
- Department of Respiratory and Critical Care MedicinePeople's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, China
| | - Paruzha Aisaiti
- Department of Respiratory and Critical Care MedicinePeople's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, China
| | - Chao Wu
- Graduate SchoolXinjiang Medical University, Urumqi 830001, China
| | - Xiaohong Yang
- Graduate SchoolXinjiang Medical University, Urumqi 830001, China
| |
Collapse
|
20
|
Kilic KD, Erisik D, Taskiran D, Turhan K, Kose T, Cetin EO, Sendemi R A, Uyanikgil Y. Protective effects of E-CG-01 (3,4-lacto cycloastragenol) against bleomycin-induced lung fibrosis in C57BL/6 mice. Biomed Pharmacother 2024; 177:117016. [PMID: 38943992 DOI: 10.1016/j.biopha.2024.117016] [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: 04/15/2024] [Revised: 06/05/2024] [Accepted: 06/17/2024] [Indexed: 07/01/2024] Open
Abstract
Idiopathic pulmonary fibrosis is an aging-related, chronic lung disease, with unclear pathogenesis and no effective treatment. One of the triggering factors in cell aging is oxidative stress and it is known to have a role in idiopathic pulmonary fibrosis. In this paper, the protective effect of the E-CG-01 (3,4-lacto-cycloastragenol) molecule in terms of its antioxidant properties was evaluated in the bleomycin induced mice lung fibrosis model. Bleomycin sulfate was administered as a single dose (2.5 U/kg body weight) intratracheally to induce lung fibrosis. E-CG-01 was administered intraperitoneally in three different doses (2 mg/kg/day, 6 mg/kg/day, and 10 mg/kg/day) for 14 days, starting three days before the bleomycin administration. Fibrosis was examined by Hematoxylin-Eosin, Masson Trichrome, and immunohistochemical staining for TGF-beta1, Type I collagen Ki-67, and gama-H2AX markers. Activity analysis of catalase and Superoxide dismutase enzymes, measurement of total oxidant, total glutathione, and Malondialdehyde levels. In histological analysis, it was determined that all three different doses of the molecule provided a prophylactic effect against the progression of fibrosis compared to the bleomycin control group. However, it was observed that only the molecule applied in the high dose decreased the total oxidant stress level. Lung weight ratio increased in the BLM group but significantly reduced with high-dose E-CG-01. E-CG-01 at all doses reduced collagen deposition, TGF-β expression, and Ki-67 expression compared to the BLM group. Intermediate and high doses of E-CG-01 also significantly reduced alveolar wall thickness and edema formation. These findings suggest that E-CG-01 has potential therapeutic effects in mitigating lung fibrosis through its antioxidant properties.
Collapse
Affiliation(s)
- Kubilay Dogan Kilic
- Ege University, Faculty of Medicine, Department of Histology and Embryology, İzmir, Turkiye; Leibniz Institute for Evolution and Biodiversity Science, Museum für Naturkunde, Berlin, Germany.
| | - Derya Erisik
- Ege University, Faculty of Medicine, Department of Histology and Embryology, İzmir, Turkiye
| | - Dilek Taskiran
- Ege University, Faculty of Medicine, Department of Physiology, İzmir, Turkiye
| | - Kutsal Turhan
- Ege University, Faculty of Medicine, Department of Thoracic Surgery, İzmir, Turkiye; Acibadem Kent Hospital, Department of Thoracic Surgery, İzmir, Türkiye
| | - Timur Kose
- Ege University, Faculty of Medicine, Department of Biostatistics and Medical Informatics, İzmir, Turkiye
| | - Emel Oyku Cetin
- Ege University, Faculty of Pharmacy, Department of Biopharmaceutics and Pharmacokinetics, İzmir, Turkiye
| | - Aylin Sendemi R
- Ege University, Faculty of Engineering, Department of Bioengineering, İzmir, Turkiye
| | - Yiğit Uyanikgil
- Ege University, Faculty of Medicine, Department of Histology and Embryology, İzmir, Turkiye; Ege University, Cord Blood Cell - Tissue Research and Application Center, İzmir, Turkiye; Ege University, Institute of Health Sciences, Department of Stem Cell, İzmir, Turkiye
| |
Collapse
|
21
|
Wei Y, Gao S, Li C, Huang X, Xie B, Geng J, Dai H, Wang C. Aldehyde Dehydrogenase 2 Deficiency Aggravates Lung Fibrosis through Mitochondrial Dysfunction and Aging in Fibroblasts. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1458-1477. [PMID: 38777148 DOI: 10.1016/j.ajpath.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/07/2024] [Accepted: 04/09/2024] [Indexed: 05/25/2024]
Abstract
Idiopathic pulmonary fibrosis, a fatal interstitial lung disease, is characterized by fibroblast activation and aberrant extracellular matrix accumulation. Effective therapeutic development is limited because of incomplete understanding of the mechanisms by which fibroblasts become aberrantly activated. Here, we show aldehyde dehydrogenase 2 (ALDH2) in fibroblasts as a potential therapeutic target for pulmonary fibrosis. A decrease in ALDH2 expression was observed in patients with idiopathic pulmonary fibrosis and bleomycin-treated mice. ALDH2 deficiency spontaneously induces collagen accumulation in the lungs of aged mice. Furthermore, young ALDH2 knockout mice exhibited exacerbated bleomycin-induced pulmonary fibrosis and increased mortality compared with that in control mice. Mechanistic studies revealed that transforming growth factor (TGF)-β1 induction and ALDH2 depletion constituted a positive feedback loop that exacerbates fibroblast activation. TGF-β1 down-regulated ALDH2 through a TGF-β receptor 1/Smad3-dependent mechanism. The subsequent deficiency in ALDH2 resulted in fibroblast dysfunction that manifested as impaired mitochondrial autophagy and senescence, leading to fibroblast activation and extracellular matrix production. ALDH2 overexpression markedly suppressed fibroblast activation, and this effect was abrogated by PTEN-induced putative kinase 1 (PINK1) knockdown, indicating that the profibrotic effects of ALDH2 are PINK1- dependent. Furthermore, ALDH2 activated by N-(1,3-benzodioxol-5-ylmethyl)-2,6-dichlorobenzamide (Alda-1) reversed the established pulmonary fibrosis in both young and aged mice. In conclusion, ALDH2 expression inhibited the pathogenesis of pulmonary fibrosis. Strategies to up-regulate or activate ALDH2 expression could be potential therapies for pulmonary fibrosis.
Collapse
Affiliation(s)
- Yanqiu Wei
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China; National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Shuwei Gao
- National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Chen Li
- National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaoxi Huang
- Department of Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Bingbing Xie
- National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jing Geng
- National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Huaping Dai
- National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
| | - Chen Wang
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China; National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
22
|
Mao X, Wu S, Huang D, Li C. Complications and comorbidities associated with antineoplastic chemotherapy: Rethinking drug design and delivery for anticancer therapy. Acta Pharm Sin B 2024; 14:2901-2926. [PMID: 39027258 PMCID: PMC11252465 DOI: 10.1016/j.apsb.2024.03.006] [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: 11/26/2023] [Revised: 01/29/2024] [Accepted: 02/10/2024] [Indexed: 07/20/2024] Open
Abstract
Despite the considerable advancements in chemotherapy as a cornerstone modality in cancer treatment, the prevalence of complications and pre-existing diseases is on the rise among cancer patients along with prolonged survival and aging population. The relationships between these disorders and cancer are intricate, bearing significant influence on the survival and quality of life of individuals with cancer and presenting challenges for the prognosis and outcomes of malignancies. Herein, we review the prevailing complications and comorbidities that often accompany chemotherapy and summarize the lessons to learn from inadequate research and management of this scenario, with an emphasis on possible strategies for reducing potential complications and alleviating comorbidities, as well as an overview of current preclinical cancer models and practical advice for establishing bio-faithful preclinical models in such complex context.
Collapse
Affiliation(s)
- Xiaoman Mao
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Shuang Wu
- Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Dandan Huang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Chong Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
- Medical Research Institute, Southwest University, Chongqing 400715, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
23
|
Samuel CS, Li Y, Wang Y, Widdop RE. Functional crosstalk between angiotensin receptors (types 1 and 2) and relaxin family peptide receptor 1 (RXFP1): Implications for the therapeutic targeting of fibrosis. Br J Pharmacol 2024; 181:2302-2318. [PMID: 36560925 DOI: 10.1111/bph.16019] [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: 08/29/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Class A, rhodopsin-like, G-protein-coupled receptors (GPCRs) are by far the largest class of GPCRs and are integral membrane proteins used by various cells to convert extracellular signals into intracellular responses. Initially, class A GPCRs were believed to function as monomers, but a growing body of evidence has emerged to suggest that these receptors can function as homodimers and heterodimers and can undergo functional crosstalk to influence the actions of agonists or antagonists acting at each receptor. This review will focus on the angiotensin type 1 (AT1) and type 2 (AT2) receptors, as well as the relaxin family peptide receptor 1 (RXFP1), each of which have their unique characteristics but have been demonstrated to undergo some level of interaction when appropriately co-expressed, which influences the function of each receptor. In particular, this receptor functional crosstalk will be discussed in the context of fibrosis, the tissue scarring that results from a failed wound-healing response to injury, and which is a hallmark of chronic disease and related organ dysfunction. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Yifang Li
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Yan Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
24
|
Rezaie F, Ghafouri Khosroshahi A, Larki-Harchegani A, Nourian A, Khosravi H. Hydroalcoholic Sumac Extract as a Protective Agent Against X-Ray-Induced Pulmonary Fibrosis. Rep Biochem Mol Biol 2024; 13:231-242. [PMID: 39995652 PMCID: PMC11847579 DOI: 10.61186/rbmb.13.2.231] [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: 04/09/2024] [Accepted: 06/23/2024] [Indexed: 02/26/2025]
Abstract
Background X-ray exposure can result in acute or chronic damage to lung tissue, leading to pneumonitis and fibrosis. Given the potent antioxidant properties of sumac, this study investigates the impact of hydroalcoholic sumac extract on X-ray-induced pulmonary fibrosis in rats. Methods In this experimental study, 36 rats were randomly divided into six groups of six rats each. The treatment and sham groups received intraperitoneal administration of the extract daily for one week before exposure to X-ray radiation. On the seventh day, all rats except those in group 3 were exposed to 2 Gy of 6 MV X-rays using an electro-linear accelerator. Lung tissue was subsequently removed to assess the subacute effects of the extract. Data analysis involved independent sample t-tests and one-way ANOVA using SPSS 26. Results A single dose of X-rays significantly increased oxidative stress and lung tissue damage in rats. However, rats receiving vitamin C and hydroalcoholic sumac extract at two different doses (100 and 400 mg/kg intraperitoneally) positively improved lung damage and decreased antioxidant parameters. Conclusions The findings demonstrate that hydroalcoholic sumac extract can mitigate oxidative stress and enhance lung repair following X-ray radiation exposure.
Collapse
Affiliation(s)
- Faezeh Rezaie
- Department of Medicinal Chemistry, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran.
| | | | - Amir Larki-Harchegani
- Department of Medicinal Chemistry, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Alireza Nourian
- Department of Pathobiology, School of Veterinary Science, Bu-Ali Sina University, Hamadan, Iran.
| | - Hossein Khosravi
- Department of Radiology, School of Allied Medical Sciences, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
25
|
Hu Y, Huang Y, Zong L, Lin J, Liu X, Ning S. Emerging roles of ferroptosis in pulmonary fibrosis: current perspectives, opportunities and challenges. Cell Death Discov 2024; 10:301. [PMID: 38914560 PMCID: PMC11196712 DOI: 10.1038/s41420-024-02078-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic interstitial lung disorder characterized by abnormal myofibroblast activation, accumulation of extracellular matrix (ECM), and thickening of fibrotic alveolar walls, resulting in deteriorated lung function. PF is initiated by dysregulated wound healing processes triggered by factors such as excessive inflammation, oxidative stress, and coronavirus disease (COVID-19). Despite advancements in understanding the disease's pathogenesis, effective preventive and therapeutic interventions are currently lacking. Ferroptosis, an iron-dependent regulated cell death (RCD) mechanism involving lipid peroxidation and glutathione (GSH) depletion, exhibits unique features distinct from other RCD forms (e.g., apoptosis, necrosis, and pyroptosis). Imbalance between reactive oxygen species (ROS) production and detoxification leads to ferroptosis, causing cellular dysfunction through lipid peroxidation, protein modifications, and DNA damage. Emerging evidence points to the crucial role of ferroptosis in PF progression, driving macrophage polarization, fibroblast proliferation, and ECM deposition, ultimately contributing to alveolar cell death and lung tissue scarring. This review provides a comprehensive overview of the latest findings on the involvement and signaling mechanisms of ferroptosis in PF pathogenesis, emphasizing potential novel anti-fibrotic therapeutic approaches targeting ferroptosis for PF management.
Collapse
Affiliation(s)
- Yixiang Hu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China
| | - Ying Huang
- Zhongshan Hospital of Traditional Chinese Medicine Afflilated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, China
| | - Lijuan Zong
- Department of Rehabilitation Medicine, Zhongda Hospital of Southeast University, Nanjing, 210096, China
| | - Jiaxin Lin
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Xiang Liu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China.
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China.
| |
Collapse
|
26
|
Edwards H, Javed K, Yadev K, Ara C, Omer AM. Therapeutic potential of salvigenin to combat atrazine induced liver toxicity in rats via regulating Nrf-2/Keap-1 and NF-κB pathway. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 202:105966. [PMID: 38879343 DOI: 10.1016/j.pestbp.2024.105966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/11/2024] [Accepted: 05/24/2024] [Indexed: 06/29/2024]
Abstract
Atrazine (ATR) is the second most extensively used herbicide which adversely affects the body organs including liver. Salvigenin (SGN) is a flavonoid which demonstrates a wide range of biological and pharmacological abilities. This study was planned to assess the protective ability of SGN to avert ATR induced liver damage in rats. Thirty-two rats (Rattus norvegicus) were divided into four groups including control, ATR (5 mg/kg), ATR (5 mg/kg) + SGN (10 mg/kg) and SGN (10 mg/kg) alone supplemented group. ATR exposure reduced the expression of Nrf-2 while instigating an upregulation in Keap-1 expression. Furthermore, the activities of catalase (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD), heme‑oxygenase-1 (HO-1) and glutathione reductase (GSR) contents were decreased while increasing reactive oxygen species (ROS) and malondialdehyde (MDA) levels after ATR treatment. Moreover, ATR poisoning increased the levels of ALT, AST, and ALP while reducing the levels of total proteins, and albumin in hepatic tissues of rats. Besides, ATR administration escalated the expressions of Bax and Caspase-3 while inducing a downregulation in the expressions of Bcl-2. Similarly, ATR intoxication increased the levels of Interleukin-6 (IL-6), Nuclear factor kappa-B (NF-κB), Interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and the activity of cyclooxygenase-2 (COX-2). Furthermore, ATR disrupted the normal histology of hepatic tissues. However, SGN treatment remarkably protected the liver tissues via regulating antioxidant, anti, inflammatory, anti-apoptotic as well as histology parameters. Therefore, it is concluded that SGN can be used as therapeutic agent to combat ATR-induced hepatotoxicity.
Collapse
Affiliation(s)
- Henry Edwards
- Department of Biology, The University of Melbourne, Australia.
| | - Khadija Javed
- School of Natural Sciences, University of Chester, England
| | - Kumar Yadev
- Department of Biology, The University of Melbourne, Australia
| | - Chaman Ara
- Department of Zoology, Ghazi University, Pakistan
| | | |
Collapse
|
27
|
Zhao Z, Yang X. Inhibition of SMYD2 attenuates paraquat-induced pulmonary fibrosis by inhibiting the epithelial-mesenchymal transition through the GLIPR2/ERK/p38 axis. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 202:105971. [PMID: 38879290 DOI: 10.1016/j.pestbp.2024.105971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/17/2024] [Accepted: 05/27/2024] [Indexed: 06/29/2024]
Abstract
Paraquat (PQ) poisoning leads to irreversible fibrosis in the lungs with high mortality and no known antidote. In this study, we investigated the effect of the SET and MYND domain containing 2 (SMYD2) on PQ-induced pulmonary fibrosis (PF) and its potential mechanisms. We established an in vivo PQ-induced PF mouse model by intraperitoneal injection of PQ (20 mg/kg) and in vitro PQ (25 μM)-injured MLE-12 cell model. On the 15th day of administration, tissue injury, inflammation, and fibrosis in mice were evaluated using various methods including routine blood counts, blood biochemistry, blood gas analysis, western blotting, H&E staining, ELISA, Masson staining, and immunofluorescence. The findings indicated that AZ505 administration mitigated tissue damage, inflammation, and collagen deposition in PQ-poisoned mice. Mechanistically, both in vivo and in vitro experiments revealed that AZ505 treatment suppressed the PQ-induced epithelial-mesenchymal transition (EMT) process by downregulating GLI pathogenesis related 2 (GLIPR2) and ERK/p38 pathway. Further investigations demonstrated that SMYD2 inhibition decreased GLIPR2 methylation and facilitated GLIPR2 ubiquitination, leading to GLIPR2 destabilization in PQ-exposed MLE-12 cells. Moreover, rescue experiments conducted in vitro demonstrated that GLIPR2 overexpression eliminated the inhibitory effect of AZ505 on the ERK/p38 pathway and EMT. Our results reveal that the SMYD2 inhibitor AZ505 may act as a novel therapeutic candidate to suppress the EMT process by modulating the GLIPR2/ERK/p38 axis in PQ-induced PF.
Collapse
Affiliation(s)
- Zheng Zhao
- Department of Emergency, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xue Yang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
28
|
Lao Q, Wang X, Zhu G, Yuan H, Ma T, Wang N. A Chinese classical prescription Maimendong decoction in treatment of pulmonary fibrosis: an overview. Front Pharmacol 2024; 15:1329743. [PMID: 38783956 PMCID: PMC11112100 DOI: 10.3389/fphar.2024.1329743] [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: 11/02/2023] [Accepted: 04/11/2024] [Indexed: 05/25/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic and progressive disease characterized by fibrosis and interstitial pneumonia. It has similar clinical symptoms to "Fei Bi" and "Fei Wei" as described in the traditional Chinese medicine (TCM) classic Jingui Yaolue written by Zhang Zhongjing in the Han Dynasty. This study explored the potential of Maimendong Decoction (MMDD). MMDD consists of Ophiopogon japonicus (L.f) (ophiopogonis), Pinellia ternata (Thunb.) Breit. (pinellia), Panax ginseng C. A. Mey. (ginseng), Glycyrrhiza uralensis Fisch. (glycyrrhiza), Zizi phus jujuba Mill. (jujuba), and Oryza sativa L. (oryza sativa), with the function of nourishing the lung and stomach, and reducing the effect of reverse qi. It has been used clinically for over two thousand years to treat conditions like "Fei Bi" and "Fei Wei". Previous research suggests that MMDD and its individual herbal extracts have anti-fibrotic effects. The main focus of MMDD in treating PF is to reduce inflammatory cytokines, inhibit pro-fibrotic factors and oxidative stress, promote differentiation and homing of bone marrow mesenchymal stem cells, and enhance cell autophagy activity. This review summarized the clinical applications, mechanisms, and pharmacological effects of MMDD in treating PF based on existing clinical applications and experimental research. It also discussed current issues and prospects, aiming to provide a reference for further research on the mechanism of PF, drug development, and clinical trials.
Collapse
Affiliation(s)
- Qiurong Lao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianbin Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guangqing Zhu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haochen Yuan
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ting Ma
- College of Rehabilitation Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ning Wang
- Research Department of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
29
|
Cao X, Yu C, Cheng S, Wang Y, Zhang Z, Huang J. Co-Delivery of Astaxanthin and si TGF-β1 via Ionizable Liposome Nanoparticles for Improved Idiopathic Pulmonary Fibrosis Therapy. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38597290 DOI: 10.1021/acsami.4c01953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Alleviating the injury of type II alveolar epithelial cells (AEC 2s) and inhibiting the activation and differentiation of fibroblasts are significant for improving the therapeutic effect of idiopathic pulmonary fibrosis (IPF). To this aim, ionizable liposome nanoparticles (ASNPs) coloaded with antioxidant drug astaxanthin (AST) and small interfering RNA targeting transforming growth factor β1 (siTGF-β1) were developed for enhanced IPF therapy. ASNPs showed high loading and intracellular delivery efficiency for AST and siTGF-β1. After the injection of ASNPs in an IPF mice model, the loaded AST largely scavenged reactive oxygen species (ROS) in the diseased lung to reduce AEC2 apoptosis, thereby ensuring the integrity of the alveolar epithelium. Meanwhile, siTGF-β1, delivered by ASNPs, significantly silenced the expression of TGF-β1 in fibroblasts, inhibiting the differentiation of fibroblasts into myofibroblasts as well as reducing the excessive deposition of extracellular matrix (ECM). The combined use of the two drugs exhibited an excellent synergistic antifibrotic effect and was conducive to minimizing alveolar epithelial damage. This work provides a codelivery strategy of AST and siTGF-β1, which shows great promise for the treatment of IPF by simultaneously reducing alveolar epithelial damage and inhibiting fibroblast activation.
Collapse
Affiliation(s)
- Xiaoling Cao
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China institution, Hefei 230026, China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Chenggong Yu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China institution, Hefei 230026, China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Shengnan Cheng
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China institution, Hefei 230026, China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yuhan Wang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China institution, Hefei 230026, China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zhijun Zhang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China institution, Hefei 230026, China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jie Huang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China institution, Hefei 230026, China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-bionics, Chinese Academy of Sciences, Suzhou 215123, China
| |
Collapse
|
30
|
Mohanan A, Washimkar KR, Mugale MN. Unraveling the interplay between vital organelle stress and oxidative stress in idiopathic pulmonary fibrosis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119676. [PMID: 38242330 DOI: 10.1016/j.bbamcr.2024.119676] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 12/22/2023] [Accepted: 01/10/2024] [Indexed: 01/21/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung disease characterized by excessive accumulation of extracellular matrix, leading to irreversible fibrosis. Emerging evidence suggests that endoplasmic reticulum (ER) stress, mitochondrial stress, and oxidative stress pathways play crucial roles in the pathogenesis of IPF. ER stress occurs when the protein folding capacity of the ER is overwhelmed, triggering the unfolded protein response (UPR) and contributing to protein misfolding and cellular stress in IPF. Concurrently, mitochondrial dysfunction involving dysregulation of key regulators, including PTEN-induced putative kinase 1 (PINK1), Parkin, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), and sirtuin 3 (SIRT3), disrupts mitochondrial homeostasis and impairs cellular energy metabolism. This leads to increased reactive oxygen species (ROS) production, release of pro-fibrotic mediators, and activation of fibrotic pathways, exacerbating IPF progression. The UPR-induced ER stress further disrupts mitochondrial metabolism, resulting in altered mitochondrial mechanisms that increase the generation of ROS, resulting in further ER stress, creating a feedback loop that contributes to the progression of IPF. Oxidative stress also plays a pivotal role in IPF, as ROS-mediated activation of TGF-β, NF-κB, and MAPK pathways promotes inflammation and fibrotic responses. This review mainly focuses on the links between ER stress, mitochondrial dysfunctions, and oxidative stress with different signaling pathways involved in IPF. Understanding these mechanisms and targeting key molecules within these pathways may offer promising avenues for intervention.
Collapse
Affiliation(s)
- Anushree Mohanan
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India
| | - Kaveri R Washimkar
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Madhav Nilakanth Mugale
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
31
|
Bahri S, Abdennabi R, Chaker A, Nahdi A, Elgheryeni A, Mlika M, Jameleddine S. Phœnix dactylifera, L. seed oil alleviates Bleomycin-induced pulmonary fibrosis and oxidative stress in Wistar rats. Biomarkers 2024; 29:45-54. [PMID: 38314578 DOI: 10.1080/1354750x.2024.2311178] [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: 08/29/2023] [Accepted: 01/23/2024] [Indexed: 02/06/2024]
Abstract
OBJECTIVE Idiopathic pulmonary fibrosis (IPF) is the most serious form of interstitial lung disease. We aimed to investigate the effect of Phœnix dactylifera, L. seed oil (DSO) on a murine model of IPF induced by bleomycin (BLM). METHODS Male Wistar rats were treated with a single intra-tracheal injection of BLM (4 mg/kg) and a daily intraperitoneal injection of DSO (75, 150 and 300 mg/kg) for 4 weeks. RESULTS Our phytochemical results showed that DSO has an important antioxidant activity with a high content of polyphenols and flavonoids. High-Performance Liquid Chromatography (HPLC) and Gas chromatography/mass spectrometry (GC-MS) analysis revealed a high amount of oleic and lauric acids and a large quantity of vitamins. Histological examination showed a significant reduction in fibrosis score and collagen bands in the group of rats treated with 75 mg/kg of DSO compared to the BLM group. DSO (75 mg/kg) reversed also the increase in catalase and malondialdehyde (MDA) levels while higher doses (150 and 300 mg/kg) are ineffective against the deleterious effects of BLM. We revealed also that DSO has no renal or hepatic cytotoxic effects. CONCLUSION DSO can play antioxidant and antifibrotic effects on rat models of pulmonary fibrosis at the lowest dose administered.
Collapse
Affiliation(s)
- Sana Bahri
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Physiopathology, Food and Biomolecules (LR-17-ES-03), Technology Center of Sidi Thabet, University of Manouba, Tunis, Tunisia
| | - Raed Abdennabi
- Laboratory of Plant Biotechnology, Faculty of Science, University of Sfax, Sfax, Tunisia
| | - Asma Chaker
- Functional Exploration and Physiotherapy Department, Abderhaman Mami Hospital, Ariana, Tunisia
| | - Afef Nahdi
- Research Unit n° 17/ES/13, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | | | - Mona Mlika
- Laboratory of Anatomy and Pathology, Abderhaman Mami Hospital, Ariana, Tunisia
| | - Saloua Jameleddine
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Physiopathology, Food and Biomolecules (LR-17-ES-03), Technology Center of Sidi Thabet, University of Manouba, Tunis, Tunisia
| |
Collapse
|
32
|
Ijaz MU, Alvi K, Hamza A, Anwar H, Al-Ghanim KA, Riaz MN. Curative effects of tectochrysin on paraquat-instigated testicular toxicity in rats: A biochemical and histopathological based study. Heliyon 2024; 10:e25337. [PMID: 38356568 PMCID: PMC10865255 DOI: 10.1016/j.heliyon.2024.e25337] [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: 06/12/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
Background Paraquat (PQ) is a herbicide that is used globally in the agriculture sector to eradicate unwanted weeds, however it also induces significant damages in various organs of the body such as testes. Tectochrysin (TEC) is an important flavonoid that shows versatile therapeutic potentials. Currently, there is no established antidote to cure PQ-induced testicular toxicity. Objective The present study was conducted to evaluate the ameliorative effects of TEC against PQ prompted testicular damage. Methods Sprague-Dawley rats (n = 48) were used to conduct the trial. Rats were allocated in to 4 groups i.e., Control, PQ administrated group (5 mgkg-1), PQ + TEC co-administrated group (5 mgkg-1 + 2.5 mgkg-1) and TEC only administrated group (2.5 mgkg-1). The trial was conducted for 8 weeks. The activity of anti-oxidants and the levels of MDA and ROS were determined by spectrophotometric method. Steroidogenic enzymes as well as apoptotic markers expressions were evaluated by qRT-PCR. The level of hormones and inflammatory indices was quantified by enzyme-linked immunosorbent assay. Results PQ exposure markedly (P < 0.05) disturbed the biochemical, spermatogenic and histological profile in the rats. Nevertheless, TEC treatment considerably (P < 0.05) increased CAT, GPx GSR and SOD activity, besides decreasing MDA and ROS contents. TEC administration also increased sperm viability, count and motility. 17β-HSD, 3β-HSD, StAR and Bcl-2 expressions were also increased following TEC administration. The supplementation of TEC substantially (P < 0.05) decreased Bax, Caspase-3 expression and the levels of inflammatory markers i.e., interleukin-1β (IL-1β), interleukin-6 (IL-6), nuclear factor kappa-B (NF-κB), tumor necrosis factor-α (TNF-α) and cyclooxygenase-2 (COX-2) activity. Additionally, the levels of plasma testosterone, follicle-stimulating hormone (FSH) and luteinizing hormone (LH) were increased following TEC supplementation. Furthermore, TEC supplementation considerably decreased sperm structural abnormalities and histomorphological damages of the testes. The mitigative role of TEC might be due to its anti-inflammatory, anti-apoptotic, androgenic and anti-oxidant potentials. Conclusion Taken together, it is concluded that TEC can be used as a potential candidate to treat testicular toxicity.
Collapse
Affiliation(s)
- Muhammad Umar Ijaz
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Kaynat Alvi
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Ali Hamza
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Haseeb Anwar
- Department of Physiology, Government College University of Faisalabad, Faisalabad, Pakistan
| | - Khalid A. Al-Ghanim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | | |
Collapse
|
33
|
Bian B, Ge C, Wu F, Fan Y, Kong J, Li K, Bian H, Miao Q. Wogonin Attenuates Bleomycin-Induced Pulmonary Fibrosis and Oxidative Stress Injury via the MAPK Signaling Pathway. Biol Pharm Bull 2024; 47:2165-2172. [PMID: 39756931 DOI: 10.1248/bpb.b24-00534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Idiopathic pulmonary fibrosis (PF) is an irreversible and chronic inflammatory condition with limited therapeutic options and a high mortality rate. We aimed to determine the possible role and mechanisms of wogonin (WGN) on PF. A rat model of PF was established with intratracheally administrated with bleomycin (BLM), followed by intravenously injecting with WGN and weekly body weight measurements for four weeks. Hematoxylin-eosin (H&E) and Masson's trichrome staining were implemented for histopathological analysis. In addition, the levels of fibrotic proteins and indicators of the mitogen-activated protein kinase (MAPK) pathway were assessed with Western blot. RT-quantitative (q)PCR experiment was conducted to investigate the fibrotic proteins' mRNA expression. Ultimately, the concentrations of glutathione peroxidase (GSH-PX), malonaldehyde (MDA), and superoxide dismutase (SOD) were ascertained with appropriate kits. The results showed that WGN administration significantly reversed BLM-induced body weight reduction, alleviated pathological fibrosis, and reduced the Ashcroft score and the lung wet-to-dry weight ratio. Additionally, WGN suppressed the rise of fibrotic protein levels in BLM-treated rat's lung tissues. Furthermore, WGN attenuated BLM-stimulated oxidative stress, as evidenced by the increased GSH-PX and SOD levels and decreased MDA levels in vivo. Finally, wogonin supplements significantly lowered the extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 MAPK phosphorylation levels in the BLM-treated rat's lung tissues. In conclusion, our study proved that PF induced by BLM administration can be mitigated by WGN treatment via suppressing the MAPK pathway, indicating that WGN is a candidate therapeutic agent for managing PF.
Collapse
Affiliation(s)
- Bo Bian
- Traditional Chinese Medical College, North China University of Science and Technology
| | - Chang Ge
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Fanwu Wu
- Traditional Chinese Medical College, North China University of Science and Technology
| | - Yiling Fan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences
| | | | - Kai Li
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology
| | - Hua Bian
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology
| | - Qing Miao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences
| |
Collapse
|
34
|
Kim NH, Kim HY, Lee JH, Chang I, Heo SH, Kim J, Kim JH, Kang JH, Lee SW. Superoxide dismutase secreting Bacillus amyloliquefaciens spores attenuate pulmonary fibrosis. Biomed Pharmacother 2023; 168:115647. [PMID: 37826939 DOI: 10.1016/j.biopha.2023.115647] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/18/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023] Open
Abstract
Superoxide dismutase (SOD) can convert active oxygen to oxygen or hydrogen peroxide, and recent research has suggested that it can protect against lung damage and fibrosis. Clinical applications based on SOD remain limited however due to costs and low stability. We here investigated a potential new therapeutic delivery system for this enzyme in the form of SOD-overexpressing Bacillus amyloliquefaciens spores which we introduced into a bleomycin-induced pulmonary fibrosis mouse model. This treatment significantly alleviated the disease, as quantified using a hydroxyproline assay, at 107 colony forming unit (CFU) of Bacillus spores per day. Exposure of the mice to the spores was further found to decrease the lung mRNA levels of CTGF, Col1a1, α-SMA, TGF-β, TNF-α, and IL-6, and the protein levels of TGF-β, Smad2/3, αSMA and Col1a1, all major indicators of pulmonary fibrosis. Survival benefits, and reduced byproducts of lipid peroxidase such as malondialdehyde and 4-hydroxynen, were also noted in the treated animals. The beneficial effects of these Bacillus spores on pulmonary fibrosis were further found to be greater than the equivalent free SOD concentration. Immunofluorescence staining of primary pulmonary fibroblasts extracted from the bleomycin-induced model showed decreased αSMA expression following the in vivo treatment with SOD-overexpressing Bacillus. Our treatment approach SOD through Bacillus spores shows beneficial effects against pulmonary fibrosis, combined with the suppression of the SMAD/TGF-β pathway, suggesting that it is an effective novel delivery route for antioxidants.
Collapse
Affiliation(s)
- Na Hyun Kim
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hee Young Kim
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; BiomLogic, Inc., Seoul, Republic of Korea
| | - Jang Ho Lee
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Inik Chang
- BiomLogic, Inc., Seoul, Republic of Korea
| | - Sun-Hee Heo
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jiseon Kim
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Pharmacology and Regnerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | | | | | - Sei Won Lee
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
35
|
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
|
36
|
Liu L, Zhang X, Zhang R, Wang L, Zhi S, Feng X, Liu X, Shen Y, Hao J. Sohlh2 promotes pulmonary fibrosis via repression of p62/Keap1/Nrf2 mediated anti-oxidative signaling pathway. Cell Death Dis 2023; 14:698. [PMID: 37875506 PMCID: PMC10598036 DOI: 10.1038/s41419-023-06179-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 09/10/2023] [Accepted: 09/26/2023] [Indexed: 10/26/2023]
Abstract
Disturbance in the redox balance of alveolar epithelial cells (AECs) was considered as a causal factor for pulmonary fibrosis. The regulatory mechanisms of redox hemostasis in the development of pulmonary fibrosis remain largely unknown. Using a type II AEC-specific Sohlh2 conditional knock-in (CKI) mouse model, we found that Sohlh2, a basic HLH transcription factor, accelerated age-related pulmonary fibrosis. High-fat diet (HFD) resulted in a tremendous increase in lung inflammation and fibrotic changes in the lung tissues of Sohlh2 CKI mice. Sohlh2 overexpression led to a significant rise of intracellular ROS and apoptosis in the lung, mouse primary AECIIs, and human A549 cells, which was attenuated by ROS inhibitor (NAC). Sohlh2 enhanced oxidative stress via repressing p62/Keap1/Nrf2 mediated anti-oxidative signaling pathway. p62, a direct target of Sohlh2, mediated Sohlh2 effects on ROS generation and apoptosis in A549 cells. Hence, our findings elucidate a pivotal mechanism underlying oxidative stress-induced pulmonary fibrosis, providing a framework for aging-related disorder interventions.
Collapse
Affiliation(s)
- Lanlan Liu
- Key Laboratory of the Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, P. R. China
| | - Xiaoli Zhang
- Key Laboratory of the Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, P. R. China
| | - Ruihong Zhang
- Key Laboratory of the Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, P. R. China
| | - Liyan Wang
- Morphological Experimental Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, P. R. China
| | - Sujuan Zhi
- Key Laboratory of the Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, P. R. China
| | - Xiaoning Feng
- Key Laboratory of the Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, P. R. China
| | - Xuyue Liu
- Key Laboratory of the Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, P. R. China
| | - Ying Shen
- Key Laboratory of the Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, P. R. China
| | - Jing Hao
- Key Laboratory of the Ministry of Education for Experimental Teratology, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, P. R. China.
| |
Collapse
|
37
|
Ijaz MU, Saher F, Aslam N, Hamza A, Anwar H, Alkahtani S, Khan HA, Riaz MN. Evaluation of possible attenuative role of chrysoeriol against polyethylene microplastics instigated testicular damage: A biochemical, spermatogenic and histological study. Food Chem Toxicol 2023; 180:114043. [PMID: 37722616 DOI: 10.1016/j.fct.2023.114043] [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: 06/02/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 09/20/2023]
Abstract
The current study was designed to evaluate the protective role of chrysoeriol against polyethylene microplastics (PE-MP) induced testicular damage. Forty eight male rats were distributed into 4 equal groups: vehicle control, PE-MP administrated, PE-MP + chrysoeriol co-administrated and only chrysoeriol supplemented group. The administration of PE-MP significantly reduced the activities of anti-oxidant enzymes, i.e., glutathione peroxidase, catalase, glutathione reductase and superoxide dismutase, whereas the levels of reactive oxygen species and malondialdehyde were increased. PE-MP exposure increased the levels of inflammatory markers (TNF-α, 1L-1β, NF-κβ, IL-6 & COX-2). Additionally, a considerable increase was observed in dead sperms number, abnormality of sperms (tail, midpiece and head), while a potential decrease was noticed in sperm motility in PE-MP treated rats. The expressions of steroidogenic enzymes were also decreased in PE-MP administrated group. The levels of plasma testosterone, luteinizing & follicle stimulating hormone were decreased in PE-MP treated group. Moreover, Bax and Caspase-3 expressions were increased, whereas Bcl-2 expressions were reduced. Furthermore, histopathological analysis showed that PE-MP exposure considerably damaged the testicular tissues. However, chrysoeriol supplementation potentially decreased all the adverse effects induced by PE-MP. Taken together, our findings indicate that chrysoeriol holds significant potential to avert PE-MP-induced testicular damage due to its androgenic, anti-apoptotic, anti-oxidant and anti-inflammatory nature.
Collapse
Affiliation(s)
- Muhammad Umar Ijaz
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad 38040, Pakistan.
| | - Faria Saher
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad 38040, Pakistan
| | - Nemra Aslam
- Faisalabad Medical University, Faisalabad 38000, Pakistan
| | - Ali Hamza
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad 38040, Pakistan
| | - Haseeb Anwar
- Department of Physiology, Government College University, Faisalabad 38000, Pakistan
| | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Hammad Ahmad Khan
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad 38040, Pakistan
| | | |
Collapse
|
38
|
Saher F, Ijaz MU, Hamza A, Ain QU, Hayat MF, Afsar T, Almajwal A, Shafique H, Razak S. Mitigative potential of rhoifolin against cisplatin prompted testicular toxicity: biochemical, spermatogenic and histological based analysis. Toxicol Res (Camb) 2023; 12:814-823. [PMID: 37915485 PMCID: PMC10615821 DOI: 10.1093/toxres/tfad073] [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: 04/11/2023] [Revised: 07/26/2023] [Accepted: 08/05/2023] [Indexed: 11/03/2023] Open
Abstract
Rhoifolin (ROF) is a naturally occurring flavonoid compound with diverse pharmacological and therapeutic benefits. The current investigation was designed to evaluate the curative potential of Rhoifolin (ROF) against Cisplatin (CP) induced testicular damage. Mature male albino rats (n = 48) were randomly distributed into 4 equal groups: control, CP (10 mg/kg), CP + ROF (10 mg/kg + 20 mg/kg) and ROF (20 mg/kg) supplemented group. Following 56 days of the trial, biochemical, inflammatory markers, spermatogenic, steroidogenic, hormonal, apoptotic, anti-apoptotic, and histopathological parameters were evaluated. The exposure to CP markedly (p < 0.05) lowered the activities of anti-oxidant enzymes, glutathione reductase (GSR), catalase (CAT), and glutathione peroxidase (GPx) as well as superoxide dismutase (SOD) in testicular tissues of male albino rats. Besides the levels of reactive oxygen species (ROS) and thiobarbituric acid reactive substances (TBARS) were considerably augmented in CP exposed rats. The administration of CP also increased the level of inflammatory cytokines i.e. IL-6, TNF-α, 1L-1β and NF-κβ as well as COX-2 activity. Additionally, a notable (p < 0.05) upsurge was observed in dead sperms count, abnormality in the tail, midpiece as well as head of sperms along with a notable decline in sperm motility in CP treated rats. Moreover, the expressions of steroidogenic enzymes were also lowered in CP administered group. The levels of follicle stimulating hormone (FSH) and plasma testosterone as well as luteinizing hormone (LH) were decreased in CP treated group. Moreover, the expression of Bax as well as Caspase-3 (apoptotic markers) were increased. On the other hand, Bcl-2 expression (anti-apoptotic marker) was reduced. Furthermore, the histopathological analysis showed that CP considerably (p < 0.05) damaged the testicular tissues. However, the administration of ROF significantly reduced the damaging effects of CP in testicular tissues. The results of our study suggested that ROF can potentially alleviate CP-induced testicular damages due to its androgenic, anti-oxidant and anti-inflammatory as well as anti-apoptotic nature.
Collapse
Affiliation(s)
- Faria Saher
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad 38040, Pakistan
| | - Muhammad Umar Ijaz
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad 38040, Pakistan
| | - Ali Hamza
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad 38040, Pakistan
| | - Qurat Ul Ain
- Department of Zoology, Government College Women University, Sialkot 51040, Pakistan
| | - Muhammad Faisal Hayat
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad 38040, Pakistan
| | - Tayyaba Afsar
- Department of Community Health Sciences, College of Applied Medical Sciences, 11433, King Saud University, Riyadh, Saudi Arabia
| | - Ali Almajwal
- Department of Community Health Sciences, College of Applied Medical Sciences, 11433, King Saud University, Riyadh, Saudi Arabia
| | - Huma Shafique
- Institute of Cellular Medicine, Newcastle University Medical School, Newcastle University, Newcastle upon Tyne, NE1 7RU, United Kingdom
| | - Suhail Razak
- Department of Community Health Sciences, College of Applied Medical Sciences, 11433, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
39
|
Rastegar-Moghaddam SH, Akbarian M, Rajabian A, Alipour F, Ebrahimzadeh bideskan A, Hosseini M. Vitamin D alleviates hypothyroidism associated liver dysfunction: Histological and biochemical evidence. Heliyon 2023; 9:e18860. [PMID: 37593614 PMCID: PMC10428045 DOI: 10.1016/j.heliyon.2023.e18860] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
There is a complex correlation between thyroid hormones (THs) and liver function. Hypothyroidism as a failure of the thyroid gland to produce adequate thyroid hormones to fulfill the metabolic requirements of the body, may perturb liver structure and function. Emerging evidence suggests the protective effects of vitamin D against liver damage. Herein, this study aimed to investigate the role of vitamin D in hypothyroidism-associated liver injury. Forty male Wistar rats were classified into 4 groups: control, hypothyroid (Hypo) group received 0.05% PTU, Hypo- Vitamin D groups were given 100 and 500 IU/kg vitamin D orally via gavage for 6 weeks. Serum level of liver function including alanine aminotransferase (ALT), aspartate aminotransferase (AST), and alkaline phosphatase (ALP) were measured. Malondialdehyde (MDA) level, superoxide dismutase (SOD) enzyme activity, and total thiol content were measured as oxidative stress indicators in the liver tissue. Furthermore, to estimate liver tissue fibrosis, Masson's trichrome staining was done. Our findings showed that hypothyroidism-induced liver fibrosis was associated with increased levels of ALT, AST and ALP. Though, vitamin D administration could significantly reduce the ALT, AST and ALP in the serum and suppress the accumulation of collagen fibers. Moreover, the activity of SOD and total thiol content was notably reduced, while the MDA content was significantly increased in the PTU- induced hypothyroid rats compared to the control group. Nonetheless, treatment with vitamin D improved mentioned oxidative stress markers in the Hypo-vitamin D groups. In conclusion, vitamin D due to its potential antioxidant and anti-fibrotic properties could be effective in the decrease of hypothyroidism-associated liver injury.
Collapse
Affiliation(s)
- Seyed Hamidreza Rastegar-Moghaddam
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahsan Akbarian
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Rajabian
- Department of Internal Medicine, Faculty of Medicine, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Alipour
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Ebrahimzadeh bideskan
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
40
|
Ijaz MU, Mustafa S, Ain QU, Hamza A, Ahmed H, Abdel-Daim MM, Albadrani GM, Najda A, Ali S. Eriodictyol attenuates Furan induced testicular toxicity in Rats: Role of oxidative stress, steroidogenic enzymes and apoptosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 259:115003. [PMID: 37224777 DOI: 10.1016/j.ecoenv.2023.115003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/26/2023]
Abstract
Furan (C4H4O) is a naturally occurring organic compound. It develops as a result of the thermal processing of food and stimulates critical impairments in male reproductive tract. Eriodictyol (Etyol) is a natural dietary flavonoid possessing diverse pharmacological potentials. The recent investigation was proposed to ascertain the ameliorative potential of eriodictyol against furan-instigated reproductive dysfunctions. Male rats (n = 48) were classified into 4 groups: untreated/control, furan (10 mg/kg), furan+ eriodictyol (10 mg/kg + 20 mg/kg) and eriodictyol (20 mg/kg). At the 56th day of the trial, the protective effects of eriodictyol were evaluated by assessing various parameters. Results of the study revealed that eriodictyol attenuated furan-induced testicular toxicity in the biochemical profile by increasing catalase (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD) along with glutathione reductase (GSR) activities, whereas reduced the reactive oxygen species (ROS) along with malondialdehyde (MDA) levels. It also restored the normal state of sperm motility, viability, the count of hypo-osmotic tail swelled sperm as well as epididymal sperm number along with reduced sperm anomalies (morphological) tail, mid-piece and head. Furthermore, it elevated the decreased levels of luteinizing hormone (LH), plasma testosterone and follicle-stimulating hormone (FSH) as well steroidogenic enzymes (17β-HSD, StAR protein & 3β-HSD) and testicular anti-apoptotic marker (Bcl-2) expression, whereas, down-regulating apoptotic markers (Bax & Caspase-3) expression. Eriodictyol treatment also effectively mitigated the histopathological damages. The outcomes of the current study provide fundamental insights into the ameliorative potential of eriodictyol against furan-instigated testicular toxicity.
Collapse
Affiliation(s)
- Muhammad Umar Ijaz
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Shama Mustafa
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Qurat Ul Ain
- Department of Zoology, Government College Women University, Sialkot, Pakistan
| | - Ali Hamza
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Hussain Ahmed
- Department of Zoology, The University of Buner, Khyber Pakhtunkhwa, Pakistan
| | - Mohamed M Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt.
| | - Ghadeer M Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, 84428, Riyadh 11671, Saudi Arabia
| | - Agnieszka Najda
- Department of Vegetable Crops and Medicinal Plants University of Life Sciences in Lublin, 50A Doświadczalna Street, 20-280 Lublin, Poland
| | - Shafaqat Ali
- Department of Environmental Sciences, Government College University, 38000, Faisalabad, Pakistan; Department of Biological Sciences and Technology, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
41
|
Mustafa S, Anwar H, Ain QU, Ahmed H, Iqbal S, Ijaz MU. Therapeutic effect of gossypetin against paraquat-induced testicular damage in male rats: a histological and biochemical study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:62237-62248. [PMID: 36940025 DOI: 10.1007/s11356-023-26469-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 03/11/2023] [Indexed: 05/10/2023]
Abstract
Paraquat (PQ) is an organic compound, which is commonly used as a herbicide in the agriculture sector, and it is also known to stimulate critical damages in the male reproductive system. Gossypetin (GPTN) is one of important members of the flavonoid family, which is an essential compound in flowers and calyx of Hibiscus sabdariffa with potential pharmacological properties. The current investigation was aimed to examine the ameliorative potential of GPTN against PQ-instigated testicular damages. Adult male Sprague-Dawley rats (n = 48) were distributed into four groups: control, PQ (5 mg/kg), PQ + GPTN (5 mg/kg + 30 mg/kg respectively), and GPTN (30 mg/kg). After 56 days of treatment, biochemical, spermatogenic indices, hormonal, steroidogenic, pro-or-anti-apoptotic, and histopathological parameters were estimated. PQ exposure disturbed the biochemical profile by reducing the activities of catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione reductase (GSR), while it increased the concentration of reactive oxygen species (ROS) and malondialdehyde (MDA) level. Furthermore, PQ exposure decreased the sperm motility, viability, number of hypo-osmotic tail swelled spermatozoa, and epididymal sperm count; additionally, it increased sperm morphological (head mid-piece and tail) abnormalities. Moreover, PQ lessened the follicle-stimulating hormone (FSH), luteinizing hormone (LH), and plasma testosterone levels. Besides, PQ-intoxication downregulated the gene expression of steroidogenic enzymes (StAR, 3β-HSD, and 17β-HSD) and anti-apoptotic marker (Bcl-2), whereas upregulated the gene expression of apoptotic markers (Bax and Caspase-3). PQ exposure led to histopathological damages in testicular tissues as well. Nonetheless, GPTN inverted all the illustrated impairments in testes. Taken together, GPTN could potently ameliorate PQ-induced reproductive dysfunctions due to its antioxidant, androgenic, and anti-apoptotic potential.
Collapse
Affiliation(s)
- Shama Mustafa
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Haseeb Anwar
- Department of Physiology, Government College University, Faisalabad, Pakistan
| | - Qurat Ul Ain
- Department of Zoology, Government College Women University, Sialkot, Pakistan
| | - Hussain Ahmed
- Department of Zoology, The University of Buner, Khyber Pakhtunkhwa, Pakistan
| | - Shabnoor Iqbal
- Department of Zoology, Government College University, Faisalabad, Pakistan
| | - Muhammad Umar Ijaz
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan.
| |
Collapse
|
42
|
Abbas NAT, Nafea OE, Mohammed HO, Samy W, Abdelmageed AF, Afifi R, Hassan HA. Repurposing of carvedilol to alleviate lung fibrosis in rats: Repressing of TGF-β1/α-SMA/Smad2/3 and STAT3 gene expressions. Life Sci 2023; 324:121692. [PMID: 37061127 DOI: 10.1016/j.lfs.2023.121692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/30/2023] [Accepted: 04/07/2023] [Indexed: 04/17/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most widely studied interstitial lung disease. IPF eventually leads to respiratory insufficiency, lung cancer, and death. Carvedilol (CAR) is a third-generation β-adrenergic receptor antagonist with α1-blocking effect. CAR demonstrates antifibrotic activities in various experimental models of organ fibrosis. AIMS This work is designed to explore the possible alleviating effects of CAR on bleomycin (BLM)-induced lung fibrosis in rats. MAIN METHODS The BLM rat model of lung fibrosis was achieved by intratracheal delivery of a single dose of 5 mg/kg of BLM. Seven days following BLM injection, either prednisolone or CAR was orally administered at doses of 10 mg/kg once daily for 21 days to rats. The actions of CAR were evaluated by lung oxidant/antioxidant parameters, protein concentration and total leucocyte count (TLC) in bronchoalveolar lavage fluid (BALF), fibrosis regulator-related genes along with the coexistent lung histological changes. KEY FINDINGS CAR effectively decreased lung malondialdehyde level, increased superoxide dismutase activity, declined both protein concentration and TLC in BALF, downregulated TGF-β1/α-SMA/Smad2/3 and STAT3 gene expressions, and repaired the damaged lung tissues. SIGNIFICANCE CAR conferred therapeutic potential against BLM-induced lung fibrosis in rats, at least in part, to its antioxidant, anti-inflammatory, and antifibrotic activities. CAR could be utilized as a prospective therapeutic option in patients with lung fibrosis in clinical practice.
Collapse
Affiliation(s)
- Noha A T Abbas
- Department of Clinical Pharmacology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Ola Elsayed Nafea
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt; Department of Clinical Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Heba Osama Mohammed
- Department of Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Walaa Samy
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig 45519, Egypt
| | - Amal Fawzy Abdelmageed
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig 45519, Egypt
| | | | - Heba A Hassan
- Department of Clinical Pharmacology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt; Department of Pharmacology, Faculty of Medicine, Mutah University, P.O. Box 7, Al-Karak 61710, Jordan
| |
Collapse
|
43
|
Secretome of hESC-Derived MSC-like Immune and Matrix Regulatory Cells Mitigate Pulmonary Fibrosis through Antioxidant and Anti-Inflammatory Effects. Biomedicines 2023; 11:biomedicines11020463. [PMID: 36830999 PMCID: PMC9953085 DOI: 10.3390/biomedicines11020463] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Oxidative stress and inflammation are major drivers in the pathogenesis and progression of pulmonary fibrosis (PF). The mesenchymal stem cell (MSC) secretome has regenerative potential and immunomodulatory functions. Human embryonic stem cell (hESC)-derived MSC-like immune and matrix regulatory cells (IMRCs) are manufacturable with large-scale good manufacturing practice (GMP) preparation. In the present study, the antioxidative and anti-inflammatory properties and the therapeutic effect of the secretome of hESC-MSC-IMRC-derived conditioned culture medium (CM) (hESC-MSC-IMRC-CM) were investigated. Results revealed the capacities of hESC-MSC-IMRC-CM to reduce bleomycin (BLM)-induced reactive oxygen species (ROS), extracellular matrix (ECM) deposition, and epithelial-mesenchymal transition (EMT) in A549 cells. The administration of concentrated hESC-MSC-IMRC-CM significantly alleviated the pathogenesis of PF in lungs of BLM-injured mice, as accessed by pathohistological changes and the expression of ECM and EMT. A mechanistic study further demonstrated that the hESC-MSC-IMRC-CM was able to inhibit BLM-induced ROS and pro-inflammatory cytokines, accompanied by a reduced expression of Nox4, Nrf2, Ho-1, and components of the Tlr4/MyD88 signaling cascade. These results provide a proof of concept for the hESC-MSC-IMRC-derived secretome treatment of PF, in part mediated by their antioxidative and anti-inflammatory effects. This study thus reinforces the development of ready-to-use, cell-free hESC-MSC-IMRC secretome biomedicine for the treatment of PF in clinical settings.
Collapse
|
44
|
Li S, Wang P, Liu Y, Yang K, Zhong R, Cheng D, He L. A mitochondrial-targeted near-infrared fluorescent probe for visualizing the fluctuation of hypochlorite acid in idiopathic pulmonary fibrosis mice. Anal Chim Acta 2023; 1239:340731. [PMID: 36628728 DOI: 10.1016/j.aca.2022.340731] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/01/2022] [Accepted: 12/17/2022] [Indexed: 12/23/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic inflammatory disease destroying lungs irreversibly with high mortality rates. There are challenges in diagnosing IPF and treating it at an early stage. Mounting evidence suggests that hypochlorous acid (HClO) can help in diagnosing inflammation and relevant conditions. Pulmonary fibrosis is linked to the mitochondrial oxidative stress where excessive HClO production is a key molecular mechanism. Measuring mitochondrial HClO levels assists in the investigations of how the mitochondrial oxidative stress affects IPF. Herein, NIR-PTZ-HClO was developed and optimized as a probe for detecting fluctuations in HClO concentrations of cells and mice models through near-infrared (NIR) fluorescence. The probe featured large Stokes shift of 150 nm, NIR turn-on signal at 650 nm, high sensitivity (45-fold) and quick HClO detection (2 s). The probe is selective for HClO in the presence of range of other analytes. NIR-PTZ-HClO visualized both endogenous and exogenous HClO in living cells (RAW264.7, H460 and A549). The probe monitored HClO in mice models with IPF and moreover the HClO profile could be tracked during the IPF process. The probe also detected precipitous decrease in HClO levels in IPF mice treated with OFEV. NIR-PTZ-HClO probe has thus the potential for earlier diagnosis of lung fibrosis, thereby improving the treatment efficacy.
Collapse
Affiliation(s)
- Songjiao Li
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Cancer Research Institute, Department of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, PR China
| | - Peipei Wang
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Cancer Research Institute, Department of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, PR China
| | - Ying Liu
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Cancer Research Institute, Department of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, PR China
| | - Ke Yang
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Cancer Research Institute, Department of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, PR China
| | - Rongbin Zhong
- Clinical Research Institute, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, PR China
| | - Dan Cheng
- Clinical Research Institute, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, PR China.
| | - Longwei He
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Cancer Research Institute, Department of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, 421001, PR China.
| |
Collapse
|
45
|
Ostovar T, Rezaei S, Shokri-Afra H, Samavarchi Tehrani S, Namvarjah F, Aliabadi M, Effatpanah H, Moradi-Sardareh H. Effect of Capparis spinosa Fruit Hydroalcoholic Extract on Paraquat-Induced Pulmonary Fibrosis in the Rat. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2023; 12:423-434. [PMID: 39006195 PMCID: PMC11240055 DOI: 10.22088/ijmcm.bums.12.4.423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/07/2023] [Accepted: 04/08/2024] [Indexed: 07/16/2024]
Abstract
Pulmonary fibrosis (PF) is a lethal inflammatory disease and there has been no effective medication for this progressive disease up to now. Paraquat is commonly used in agricultural settings to control weed growth and is one of the important risk factors for PF. Additionally, emerging evidence has demonstrated Capparis spinosa (C. spinose) fruit extract has anti-fibrotic, anti-inflammatory, and antioxidant properties. We aimed to evaluate whether C. spinose fruit hydroalcoholic extract has a positive effect against Paraquat-induced PF in rats. 30 male Wistar rats were randomly divided into 5 groups, which included: a control group, a Paraquat control group, a C. spinose group with a dose of 20 mg/kg, a C. spinose group with a dose of 30 mg/kg, a C. spinose group with a dose of 50 mg/kg. After 21 days of the treatment, levels of hydroxyproline and malondialdehyde (MDA) in lung tissue were assessed and lung indices and semi-quantitative histopathological changes were determined. The results showed that treatment with C. spinose, led to increased weight gain, whereas reduced lung weight. C. spinose demonstrated a decreasing effect on levels of MDA, and hydroxyproline in lung tissue. Moreover, histopathological data and the number of lung indices indicated the preventive role of C. spinose Paraquat-induced PF in rats.
Collapse
Affiliation(s)
- Tahmine Ostovar
- International Campus, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Sahar Rezaei
- Department of Clinical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hajar Shokri-Afra
- Gut and Liver Research Center, Non-communicable Disease Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Sadra Samavarchi Tehrani
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Science, Tehran, Iran.
| | - Fatemeh Namvarjah
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Masoume Aliabadi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hosein Effatpanah
- Department of Public Health, Asadabad School of Medical Sciences, Asadabad, Iran.
| | | |
Collapse
|
46
|
Zhao C, Pu W, Wazir J, Jin X, Wei L, Song S, Su Z, Li J, Deng Y, Wang H. Long-term exposure to PM2.5 aggravates pulmonary fibrosis and acute lung injury by disrupting Nrf2-mediated antioxidant function. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 313:120017. [PMID: 36007796 DOI: 10.1016/j.envpol.2022.120017] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
Epidemiological studies have indicated that exposure to ambient air-borne fine particulate matter (PM2.5) is associated with many cardiopulmonary diseases; however, the underlying pathological mechanisms of PM2.5-induced lung injury remain unknown. In this study, we aimed to assess the impact of acute or prolonged exposure to water-insoluble fractions of PM2.5 (PM2.5 particulate) on lung injury and its molecular mechanisms. Balb/c mice were randomly exposed to PM2.5 once (acute exposure) or once every three days for a total of 6 times (prolonged exposure). Lung, BALF and blood samples were collected, and pulmonary pathophysiological alterations were analyzed. Nrf2 knockout mice were adapted to assess the involvement of Nrf2 in lung injury, and transcriptomic analysis was performed to delineate the mechanisms. Through transcriptomic analysis and validation of Nrf2 knockout mice, we found that acute exposure to PM2.5 insoluble particulates induced neutrophil infiltration-mediated airway inflammation, whereas prolonged exposure to PM2.5 insoluble particulate triggered lung fibrosis by decreasing the transcriptional activity of Nrf2, which resulted in the downregulated expression of antioxidant-related genes. In response to secondary LPS exposure, prolonged PM2.5 exposure induced more severe lung injury, indicating that prolonged PM2.5 exposure induced Nrf2 inhibition weakened its antioxidative defense capacity against oxidative stress injury, leading to the formation of pulmonary fibrosis and increasing its susceptibility to secondary bacterial infection.
Collapse
Affiliation(s)
- Chen Zhao
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Wenyuan Pu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Xiaolu Jin
- The First People's Hospital of Yancheng, The Affiliated Hospital of Nanjing University Medical School, Yancheng, 224006, China
| | - Lulu Wei
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Shiyu Song
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China
| | - Zhonglan Su
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jiabin Li
- The First People's Hospital of Yancheng, The Affiliated Hospital of Nanjing University Medical School, Yancheng, 224006, China
| | - Yijun Deng
- The First People's Hospital of Yancheng, The Affiliated Hospital of Nanjing University Medical School, Yancheng, 224006, China
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
47
|
Nie H, Liu H, Shi Y, Lai W, Liu X, Xi Z, Lin B. Effects of Different Concentrations of Oil Mist Particulate Matter on Pulmonary Fibrosis In Vivo and In Vitro. TOXICS 2022; 10:647. [PMID: 36355939 PMCID: PMC9695344 DOI: 10.3390/toxics10110647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
Oil-mist particulate matter (OMPM) refers to oily particles with a small aerodynamic equivalent diameter in ambient air. Since the pathogenesis of pulmonary fibrosis (PF) has not been fully elucidated, this study aims to explore the potential molecular mechanisms of the adverse effects of exposure to OMPM at different concentrations in vivo and in vitro on PF. In this study, rats and cell lines were treated with different concentrations of OMPM in vivo and in vitro. Sirius Red staining analysis shows that OMPM exposure could cause pulmonary lesions and fibrosis symptoms. The expression of TGF-β1, α-SMA, and collagen I was increased in the lung tissue of rats. The activities of MMP2 and TIMP1 were unbalanced, and increased N-Cadherin and decreased E-Cadherin upon OMPM exposure in a dose-dependent manner. In addition, OMPM exposure could activate the TGF-β1/Smad3 and TGF-β1/MAPK p38 signaling pathways, and the differentiation of human lung fibroblast HFL-1 cells. Therefore, OMPM exposure could induce PF by targeting the lung epithelium and fibroblasts, and activating the TGF-β1/Smad3 and TGF-β1/MAPK p38 signaling pathways.
Collapse
|
48
|
Ye Q, Taleb SJ, Wang H, Parinandi NL, Kass DJ, Rojas M, Wang C, Ma Q, Zhao J, Zhao Y. Molecular Regulation of Heme Oxygenase-1 Expression by E2F Transcription Factor 2 in Lung Fibroblast Cells: Relevance to Idiopathic Pulmonary Fibrosis. Biomolecules 2022; 12:biom12101531. [PMID: 36291740 PMCID: PMC9599643 DOI: 10.3390/biom12101531] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/26/2022] [Accepted: 10/20/2022] [Indexed: 01/05/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal chronic lung disease. Heme oxygenase-1 (HMOX1/HO-1) is an enzyme that catalyzes the degradation of heme. The role of HO-1 in the pathogenesis of IPF has been studied; however, the molecular regulation of HO-1 and its role in IPF are still unclear. In this study, we found that HO-1 protein levels significantly increased in lung myofibroblasts in IPF patients and in lungs in a murine model of bleomycin-induced lung fibrosis. In addition, we observed that administration of a E2F transcription factor inhibitor elevated HO-1 mRNA and protein levels in lung fibroblasts. Downregulation of E2F2 by siRNA transfection increased HO-1 mRNA and protein levels, while overexpression of E2F2 reduced HO-1 levels. However, overexpression of E2F2 did not alter hemin-induced HO-1 protein levels. Furthermore, modulation of HO-1 levels regulated TGF-β1-induced myofibroblast differentiation without altering the phosphorylation of Smad2/3 in lung fibroblast cells. Moreover, the phosphorylation of protein kinase B (Akt) was significantly upregulated in HO-1-depleted lung fibroblast cells. In summary, this study demonstrated that E2F2 regulates the baseline expression of HO-1, but has no effect on modulating HO-1 expression by hemin. Finally, elevated HO-1 expression contributes to the TGF-β1-induced lung myofibroblast differentiation through the activation of the serine/threonine kinase AKT pathway. Overall, our findings suggest that targeting E2F2/HO-1 might be a new therapeutic strategy to treat fibrotic diseases such as IPF.
Collapse
Affiliation(s)
- Qinmao Ye
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Sarah J. Taleb
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Heather Wang
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Narasimham L. Parinandi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel J. Kass
- Department of Medicine, The University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Mauricio Rojas
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Cankun Wang
- Department of Bioinformatics, The Ohio State University, Columbus, OH 43210, USA
| | - Qin Ma
- Department of Bioinformatics, The Ohio State University, Columbus, OH 43210, USA
| | - Jing Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Yutong Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: ; Tel.: +1-614-685-0360
| |
Collapse
|
49
|
Suzuki T, Kropski JA, Chen J, Carrier EJ, Chen X, Sherrill TP, Winters NI, Camarata JE, Polosukhin VV, Han W, Rathinasabapathy A, Gutor S, Gulleman P, Sabusap C, Banovich NE, Tanjore H, Freeman ML, Tada Y, Young LR, Gokey JJ, Blackwell TS, West JD. Thromboxane-Prostanoid Receptor Signaling Drives Persistent Fibroblast Activation in Pulmonary Fibrosis. Am J Respir Crit Care Med 2022; 206:596-607. [PMID: 35728047 PMCID: PMC9716913 DOI: 10.1164/rccm.202106-1503oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Rationale: Although persistent fibroblast activation is a hallmark of idiopathic pulmonary fibrosis (IPF), mechanisms regulating persistent fibroblast activation in the lungs have not been fully elucidated. Objectives: On the basis of our observation that lung fibroblasts express TBXA2R (thromboxane-prostanoid receptor) during fibrosis, we investigated the role of TBXA2R signaling in fibrotic remodeling. Methods: We identified TBXA2R expression in lungs of patients with IPF and mice and studied primary mouse and human lung fibroblasts to determine the impact of TBXA2R signaling on fibroblast activation. We used TBXA2R-deficient mice and small-molecule inhibitors to investigate TBXA2R signaling in preclinical lung fibrosis models. Measurements and Main Results: TBXA2R expression was upregulated in fibroblasts in the lungs of patients with IPF and in mouse lungs during experimental lung fibrosis. Genetic deletion of TBXA2R, but not inhibition of thromboxane synthase, protected mice from bleomycin-induced lung fibrosis, thereby suggesting that an alternative ligand activates profibrotic TBXA2R signaling. In contrast to thromboxane, F2-isoprostanes, which are nonenzymatic products of arachidonic acid induced by reactive oxygen species, were persistently elevated during fibrosis. F2-isoprostanes induced TBXA2R signaling in fibroblasts and mediated a myofibroblast activation profile due, at least in part, to potentiation of TGF-β (transforming growth factor-β) signaling. In vivo treatment with the TBXA2R antagonist ifetroban reduced profibrotic signaling in the lungs, protected mice from lung fibrosis in three preclinical models (bleomycin, Hermansky-Pudlak mice, and radiation-induced fibrosis), and markedly enhanced fibrotic resolution after bleomycin treatment. Conclusions: TBXA2R links oxidative stress to fibroblast activation during lung fibrosis. TBXA2R antagonists could have utility in treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Toshio Suzuki
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
- Department of Medical Oncology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Medicine, Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Jingyuan Chen
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Erica J. Carrier
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Xinping Chen
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Taylor P. Sherrill
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Nichelle I. Winters
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Jane E. Camarata
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Vasiliy V. Polosukhin
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Wei Han
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | | | - Sergey Gutor
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Peter Gulleman
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Carleen Sabusap
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | | | - Harikrishna Tanjore
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Michael L. Freeman
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yuji Tada
- Department of Pulmonary Medicine, School of Medicine, International University of Health and Welfare, Chiba, Japan; and
| | - Lisa R. Young
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
- Division of Pulmonary Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jason J. Gokey
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Timothy S. Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Medicine, Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - James D. West
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| |
Collapse
|
50
|
Contribution of Adiponectin/Carnitine Palmityl Transferase 1A-Mediated Fatty Acid Metabolism during the Development of Idiopathic Pulmonary Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5265616. [PMID: 36035217 PMCID: PMC9402305 DOI: 10.1155/2022/5265616] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/14/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease that leads rapidly to death. The present study is aimed at discovering the in-depth pathogenesis of IPF, exploring the role of adiponectin/carnitine palmityl transferase 1A- (APN/CPT1A-) mediated fatty acid metabolism during the development of IPF, and excavating its potential mechanism. Here, THP-1 cells were differentiated into M0 macrophages, followed by polarization to M1 macrophages upon hypoxia. Subsequently, lung fibroblast HFL-1 cells were stimulated by M1 macrophages to simulate hypoxia-related IPF condition in vitro. It was discovered that the stimulation of M1 macrophages promoted fibroblast proliferation and fibrosis formation in vitro, accompanied with a disorder of the APN/CPT1A pathway, an overproduction of lipid peroxides, and a low level of autophagy in HFL-1 cells. Thereafter, APN treatment or CPT1A overexpression greatly suppressed above lipid peroxide accumulation, fibroblast proliferation, and fibrosis but activated autophagy in vitro. Furthermore, an in vivo IPF rat model was established by injection of bleomycin (BLM). Consistently, CPT1A overexpression exerted a protective role against pulmonary fibrosis in vivo; however, the antifibrosis property of CPT1A was partly abolished by 3-methyladenine (an autophagy inhibitor). In summary, APN/CPT1A-mediated fatty acid metabolism exerted its protective role in IPF partly through activating autophagy, shedding a new prospective for the treatment of IPF.
Collapse
|