1
|
Wu J, Wei Y, Kang H, Yu X, Wei S, Xue T, Kong X. Integrated bioinformatics analysis screened the key genes and pathways of idiopathic pulmonary fibrosis. Sci Rep 2025; 15:14448. [PMID: 40280949 PMCID: PMC12032202 DOI: 10.1038/s41598-025-97037-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
To investigate the molecular mechanisms underlying idiopathic pulmonary fibrosis (IPF), we analyzed the GSE173355 and GSE173356 datasets obtained from the NCBI-GEO database. We identified differentially expressed genes (DEGs) and differentially methylated sites. Functional enrichment analysis was conducted for both DEGs and differentially methylated sites. Functional enrichment analysis was performed for both DEGs and differentially methylated sites, alongside an examination of immune-related scores, proportions, and GSVA enrichment scores of immune cells in IPF versus control samples. An integrated gene-methylation association analysis revealed 8 genes with expression levels negatively influenced by methylation. The Rap1 pathway, Focal adhesion, and Axon guidance were significantly enriched among both DEGs and differentially methylated sites. Immune-related scores were notably lower in the IPF group compared to the control group, with marked differences in immune cell proportions and GSVA enrichment scores. Screening of DEGs identified 361 differentially expressed immune-related genes (IRGs). Protein-protein interaction (PPI) network analysis using the STRING database and Cytoscape software unveiled 10 key genes and 3 core subnetworks. RT-qPCR validation in bleomycin-induced IPF mouse model and A549 EMT model confirmed the reliability of most findings. These results provide new insights into IPF pathogenesis and potential therapeutic strategies, necessitating further functional validation.
Collapse
Affiliation(s)
- Juan Wu
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yangyang Wei
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hong Kang
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiao Yu
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuting Wei
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ting Xue
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaomei Kong
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
2
|
Liu X, Dong X, Peng Z, Wang C, Wan J, Chen M, Zheng C. Collagenase-functionalized Liposomes Based on Enhancing Penetration into the Extracellular Matrix Augment Therapeutic Effect on Idiopathic Pulmonary Fibrosis. AAPS PharmSciTech 2025; 26:113. [PMID: 40281247 DOI: 10.1208/s12249-025-03112-9] [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: 02/18/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
In this study, a quercetin-loaded liposome system modified with collagenase was developed to increase QU penetration in the ECM and improve IPF treatment. Quercetin-loaded long circulation liposome (QU-LP) and quercetin-loaded liposome modified with collagenase type I (QU-CLP) were prepared, followed by characterization of the encapsulation efficiency, particle size, morphology, and in vitro drug release. Their effect on the cytotoxicity of A549 cells was detected by the Cell Counting Kit-8, and the cellular uptake was investigated using cellular fluorescence imaging and flow cytometry. TGF-β1 induced A549 cell model was established to mimic pulmonary fibrosis to explore further the anti-pulmonary fibrosis effect of QU-CLP by CCK8 experiment. QU-CLP significantly improves the solubility and bioavailability of QU by encapsulating it in the internal cavity with a high encapsulation efficiency (EE%) of 92.86 ± 1.03%. Liposomes alleviate the influence of QU on normal A549 cell growth. Enhanced fluorescence intensity was observed in A549 cells treated with coumarin 6-labeled and collagenase-modified nanoliposomes (C6-CLP) after 4 h of incubation on the collagen matrix, confirming that collagenase-loaded liposomes could penetrate the collagen barrier and cells internalized more hydrophobic drug. The mean fluorescence intensity (MFI) of the C6-CLP group was 2.88 times that of the C6-labeled nanoliposomes (C6-LP). Moreover, QU-CLP significantly (**P < 0.01) inhibited the proliferation of A549 cells stimulated by TGF-β1. QU-CLP has excellent potential for delivering QU with enhanced bioavailability, high cellular uptake efficiency, and improved therapeutic efficacy in IPF.
Collapse
Affiliation(s)
- Xiaoqing Liu
- Department of Pharmacy, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Xiaoling Dong
- Shandong Hubble Kisen Biological Technology Co.,Ltd., Jinan, 250100, China
| | - Zhen Peng
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Cuihong Wang
- Department of Pharmacy, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Jianwei Wan
- Department of Pharmacy, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Min Chen
- Department of Pharmacy, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China.
| | - Chunli Zheng
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
3
|
Khoury M, Faiz SA, Sheshadri A. Immune checkpoint inhibitor-associated pneumonitis: focus on diagnosis and underlying mechanisms. Curr Opin Pulm Med 2025:00063198-990000000-00239. [PMID: 40265506 DOI: 10.1097/mcp.0000000000001175] [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: 04/24/2025]
Abstract
PURPOSE OF REVIEW This review aims to provide an updated overview of the diagnosis, risk factors, and treatment strategies for immune checkpoint inhibitor (ICI) pneumonitis, with a particular emphasis on its underlying pathophysiology. RECENT FINDINGS Recent advances, such as single-cell RNA sequencing of bronchoalveolar lavage fluid and the identification of biomarkers, including autoantibodies, are enhancing our understanding of ICI-related pneumonitis. These findings suggest that both cell-mediated and humoral mechanisms contribute to the pathophysiology of the condition. SUMMARY Pneumonitis can significantly limit the efficacy of life-saving cancer treatments, such as ICIs. Although corticosteroids are the first-line treatment according to guidelines, steroid-refractory pneumonitis remains common and is associated with high mortality. Emerging data is providing a more detailed understanding of the dysregulated immune response responsible for pneumonitis, which may guide the development of targeted therapies and direct future research efforts.
Collapse
Affiliation(s)
- Mtanis Khoury
- Divisions of Pulmonary, Critical Care Medicine and Sleep Medicine, McGovern Medical School at University of Texas Health
| | - Saadia A Faiz
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
4
|
Lu J, Wang Z, Zhang L. Single-cell transcriptome analysis revealing mechanotransduction via the Hippo/YAP pathway in promoting fibroblast-to-myofibroblast transition and idiopathic pulmonary fibrosis development. Gene 2025; 943:149271. [PMID: 39855369 DOI: 10.1016/j.gene.2025.149271] [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: 09/27/2024] [Revised: 12/12/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
OBJECTIVE Idiopathic pulmonary fibrosis (IPF) is an irreversible and fatal interstitial lung disease, characterized by excessive extracellular matrix (ECM) secretion that disrupts normal alveolar structure. This study aims to explore the potential molecular mechanisms underlying the promotion of IPF development. METHODS Firstly, we compared the transcriptome and single-cell sequencing data from lung tissue samples of patients with IPF and healthy individuals. Subsequently, we conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses on the differentially expressed genes (DEGs). Furthermore, we employed sodium alginate hydrogels with varying degrees of crosslinking to provide differential mechanical stress, mimicking the mechanical microenvironment in vivo during lung fibrosis. On this basis, we examined cytoskeletal remodeling in fibroblasts MRC-5, mRNA expression of multiple related genes, immunofluorescence localization, and cellular proliferation capacity. RESULTS Bioinformatics analysis revealed a series of DEGs associated with IPF. Further functional and pathway enrichment analyses indicated that these DEGs were primarily enriched in ECM-related biological processes. Single-cell sequencing data revealed that fibroblasts and myofibroblasts are the main contributors to excessive ECM secretion and suggested activation of mechanotransduction and the Hippo/YAP signaling pathway in myofibroblasts. Cellular experiments demonstrated that sodium alginate hydrogels with different stiffness can simulate different mechanical stress environments, thereby affecting cytoskeletal rearrangement and Hippo/YAP pathway activity in MRC-5 lung fibroblasts. Notably, high levels of mechanical stress promoted YAP nuclear translocation, increased expression of type I collagen and α-SMA, and enhanced proliferative capacity. Additionally, we also found that fibroblasts primarily participate in mechanotransduction through the Rho/ROCK and Integrin/FAK pathways under high mechanical stress conditions, ultimately upregulating the gene expression of CCNE1/2, CTGF, and FGF1. CONCLUSION Our study uncovers the crucial role of cytoskeletal mechanotransduction in myofibroblast transformation and IPF development through activation of the Hippo/YAP pathway, providing new insights into understanding the pathogenesis of IPF.
Collapse
Affiliation(s)
- Jiaqi Lu
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China.
| | - Zhenhua Wang
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China
| | - Liguo Zhang
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China
| |
Collapse
|
5
|
Huang P, Qin D, Qin Y, Tao S, Liu G. SIRT3/6/7: promising therapeutic targets for pulmonary fibrosis. Front Cell Dev Biol 2025; 13:1557384. [PMID: 40241794 PMCID: PMC12000143 DOI: 10.3389/fcell.2025.1557384] [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: 01/08/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Pulmonary fibrosis is a chronic progressive fibrosing interstitial lung disease of unknown cause, characterized by excessive deposition of extracellular matrix, leading to irreversible decline in lung function and ultimately death due to respiratory failure and multiple complications. The Sirtuin family is a group of nicotinamide adenine dinucleotide (NAD+) -dependent histone deacetylases, including SIRT1 to SIRT7. They are involved in various biological processes such as protein synthesis, metabolism, cell stress, inflammation, aging and fibrosis through deacetylation. This article reviews the complex molecular mechanisms of the poorly studied SIRT3, SIRT6, and SIRT7 subtypes in lung fibrosis and the latest research progress in targeting them to treat lung fibrosis.
Collapse
Affiliation(s)
- Pingping Huang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Dan Qin
- Department of Endocrinology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yanling Qin
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Sha Tao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guangnan Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
6
|
Fang YF, Zhang C, Han MM, Wang Y, Zhou TJ, Xing L, Wei N, Wang J, Jeong JH, Zhou F, Wang GJ, Jiang HL. Engineered MSCs Break Endothelial-Myofibroblast Crosstalk in Pulmonary Fibrosis: Reconstructing the Vascular Niche. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414601. [PMID: 40018848 DOI: 10.1002/adma.202414601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/26/2025] [Indexed: 03/01/2025]
Abstract
In the progress of pulmonary fibrosis (PF), the normal vascular niche plays a crucial role in alveolar regeneration by secreting angiocrine factors. However, the malignant interaction between myofibroblasts and vascular endothelial cells results in significant loss of pulmonary capillaries in fibroblast foci, which promotes continuous deterioration of fibrosis. Herein, an engineered mesenchymal stem cell (MSC) therapeutic named MSC-MM@LPHN is developed for reconstructing the vascular niche, which is formed by modifying the surface of MSC with ROS-responsive lipid polymeric hybrid nanoparticles encapsulating the metformin and macitentan. Due to the homing ability of MSC, the MSC-MM@LPHN can effectively target lung tissue, then induce myofibroblast dedifferentiation to reduce the secretion of cytokines that cause endothelial cell damage and preventing endothelial cells from turning into a fibrotic phenotype, leading to recovery of the vascular endothelial cells function. Combined with the role of MSC-secreted growth factors promoting angiogenesis, the MSC-MM@LPHN ultimately constructs normal vascular structure in the fibroblast area and reverses bleomycin-induced PF. The findings suggest targeting the cell network in the vascular niche can effectively treat PF, which provides a novel therapeutic strategy for fibrosis-related diseases.
Collapse
Affiliation(s)
- Yue-Fei Fang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Chen Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Meng-Meng Han
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, 210009, China
| | - Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, 210009, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, 210009, China
| | - Ning Wei
- Jiangsu Renocell Biotech Co., Ltd, Nanjing, 211100, China
| | - Jing Wang
- Jiangsu Renocell Biotech Co., Ltd, Nanjing, 211100, China
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Guang-Ji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, 210009, China
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, 16419, South Korea
- College of Pharmacy, Yanbian University, Yanji, 133002, China
| |
Collapse
|
7
|
Mutlu S, Fytianos K, Ferrié C, Scalise M, Mykoniati S, Gazdhar A, Blank F. Adoptive Transfer of T Cells as a Potential Therapeutic Approach in the Bleomycin-Injured Mouse Lung. J Gene Med 2025; 27:e70018. [PMID: 40159455 PMCID: PMC11955259 DOI: 10.1002/jgm.70018] [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: 11/18/2024] [Revised: 02/21/2025] [Accepted: 03/15/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a lethal disease with an unknown etiology and complex pathophysiology that are not fully understood. The disease involves intricate cellular interplay, particularly among various immune cells. Currently, there is no treatment capable of reversing the fibrotic process or aiding lung regeneration. Hepatocyte growth factor (HGF) has demonstrated antifibrotic properties, whereas the adoptive transfer of modified T cells is a well-established treatment for various malignancies. We aimed to understand the dynamics of T cells in the progression of lung fibrosis and to study the therapeutic benefit of adoptive T cell transfer in a bleomycin-injured mouse lung (BLM) model. METHODS T cells were isolated from the spleen of naïve mice and transfected in vitro with mouse HGF plasmid and were administered intratracheally to the mice lungs 7 days post-bleomycin injury to the lung. Lung tissue and bronchoalveolar lavage were collected and analyzed using flow cytometry, histology, qRT-PCR, ELISA, and hydroxyproline assay. RESULTS Our findings demonstrate the successful T cell therapy of bleomycin-induced lung injury through the adoptive transfer of HGF-transfected T cells in mice. This treatment resulted in decreased collagen deposition and a balancing of immune cell exhaustion and cytokine homeostasis compared with untreated controls. In vitro testing showed enhanced apoptosis in myofibroblasts induced by HGF-overexpressing T cells. CONCLUSIONS Taken together, our data highlight the great potential of adoptive T cell transfer as an emerging therapy to counteract lung fibrosis.
Collapse
Affiliation(s)
- Seyran Mutlu
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR)University of BernBernSwitzerland
- Graduate School for Cellular and Biomedical SciencesUniversity of BernBernSwitzerland
| | - Kleanthis Fytianos
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR)University of BernBernSwitzerland
| | - Céline Ferrié
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR)University of BernBernSwitzerland
| | - Melanie Scalise
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR)University of BernBernSwitzerland
| | | | - Amiq Gazdhar
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR)University of BernBernSwitzerland
| | - Fabian Blank
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR)University of BernBernSwitzerland
| |
Collapse
|
8
|
Parvin S, Bagheri H, Halabian R, Arabfard M, Ghazvini A, Vahedi E, Najafi A, Ghanei M. Comprehensive transcriptomics analysis of peripheral blood mononuclear cells in exposure to mustard gas. Int Immunopharmacol 2025; 150:114197. [PMID: 39946765 DOI: 10.1016/j.intimp.2025.114197] [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: 12/25/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 03/03/2025]
Abstract
INTRODUCTION Sulfur mustard (SM) is a substance that causes blisters and has been repeatedly used by Iraq in chemical warfare against more than 100,000 Iranians. The main issue for these people is various pulmonary problems similar to chronic obstructive pulmonary disease (COPD). MATERIALS AND METHODS Our study analyzed the total RNA profile extracted using the RNA-seq technique from peripheral blood mononuclear cells (PBMCs) isolated from Mustard Lung (ML) patients of all three groups (Severe, Moderate, and Mild) in terms of disease in healthy control (HC) subjects on the BGISEQ platform (Paired-end, 7 GB data, and rRNA depletion). However, given the severe group's importance in clinical problems, we prioritized studying this group. Differentially expressed genes (DEGs) of the severe group versus HC were obtained using the limma package. DEGs were analyzed through bioinformatics tools, and their gene ontology (GO) and enrichment analysis (EA) were evaluated. Then, String-db and Cytoscape tools were used to search for the most important functional genes. RESULTS We identified SERPINA1, MAPK3, MMP9, FOXO3, SLC4A1, FCGR3B, CXCR2, PTGS2, HBA2, GPX1, IL1RN, IFNG, RPS29, CXCL1, FPR1, and RPS9 genes using hub and bottleneck criteria. Based on the analysis of important genes, several biological pathways were identified, including innate immunity, inflammatory response, and activation of neutrophils, cellular response to cytokines, and cellular response to oxidative stress, lipoxygenase pathway, and macrophage differentiation. CONCLUSION Innate immunity and neutrophils play a crucial role in the pathogenesis of these individuals. The signaling pathways of interleukins 4, 10, and 13 stimulate the differentiation of lung macrophages (MQs) into M2, essential for repair, remodeling, and inflammation. Additionally, reactive oxygen species (ROS) activate Protein kinase B (PKB), also known as AKT, through Phosphoinositide 3-kinases (PI3K) and increase the activity of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), which results in decreased histone deacetylase 2 (HDAC2) being one of the important pathways of pathophysiology in these patients.
Collapse
Affiliation(s)
- Shahram Parvin
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hasan Bagheri
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Biomedicine Technologies Institute Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Masoud Arabfard
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Ghazvini
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ensieh Vahedi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Najafi
- Molecular Biology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Sánchez SV, Otavalo GN, Gazeau F, Silva AKA, Morales JO. Intranasal delivery of extracellular vesicles: A promising new approach for treating neurological and respiratory disorders. J Control Release 2025; 379:489-523. [PMID: 39800240 DOI: 10.1016/j.jconrel.2025.01.018] [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: 09/13/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Extracellular vesicles (EVs) are membrane vesicles secreted by all types of cells, including bacteria, animals, and plants. These vesicles contain proteins, nucleic acids, and lipids from their parent cells and can transfer these components between cells. EVs have attracted attention for their potential use in diagnosis and therapy due to their natural properties, such as low immunogenicity, high biocompatibility, and ability to cross the blood-brain barrier. They can also be engineered to carry therapeutic molecules. EVs can be delivered via various routes. The intranasal route is particularly advantageous for delivering them to the central nervous system, making it a promising approach for treating neurological disorders. SCOPE OF REVIEW This review delves into the promising potential of intranasally administered EVs-based therapies for various medical conditions, with a particular focus on those affecting the brain and central nervous system. Additionally, the potential use of these therapies for pulmonary conditions, cancer, and allergies is examined, offering a hopeful outlook for the future of medical treatments. MAJOR CONCLUSIONS The intranasal administration of EVs offers significant advantages over other delivery methods. By directly delivering EVs to the brain, specifically targeting areas that have been injured, this administration proves to be highly efficient and effective, providing reassurance about the progress in medical treatments. Intranasal delivery is not limited to brain-related conditions. It can also benefit other organs like the lungs and stimulate a mucosal immune response against various pathogens due to the highly vascularized nature of the nasal cavity and airways. Moreover, it has the added benefit of minimizing toxicity to non-targeted organs and allows the EVs to remain longer in the body. As a result, there is a growing emphasis on conducting clinical trials for intranasal administration of EVs, particularly in treating respiratory tract pathologies such as coronavirus disease.
Collapse
Affiliation(s)
- Sofía V Sánchez
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile; Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile
| | - Gabriela N Otavalo
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile; Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile
| | - Florence Gazeau
- Université Paris Cité, CNRS UMR8175, INSERM U1334, Laboratory NABI (Nanomédecine, Biologie Extracellulaire, Intégratome et Innovations en santé), Paris, France
| | - Amanda K A Silva
- Université Paris Cité, CNRS UMR8175, INSERM U1334, Laboratory NABI (Nanomédecine, Biologie Extracellulaire, Intégratome et Innovations en santé), Paris, France
| | - Javier O Morales
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile; Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile.
| |
Collapse
|
10
|
He GS, Xia JK, Li QH, Zheng Y, Shi CR, Li R, Hong Q, Chen XM. Specnuezhenide: Comprehensive review of pharmacology, pharmacokinetics and ethnomedicinal uses. Fitoterapia 2025; 181:106389. [PMID: 39805507 DOI: 10.1016/j.fitote.2025.106389] [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: 10/14/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
BACKGROUND Specnuezhenide (SPN) is a bioactive iridoid terpenoid compound mainly found in Ligustri Lucidi Fructus (LLF), that has a broad spectrum of pharmacological effects, including anti-neoplastic, hepatoprotective, anti-aging, anti-inflammatory, immune-modulatory properties. PURPOSE The present review provides a comprehensive summary of natural medicinal plants, traditional Chinese medicine compounds containing SPN, and their corresponding pharmacological mechanisms. METHODS Using several globally recognized databases such as Web of Science, Google Scholar, PubMed, ScienceDirect, Wiley, ACS, Springer, and CNKI until December 2024, A comprehensive literature search and analysis was carried out with the keywords "Specnuezhenide", " Pharmacology ", "Pharmacokinetics" and " Chinese herbal compound". RESULTS The results indicated that SPN is present in a diverse range of plants, including LLF, Osmanthus fragrans seeds and Naked barley. SPN plays an anti-inflammatory role by regulating the NF-κB and MAPK signaling pathways, down-regulating the expression of TNF-α, IL-1β, IL-6 and other cytokines. Furthermore, many Chinese herbal compounds have been found to contain SPN, such as treatment of spleen and kidney deficiency of compound Shenhua tablet, treatment of liver-kidney Yin deficiency of Er Zhi Wan, treatment of pulmonray abscess of Qidongning and treatment of stagnation of QI due to depression of the liver of Shuganzhi Tablet. SPN is primarily distributed in the stomach, intestine, and liver. However, due to its limited absorption in the gastrointestinal tract and low blood concentration, its bioavailability is significantly reduced. CONCLUSIONS Thereby, SPN holds immense potential in the prevention and treatment of liver, lung and kidney complications. This review intends to provide a novel insight for further development of SPN, hoping to reveal the potential of SPN and necessity of further studies in this field.
Collapse
Affiliation(s)
- Guo-Sen He
- The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China
| | - Ji-Kai Xia
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China; School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qi-Hu Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China; School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Zheng
- The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China
| | - Chun-Ru Shi
- The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China
| | - Run Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China
| | - Quan Hong
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China.
| | - Xiang-Mei Chen
- The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, State Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Key Disciplines of National Administration of Traditional Chinese Medicine(zyyzdxk-2023310), Beijing 100853, China.
| |
Collapse
|
11
|
Cao W, Huang L, Yu H, Qian Y, Liu L, Xu M, Li S, Zhou H, Li F. Calycosin extracted from Astragali Radix reduces NETs formation to improve renal fibrosis via TLR4/NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119391. [PMID: 39855434 DOI: 10.1016/j.jep.2025.119391] [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: 08/16/2024] [Revised: 12/20/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragali Radix (A. Radix) is the dry root of the leguminous plants Astragalus membranaceus (Fisch) Beg. var. mongholicus (Beg) Hsiao, and Astragalus membranaceus (Fisch) Bge., being used as a medicinal and edible resource. AR is used in traditional Chinese medicine prescriptions to treat chronic nephritis. Calycosin (CA), the primary active compound derived from Astragali Radix, shows significant antifibrotic effects in multiple organs, but the anti-renal fibrosis effect of CA is rarely reported, and the associated mechanism of action is still need to be elucidated. AIM OF THE STUDY The objective of this study was to investigate the protective effects of CA on the kidney against renal fibrosis and the underlying molecular mechanisms. Evaluation of the effects of CA on renal fibrosis using unilateral ureteral obstruction (UUO) mice and transforming growth factor β1 (TGF β1)-induced cell fibrosis. The mechanism of action supporting the investigated anti-renal fibrosis effects were studied by a series of biochemical experiments. RESULTS Our research demonstrated that CA reduced kidney cell fibrosis in mice with UUO and in TGF-β1-stimulated NRK-52E cells. Additionally, CA mitigated renal fibrosis via toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) and had a synergistic effect with resatorvid (TAK-242). Our findings revealed an unobserved impact of CA in inhibiting neutrophil extracellular traps (NETs) formation in UUO mice and neutrophils activated by phorbol 12-myristate 13-acetate. CONCLUSIONS Our findings revealed that calycosin reduces NETs production to alleviate renal fibrosis via TLR4 and NF-κB, supporting its potential as a strategy for treating renal fibrosis.
Collapse
Affiliation(s)
- Wenjie Cao
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Linsheng Huang
- Department of Hepatopancreatobiliary Surgery, Taihe Hospital, Shiyan, 442000, China
| | - Huifan Yu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, 442000, China; Institute of Biomedicine, Hubei University of Medicine, Shiyan, 442000, China
| | - Yongshuai Qian
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Li Liu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Mao Xu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Siyi Li
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Hong Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Fei Li
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, 442000, China.
| |
Collapse
|
12
|
Liu L, Wu P, Wei Y, Lu M, Ge H, Wang P, Sun J, Horng T, Liu X, Shen X, Sun L, Xi Y. TWEAK-Fn14 signaling protects mice from pulmonary fibrosis by inhibiting fibroblast activation and recruiting pro-regenerative macrophages. Cell Rep 2025; 44:115220. [PMID: 39827460 DOI: 10.1016/j.celrep.2024.115220] [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: 04/08/2024] [Revised: 11/11/2024] [Accepted: 12/26/2024] [Indexed: 01/22/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease characterized by excess accumulation of the extracellular matrix (ECM). The role of macrophage-fibroblast crosstalk in lung fibrogenesis is incompletely understood. Here we found that fibroblast growth factor-inducible molecule 14 (Fn14), the receptor for tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is highly induced in myofibroblasts in the lungs of IPF patients and the bleomycin-induced lung fibrosis model. TWEAK-Fn14 signaling inhibits fibroblast activation and ECM synthesis and induces chemokine expression to recruit monocytes/macrophages into the lung. Fn14 deficiency increases ECM production and impairs macrophage infiltration and differentiation, leading to exacerbated lung fibrosis and impaired alveolar regeneration in a bleomycin model. Interestingly, Fn14 deficiency diminishes an injury-induced SiglecF- CD11b- MHCIIlo intermediate macrophage (IntermM) subpopulation, which promotes alveolar type II (AT2) cell proliferation in organoid cultures. These results collectively demonstrate a protective role of TWEAK-Fn14 signaling in lung fibrosis, highlighting the complexities and multilayered regulation of macrophage-fibroblast crosstalk.
Collapse
Affiliation(s)
- Li Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Pei Wu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Yuqi Wei
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Meng Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Haiyan Ge
- Department of Pulmonary and Critical Care Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Ping Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Jianlong Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Tiffany Horng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiucheng Liu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China.
| | - Xiaoyong Shen
- Department of Thoracic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - Ying Xi
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
13
|
Xia J, Dong R, Fang Y, Guo J, Xiong Z, Zhang T, Sun W. A micro-lung chip with macrophages for targeted anti-fibrotic therapy. Biofabrication 2025; 17:025020. [PMID: 39914008 DOI: 10.1088/1758-5090/adb338] [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: 09/10/2024] [Accepted: 02/06/2025] [Indexed: 02/26/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease of unknown etiology. Macrophages are implicated in the fibrotic process, but exhibit remarkable plasticity in the activated immune environmentin vivo, presenting significant challenges as therapeutic targets. To explore the influence of macrophages on IPF and develop macrophage-targeted therapies, we engineered a micro-lung chip with a lung epithelium-interstitium tissue unit to establish a controlled immune environment containing only macrophages. We discovered that macrophages exacerbated inflammation and fibrosis by comparing microchips treated with bleomycin (BLM) in the presence and absence of macrophages. Based on the duration of BLM treatment, we established pathological models corresponding to inflammation and fibrosis stages. Transcriptome analysis revealed that activation of the PI3K-AKT signalling pathway facilitates the transition from inflammation to fibrosis. However, LY294002, a PI3K inhibitor, not only suppressed fibrosis and decreased the accumulation of M2 macrophages but also intensified the severity of inflammation. These findings suggest that macrophages play a pivotal role in the potential development at the tissue level. The micro-lung chip co-cultured with macrophages holds significant potential for exploring the pathological progression of IPF and elucidating the mechanisms of anti-fibrotic drugs.
Collapse
Affiliation(s)
- Jingjing Xia
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
- 'Biomanufacturing and Engineering Living Systems' Innovation International Talents Base (111 Base), Beijing 100084, People's Republic of China
| | - Ruming Dong
- School of Public Health, China Medical University, Shenyang 110122, People's Republic of China
| | - Yongcong Fang
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
- 'Biomanufacturing and Engineering Living Systems' Innovation International Talents Base (111 Base), Beijing 100084, People's Republic of China
| | - Jiabin Guo
- School of Public Health, China Medical University, Shenyang 110122, People's Republic of China
| | - Zhuo Xiong
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
- 'Biomanufacturing and Engineering Living Systems' Innovation International Talents Base (111 Base), Beijing 100084, People's Republic of China
| | - Ting Zhang
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
- 'Biomanufacturing and Engineering Living Systems' Innovation International Talents Base (111 Base), Beijing 100084, People's Republic of China
| | - Wei Sun
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing 100084, People's Republic of China
- 'Biomanufacturing and Engineering Living Systems' Innovation International Talents Base (111 Base), Beijing 100084, People's Republic of China
- Department of Mechanical Engineering, Drexel University, Philadelphia, PA 19104, United States of America
| |
Collapse
|
14
|
Yu J, Li Y, Li Y, Liu X, Huo Q, Wu N, Zhang Y, Zeng T, Zhang Y, Li HY, Lian J, Zhou J, Moses EJ, Geng J, Lin J, Li W, Zhu X. Phosphorylation of FOXN3 by NEK6 promotes pulmonary fibrosis through Smad signaling. Nat Commun 2025; 16:1865. [PMID: 39984467 PMCID: PMC11845461 DOI: 10.1038/s41467-025-56922-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025] Open
Abstract
The transcriptional repressor FOXN3 plays a key role in regulating pulmonary inflammatory responses, which are crucial in the development of pulmonary fibrosis. However, its specific regulatory function in lung fibrosis remains unclear. Here, we show that FOXN3 suppresses pulmonary fibrosis by inhibiting Smad transcriptional activity. FOXN3 targets a substantial number of Smad response gene promoters, facilitating Smad4 ubiquitination, which disrupts the association of the Smad2/3/4 complex with chromatin and abolishes its transcriptional response. In response to pro-fibrotic stimuli, NEK6 phosphorylates FOXN3 at S412 and S416, leading to its degradation. The loss of FOXN3 inhibits β-TrCP-mediated ubiquitination of Smad4, stabilizing the Smad complex's association with its responsive elements and promoting transcriptional activation, thus contributing to the development of pulmonary fibrosis. Notably, we found a significant inverse expression pattern between FOXN3 and Smad4 in clinical pulmonary fibrosis cases, underscoring the importance of the NEK6-FOXN3-Smad axis in the pathological process of pulmonary fibrosis.
Collapse
Affiliation(s)
- Jinjin Yu
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Bengbu Medical University, Bengbu, China
- Regenerative Medicine Sciences Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
- Molecular Diagnosis Center, First Affiliated Hospital, Bengbu Medical University, Bengbu, China
| | - Yingke Li
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Yiming Li
- Research Center of Clinical Laboratory Science, School of Laboratory Medicine, Bengbu Medical University, Bengbu, China
| | - Xiaotian Liu
- Research Center of Clinical Laboratory Science, School of Laboratory Medicine, Bengbu Medical University, Bengbu, China
| | - Qingyang Huo
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Nan Wu
- Molecular Diagnosis Center, First Affiliated Hospital, Bengbu Medical University, Bengbu, China
| | - Yangxia Zhang
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Taoling Zeng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Yong Zhang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Bengbu Medical University, Bengbu, China
| | - Henry You Li
- Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand
| | - Jie Lian
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Jihong Zhou
- Research Center of Clinical Laboratory Science, School of Laboratory Medicine, Bengbu Medical University, Bengbu, China
| | - Emmanuel Jairaj Moses
- Regenerative Medicine Sciences Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia.
| | - Jian Geng
- Research Center of Clinical Laboratory Science, School of Laboratory Medicine, Bengbu Medical University, Bengbu, China.
| | - Juntang Lin
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, China.
| | - Wei Li
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Bengbu Medical University, Bengbu, China.
| | - Xinxing Zhu
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Bengbu Medical University, Bengbu, China.
| |
Collapse
|
15
|
Liu H, Cui H, Liu G. The Intersection between Immune System and Idiopathic Pulmonary Fibrosis-A Concise Review. FIBROSIS (HONG KONG, CHINA) 2025; 3:10004. [PMID: 40124525 PMCID: PMC11928166 DOI: 10.70322/fibrosis.2025.10004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is marked by progressive alveolar destruction, impaired tissue regeneration, and relentless fibrogenesis, culminating in respiratory failure and death. A diverse array of resident and non-resident cells within the lung contribute to disease pathogenesis. Notably, immune cells, both resident and recruited, respond to cues from sites of lung injury by undergoing phenotypic transitions and producing a wide range of mediators that influence, initiate, or dictate the function, or dysfunction, of key effector cells in IPF pathology, such as alveolar epithelial cells, lung fibroblasts, and capillary endothelial cells. The role of the immune system in IPF has undergone an interesting evolution, oscillating from initial enthusiasm to skepticism, and now to a renewed focus. This shift reflects both the past failures of immune-targeting therapies for IPF and the unprecedented insights into immune cell heterogeneity provided by emerging technologies. In this article, we review the historical evolution of perspectives on the immune system's role in IPF pathogenesis and examine the lessons learned from previous therapeutic failures targeting immune responses. We discuss the major immune cell types implicated in IPF progression, highlighting their phenotypic transitions and mechanisms of action. Finally, we identify key knowledge gaps and propose future directions for research on the immune system in IPF.
Collapse
Affiliation(s)
- Hongli Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Huachun Cui
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
16
|
Jin C, Li J, Li Q, Zhang L, Zheng S, Feng Q, Li Y, Zheng Y, Nie Q, Liang J, Wang J. Contribution of cuproptosis and immune-related genes to idiopathic pulmonary fibrosis disease. Front Immunol 2025; 16:1458341. [PMID: 39991151 PMCID: PMC11842377 DOI: 10.3389/fimmu.2025.1458341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/14/2025] [Indexed: 02/25/2025] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a degenerative respiratory condition characterized by significant mortality rates and a scarcity of available treatment alternatives. Cuproptosis, a novel form of copper-induced cell death, has garnered attention for its potential implications. The study aimed to explore the diagnostic value of cuproptosis-related hub genes in patients with IPF. Additionally, multiple bioinformatics analyses were employed to identify immune-related biomarkers associated with the diagnosis of IPF, offering valuable insights for future treatment strategies. Methods Four microarray datasets were selected from the Gene Expression Omnibus (GEO) collection for screening. Differentially expressed genes (DEGs) associated with IPF were analyzed. Additionally, weighted gene coexpression network analysis (WGCNA) was employed to identify the DEGs most associated with IPF. Ultimately, we analyzed five cuproptosis-related hub genes and assessed their diagnostic value for IPF in both the training and validation sets. Additionally, four immune-related hub genes were screened using a protein-protein interaction (PPI) network and evaluated through the receiver operating characteristic (ROC) curve. Lastly, single-cell RNA-seq was employed to further investigate differential gene distribution. Results We identified a total of 92 DEGs. Bioinformatics analysis highlighted five cuproptosis-related genes as candidate biomarkers, including three upregulated genes (CFH, STEAP1, and HDC) and two downregulated genes (NUDT16 and FMO5). The diagnostic accuracy of these five genes in the cohort was confirmed to be reliable. Additionally, we identified four immune-related hub genes that demonstrated strong diagnostic performance for IPF, with CXCL12 showing an AUROC of 0.90. We also examined the relationship between these four genes and immune cells. CXCL12 was significantly negatively associated with neutrophils, while CXCR2 was associated exclusively with neutrophils, consistent with our single-cell sequencing results. CTSG showed a primarily positive association with follicular helper T, and SPP1 was most strongly associated with macrophages. Finally, our single-cell sequencing data revealed that in patients with IPF, CXCL12 was highly expressed in the endothelial cell subset (ECs), while SPP1 exhibited high expression in multiple cellular populations. The expression of the CTSG showed statistically significant differences in monocyte macrophages. Conclusion The research methodically depicted the intricate interplay among five cuproptosis-related genes, four immune-related hub genes, and IPF, offering new ideas for diagnosing and treating patients with IPF.
Collapse
Affiliation(s)
- Chengji Jin
- Department of Respiratory Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Jia Li
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Qiaoyu Li
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Lipeng Zhang
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Shaomao Zheng
- Department of Respiratory Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Qiong Feng
- Department of Respiratory Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Yongjie Li
- Department of Thoracic Surgery, The Second Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Yu Zheng
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Qiuli Nie
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Jin Liang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Jing Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou, China
- National Health Commission (NHC) Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, China
| |
Collapse
|
17
|
Tian L, Song W, Wu J, Lan Y, Chen L. Diagnostic and predictive values of m5C‑associated genes in idiopathic pulmonary fibrosis. Mol Med Rep 2025; 31:53. [PMID: 39704195 DOI: 10.3892/mmr.2024.13418] [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: 06/22/2024] [Accepted: 10/24/2024] [Indexed: 12/21/2024] Open
Abstract
In patients with idiopathic pulmonary fibrosis (IPF), the role of 5‑methylcytosine (m5C)‑associated genes in the pathogenesis and development of the disease remains unclear. The present study aimed to identify reliable diagnostic markers based on the expression of m5C‑associated genes for the early detection of IPF. Count data were obtained by screening the IPF genome‑wide assay in the Gene Expression Omnibus database, followed by a comparison of m5C gene expression in patients with IPF and controls. The GSE150910 and GSE173355 datasets yielded a total of 23 differentially expressed m5C‑associated genes, which were then investigated for their functions. A diagnostic model was built using eight m5C genes and validated with training sets and the GSE124685 dataset. IPF subtypes were identified based on expression of m5C‑related genes as well as clinical and immunological characteristics. Furthermore, a pulmonary fibrosis model was established in mice by administering bleomycin into the trachea. Lungs were harvested and analyzed using quantitative PCR to determine the expression of m5C‑related genes. The Kyoto Encyclopedia of Genes and Genomes enrichment analysis revealed that these genes were significantly enriched in 'base excision repair'. Immunoassay results revealed that 13 immune cell markers (naive, memory and B cell plasma, T cell CD4 naive, T cell CD4 memory resting, T cell follicular helper, T cell regulatory Tregs, NK cell resting, Monocyte, Macrophage M0, Mast cell activated, Eosinophil, and Neutrophil) were significantly associated with IPF. Patients with IPF had lower levels of resting memory CD4+ T cells, which were positively associated with Tet methylcytosine dioxygenase2 (TET2) and Thymine‑DNA glycosylase (TDG) but negatively correlated with NOP2/Sun RNA methyltransferase5 (NSUN5) expression. All samples were classified into based on the levels of the eight diagnostic m5C genes. Samples with high m5C scores are subtype 1, and those with low m5C scores are subtype 2. In subtype 2, male patients had lower levels of CD27 and CD70 but higher levels of CD274, CD86, Cytotoxic T‑lymphocyte‑associated protein4 and Hepatitis A virus cellular receptor2 (HAVCR2). When compared with normal mouse lung tissue samples, expression levels of NOP2/Sun RNA methyltransferase6 (NSUN6), Ubiquitin‑like with PHD and RING Finger Domains1, TDG and TET2 in lung fibrosis tissue samples were significantly higher, while expression levels of NSUN5, NTH‑like DNA glycosylase1, DNA (cytosine‑5‑)‑methyltransferase3 β and Methyl‑CpG binding domain protein 3) were lower. It is possible that m5C‑associated genes play an important role in the diagnosis and typing of IPF. These genes may facilitate investigation of the pathophysiology of IPF and identification of potential treatment targets.
Collapse
Affiliation(s)
- Lan Tian
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Wanting Song
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Jiabao Wu
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Yi Lan
- Department of General Medicine, Nanping First Hospital Affiliated to Fujian Medical University, Nanping, Fujian 353000, P.R. China
| | - Limin Chen
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
18
|
Zhang T, Hou Z, Ding Z, Wang P, Pan X, Li X. Single Cell RNA-Seq Identifies Cell Subpopulations Contributing to Idiopathic Pulmonary Fibrosis in Humans. J Cell Mol Med 2025; 29:e70402. [PMID: 39928535 PMCID: PMC11809556 DOI: 10.1111/jcmm.70402] [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: 10/31/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/12/2025] Open
Abstract
The cell populations, particularly subpopulations, involved in the onset and progression of idiopathic pulmonary fibrosis (IPF) remain incompletely understood. This study employed single-cell RNA-seq to identify cell populations and subpopulations with significantly altered proportions in the lungs of patients with IPF. In IPF lungs, endothelial cell proportions were significantly increased, while alveolar epithelial cell proportions were markedly decreased. Among the three identified fibroblast subpopulations, the proportion of myofibroblasts was significantly increased, while the proportions of the other two fibroblast subtypes were reduced. Similarly, within the three macrophage subpopulations, the macrophage_SPP1 subpopulation, localised to fibroblastic foci, showed a significant increase in proportion, while the alveolar macrophage subpopulation was significantly reduced. Trajectory analysis revealed that fibroblasts in IPF lungs could differentiate into myofibroblasts, and alveolar macrophages could transition into the macrophage_SPP1 subpopulation. Among T-cell subpopulations, only the CD4 T_FOXP3 subpopulation exhibited a significant change, whereas all four B-cell subpopulations showed significant proportional shifts. These findings provide a comprehensive view of the cellular alterations contributing to IPF pathogenesis. Extensive interactions among various cell populations and subpopulations were identified. The proportions of various cell populations and subpopulations in IPF lungs, including endothelial cells, fibroblasts, macrophages and B cells, were significantly altered. Further in-depth investigation into the roles of cell subpopulations with significantly altered proportions in the onset and progression of IPF will provide valuable insights into the pathological mechanisms underlying the disease. This understanding could facilitate the development of novel therapeutic strategies and medications for IPF treatment.
Collapse
Affiliation(s)
- Tangjuan Zhang
- Department of EmergencyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhichao Hou
- Department of Thoracic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zheng Ding
- Department of Thoracic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Peng Wang
- School of Nursing and HealthZhengzhou UniversityZhengzhouChina
| | - Xue Pan
- School of Nursing and HealthZhengzhou UniversityZhengzhouChina
| | - Xiangnan Li
- Department of Thoracic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
19
|
Chang SY, Chang WH, Yang DC, Hong QS, Hsu SW, Wu R, Chen CH. Autologous precision-cut lung slice co-culture models for studying macrophage-driven fibrosis. Front Physiol 2025; 16:1526787. [PMID: 39958688 PMCID: PMC11825446 DOI: 10.3389/fphys.2025.1526787] [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: 11/12/2024] [Accepted: 01/15/2025] [Indexed: 02/18/2025] Open
Abstract
Precision-cut lung slices (PCLS) are commonly used as an ex vivo model to study lung fibrosis; however, traditional models lack immune cell infiltration, including the recruitment of monocytes and macrophages, which are critical for inflammation and fibrosis. To address this limitation, we developed novel autologous PCLS-immune co-culture models that better replicate the processes of inflammation, repair, and immune cell recruitment associated with fibrosis. Fibrotic responses to nicotine, cigarette smoke extract (CSE), and a fibrosis-inducing cocktail (FC) were first evaluated in PCLS containing only tissue-resident macrophages, with upregulation of α-SMA-expressing fibroblasts confirmed by immunofluorescence and Western blotting, and collagen deposition quantified using Sirius Red staining. To study macrophage recruitment, we employed an indirect co-culture model using transwells to approximate blood vessel function. Chemotactic studies revealed increased migration of autologous bone marrow-derived macrophages (BMDMs) toward and infiltration into CSE-injured PCLS. In a direct co-culture model simulating the repair phase of fibrosis, PCLS exposed to CSE and FC showed further increased collagen deposition in the presence of autologous BMDMs, but not heterologous ones. These findings suggest that our novel PCLS-immune co-culture models provide a platform for studying macrophage involvement in fibrosis and offer potential for developing macrophage-targeted therapeutic strategies in pulmonary fibrosis.
Collapse
Affiliation(s)
- So-Yi Chang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| | - Wen-Hsin Chang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| | - David C. Yang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| | - Qi-Sheng Hong
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| | - Ssu-Wei Hsu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| | - Reen Wu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| | - Ching-Hsien Chen
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, United States
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, United States
| |
Collapse
|
20
|
Sung EA, Dozmorov MG, Song S, Aung T, Park MH, Sime PJ, Chae WJ. Ablation of LRP6 in alpha-smooth muscle actin-expressing cells abrogates lung inflammation and fibrosis upon bleomycin-induced lung injury. FEBS Lett 2025. [PMID: 39873304 DOI: 10.1002/1873-3468.15106] [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: 09/16/2024] [Revised: 12/20/2024] [Accepted: 01/05/2025] [Indexed: 01/30/2025]
Abstract
Tissue fibrosis is a progressive pathological process with excessive deposition of extracellular matrix proteins (ECM). Myofibroblasts, identified by alpha-smooth muscle actin (αSMA) expression, play an important role in tissue fibrosis by producing ECM. Here, we found that the Wnt antagonist Dickkopf1 (DKK1) induced gene expressions associated with inflammation and fibrosis in lung fibroblasts. We demonstrated that genetic deletion of LRP6, a receptor for Wnt ligands and DKK1, in αSMA-expressing cells using Acta2-cre Lrp6fl/fl (Lrp6AKO) mice abrogated the bleomycin (BLM)-induced lung inflammation and fibrosis phenotype, suggesting an important role for LRP6 in modulating inflammation and fibrotic processes in the lung. Our results highlight the crucial role of LRP6 in fibroblasts in regulating inflammation and fibrosis upon BLM-induced lung injury.
Collapse
Affiliation(s)
- Eun-Ah Sung
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Mikhail G Dozmorov
- Department of Biostatistics, Department of Pathology, Virginia Commonwealth University, Richmond, VA, USA
| | - SuJeong Song
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Theingi Aung
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Min Hee Park
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Patricia J Sime
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Wook-Jin Chae
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
- Phillips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University School of Dentistry, Richmond, VA, USA
| |
Collapse
|
21
|
Ding C, Liao Q, Zuo R, Zhang S, Guo Z, He J, Ye Z, Chen W, Ke S. Machine learning potential predictor of idiopathic pulmonary fibrosis. Front Genet 2025; 15:1464471. [PMID: 39935693 PMCID: PMC11811625 DOI: 10.3389/fgene.2024.1464471] [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: 07/24/2024] [Accepted: 12/26/2024] [Indexed: 02/13/2025] Open
Abstract
Introduction Idiopathic pulmonary fibrosis (IPF) is a severe chronic respiratory disease characterized by treatment challenges and poor prognosis. Identifying relevant biomarkers for effective early-stage risk prediction is therefore of critical importance. Methods In this study, we obtained gene expression profiles and corresponding clinical data of IPF patients from the GEO database. GO enrichment and KEGG pathway analyses were performed using R software. To construct an IPF risk prediction model, we employed LASSO-Cox regression analysis and the SVM-RFE algorithm. PODNL1 and PIGA were identified as potential biomarkers associated with IPF onset, and their predictive accuracy was confirmed using ROC curve analysis in the test set. Furthermore, GSEA revealed enrichment in multiple pathways, while immune function analysis demonstrated a significant correlation between IPF onset and immune cell infiltration. Finally, the roles of PODNL1 and PIGA as biomarkers were validated through in vivo and in vitro experiments using qRT-PCR, Western blotting, and immunohistochemistry. Results These findings suggest that PODNL1 and PIGA may serve as critical biomarkers for IPF onset and contribute to its pathogenesis. Discussion This study highlights their potential for early biomarker discovery and risk prediction in IPF, offering insights into disease mechanisms and diagnostic strategies.
Collapse
Affiliation(s)
- Chenchun Ding
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Quan Liao
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Renjie Zuo
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shichao Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhenzhen Guo
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Junjie He
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Ziwei Ye
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Weibin Chen
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Sunkui Ke
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
22
|
Maciejewska A, Czernia P, Piotrowska-Mieczkowska M, Wajda B, Słomiński B, Romantowski J, Sudoł A, Dąbrowska M, Górska L, Smiatacz T, Niedoszytko M, Jassem E, Skrzypkowska M, Trzonkowski P. Comprehensive analyses of immune activity in COVID-19-vaccinated idiopathic pulmonary fibrosis patients. Front Immunol 2025; 15:1436491. [PMID: 39845961 PMCID: PMC11750670 DOI: 10.3389/fimmu.2024.1436491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal disease, characterized by impaired wound repair, tissue remodeling and fibrosis. Immune system may participate in the development and progression of the disease as indicated by altered activity in IPF sufferers. This study investigates the immune response to the BNT162b2 COVID-19 vaccine in patients with IPF compared to healthy controls, with a particular focus on evaluation of antibody responses, interferon-gamma release, cytokine profiling and a broad panel of immune cell subpopulations. IPF patients without prior exposure to SARS-CoV-2 had undetectable levels of anti-N IgG antibodies, highlighting their lack of previous infection. After vaccination, IPF patients showed a significant increase in anti-S1 IgG and IgA antibodies, though their levels were lower compared to healthy controls and convalescent IPF patients. Additionally, IPF patients exhibited altered proportions of regulatory T cells (Tregs) and effector T lymphocytes (Teffs) before and after vaccination. Specifically, IPF patients had higher percentages of Tregs with a Th2 phenotype and Th17 Tregs, along with reduced proportions of Th1/17 Tregs. Teffs in IPF patients showed a decrease in Th1-like and Th2-like populations after vaccination. Moreover, IPF patients demonstrated elevated populations of cytotoxic T lymphocytes (Tc) before vaccination and increased levels of γδ Tc cells throughout the study. Alterations in cytokine profiles were also observed, IPF patients showed higher levels of IL-6 and IL-22 compared to healthy controls. These findings suggest a distinct immune response in IPF patients to the COVID-19 vaccine, characterized by differences in antibody production, T cell differentiation and cytokine secretion compared to healthy individuals.
Collapse
Affiliation(s)
- Agata Maciejewska
- Department of Medical Immunology, Medical University of Gdansk, Gdansk, Poland
| | - Piotr Czernia
- Department of Pneumonology, Medical University of Gdansk, Gdansk, Poland
| | | | - Beata Wajda
- Department of Pneumonology, Medical University of Gdansk, Gdansk, Poland
| | - Bartosz Słomiński
- Department of Medical Immunology, Medical University of Gdansk, Gdansk, Poland
| | - Jan Romantowski
- Department of Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Adam Sudoł
- Central Clinical Laboratory, University Clinical Centre, Gdansk, Poland
| | | | - Lucyna Górska
- Department of Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Tomasz Smiatacz
- Department of Infectious Diseases, Medical University of Gdansk, Gdansk, Poland
| | - Marek Niedoszytko
- Department of Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Ewa Jassem
- Department of Pneumonology, Medical University of Gdansk, Gdansk, Poland
| | - Maria Skrzypkowska
- Department of Medical Immunology, Medical University of Gdansk, Gdansk, Poland
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
23
|
Fischer A, Han W, Hu S, Mück-Häusl M, Wannemacher J, Kadri S, Lin Y, Dai R, Christ S, Su Y, Dasgupta B, Sardogan A, Deisenhofer C, Dutta S, Kadri A, Güney TG, Correa-Gallegos D, Mayr CH, Hatz R, Stoleriu MG, Lindner M, Hilgendorff A, Adler H, Machens HG, Schiller HB, Hauck SM, Rinkevich Y. Targeting pleuro-alveolar junctions reverses lung fibrosis in mice. Nat Commun 2025; 16:173. [PMID: 39747171 PMCID: PMC11696612 DOI: 10.1038/s41467-024-55596-x] [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: 11/07/2023] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Lung fibrosis development utilizes alveolar macrophages, with mechanisms that are incompletely understood. Here, we fate map connective tissue during mouse lung fibrosis and observe disassembly and transfer of connective tissue macromolecules from pleuro-alveolar junctions (PAJs) into deep lung tissue, to activate fibroblasts and fibrosis. Disassembly and transfer of PAJ macromolecules into deep lung tissue occurs by alveolar macrophages, activating cysteine-type proteolysis on pleural mesothelium. The PAJ niche and the disassembly cascade is active in patient lung biopsies, persists in chronic fibrosis models, and wanes down in acute fibrosis models. Pleural-specific viral therapeutic carrying the cysteine protease inhibitor Cystatin A shuts down PAJ disassembly, reverses fibrosis and regenerates chronic fibrotic lungs. Targeting PAJ disassembly by targeting the pleura may provide a unique therapeutic avenue to treat lung fibrotic diseases.
Collapse
Affiliation(s)
- Adrian Fischer
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
| | - Wei Han
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany.
- Member of the German Center of Lung Research (DZL), Munich, Germany.
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany.
| | - Shaoping Hu
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
- Zhangzhou Health Vocational College, Zhangzhou, China
| | - Martin Mück-Häusl
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Helmholtz Munich, Research Unit for Precision Regenerative Medicine (PRM), Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Juliane Wannemacher
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Safwen Kadri
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Helmholtz Munich, Research Unit for Precision Regenerative Medicine (PRM), Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Yue Lin
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Ruoxuan Dai
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Simon Christ
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Yiqun Su
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Bikram Dasgupta
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Aydan Sardogan
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Christoph Deisenhofer
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Subhasree Dutta
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Amal Kadri
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Tankut Gökhan Güney
- Institute of Regenerative Biology and Medicine(IRBM), Helmholtz Zentrum München, Munich, Germany
- Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Donovan Correa-Gallegos
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Christoph H Mayr
- Helmholtz Munich, Research Unit for Precision Regenerative Medicine (PRM), Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Rudolf Hatz
- Asklepios Fachkliniken in Munich-Gauting, Munich, Germany
| | | | - Michael Lindner
- Asklepios Fachkliniken in Munich-Gauting, Munich, Germany
- University Department of Visceral and Thoracic Surgery Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Anne Hilgendorff
- Helmholtz Zentrum München, Institute of Lung Biology & Disease, Group Mechanism of Neonatal Chronic Lung Disease, Member of the German Center of Lung Research (DZL), Munich, Germany
- Comprehensive Pneumology Center with the CPC-M bioArchive and Institute of Lung Health and Immunity, Helmholtz-Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Heiko Adler
- Member of the German Center of Lung Research (DZL), Munich, Germany
- Institute of Asthma and Allergy Prevention, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Hans-Günther Machens
- Department of Plastic and Hand Surgery, Technical University of Munich, School of Medicine and Health, Klinikum rechts der Isar, Munich, Germany
| | - Herbert B Schiller
- Helmholtz Munich, Research Unit for Precision Regenerative Medicine (PRM), Member of the German Center for Lung Research (DZL), Munich, Germany
- Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Munich, Germany
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Chinese Institutes for Medical Research, Beijing, China.
- Capital Medical University, Beijing, China.
| |
Collapse
|
24
|
Spagnolo P, Tonelli R, Mura M, Reisman W, Sotiropoulou V, Tzouvelekis A. Investigational gene expression inhibitors for the treatment of idiopathic pulmonary fibrosis. Expert Opin Investig Drugs 2025; 34:61-80. [PMID: 39916340 DOI: 10.1080/13543784.2025.2462592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 01/31/2025] [Indexed: 02/12/2025]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive fibrosing interstitial lung disease of unknown cause that occurs primarily in older adults and is associated with poor quality of life and substantial healthcare utilization. IPF has a dismal prognosis. Indeed, first-line therapy, which includes nintedanib and pirfenidone, does not stop disease progression and is often associated with tolerability issues. Therefore, there remains a high medical need for more efficacious and better tolerated treatments. AREAS COVERED Gene therapy is a relatively unexplored field of research in IPF that has the potential to mitigate a range of profibrotic pathways by introducing genetic material into cells. Here, we summarize and critically discuss publications that have explored the safety and efficacy of gene therapy in experimentally-induced pulmonary fibrosis in animals, as clinical studies in humans have not been published yet. EXPERT OPINION The application of gene therapy in pulmonary fibrosis requires further investigation to address several technical and biological hurdles, improve vectors' design, drug delivery, and target selection, mitigate off-target effects and develop markers of gene penetration into target cells. Long-term clinical data are needed to bring gene therapy in IPF one step closer to practice.
Collapse
Affiliation(s)
- Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Roberto Tonelli
- Respiratory Disease Unit, Department of Medical and Surgical Sciences, University Hospital of Modena and Reggio Emilia, Modena, Italy
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Modena, Italy
| | - Marco Mura
- Division of Respirology, Western University, London, Ontario, Canada
| | - William Reisman
- Division of Respirology, Western University, London, Ontario, Canada
| | | | - Argyrios Tzouvelekis
- Department of Respiratory Medicine, University Hospital of Patras, Patras, Greece
| |
Collapse
|
25
|
Chen L, Hou T, Ge F, Jiang H, Liu F, Tian J, Zheng M. Idiopathic Pulmonary Fibrosis Is Associated With Type 1 Diabetes: A Two-Sample Mendelian Randomization Study. J Gene Med 2025; 27:e70008. [PMID: 39822044 DOI: 10.1002/jgm.70008] [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: 08/12/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND The pathogenesis of idiopathic pulmonary fibrosis (IPF) remains unclear; previous studies revealed the underlying connection between IPF and diabetes, but there is no consensual opinion. This study is aimed at examining the association between Type 1 diabetes (T1D) and IPF using Mendelian randomization (MR). METHOD In our two-sample MR study, we selected single nucleotide polymorphisms (SNPs) that are strongly associated with T1D in a genome-wide association study (GWAS) from IEU (dataset: ebi-a-GCST005536) and obtained their corresponding effect estimates on T1D risk in an IPF GWAS from IEU (dataset: finn-b-IPF). We conducted a multivariable Mendelian randomization (MVMR) analysis to eliminate the interference of aging. RESULT In the outcome of inverse-variance weighted (IVW) method, T1D showed a promoting effect on IPF (odds ratio (OR): 1.132, p = 0.005). The statistics passed the MR-PRESSO test, and no outliers were observed (global test p = 0.238). MVMR study was performed, and the aging-adjusted result remains almost the same (OR = 1.132, OR_95% CI: 1.034-1.239, p = 0.007). CONCLUSION Our study shows a causal relation between T1D and IPF; further investigation should be conducted.
Collapse
Affiliation(s)
- Leyan Chen
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Tianzhichao Hou
- Department of Medical Biophysics, Princess Margaret Cancer Centre-University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Feifan Ge
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Huachi Jiang
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Feng Liu
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Jingyan Tian
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingfeng Zheng
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| |
Collapse
|
26
|
Zhao T, Zhou ZR, Wan HQ, Feng T, Hu XH, Li XQ, Zhao SM, Li HL, Hou JW, Li W, Lu DY, Qian MY, Shen X. Otilonium bromide ameliorates pulmonary fibrosis in mice through activating phosphatase PPM1A. Acta Pharmacol Sin 2025; 46:107-121. [PMID: 39160244 PMCID: PMC11695943 DOI: 10.1038/s41401-024-01368-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/24/2024] [Indexed: 08/21/2024]
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive and irreversible interstitial lung disease characterized by unremitting pulmonary myofibroblasts activation, extracellular matrix (ECM) deposition and inflammatory recruitment. PF has no curable medication yet. In this study we investigated the molecular pathogenesis and potential therapeutic targets of PF and discovered drug lead compounds for PF therapy. A murine PF model was established in mice by intratracheal instillation of bleomycin (BLM, 5 mg/kg). We showed that the protein level of pulmonary protein phosphatase magnesium-dependent 1A (PPM1A, also known as PP2Cα) was significantly downregulated in PF patients and BLM-induced PF mice. We demonstrated that TRIM47 promoted ubiquitination and decreased PPM1A protein in PF progression. By screening the lab in-house compound library, we discovered otilonium bromide (OB, clinically used for treating irritable bowel syndrome) as a PPM1A enzymatic activator with an EC50 value of 4.23 μM. Treatment with OB (2.5, 5 mg·kg-1·d-1, i.p., for 20 days) significantly ameliorated PF-like pathology in mice. We constructed PF mice with PPM1A-specific knockdown in the lung tissues, and determined that by targeting PPM1A, OB treatment suppressed ECM deposition through TGF-β/SMAD3 pathway in fibroblasts, repressed inflammatory responses through NF-κB/NLRP3 pathway in alveolar epithelial cells, and blunted the crosstalk between inflammation in alveolar epithelial cells and ECM deposition in fibroblasts. Together, our results demonstrate that pulmonary PPM1A activation is a promising therapeutic strategy for PF and highlighted the potential of OB in the treatment of the disease.
Collapse
Affiliation(s)
- Tong Zhao
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhi-Ruo Zhou
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Hui-Qi Wan
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tian Feng
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xu-Hui Hu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiao-Qian Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shi-Mei Zhao
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hong-Lin Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ji-Wei Hou
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, China
| | - Wei Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Da-Yun Lu
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Min-Yi Qian
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xu Shen
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
27
|
Svobodová B, Löfdahl A, Nybom A, Wigén J, Hirdman G, Olm F, Brunnström H, Lindstedt S, Westergren-Thorsson G, Elowsson L. Overlapping Systemic Proteins in COVID-19 and Lung Fibrosis Associated with Tissue Remodeling and Inflammation. Biomedicines 2024; 12:2893. [PMID: 39767799 PMCID: PMC11727205 DOI: 10.3390/biomedicines12122893] [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: 11/14/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Background/Objectives: A novel patient group with chronic pulmonary fibrosis is emerging post COVID-19. To identify patients at risk of developing post-COVID-19 lung fibrosis, we here aimed to identify systemic proteins that overlap with fibrotic markers identified in patients with idiopathic pulmonary fibrosis (IPF) and may predict COVID-19-induced lung fibrosis. Methods: Ninety-two proteins were measured in plasma samples from hospitalized patients with moderate and severe COVID-19 in Sweden, before the introduction of the vaccination program, as well as from healthy individuals. These measurements were conducted using proximity extension assay (PEA) technology with a panel including inflammatory and remodeling proteins. Histopathological alterations were evaluated in explanted lung tissue. Results: Connecting to IPF pathology, several proteins including decorin (DCN), tumor necrosis factor receptor superfamily member 12A (TNFRSF12A) and chemokine (C-X-C motif) ligand 13 (CXCL13) were elevated in COVID-19 patients compared to healthy subjects. Moreover, we found incrementing expression of monocyte chemotactic protein-3 (MCP-3) and hepatocyte growth factor (HGF) when comparing moderate to severe COVID-19. Conclusions: Both extracellular matrix- and inflammation-associated proteins were identified as overlapping with pulmonary fibrosis, where we found DCN, TNFRSF12A, CXCL13, CXCL9, MCP-3 and HGF to be of particular interest to follow up on for the prediction of disease severity.
Collapse
Affiliation(s)
- Barbora Svobodová
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden; (B.S.); (J.W.); (G.W.-T.)
| | - Anna Löfdahl
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden; (B.S.); (J.W.); (G.W.-T.)
| | - Annika Nybom
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden; (B.S.); (J.W.); (G.W.-T.)
| | - Jenny Wigén
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden; (B.S.); (J.W.); (G.W.-T.)
| | - Gabriel Hirdman
- Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, 222 42 Lund, Sweden; (G.H.); (F.O.); (S.L.)
- Wallenberg Center for Molecular Medicine, Lund University, 221 84 Lund, Sweden
- Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden;
| | - Franziska Olm
- Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, 222 42 Lund, Sweden; (G.H.); (F.O.); (S.L.)
- Wallenberg Center for Molecular Medicine, Lund University, 221 84 Lund, Sweden
- Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden;
| | - Hans Brunnström
- Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden;
| | - Sandra Lindstedt
- Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, 222 42 Lund, Sweden; (G.H.); (F.O.); (S.L.)
- Wallenberg Center for Molecular Medicine, Lund University, 221 84 Lund, Sweden
- Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden;
| | - Gunilla Westergren-Thorsson
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden; (B.S.); (J.W.); (G.W.-T.)
| | - Linda Elowsson
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden; (B.S.); (J.W.); (G.W.-T.)
| |
Collapse
|
28
|
Yang J, Lu D, Sun Y, Qiu M, Zhao T, Yan B, Wang S, Shao Z, Wang D, Li T, Xiao Q, Fu T. Cell Membrane Hybrid Liposome-Targeted Delivery of the Heat Shock Protein 90 C-Terminal Inhibitor for the Treatment of Idiopathic Pulmonary Fibrosis. ACS Pharmacol Transl Sci 2024; 7:4083-4095. [PMID: 39698274 PMCID: PMC11651165 DOI: 10.1021/acsptsci.4c00524] [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: 08/30/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 12/20/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) represents a grave challenge as it is characterized by high fatality rates and irreversible progression without effective clinical interventions available at present. Previous studies have demonstrated that inhibition of heat shock protein 90 (HSP90) by an N-terminal inhibitor disrupts its interaction with TGFβRII, leading to the instability of TGFβRII, thus blocking the role of transforming growth factor-β1 (TGF-β1), which could potentially ameliorate IPF symptoms. However, given that the broad spectrum of HSP90 N-terminal inhibitors may lead to unanticipated side effects, we hypothesize that C-terminal inhibitors of HSP90 can interfere with TGFβRII while minimizing adverse reactions. In this study, silybin, a C-terminal inhibitor of HSP90, was separated into monomers, and silybin A was screened for its superior efficacy against TGFβRII. To facilitate targeted therapy for treating IPF, a cell membrane hybrid liposome loaded with silybin A (Cm-A-Lip) was developed to deliver silybin A to lung fibroblasts through pulmonary drug delivery. A bleomycin-induced IPF mouse model was used to evaluate the efficacy of Cm-A-Lip. By examination of lung hydroxyproline content, wet weight, histology, and inflammatory factor expression, the results showed that pulmonary delivery of Cm-A-Lip could increase the drug retention time in lung tissue compared with intravenous injection. Furthermore, Cm-A-Lip exhibited superior antifibrotic activity relative to conventional liposmomes loaded with silybin A (A-Lip) while concurrently mitigating systemic inflammatory responses associated with silybin A administration, thus enhancing the overall safety profile.
Collapse
Affiliation(s)
| | | | - Yuping Sun
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengmeng Qiu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tianlong Zhao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Baofei Yan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Siting Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhitao Shao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Demei Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ting Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qingqing Xiao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tingming Fu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
29
|
Russo RC, Ryffel B. The Chemokine System as a Key Regulator of Pulmonary Fibrosis: Converging Pathways in Human Idiopathic Pulmonary Fibrosis (IPF) and the Bleomycin-Induced Lung Fibrosis Model in Mice. Cells 2024; 13:2058. [PMID: 39768150 PMCID: PMC11674266 DOI: 10.3390/cells13242058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and lethal interstitial lung disease (ILD) of unknown origin, characterized by limited treatment efficacy and a fibroproliferative nature. It is marked by excessive extracellular matrix deposition in the pulmonary parenchyma, leading to progressive lung volume decline and impaired gas exchange. The chemokine system, a network of proteins involved in cellular communication with diverse biological functions, plays a crucial role in various respiratory diseases. Chemokine receptors trigger the activation, proliferation, and migration of lung-resident cells, including pneumocytes, endothelial cells, alveolar macrophages, and fibroblasts. Around 50 chemokines can potentially interact with 20 receptors, expressed by both leukocytes and non-leukocytes such as tissue parenchyma cells, contributing to processes such as leukocyte mobilization from the bone marrow, recirculation through lymphoid organs, and tissue influx during inflammation or immune response. This narrative review explores the complexity of the chemokine system in the context of IPF and the bleomycin-induced lung fibrosis mouse model. The goal is to identify specific chemokines and receptors as potential therapeutic targets. Recent progress in understanding the role of the chemokine system during IPF, using experimental models and molecular diagnosis, underscores the complex nature of this system in the context of the disease. Despite advances in experimental models and molecular diagnostics, discovering an effective therapy for IPF remains a significant challenge in both medicine and pharmacology. This work delves into microarray results from lung samples of IPF patients and murine samples at different stages of bleomycin-induced pulmonary fibrosis. By discussing common pathways identified in both IPF and the experimental model, we aim to shed light on potential targets for therapeutic intervention. Dysregulation caused by abnormal chemokine levels observed in IPF lungs may activate multiple targets, suggesting that chemokine signaling plays a central role in maintaining or perpetuating lung fibrogenesis. The highlighted chemokine axes (CCL8-CCR2, CCL19/CCL21-CCR7, CXCL9-CXCR3, CCL3/CCL4/CCL5-CCR5, and CCL20-CCR6) present promising opportunities for advancing IPF treatment research and uncovering new pharmacological targets within the chemokine system.
Collapse
Affiliation(s)
- Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais-UFMG, Belo Horizonte 31270-901, MG, Brazil
| | - Bernhard Ryffel
- Laboratory of Immuno-Neuro Modulation (INEM), UMR7355 Centre National de la Recherche Scientifique (CNRS), University of Orleans, 45071 Orleans, France
| |
Collapse
|
30
|
Liu Y, Wang D, Liu X, Yuan H, Liu D, Hu Y, Ning S. Biological and pharmacological roles of pyroptosis in pulmonary inflammation and fibrosis: recent advances and future directions. Cell Commun Signal 2024; 22:586. [PMID: 39639365 PMCID: PMC11619304 DOI: 10.1186/s12964-024-01966-3] [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: 09/08/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Pyroptosis, an inflammatory regulated cell death (RCD) mechanism, is characterized by cellular swelling, membrane rupture, and subsequent discharge of cellular contents, exerting robust proinflammatory effects. Recent studies have significantly advanced our understanding of pyroptosis, revealing that it can be triggered through inflammasome- and caspase-independent pathways, and interacts intricately with other RCD pathways (e.g., pyroptosis, necroptosis, ferroptosis, and cuproptosis). The pathogenesis of pulmonary fibrosis (PF), including idiopathic pulmonary fibrosis (IPF) and other interstitial lung diseases, involves a multifaceted interplay of factors such as pathogen infections, environmental pollutants, genetic variations, and immune dysfunction. This chronic and progressive interstitial lung disease is characterized by persistent inflammation, extracellular matrix (ECM) accumulation, and fibrotic alveolar wall thickening, which potentially contribute to deteriorated lung function. Despite recent advances in understanding pyroptosis, the mechanisms by which it regulates PF are not entirely elucidated, and effective strategies to improve clinical outcomes remain unclear. This review strives to deliver a comprehensive overview of the biological functions and molecular mechanisms of pyroptosis, exploring its roles in the pathogenesis of PF. Furthermore, it examines potential biomarkers and therapeutic agents for anti-fibrotic treatments.
Collapse
Affiliation(s)
- Ya Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, 411100, China
| | - Danxia Wang
- Department of Pharmacy, People's Hospital of Ningxiang City, Hunan University of Chinese Medicine, Changsha, 410600, China
| | - Xiang Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, 411100, China
| | - Haibin Yuan
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Dan Liu
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Yixiang Hu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated 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
|
31
|
Morikka J, Federico A, Möbus L, Inkala S, Pavel A, Sani S, Vaani M, Peltola S, Serra A, Greco D. Toxicogenomic assessment of in vitro macrophages exposed to profibrotic challenge reveals a sustained transcriptomic immune signature. Comput Struct Biotechnol J 2024; 25:194-204. [PMID: 39430886 PMCID: PMC11490883 DOI: 10.1016/j.csbj.2024.10.010] [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: 06/20/2024] [Revised: 09/27/2024] [Accepted: 10/05/2024] [Indexed: 10/22/2024] Open
Abstract
Immune signalling is a crucial component in the progression of fibrosis. However, approaches for the safety assessment of potentially profibrotic substances, that provide information on mechanistic immune responses, are underdeveloped. This study aimed to develop a novel framework for assessing the immunotoxicity of fibrotic compounds. We exposed macrophages in vitro to multiple sublethal concentrations of the profibrotic agent bleomycin, over multiple timepoints, and generated RNA sequencing data. Using a toxicogenomic approach, we performed dose-dependent analysis to discover genes dysregulated by bleomycin exposure in a dose-responsive manner. A subset of immune genes displayed a sustained dose-dependent and differential expression response to profibrotic challenge. An immunoassay revealed cytokines and proteinases responding to bleomycin exposure that closely correlated to transcriptomic alterations, underscoring the integration between transcriptional immune response and external immune signalling activity. This study not only increases our understanding of the immunological mechanisms of fibrosis, but also offers an innovative framework for the toxicological evaluation of substances with potential fibrogenic effects on macrophage signalling. Our work brings a new immunotoxicogenomic direction for hazard assessment of fibrotic compounds, through the implementation of a time and resource efficient in vitro methodology.
Collapse
Affiliation(s)
- Jack Morikka
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
| | - Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Lena Möbus
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Simo Inkala
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Alisa Pavel
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Copenhagen, Denmark
| | - Saara Sani
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Maaret Vaani
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sanna Peltola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
32
|
Weng C, Zhao Y, Song M, Shao Z, Pang Y, Yu C, Pei P, Yang L, Millwood IY, Walters RG, Chen Y, Du H, Chen J, Chen Z, Genovese G, Terao C, Lv J, Li L, Sun D. Mosaic loss of chromosome Y, tobacco smoking and risk of age-related lung diseases: insights from two prospective cohorts. Eur Respir J 2024; 64:2400968. [PMID: 39401857 DOI: 10.1183/13993003.00968-2024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/11/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Little is known about the underlying relationship between mosaic loss of chromosome Y (mLOY), the most common chromosomal alterations in older men, and the risk of age-related lung diseases. METHODS We included 217 780 participants from the UK Biobank (UKB) and 42 859 participants from the China Kadoorie Biobank. The mLOY events were detected using the Mosaic Chromosomal Alterations (MoChA) pipeline. Outcomes included all lung diseases, COPD, lung cancer and idiopathic pulmonary fibrosis (IPF). Cox proportional hazard models were fitted to estimate the hazard ratios and 95% confidence intervals of mLOY with lung diseases in both cohorts. The combined hazard ratios were derived from meta-analysis. RESULTS Results from the two cohorts showed that expanded mLOY was associated with increased risks of all lung diseases (HR 1.19 (95% CI 1.04-1.36)), COPD (HR 1.20 (95% CI 1.13-1.28)), lung cancer (HR 1.34 (95% CI 1.21-1.48)) and IPF (HR 1.34 (95% CI 1.16-1.56) in the UKB). There was evidence of positive interactions between mLOY and smoking behaviour (relative excess risk due to interaction (97.5% CI) >0). Additionally, we observed that current smokers with expanded mLOY had the highest risk of incident lung diseases in both cohorts. CONCLUSIONS mLOY may be a novel predictor for age-related lung diseases. For current smokers carrying mLOY, adopting quitting smoking behaviour may contribute to substantially reducing their risk of incident lung diseases.
Collapse
Affiliation(s)
- Chenghao Weng
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- These authors contributed equally to this work
| | - Yuxuan Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- These authors contributed equally to this work
| | - Mingyu Song
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Zilun Shao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yuanjie Pang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Pei Pei
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
| | - Ling Yang
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Iona Y Millwood
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Robin G Walters
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yiping Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Huaidong Du
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Junshi Chen
- China National Center for Food Safety Risk Assessment, Beijing, China
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Giulio Genovese
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Dianjianyi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
33
|
Thapa R, Gupta S, Gupta G, Bhat AA, Smriti, Singla M, Ali H, Singh SK, Dua K, Kashyap MK. Epithelial-mesenchymal transition to mitigate age-related progression in lung cancer. Ageing Res Rev 2024; 102:102576. [PMID: 39515620 DOI: 10.1016/j.arr.2024.102576] [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: 09/05/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Epithelial-Mesenchymal Transition (EMT) is a fundamental biological process involved in embryonic development, wound healing, and cancer progression. In lung cancer, EMT is a key regulator of invasion and metastasis, significantly contributing to the fatal progression of the disease. Age-related factors such as cellular senescence, chronic inflammation, and epigenetic alterations exacerbate EMT, accelerating lung cancer development in the elderly. This review describes the complex mechanism among EMT and age-related pathways, highlighting key regulators such as TGF-β, WNT/β-catenin, NOTCH, and Hedgehog signalling. We also discuss the mechanisms by which oxidative stress, mediated through pathways involving NRF2 and ROS, telomere attrition, regulated by telomerase activity and shelterin complex, and immune system dysregulation, driven by alterations in cytokine profiles and immune cell senescence, upregulate or downregulate EMT induction. Additionally, we highlighted pathways of transcription such as SNAIL, TWIST, ZEB, SIRT1, TP53, NF-κB, and miRNAs regulating these processes. Understanding these mechanisms, we highlight potential therapeutic interventions targeting these critical molecules and pathways.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Smriti
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhav Singla
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
34
|
Karampitsakos T, Herazo-Maya JD. GZM hi Cytotoxic T Cells: A Key Discovery in Fibrotic Hypersensitivity Pneumonitis. Am J Respir Crit Care Med 2024; 210:1180-1182. [PMID: 39078173 PMCID: PMC11568438 DOI: 10.1164/rccm.202406-1194ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024] Open
Affiliation(s)
| | - Jose D Herazo-Maya
- Pulmonary, Critical Care and Sleep Medicine University of South Florida Tampa, Florida
| |
Collapse
|
35
|
Feng X, Yu F, He XL, Cheng PP, Niu Q, Zhao LQ, Li Q, Cui XL, Jia ZH, Ye SY, Liang LM, Song LJ, Xiong L, Xiang F, Wang X, Ma WL, Ye H. CD8 + tissue-resident memory T cells are essential in bleomycin-induced pulmonary fibrosis. Am J Physiol Cell Physiol 2024; 327:C1178-C1191. [PMID: 39246141 DOI: 10.1152/ajpcell.00368.2024] [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: 08/19/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Human tissue-resident memory T (TRM) cells play a crucial role in protecting the body from infections and cancers. Recent research observed increased numbers of TRM cells in the lung tissues of idiopathic pulmonary fibrosis patients. However, the functional consequences of TRM cells in pulmonary fibrosis remain unclear. Here, we found that the numbers of TRM cells, especially the CD8+ subset, were increased in the mouse lung with bleomycin-induced pulmonary fibrosis. Increasing or decreasing CD8+ TRM cells in mouse lungs accordingly altered the severity of fibrosis. In addition, the adoptive transfer of CD8+ T cells containing a large number of CD8+ TRM cells from fibrotic lungs was sufficient to induce pulmonary fibrosis in control mice. Treatment with chemokine CC-motif ligand (CCL18) induced CD8+ TRM cell expansion and exacerbated fibrosis, whereas blocking C-C chemokine receptor 8 (CCR8) prevented CD8+ TRM recruitment and inhibited pulmonary fibrosis. In conclusion, CD8+ TRM cells are essential for bleomycin-induced pulmonary fibrosis, and targeting CCL18/CCR8/CD8+ TRM cells may be a potential therapeutic approach. NEW & NOTEWORTHY The role of CD8+ TRM cells in the development of pulmonary fibrosis was validated and studied in the classic model of pulmonary fibrosis. It was proposed for the first time that CCL18 has a chemotactic effect on CD8+ TRM cells, thereby exacerbating pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiao Feng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fan Yu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, People's Republic of China
| | - Xin-Liang He
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, People's Republic of China
| | - Pei-Pei Cheng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qian Niu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Li-Qin Zhao
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qian Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiao-Lin Cui
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zi-Heng Jia
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shu-Yi Ye
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Li-Mei Liang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, People's Republic of China
| | - Lin-Jie Song
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, People's Republic of China
| | - Liang Xiong
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, People's Republic of China
| | - Fei Xiang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, People's Republic of China
| | - Xiaorong Wang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, People's Republic of China
| | - Wan-Li Ma
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, People's Republic of China
| | - Hong Ye
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, People's Republic of China
| |
Collapse
|
36
|
Lopes GAO, Lima BHF, Freitas CS, Peixoto AC, Soriani FM, Cassali GD, Ryffel B, Teixeira MM, Machado FS, Russo RC. Opposite effects of systemic and local conditional CD11c+ myeloid cell depletion during bleomycin-induced inflammation and fibrosis in mice. Immun Inflamm Dis 2024; 12:e70042. [PMID: 39582275 PMCID: PMC11586507 DOI: 10.1002/iid3.70042] [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: 05/22/2024] [Revised: 09/19/2024] [Accepted: 10/01/2024] [Indexed: 11/26/2024] Open
Abstract
RATIONALE Elevated levels of CD11c+ myeloid cells are observed in various pulmonary disorders, including Idiopathic Pulmonary Fibrosis (IPF). Dendritic cells (DCs) and macrophages (MΦ) are critical antigen-presenting cells (APCs) that direct adaptive immunity. However, the role of CD11c+ myeloid cells in lung extracellular matrix (ECM) accumulation and pulmonary fibrosis is poorly understood. OBJECTIVE We aimed to investigate the impact of depleting CD11c+ myeloid cells, including DCs and macrophages, during bleomycin-induced pulmonary fibrosis in mice. METHODS We used a diphtheria toxin (DTx) receptor (DTR) transgenic mouse model (CD11c-DTR-Tg) to deplete CD11c+ myeloid cells through two methods: Systemic Depletion (SD) via intraperitoneal injection (i.p.) and local depletion (LD) via intranasal instillation (i.n.). We then assessed the effects of CD11c+ cell depletion during bleomycin-induced lung inflammation and fibrosis. RESULTS Fourteen days after bleomycin instillation, there was a progressive accumulation of myeloid cells, specifically F4/80-MHCII+CD11c+ DCs and F4/80 + MHCII+CD11c+ MΦ, preceding mortality and pulmonary fibrosis. Systemic depletion of CD11c+ DCs and MΦ via i.p. DTx administration in CD11c-DTR-Tg mice protected against bleomycin-induced mortality and pulmonary fibrosis compared to wild-type (WT) mice. Systemic depletion reduced myeloid cells, airway inflammation (total leukocytes, neutrophils, and CD4+ lymphocytes in bronchoalveolar lavage (BAL), inflammatory and fibrogenic mediators, and fibrosis-related mRNAs (Collagen-1α1 and α-SMA). Increased anti-inflammatory cytokine IL-10 and CXCL9 levels were observed, resulting in lower lung hydroxyproline content and Ashcroft fibrosis score. Conversely, local depletion of CD11c+ cells increased mortality by acute leukocyte influx (predominantly neutrophils, DCs, and MΦ in BAL) correlated to IL-1β, with lung hyper-inflammation and early fibrosis development. CONCLUSION Systemic depletion of CD11c+ cells confers protection against inflammation and fibrosis induced by Bleomycin, underscoring the significance of myeloid cells expressing F4/80-MHCII+CD11c+ DCs and F4/80 + MHCII+CD11c+ MΦ orchestrating the inflammatory milieu within the lungs, potentially as a source of cytokines sustaining pulmonary chronic inflammation leading to progressive fibrosis and mortality.
Collapse
Affiliation(s)
- Gabriel Augusto Oliveira Lopes
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological SciencesUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Braulio Henrique Freire Lima
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological SciencesUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
- Department of Biochemistry and Immunology, Institute of Biological SciencesUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Camila Simões Freitas
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological SciencesUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Andiara Cardoso Peixoto
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological SciencesUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Frederico Marianetti Soriani
- Department of Genetics, Ecology, and Evolution, Institute of Biological SciencesUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Geovanni Dantas Cassali
- Department of General Pathology, Institute of Biological SciencesUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Bernhard Ryffel
- Experimental and Molecular Immunology and NeurogeneticsUniversity of Orleans, CNRS UMR7355OrleansFrance
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological SciencesUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Fabiana Simão Machado
- Laboratory of Immunoregulation of Infectious Diseases, Department of Biochemistry and Immunology, Institute of Biological SciencesUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological SciencesUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| |
Collapse
|
37
|
Hu X, Wei Z, Wu Y, Zhao M, Zhou L, Lin Q. Pathogenesis and Therapy of Hermansky-Pudlak Syndrome (HPS)-Associated Pulmonary Fibrosis. Int J Mol Sci 2024; 25:11270. [PMID: 39457053 PMCID: PMC11508683 DOI: 10.3390/ijms252011270] [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: 08/27/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Hermansky-Pudlak syndrome (HPS)-associated pulmonary fibrosis (HPS-PF) is a progressive lung disease that is a major cause of morbidity and mortality in HPS patients. Previous studies have demonstrated that the HPS proteins play an essential role in the biogenesis and function of lysosome-related organelles (LROs) in alveolar epithelial type II (AT2) cells and found that HPS-PF is associated with dysfunction of AT2 cells and abnormal immune reactions. Despite recent advances in research on HPS and the pathology of HPS-PF, the pathological mechanisms underlying HPS-PF remain poorly understood, and no effective treatment has been established. Therefore, it is necessary to refresh the progress in the pathogenesis of HPS-PF to increase our understanding of the pathogenic mechanism of HPS-PF and develop targeted therapeutic strategies. This review summarizes the recent progress in the pathogenesis of HPS-PF provides information about the current treatment strategies for HPS-PF, and hopefully increases our understanding of the pathogenesis of HPS-PF and offers thoughts for new therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | | | - Qiong Lin
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China; (X.H.); (Z.W.); (Y.W.); (M.Z.); (L.Z.)
| |
Collapse
|
38
|
Cocconcelli E, Balestro E, Turato G, Fiorentù G, Bazzan E, Biondini D, Tinè M, Bernardinello N, Pezzuto F, Baraldo S, Calabrese F, Rea F, Sanduzzi Zamparelli A, Spagnolo P, Cosio MG, Saetta M. Tertiary lymphoid structures and B-cell infiltration are IPF features with functional consequences. Front Immunol 2024; 15:1437767. [PMID: 39464888 PMCID: PMC11502372 DOI: 10.3389/fimmu.2024.1437767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/19/2024] [Indexed: 10/29/2024] Open
Abstract
Background Recent literature has shown the presence of B cells and autoantibodies in idiopathic pulmonary fibrosis (IPF) which would imply the presence of tertiary lymphoid structures (TLS, sites where the immune response is triggered), yet TLS are not considered features of the histological characteristics of IPF. Aim This study aims to quantify the presence, size, and degree of activation of TLS in biopsied and explanted lungs from patients with early- and late-IPF, never treated with antifibrotics, and relate their presence and activity to the clinical course, disease progression, and lung inflammation. Methods Immunohistochestry for B cells and CD4, CD8, and CD45 cells was performed in lung tissue from IPF patients: 18 at diagnosis (early), 39 explanted (end-stage), and 12 smoking controls. TLS activation was assessed by CD40 expression. Spirometry along 31 (12-72) months of follow-up was used to characterize end-stage IPF as slow progressors or rapid progressors. Results B cells, along with other inflammatory cells, were higher in early- and end-stage IPF than in controls (p < 0.001). In rapid progressors, all inflammatory cells were higher than in slow progressors (p < 0.05). TLS were present in 100% of early- and end-stage IPF and in 50% of controls. In end-stage IPF, the TLS area and activation score were higher than in early IPF (p < 0.0001; p = 0.005) and controls (p < 0.04; p < 0.002). TLS activation score correlated with FVC decline during follow-up in rapid progressors (r = 0.73; p = 0.007) but not in slow progressors. Conclusions A prominent B-cell infiltration, along with the presence of TLS, the activity of which correlates with FVC decline, is an important component of IPF from the beginning of the disease, likely playing an important role on its mechanism and progression.
Collapse
Affiliation(s)
- Elisabetta Cocconcelli
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Elisabetta Balestro
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Graziella Turato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Giordano Fiorentù
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Erica Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Davide Biondini
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | - Mariaenrica Tinè
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Nicol Bernardinello
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Simonetta Baraldo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Federico Rea
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Alessandro Sanduzzi Zamparelli
- Respiratory Medicine Unit at the Monaldi Hospital, AO dei Colli, Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Paolo Spagnolo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Manuel G. Cosio
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Meakins-Christie Laboratories, Respiratory Division, McGill University, Montreal, QC, Canada
| | - Marina Saetta
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
39
|
Zhang X, Xu Z, Chen Q, Zhou Z. Notch signaling regulates pulmonary fibrosis. Front Cell Dev Biol 2024; 12:1450038. [PMID: 39450276 PMCID: PMC11499121 DOI: 10.3389/fcell.2024.1450038] [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: 06/16/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Pulmonary fibrosis is a progressive interstitial lung disease associated with aging. The pathogenesis of pulmonary fibrosis remains unclear, however, alveolar epithelial cell injury, myofibroblast activation, and extracellular matrix (ECM) accumulation are recognized as key contributors. Moreover, recent studies have implicated cellular senescence, endothelial-mesenchymal transition (EndMT), and epigenetic modifications in the pathogenesis of fibrotic diseases. Various signaling pathways regulate pulmonary fibrosis, including the TGF-β, Notch, Wnt, Hedgehog, and mTOR pathways. Among these, the TGF-β pathway is extensively studied, while the Notch pathway has emerged as a recent research focus. The Notch pathway influences the fibrotic process by modulating immune cell differentiation (e.g., macrophages, lymphocytes), inhibiting autophagy, and promoting interstitial transformation. Consequently, inhibiting Notch signaling represents a promising approach to mitigating pulmonary fibrosis. In this review, we discuss the role of Notch signaling pathway in pulmonary fibrosis, aiming to offer insights for future therapeutic investigations.
Collapse
Affiliation(s)
| | - Zhihao Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | | | | |
Collapse
|
40
|
Moneke I, Ogutur ED, Kornyeva A, Fähndrich S, Schibilsky D, Bierbaum S, Czerny M, Stolz D, Passlick B, Jungraithmayr W, Frye BC. Donor age over 55 is associated with worse outcome in lung transplant recipients with idiopathic pulmonary fibrosis. BMC Pulm Med 2024; 24:499. [PMID: 39385110 PMCID: PMC11465681 DOI: 10.1186/s12890-024-03317-x] [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: 11/26/2023] [Accepted: 10/01/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Lung transplantation (LTx) remains the only efficient treatment for selected patients with end-stage pulmonary disease. The age limit for the acceptance of donor organs in LTx is still a matter of debate. We here analyze the impact of donor organ age and the underlying pulmonary disease on short- and long-term outcome and survival after LTx. METHODS Donor and recipient characteristics of LTx recipients at our institution between 03/2003 and 12/2021 were analyzed. Statistical analysis was performed using SPSS and GraphPad software. RESULTS In 230 patients analyzed, donor age ≥ 55 years was associated with a higher incidence of severe primary graft dysfunction (PGD2/3) (46% vs. 31%, p = 0.03) and reduced long-term survival after LTx (1-, 5- and 10-year survival: 75%, 54%, 37% vs. 84%, 76%, 69%, p = 0.006). Notably, this was only significant in recipients with idiopathic pulmonary fibrosis (IPF) (PGD: 65%, vs. 37%, p = 0.016; 1-, 5-, and 10-year survival: 62%, 38%, 16% vs. 80%, 76%, 70%, p = 0.0002 respectively). In patients with chronic obstructive pulmonary disease (COPD), donor age had no impact on the incidence of PGD2/3 or survival (21% vs. 27%, p = 0.60 and 68% vs. 72%; p = 0.90 respectively). Moreover, we found higher Torque-teno virus (TTV)-DNA levels after LTx in patients with IPF compared to COPD (X2 = 4.57, p = 0.033). Donor age ≥ 55 is an independent risk factor for reduced survival in the whole cohort and patients with IPF specifically. CONCLUSIONS In recipients with IPF, donor organ age ≥ 55 years was associated with a higher incidence of PGD2/3 and reduced survival after LTx. The underlying pulmonary disease may thus be a relevant factor for postoperative graft function and survival. TRIAL REGISTRATION NUMBER DKRS DRKS00033312.
Collapse
Affiliation(s)
- Isabelle Moneke
- Faculty of Medicine, Department of Thoracic Surgery, Medical Center, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.
| | - Ecem Deniz Ogutur
- Faculty of Medicine, Department of Thoracic Surgery, Medical Center, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Anastasiya Kornyeva
- Faculty of Medicine, Department of Thoracic Surgery, Medical Center, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Sebastian Fähndrich
- Faculty of Medicine, Department of Pneumology Medical Center, University of Freiburg, Freiburg, Germany
| | - David Schibilsky
- Faculty of Medicine, Clinic for Cardiovascular Surgery, University Heart Centre Freiburg - Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Sibylle Bierbaum
- Faculty of Medicine, Institute of Virology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Martin Czerny
- Faculty of Medicine, Clinic for Cardiovascular Surgery, University Heart Centre Freiburg - Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Daiana Stolz
- Faculty of Medicine, Department of Pneumology Medical Center, University of Freiburg, Freiburg, Germany
| | - Bernward Passlick
- Faculty of Medicine, Department of Pneumology Medical Center, University of Freiburg, Freiburg, Germany
| | - Wolfgang Jungraithmayr
- Faculty of Medicine, Department of Thoracic Surgery, Medical Center, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Bjoern Christian Frye
- Faculty of Medicine, Department of Pneumology Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
41
|
Bai Y, Gao L, Han T, Liang C, Zhou J, Liu Y, Guo J, Wu J, Hu D. 18β-glycyrrhetinic acid ameliorates bleomycin-induced idiopathic pulmonary fibrosis via inhibiting TGF-β1/JAK2/STAT3 signaling axis. J Steroid Biochem Mol Biol 2024; 243:106560. [PMID: 38917955 DOI: 10.1016/j.jsbmb.2024.106560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/25/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a debilitating and progressive lung disease with an unknown cause that has few treatment options. 18β-Glycyrrhetinic acid (18β-GA) is the main bioactive component in licorice, exhibiting anti-inflammatory and antioxidant effects, while also holding certain application value in the metabolism and regulation of steroids. In this study, we demonstrated that 18β-GA effectively alleviates bleomycin (BLM)-induced IPF by inhibiting the TGF-β1/JAK2/STAT3 signaling axis. In vivo experiments demonstrate that 18β-GA significantly attenuates pulmonary fibrosis progression by reducing lung inflammation, improving lung function, and decreasing collagen deposition. In vitro experiments reveal that 18β-GA inhibits the activation and migration of TGF-β1-induced fibroblasts. Furthermore, it regulates the expression of vimentin, N-cadherin and E-cadherin proteins, thereby inhibiting TGF-β1-induced epithelial-mesenchymal transition (EMT) in lung alveolar epithelial cells. Mechanistically, 18β-GA ameliorates pulmonary fibrosis by modulating the TGF-β1/JAK2/STAT3 signaling pathway in activated fibroblasts. Taken together, our findings demonstrate the potential and underlying mechanisms of 18β-GA in ameliorating IPF, emphasizing its potential as a novel therapeutic drug for the treatment of this devastating disease.
Collapse
Affiliation(s)
- Ying Bai
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Lu Gao
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Tao Han
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Chao Liang
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Jing Wu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institute, Huainan, Anhui, China; Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui, China.
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institute, Huainan, Anhui, China; Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui, China.
| |
Collapse
|
42
|
Karampitsakos T, Tourki B, Jia M, Perrot CY, Visinescu B, Zhao A, Unterman A, Tzouvelekis A, Bandyopadhyay D, Juan-Guardela BM, Prasse A, Noth I, Liggett S, Kaminski N, Benos PV, Herazo-Maya JD. The transcriptome of CD14 + CD163 - HLA-DR low monocytes predicts mortality in Idiopathic Pulmonary Fibrosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.07.24311386. [PMID: 39211854 PMCID: PMC11361223 DOI: 10.1101/2024.08.07.24311386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Rationale The association between immune-cell-specific transcriptomic profiles and Idiopathic Pulmonary Fibrosis (IPF) mortality is unknown. Objectives To determine immune-cell-specific transcriptomic profiles associated with IPF mortality. Methods We profiled peripheral blood mononuclear cells (PBMC) in 18 participants [University of South Florida: IPF, COVID-19, post-COVID-19 Interstitial Lung Disease (Post-COVID-19 ILD), controls] by single-cell RNA sequencing (scRNA-seq) and identified 16 immune-cell-specific transcriptomic profiles. The Scoring Algorithm of Molecular Subphenotypes (SAMS) was used to calculate Up-scores based on these 16 gene profiles. Their association with outcomes was investigated in peripheral blood, Bronchoalveolar Lavage (BAL) and lung tissue of N=416 IPF patients from six cohorts. Findings were validated in an independent IPF, PBMC scRNA-seq dataset (N=38). Measurements and main results Cox-regression models demonstrated that 230 genes from CD14 + CD163 - HLA-DR low circulating monocytes predicted IPF mortality [Pittsburgh (p=0.02), Chicago (p=0.003)]. PBMC proportions of CD14 + CD163 - HLA-DR low monocytes were higher in progressive versus stable IPF (Yale, 0.13±0.05 versus 0.09±0.05, p=0.034). Receiving operating characteristic identified a 230 gene, Up-score >41.84 (Pittsburgh) predictive of mortality in Chicago (HR: 6.58, 95%CI: 2.15-20.13, p=0.001) and in pooled analysis of BAL cohorts (HR: 2.20, 95%CI: 1.44-3.37, p=0.0003). High-risk patients had decreased expression of the T-cell co-stimulatory genes CD28 , ICOS , ITK and LCK (Pittsburgh and Chicago, p<0.01). 230 gene-up-scores negatively correlated with Forced Vital Capacity (FVC) in IPF lung tissues (LGRC, rho=-0.2, p=0.02). Results were replicated using a subset of 13 genes from the 230-gene signature (pooled PBMC cohorts - HR: 5.34, 95%CI: 2.83-10.06, p<0.0001). Conclusions The transcriptome of CD14 + CD163 - HLA-DR low monocytes is associated with increased IPF mortality.
Collapse
|
43
|
Sung EA, Dozmorov MG, Song S, Aung T, Park MH, Sime PJ, Chae WJ. Ablation of LRP6 in alpha-smooth muscle actin-expressing cells abrogates lung inflammation and fibrosis upon bleomycin-induced lung injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611327. [PMID: 39314349 PMCID: PMC11418957 DOI: 10.1101/2024.09.05.611327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Low-density lipoprotein receptor-related protein 6 (LRP6) is a receptor for Wnt ligands. Tissue fibrosis is a progressive pathological process with excessive extracellular matrix proteins (ECM) deposition. Myofibroblasts, identified by alpha-smooth muscle actin (αSMA) expression, play an important role in tissue fibrosis by producing ECM production. Here we found that Wnt antagonist Dickkopf1 (DKK1) induced gene expressions associated with inflammation and fibrosis in lung fibroblasts. We demonstrated that genetic deletion of LRP6 in αSMA-expressing cells using Acta2 -cre Lrp6 fl/fl ( Lrp6 AKO ) mice abrogated bleomycin (BLM)-induced lung inflammation and fibrosis phenotype, suggesting an important role of LRP6 in modulating inflammation and fibrotic processes in the lung. Our results highlight the crucial role of LRP6 in fibroblasts in regulating inflammation and fibrosis upon BLM-induced lung injury.
Collapse
|
44
|
Mohammed SM, Al-Saedi HFS, Mohammed AQ, Amir AA, Radi UK, Sattar R, Ahmad I, Ramadan MF, Alshahrani MY, Balasim HM, Alawadi A. Mechanisms of Bleomycin-induced Lung Fibrosis: A Review of Therapeutic Targets and Approaches. Cell Biochem Biophys 2024; 82:1845-1870. [PMID: 38955925 DOI: 10.1007/s12013-024-01384-9] [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] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
Pulmonary toxicity is a serious side effect of some specific anticancer drugs. Bleomycin is a well-known anticancer drug that triggers severe reactions in the lungs. It is an approved drug that may be prescribed for the treatment of testicular cancers, Hodgkin's and non-Hodgkin's lymphomas, ovarian cancer, head and neck cancers, and cervical cancer. A large number of experimental studies and clinical findings show that bleomycin can concentrate in lung tissue, leading to massive oxidative stress, alveolar epithelial cell death, the proliferation of fibroblasts, and finally the infiltration of immune cells. Chronic release of pro-inflammatory and pro-fibrotic molecules by immune cells and fibroblasts leads to pneumonitis and fibrosis. Both fibrosis and pneumonitis are serious concerns for patients who receive bleomycin and may lead to death. Therefore, the management of lung toxicity following cancer therapy with bleomycin is a critical issue. This review explains the cellular and molecular mechanisms of pulmonary injury following treatment with bleomycin. Furthermore, we review therapeutic targets and possible promising strategies for ameliorating bleomycin-induced lung injury.
Collapse
Affiliation(s)
- Shaimaa M Mohammed
- Department of Pharmacy, Al- Mustaqbal University College, 51001, Hilla, Babylon, Iraq
| | | | | | - Ahmed Ali Amir
- Department of Medical Laboratories Technology, Al-Nisour University College, Baghdad, Iraq
| | - Usama Kadem Radi
- College of Pharmacy, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Ruaa Sattar
- Al-Hadi University College, Baghdad, 10011, Iraq
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Halah Majeed Balasim
- Department of Medical Laboratory Technologies, Al Rafidain University College, Bagdad, Iraq
| | - Ahmed Alawadi
- College of technical engineering, the Islamic University, Najaf, Iraq
- College of technical engineering, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of technical engineering, the Islamic University of Babylon, Hilla, Iraq
| |
Collapse
|
45
|
Wei X, Jin C, Li D, Wang Y, Zheng S, Feng Q, Shi N, Kong W, Ma X, Wang J. Single-cell transcriptomics reveals CD8 + T cell structure and developmental trajectories in idiopathic pulmonary fibrosis. Mol Immunol 2024; 172:85-95. [PMID: 38936318 DOI: 10.1016/j.molimm.2024.06.008] [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: 03/10/2024] [Revised: 06/20/2024] [Accepted: 06/23/2024] [Indexed: 06/29/2024]
Abstract
Immune cells in the human lung are associated with idiopathic pulmonary fibrosis. However, the contribution of different immune cell subpopulations to the pathogenesis of pulmonary fibrosis remains unclear. We used single-cell RNA sequencing data to investigate the transcriptional profiles of immune cells in the lungs of 5 IPF patients and 3 subjects with non-fibrotic lungs. In an identifiable population of immune cells, we found increased percentage of CD8+ T cells in the T cell subpopulation in IPF. Monocle analyzed the dynamic immune status and cell transformation of CD8+ T cells, as well as the cytotoxicity and exhausted status of CD8+ T cell subpopulations at different stages. Among CD8+ T cells, we found differences in metabolic pathways in IPF and Ctrl, including lipid, amino acid and carbohydrate metabolic. By analyzing the metabolites of CD8+ T cells, we found that different populations of CD8+ T cells in IPF have unique metabolic characteristics, but they also have multiple identical up-regulated or down-regulated metabolites. In IPF, signaling pathways associated with fibrosis were enriched in CD8+ T cells, suggesting that CD8+ T cells may have an important contribution to fibrosis. Finally, we analyzed the interactions between CD8+ T cells and other cells. Together, these studies highlight key features of CD8+ T cells in the pathogenesis of IPF and help to develop effective therapeutic targets.
Collapse
Affiliation(s)
- Xuemei Wei
- Center of Respiratory and Critical Care Medicine, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, China; State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830000, China
| | - Chengji Jin
- Department of Respiratory Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou 570100, China
| | - Dewei Li
- Center of Respiratory and Critical Care Medicine, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang 830001, China
| | - Yujie Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou 570100, China
| | - Shaomao Zheng
- Department of Respiratory Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou 570100, China
| | - Qiong Feng
- Department of Respiratory Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou 570100, China
| | - Ning Shi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830000, China
| | - Weina Kong
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830000, China
| | - Xiumin Ma
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830000, China.
| | - Jing Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou 570100, China; NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou 571199, China.
| |
Collapse
|
46
|
Nishioka Y, Araya J, Tanaka Y, Kumanogoh A. Pathological mechanisms and novel drug targets in fibrotic interstitial lung disease. Inflamm Regen 2024; 44:34. [PMID: 39026335 PMCID: PMC11264521 DOI: 10.1186/s41232-024-00345-2] [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: 05/14/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Interstitial lung diseases (ILDs) are a diverse group of conditions characterized by inflammation and fibrosis in the lung. In some patients with ILD, a progressive fibrotic phenotype develops, which is associated with an irreversible decline in lung function and a poor prognosis. MAIN BODY The pathological mechanisms that underlie this process culminate in fibroblast activation, proliferation, and differentiation into myofibroblasts, which deposit extracellular matrix proteins and result in fibrosis. Upstream of fibroblast activation, epithelial cell injury and immune activation are known initiators of fibrosis progression, with multiple diverse cell types involved. Recent years have seen an increase in our understanding of the complex and interrelated processes that drive fibrosis progression in ILD, in part due to the advent of single-cell RNA sequencing technology and integrative multiomics analyses. Novel pathological mechanisms have been identified, which represent new targets for drugs currently in clinical development. These include phosphodiesterase 4 inhibitors and other molecules that act on intracellular cyclic adenosine monophosphate signaling, as well as inhibitors of the autotaxin-lysophosphatidic acid axis and α v integrins. Here, we review current knowledge and recent developments regarding the pathological mechanisms that underlie progressive fibrotic ILD, including potential therapeutic targets. CONCLUSION Knowledge of the pathological mechanisms that drive progressive fibrosis in patients with ILD has expanded, with the role of alveolar endothelial cells, the immune system, and fibroblasts better elucidated. Drugs that target novel mechanisms hold promise for expanding the future therapeutic armamentarium for progressive fibrotic ILD.
Collapse
Affiliation(s)
| | - Jun Araya
- The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshiya Tanaka
- University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
47
|
Li C, Abdurehim A, Zhao S, Sun Q, Xu J, Xie J, Zhang Y. Research on the potential mechanism of Deapioplatycodin D against pulmonary fibrosis based on bioinformatics and experimental verification. Eur J Pharmacol 2024; 974:176603. [PMID: 38679121 DOI: 10.1016/j.ejphar.2024.176603] [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: 09/27/2023] [Revised: 03/27/2024] [Accepted: 04/18/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a group of respiratory diseases that are extremely complex and challenging to treat. Due to its high mortality rate and short survival, it's often referred to as a "tumor-like disease" that poses a serious threat to human health. OBJECTIVE We aimed validate the potential of Deapioplatycodin D (DPD) to against PF and clarify the underlying mechanism of action of DPD for the treatment of PF based on bioinformatics and experimental verification. This finding provides a basis for the development of safe and effective therapeutic PF drugs based on DPD. METHODS We used LPS-induced early PF rats as a PF model to test the overall efficacy of DPD in vivo. Then, A variety of bioinformatics methods, such as WGCNA, LASSO algorithm and immune cell infiltration (ICI), were applied to analyze the gene microarray related to PF obtained from Gene Expression Omnibus (GEO) to obtained key targets of PF. Finally, an in vitro PF model was constructed based on BEAS-2B cells while incorporating rat lung tissues to validate the regulatory effects of DPD on critical genes. RESULTS DPD can effectively alleviate inflammatory and fibrotic markers in rat lungs. WGCNA analysis resulted in a total of six expression modules, with the brown module having the highest correlation with PF. Subsequently, seven genes were acquired by intersecting the genes in the brown module with DEGs. Five key genes were identified as potential biomarkers of PF by LASSO algorithm and validation dataset verification analysis. In the ICI analysis, infiltration of activated B cell, immature B cell and natural killer cells were found to be more crucial in PF. Ultimately, it was observed that DPD could modulate key genes to achieve anti-PF effects. CONCLUSION In short, these comprehensive analysis methods were employed to identify critical biomarkers closely related to PF, which helps to elucidate the pathogenesis and potential immunotherapy targets of PF. It also provides essential support for the potential of DPD against PF.
Collapse
Affiliation(s)
- Chao Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin, China.
| | - Aliya Abdurehim
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin, China.
| | - Shuang Zhao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Qing Sun
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin, China.
| | - Jiawen Xu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin, China.
| | - Junbo Xie
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin, China.
| | - Yanqing Zhang
- Biotechnology & Food Science College, Tianjin University of Commerce, Tianjin, 300134, China.
| |
Collapse
|
48
|
Shaikh TB, Chandra Y, Andugulapati SB, Sistla R. Vistusertib improves pulmonary inflammation and fibrosis by modulating inflammatory/oxidative stress mediators via suppressing the mTOR signalling. Inflamm Res 2024; 73:1223-1237. [PMID: 38789791 DOI: 10.1007/s00011-024-01894-5] [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: 03/25/2024] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
INTRODUCTION Inflammation and oxidative stress are key factors in the development of pulmonary fibrosis (PF) by promoting the differentiation of fibroblasts through modulating various pathways including Wnt/β-catenin, TGF-β and mTOR signalling. OBJECTIVE AND METHODS This study aimed to evaluate the effects and elucidate the mechanisms of vistusertib (VSB) in treating pulmonary inflammation/fibrosis, specifically by targeting the mTOR pathway using various in vitro and in vivo models. RESULTS Lipopolysaccharide (LPS)-induced inflammation model in macrophages (RAW 264.7), epithelial (BEAS-2B) and endothelial (HMVEC-L) cells revealed that treatment with VSB significantly reduced the IL-6, TNF-α, CCL2, and CCL7 expression. TGF-β induced differentiation was also significantly reduced upon VSB treatment in fibrotic cells (LL29 and DHLF). Further, bleomycin-induced inflammation and fibrosis models demonstrated that treatment with VSB significantly ameliorated the severe inflammation, and lung architectural distortion, by reducing the inflammatory markers expression/levels, inflammatory cells and oxidative stress indicators. Further, fibrosis model results exhibited that, VSB treatment significantly reduced the α-SMA, collagen and TGF-β expressions, improved the lung architecture and restored lung functions. CONCLUSION Overall, this study uncovers the anti-inflammatory/anti-fibrotic effects of VSB by modulating the mTOR activation. Although VSB was tested for lung fibrosis, it can be tested for other fibrotic disorders to improve the patient's survival and quality of life.
Collapse
Affiliation(s)
- Taslim B Shaikh
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India
| | - Yogesh Chandra
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India
| | - Sai Balaji Andugulapati
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India.
| | - Ramakrishna Sistla
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India.
| |
Collapse
|
49
|
Hu Y, Huang Y, Zong L, Lin J, Liu X, Ning S. Emerging roles of ferroptosis in pulmonary fibrosis: current perspectives, opportunities and challenges. Cell Death Discov 2024; 10:301. [PMID: 38914560 PMCID: PMC11196712 DOI: 10.1038/s41420-024-02078-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic interstitial lung disorder characterized by abnormal myofibroblast activation, accumulation of extracellular matrix (ECM), and thickening of fibrotic alveolar walls, resulting in deteriorated lung function. PF is initiated by dysregulated wound healing processes triggered by factors such as excessive inflammation, oxidative stress, and coronavirus disease (COVID-19). Despite advancements in understanding the disease's pathogenesis, effective preventive and therapeutic interventions are currently lacking. Ferroptosis, an iron-dependent regulated cell death (RCD) mechanism involving lipid peroxidation and glutathione (GSH) depletion, exhibits unique features distinct from other RCD forms (e.g., apoptosis, necrosis, and pyroptosis). Imbalance between reactive oxygen species (ROS) production and detoxification leads to ferroptosis, causing cellular dysfunction through lipid peroxidation, protein modifications, and DNA damage. Emerging evidence points to the crucial role of ferroptosis in PF progression, driving macrophage polarization, fibroblast proliferation, and ECM deposition, ultimately contributing to alveolar cell death and lung tissue scarring. This review provides a comprehensive overview of the latest findings on the involvement and signaling mechanisms of ferroptosis in PF pathogenesis, emphasizing potential novel anti-fibrotic therapeutic approaches targeting ferroptosis for PF management.
Collapse
Affiliation(s)
- Yixiang Hu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China
| | - Ying Huang
- Zhongshan Hospital of Traditional Chinese Medicine Afflilated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, China
| | - Lijuan Zong
- Department of Rehabilitation Medicine, Zhongda Hospital of Southeast University, Nanjing, 210096, China
| | - Jiaxin Lin
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Xiang Liu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China.
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China.
| |
Collapse
|
50
|
Mannes PZ, Adams TS, Farsijani S, Barnes CE, Latoche JD, Day KE, Nedrow JR, Ahangari F, Kaminski N, Lee JS, Tavakoli S. Noninvasive assessment of the lung inflammation-fibrosis axis by targeted imaging of CMKLR1. SCIENCE ADVANCES 2024; 10:eadm9817. [PMID: 38896611 PMCID: PMC11186491 DOI: 10.1126/sciadv.adm9817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
Precision management of fibrotic lung diseases is challenging due to their diverse clinical trajectories and lack of reliable biomarkers for risk stratification and therapeutic monitoring. Here, we validated the accuracy of CMKLR1 as an imaging biomarker of the lung inflammation-fibrosis axis. By analyzing single-cell RNA sequencing datasets, we demonstrated CMKLR1 expression as a transient signature of monocyte-derived macrophages (MDMφ) enriched in patients with idiopathic pulmonary fibrosis (IPF). Consistently, we identified MDMφ as the major driver of the uptake of CMKLR1-targeting peptides in a murine model of bleomycin-induced lung fibrosis. Furthermore, CMKLR1-targeted positron emission tomography in the murine model enabled quantification and spatial mapping of inflamed lung regions infiltrated by CMKLR1-expressing macrophages and emerged as a robust predictor of subsequent lung fibrosis. Last, high CMKLR1 expression by bronchoalveolar lavage cells identified an inflammatory endotype of IPF with poor survival. Our investigation supports the potential of CMKLR1 as an imaging biomarker for endotyping and risk stratification of fibrotic lung diseases.
Collapse
Affiliation(s)
- Philip Z. Mannes
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Taylor S. Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Samaneh Farsijani
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Aging and Population Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Clayton E. Barnes
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joseph D. Latoche
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kathryn E. Day
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jessie R. Nedrow
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Farida Ahangari
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Janet S. Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Sina Tavakoli
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|