1
|
Li J, Huang N, Zhang X, Wang H, Chen J, Wei Q. Functional analysis of yak alveolar type II epithelial cells at high and low altitudes based on single-cell sequencing. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119889. [PMID: 39681250 DOI: 10.1016/j.bbamcr.2024.119889] [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: 09/13/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024]
Abstract
The adaptation of lung cells to high-altitude environments represents a notable gap in our understanding of how animals cope with hypoxic conditions. Alveolar epithelial cells type II (AEC II) are crucial for lung development and repair. However, their, specific role in the adaptation of yaks to high-altitude environments remains unclear. In this study, we aimed to address this gap by investigating the differential responses of AEC II in yaks at high and low altitudes (4000 m and 2600 m, respectively). We used the 10 × scRNA-seq technology to construct a comprehensive cell atlas of yak lung tissue, and identified 15 distinct cell classes. AEC II in high-altitude yaks revealed increased immunomodulatory, adhesive, and metabolic activities, which are crucial for maintaining lung tissue stability and energy supply under hypoxic conditions. Furthermore, alveolar epithelial progenitor cells within AEC II can differentiate into both Alveolar epithelial cell type I (AEC I) and AEC II. SHIP1 and other factors are promoters of AEC I transdifferentiation, whereas SFTPC and others promote AEC II transdifferentiation. This study provides new insights into the evolutionary adaptation of lung cells in plateau animals by elucidating the molecular mechanisms underlying AEC II adaptation to high-altitude environments.
Collapse
Affiliation(s)
- Jingyi Li
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Nating Huang
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Xun Zhang
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Huizhen Wang
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Jiarui Chen
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China
| | - Qing Wei
- College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai, China; State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, China.
| |
Collapse
|
2
|
Shan N, Shang Y, He Y, Wen Z, Ning S, Chen H. Common biomarkers of idiopathic pulmonary fibrosis and systemic sclerosis based on WGCNA and machine learning. Sci Rep 2025; 15:610. [PMID: 39753882 PMCID: PMC11699037 DOI: 10.1038/s41598-024-84820-3] [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] [Accepted: 12/27/2024] [Indexed: 01/06/2025] Open
Abstract
Interstitial lung disease (ILD) is known to be a major complication of systemic sclerosis (SSc) and a leading cause of death in SSc patients. As the most common type of ILD, the pathogenesis of idiopathic pulmonary fibrosis (IPF) has not been fully elucidated. In this study, weighted correlation network analysis (WGCNA), protein‒protein interaction, Kaplan-Meier curve, univariate Cox analysis and machine learning methods were used on datasets from the Gene Expression Omnibus database. CCL2 was identified as a common characteristic gene of IPF and SSc. The genes associated with CCL2 expression in both diseases were enriched mainly in chemokine-related pathways and lipid metabolism-related pathways according to Gene Set Enrichment Analysis. Single-cell RNA sequencing (sc-RNAseq) revealed a significant difference in CCL2 expression in alveolar epithelial type 1/2 cells, mast cells, ciliated cells, club cells, fibroblasts, M1/M2 macrophages, monocytes and plasma cells between IPF patients and healthy donors. Statistical analyses revealed that CCL2 was negatively correlated with lung function in IPF patients and decreased after mycophenolate mofetil (MMF) treatment in SSc patients. Finally, we identified CCL2 as a common biomarker from IPF and SSc, revealing the common mechanism of these two diseases and providing clues for the study of the treatment and mechanism of these two diseases.
Collapse
Affiliation(s)
- Ning Shan
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yu Shang
- The Second Hospital of Heilongjiang Province, Harbin, Heilongjiang Province, China
| | - Yaowu He
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Zhe Wen
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Shangwei Ning
- Harbin Medical University, Harbin, Heilongjiang Province, China.
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province, China.
| | - Hong Chen
- Harbin Medical University, Harbin, Heilongjiang Province, China.
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
3
|
Li Y, Qi J, Guo L, Jiang X, He G. Organellar quality control crosstalk in aging-related disease: Innovation to pave the way. Aging Cell 2025; 24:e14447. [PMID: 39668579 PMCID: PMC11709098 DOI: 10.1111/acel.14447] [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: 09/15/2024] [Revised: 11/04/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024] Open
Abstract
Organellar homeostasis and crosstalks within a cell have emerged as essential regulatory and determining factors for the survival and functions of cells. In response to various stimuli, cells can activate the organellar quality control systems (QCS) to maintain homeostasis. Numerous studies have demonstrated that dysfunction of QCS can lead to various aging-related diseases such as neurodegenerative, pulmonary, cardiometabolic diseases and cancers. However, the interplay between QCS and their potential role in these diseases are poorly understood. In this review, we present an overview of the current findings of QCS and their crosstalk, encompassing mitochondria, endoplasmic reticulum, Golgi apparatus, ribosomes, peroxisomes, lipid droplets, and lysosomes as well as the aberrant interplays among these organelles that contributes to the onset and progression of aging-related disorders. Furthermore, potential therapeutic approaches based on these quality control interactions are discussed. Our perspectives can enhance insights into the regulatory networks underlying QCS and the pathology of aging and aging-related diseases, which may pave the way for the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Yu Li
- Department of Dermatology & VenerologyWest China Hospital, Sichuan UniversityChengduChina
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease‐Related Molecular Network, State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Jinxin Qi
- Department of Dermatology & VenerologyWest China Hospital, Sichuan UniversityChengduChina
| | - Linhong Guo
- Department of Dermatology & VenerologyWest China Hospital, Sichuan UniversityChengduChina
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease‐Related Molecular Network, State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xian Jiang
- Department of Dermatology & VenerologyWest China Hospital, Sichuan UniversityChengduChina
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease‐Related Molecular Network, State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Gu He
- Department of Dermatology & VenerologyWest China Hospital, Sichuan UniversityChengduChina
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease‐Related Molecular Network, State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
4
|
Liu M, Sheng Y, Li M, Pan T, Jiang W, Zhang Y, Pan X, Huang C, Li J, Wang Y. METTL3-Dependent YTHDF2 Mediates TSC1 Expression to Regulate Alveolar Epithelial Mesenchymal Transition and Promote Idiopathic Pulmonary Fibrosis. J Cell Physiol 2025; 240:e31473. [PMID: 39606797 DOI: 10.1002/jcp.31473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 11/29/2024]
Abstract
Diffuse, progressive interstitial lung disease with few treatment options and low survival rates is known as idiopathic pulmonary fibrosis (IPF). Alveolar epithelial cell damage and dysfunction are the main features of IPF. TSC1 has been documented to exert a pivotal function in governing cellular growth, proliferation, and ontogenesis. This work investigated TSC1's function and mechanism in IPF. Mice were given BLM to cause pulmonary fibrosis, and A549 cells underwent epithelial mesenchymal transition (EMT) in response to TGF-β1. According to the data, TSC1 expression was reduced in IPF. Overexpression of TSC1 was established by adenopathy-associated virus in vivo and adenovirus in vitro to significantly block the EMT process. Besides, the findings from the RNA-sequencing analysis indicate that overexpression of TSC1 mitigated the EMT process by suppressing the activation of the AKT/mTOR pathway via downregulation of ACTN4 expression. To examine the upstream regulatory mechanism, we employed the SRAMP database to predict m6A modification of TSC1 mRNA, followed by verification of m6A modification levels and expression using MERIP-qPCR, Dot blot, RT-qPCR, and WB. The results indicated a high degree of m6A modification in TSC1 mRNA in pulmonary fibrosis. The expression of METTL3 was further found to be significantly elevated. METTL3 knockdown impeded EMT progression. METTL3 inhibits TSC1 expression by increasing TSC1 m6A modification through the reading protein YTHDF2. In conclusion, our study elucidated that the METTL3/YTHDF2/TSC1 signaling axis activates the AKT/mTOR pathway to promote the development of IPF. This study provides potential molecular-level therapeutic targets for IPF disease.
Collapse
Affiliation(s)
- Min Liu
- Key Laboratory of Inflammation and Immune-Mediated Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui Province, China
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Yingying Sheng
- Key Laboratory of Inflammation and Immune-Mediated Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui Province, China
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Mengyu Li
- Key Laboratory of Inflammation and Immune-Mediated Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui Province, China
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Tianyu Pan
- Key Laboratory of Inflammation and Immune-Mediated Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui Province, China
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Wei Jiang
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Yafei Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Xin Pan
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Cheng Huang
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Jun Li
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Yuanyuan Wang
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
5
|
Yang J, Li Y, Huang Y, Chen H, Sui P. Unlocking lung regeneration: insights into progenitor cell dynamics and metabolic control. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:31. [PMID: 39676102 DOI: 10.1186/s13619-024-00212-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/17/2024]
Abstract
Regenerative responses are particularly important in the lungs, which are critical for gas exchange and frequently challenged by environmental insults. The lung progenitor cells play a central role in the lung regeneration response, and their dysfunction is associated with various lung diseases. Understanding the mechanisms regulating lung progenitor cell function is essential for developing new therapeutic approaches to promote lung regeneration. This review summarizes recent advancements in the field of lung regeneration, focusing on the metabolic control of lung progenitor cell function. We discuss cell lineage plasticity and cell-cell signaling under different physiological conditions. Additionally, we highlight the connection between progenitor cell dysfunction and lung diseases, emphasizing the need to develop new therapeutic strategies in regenerative medicine to improve lung regenerative capacity.
Collapse
Affiliation(s)
- Jiaying Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yawen Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ying Huang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Huaiyong Chen
- Department of Basic Medicine, Tianjin University Haihe Hospital, Tianjin, 300350, China.
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China.
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China.
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin, China.
| | - Pengfei Sui
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
6
|
Wang JY, Michki SN, Sitaraman S, Banaschewski BJ, Jamal R, Gokey JJ, Lin SM, Katzen JB, Basil MC, Cantu E, Kropski JA, Zepp JA, Frank DB, Young LR. Dysregulated alveolar epithelial cell progenitor function and identity in Hermansky-Pudlak syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.17.545390. [PMID: 38496421 PMCID: PMC10942273 DOI: 10.1101/2023.06.17.545390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Hermansky-Pudlak syndrome (HPS) is a genetic disorder of endosomal protein trafficking associated with pulmonary fibrosis in specific subtypes, including HPS-1 and HPS-2. Single mutant HPS1 and HPS2 mice display increased fibrotic sensitivity while double mutant HPS1/2 mice exhibit spontaneous fibrosis with aging, which has been attributed to HPS mutations in alveolar epithelial type II (AT2) cells. We utilized HPS mouse models and human lung tissue to investigate mechanisms of AT2 cell dysfunction driving fibrotic remodeling in HPS. Starting at 8 weeks of age, HPS mice exhibited progressive loss of AT2 cell numbers. HPS AT2 cell function was impaired ex vivo and in vivo . Incorporating AT2 cell lineage tracing in HPS mice, we observed aberrant differentiation with increased AT2-derived alveolar epithelial type I cells. Transcriptomic analysis of HPS AT2 cells revealed elevated expression of genes associated with aberrant differentiation and p53 activation. Lineage tracing and organoid modeling studies demonstrated that HPS AT2 cells were primed to persist in a Krt8 + reprogrammed transitional state, mediated by p53 activity. Intrinsic AT2 progenitor cell dysfunction and p53 pathway dysregulation are novel mechanisms of disease in HPS-related pulmonary fibrosis, with the potential for early targeted intervention before the onset of fibrotic lung disease.
Collapse
|
7
|
Cen Z, Cen T, Ding Q, Zhang Y, Tang P, Lv C, Wu T. Outcomes and predictors of progression in progressive pulmonary fibrosis. Ann Med 2024; 56:2406439. [PMID: 39310989 PMCID: PMC11421158 DOI: 10.1080/07853890.2024.2406439] [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: 12/16/2023] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Progressive pulmonary fibrosis (PPF) is a general term for a class of interstitial lung diseases (ILDs) characterized by a progressive fibrosing (PF) phenotype. Patients with PPF have decreased lung function, exercise ability, and quality of life. The purpose of this study was to investigate the clinical characteristics, potential associated factors for disease progression, and survival outcomes of patients in the PPF population. METHODS This study retrospectively reviewed the data of patients diagnosed with ILD between January 2011 and December 2022 at The First Affiliated Hospital of Ningbo University. A PF phenotype was defined based on the criteria that were used in the PPF clinical practice guidelines, which led to the identification of 92 patients with a PF phenotype among the 177 patients with fibrotic ILD. Baseline clinical information and laboratory parameters were collected and analysed in our cohort. RESULTS Patients in the PPF group had higher tumour marker levels and lower pulmonary function test results at baseline than did those in the non-PPF group. According to the multivariate logistic regression analysis, age >65 years (OR 2.71, 95% CI 1.26-5.89; p = 0.011), LDH >245 U/L (OR 3.07, 95% CI 1.39-6.78; p = 0.006), CA-153 > 35 U/mL (OR 3.16, 95% CI 1.25-7.97; p = 0.015), FVC <60% predicted (OR 4.82, 95% CI 1.60-14.51; p = 0.005), DLCO <50% predicted (OR 3.21, 95% CI 1.43-7.21; p = 0.005), and the UIP-like pattern on chest HRCT (OR 3.65, 95% CI 1.33-10.07; p = 0.012) were potentially associated with the progression of fibrotic interstitial lung diseases (f-ILDs) to PPF. Furthermore, the PPF group had a poorer survival rate than the non-PPF group (p = 0.0045). According to the multivariate Cox regression analysis, an SPAP ≥ 37 mmHg (HR 2.33, 95% CI 1.09-5.00; p = 0.030) and acute exacerbation (HR 2.88, 95% CI 1.26-6.59; p = 0.012) were identified as significant prognostic factors for mortality in patients with PPFs. CONCLUSIONS Patients who were older, had high CA-153 and LDH levels, had poor pulmonary function test results, or had a UIP-like pattern on chest HRCT were more likely to have indications for the progression of f-ILD to PPF. Increased SPAP and AE are independent risk factors for the prognosis of PPF patients, so additional attention should be given to such patients.
Collapse
Affiliation(s)
- Zekai Cen
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Tiantian Cen
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qunli Ding
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yun Zhang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Pan Tang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Chengna Lv
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Tingting Wu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
8
|
Wang Y, Hu LF, Liu NH, Yang JS, Xing L, Jeong JH, Li L, Jiang HL. Mitophagy-Enhanced Nanoparticle-Engineered Mitochondria Restore Homeostasis of Mitochondrial Pool for Alleviating Pulmonary Fibrosis. ACS NANO 2024; 18:32705-32722. [PMID: 39546755 DOI: 10.1021/acsnano.4c10328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Pulmonary fibrosis (PF) is an interstitial lung disease tightly associated with the disruption of mitochondrial pool homeostasis, a delicate balance influenced by functional and dysfunctional mitochondria within lung cells. Mitochondrial transfer is an emerging technology to increase functional mitochondria via exogenous mitochondrial delivery; however, the therapeutic effect on mitochondrial transfer is hampered during the PF process by the persistence of dysfunctional mitochondria, which is attributed to impaired mitophagy. Herein, we reported engineering mitochondria mediated by mitophagy-enhanced nanoparticle (Mito-MEN), which promoted synchronal regulation of functional and dysfunctional mitochondria for treating PF. Mitophagy-enhanced nanoparticles (MENs) were fabricated through the encapsulation of Parkin mRNA, and the electrostatic interaction favored MENs to anchor isolated healthy mitochondria for the construction of Mito-MEN. Mito-MEN increased the load of functional exogenous mitochondria by enhancing mitochondrial delivery efficiency and promoted mitophagy of dysfunctional endogenous mitochondria. In a bleomycin (BLM)-induced PF mouse model, Mito-MEN repaired mitochondrial function and efficiently relieved PF-related phenotypes. This study provides a powerful tool for synchronal adjustment of mitochondrial pool homeostasis and offers a translational approach for pan-mitochondrial disease therapies.
Collapse
Affiliation(s)
- Yi Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Li-Fan Hu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Na-Hui Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jing-Song Yang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
- College of Pharmacy, Yanbian University, Yanji 133002, China
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
9
|
Niayesh-Mehr R, Kalantar M, Bontempi G, Montaldo C, Ebrahimi S, Allameh A, Babaei G, Seif F, Strippoli R. The role of epithelial-mesenchymal transition in pulmonary fibrosis: lessons from idiopathic pulmonary fibrosis and COVID-19. Cell Commun Signal 2024; 22:542. [PMID: 39538298 PMCID: PMC11558984 DOI: 10.1186/s12964-024-01925-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Despite the tremendous advancements in the knowledge of the pathophysiology and clinical aspects of SARS-CoV-2 infection, still many issues remain unanswered, especially in the long-term effects. Mounting evidence suggests that pulmonary fibrosis (PF) is one of the most severe complications associated with COVID-19. Therefore, understanding the molecular mechanisms behind its development is helpful to develop successful therapeutic strategies. Epithelial to mesenchymal transition (EMT) and its cell specific variants endothelial to mesenchymal transition (EndMT) and mesothelial to mesenchymal transition (MMT) are physio-pathologic cellular reprogramming processes induced by several infectious, inflammatory and biomechanical stimuli. Cells undergoing EMT acquire invasive, profibrogenic and proinflammatory activities by secreting several extracellular mediators. Their activity has been implicated in the pathogenesis of PF in a variety of lung disorders, including idiopathic pulmonary fibrosis (IPF) and COVID-19. Aim of this article is to provide an updated survey of the cellular and molecular mechanisms, with emphasis on EMT-related processes, implicated in the genesis of PF in IFP and COVID-19.
Collapse
Affiliation(s)
- Reyhaneh Niayesh-Mehr
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mojtaba Kalantar
- Department of Occupational Health, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | - Giulio Bontempi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Claudia Montaldo
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Saeedeh Ebrahimi
- Department of Medical Microbiology (Bacteriology and Virology), Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ghader Babaei
- Department of Clinical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Faezeh Seif
- Department of Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran.
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy.
| |
Collapse
|
10
|
Sun Z, He W, Meng H, Ji Z, Qu J, Yu G. Lactate activates ER stress to promote alveolar epithelial cells apoptosis in pulmonary fibrosis. Respir Res 2024; 25:401. [PMID: 39522031 PMCID: PMC11550544 DOI: 10.1186/s12931-024-03016-5] [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: 06/01/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive lung disease characterized by fibroblast proliferation, extensive extracellular matrix and collagen deposition, accompanied by inflammatory damage, ultimately leading to death due to respiratory failure. Endoplasmic reticulum (ER) stress in pulmonary fibrotic tissue is indeed recognized as a significant factor exacerbating PF development. Emerging evidences indicated a potential association between ER stress induced by lactate and cellular apoptosis in PF. However, the mechanisms in this process need further elucidation. In this paper, pulmonary fibrosis model was induced by bleomycin (BLM) intratracheally in mice. In the cellular model, type II epithelial cells were treated by lactate and TGF-β to detect ER stress and apoptosis markers. Lactate could promote ER stress response and apoptosis. Mechanically, lactate activated Caspase-12 via ATF4-Chop axis to induce cell apoptosis and promote fibrosis. ER stress inhibitor could effectively suppress alveolar epithelial cells apoptosis and pulmonary fibrosis. We concluded that pro-fibrotic properties of lactate are associated with alveolar epithelial cells apoptosis by causing ER stress and thus provide new potential therapeutic targets for pulmonary fibrosis.
Collapse
Affiliation(s)
- Zhiheng Sun
- College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China.
- State Key Laboratory of Cell Differentiation and Regulation, Xinxiang, Henan, China.
| | - Wanyu He
- College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
- State Key Laboratory of Cell Differentiation and Regulation, Xinxiang, Henan, China
| | - Huiwen Meng
- College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
- State Key Laboratory of Cell Differentiation and Regulation, Xinxiang, Henan, China
| | - Zhihua Ji
- College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
- State Key Laboratory of Cell Differentiation and Regulation, Xinxiang, Henan, China
| | - Junxing Qu
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, Henan, China.
- Xinxiang Key Laboratory for Tumor Drug Screening and Targeted Therapy, Xinxiang, Henan, China.
| | - Guoying Yu
- College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China.
- State Key Laboratory of Cell Differentiation and Regulation, Xinxiang, Henan, China.
| |
Collapse
|
11
|
Ren Y, Qin S, Liu X, Feng B, Liu J, Zhang J, Yuan P, Yu K, Mei H, Chen M. Hyperoxia can Induce Lung Injury by Upregulating AECII Autophagy and Apoptosis Via the mTOR Pathway. Mol Biotechnol 2024; 66:3357-3368. [PMID: 37938537 PMCID: PMC11549204 DOI: 10.1007/s12033-023-00945-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023]
Abstract
Oxygen therapy is a crucial medical intervention, but it is undeniable that it can lead to lung damage. The mTOR pathway plays a pivotal role in governing cell survival, including autophagy and apoptosis, two phenomena deeply entwined with the evolution of diseases. However, it is unclarified whether the mTOR pathway is involved in hyperoxic acute lung injury (HALI). The current study aims to clarify the molecular mechanism underlying the pathogenesis of HALI by constructing in vitro and in vivo models using H2O2 and hyperoxia exposure, respectively. To investigate the role of mTOR, the experiment was divided into five groups, including normal group, injury group, mTOR inhibitor group, mTOR activator group, and DMSO control group. Western blotting, Autophagy double labeling, TUNEL staining, and HE staining were applied to evaluate protein expression, autophagy activity, cell apoptosis, and pathological changes in lung tissues. Our data revealed that hyperoxia can induce autophagy and apoptosis in Type II alveolar epithelial cell (AECII) isolated from the treated rats, as well as injuries in the rat lung tissues; also, H2O2 stimulation increased autophagy and apoptosis in MLE-12 cells. Noticeably, the experiments performed in both in vitro and in vivo models proved that the mTOR inhibitor Rapamycin (Rapa) functioned synergistically with hyperoxia or H2O2 to promote AECII autophagy, which led to increased apoptosis and exacerbated lung injury. On the contrary, activation of mTOR with MHY1485 suppressed autophagy activity, consequently resulting in reduced apoptosis and lung injury in H2O2-challenged MLE-12 cells and hyperoxia-exposed rats. In conclusion, hyperoxia caused lung injury via mTOR-mediated AECII autophagy.
Collapse
Affiliation(s)
- Yingcong Ren
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Song Qin
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Xinxin Liu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Banghai Feng
- Department of Critical Care Medicine, Zunyi Hospital of Traditional Chinese Medicine, Zunyi, 563000, Guizhou, China
| | - Junya Liu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Jing Zhang
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Ping Yuan
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Kun Yu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Hong Mei
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Miao Chen
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
12
|
Puthusseri B, Bhagavath AK, Korir D, Shetty R, Johnson GA, Stankowska DL, Konduru NV, Tang H, Bolla S, Criner GJ, Marchetti N, Das DN, Shetty S. CSP7 Protects Alveolar Epithelial Cells by Targeting p53-Fibrinolytic Pathways During Lung Injuries. Am J Respir Cell Mol Biol 2024; 72:97-108. [PMID: 39269467 PMCID: PMC11707667 DOI: 10.1165/rcmb.2023-0453oc] [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: 12/22/2023] [Accepted: 09/13/2024] [Indexed: 09/15/2024] Open
Abstract
Impaired alveolar epithelial regeneration in patients with idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD) is attributed to telomere dysfunction in type II alveolar epithelial cells (A2Cs). Genetic susceptibility, aging, and toxicant exposures, including tobacco smoke (TS), contribute to telomere dysfunction in A2Cs. Here we investigated whether improvement of telomere function plays a role in CSP7-mediated protection of A2Cs against ongoing senescence and apoptosis during bleomycin (BLM)-induced pulmonary fibrosis (PF) as well as alveolar injury caused by chronic TS exposure. We found a significant telomere shortening in A2Cs isolated from IPF and COPD lungs in line with other studies. These cells showed increased p53 in addition to its post-translational modification with induction of activated caspase-3 and β-galactosidase, suggesting a p53-mediated loss of A2C renewal. Further, we found increased expression of SIAH-1, a p53-inducible E3 ubiquitin ligase known to down-regulate telomere repeats binding factor 2 (TRF2). Consistent with the loss of TRF2 and upregulation of TRF1, telomerase reverse transcriptase (TERT) was downregulated in A2Cs. A2Cs from fibrotic lungs of mice either repeatedly instilled with BLM or isolated from chronic TS exposure-induced lung injury model showed reduced telomere length along with induction of p53, PAI-1, SIAH1 and TRF1 as well as loss of TRF2 and TERT, which were reversed in wild-type mice after treatment with CSP7. Interestingly, PAI-1-/- mice, or those lacking microRNA-34a expression in A2Cs, resisted telomere dysfunction, while uPA-/- mice failed to respond to CSP7 treatment, suggesting p53-microRNA-34a feed-forward induction and p53-uPA pathway contributes to telomere dysfunction.
Collapse
Affiliation(s)
- Bijesh Puthusseri
- The University of Texas Health Science Center at Tyler, Tyler, Texas, United States
| | | | - Daniel Korir
- The University of Texas Health Science Center at Tyler, Tyler, Texas, United States
| | - Rashmi Shetty
- The University of Texas Health Science Center at Tyler, Tyler, Texas, United States
| | - Gretchen A Johnson
- University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Dorota L Stankowska
- University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Nagarjun V Konduru
- The University of Texas Health Science Center at Tyler, Tyler, Texas, United States
| | - Hua Tang
- The University of Texas Health Science Center at Tyler, Tyler, Texas, United States
| | - Sudhir Bolla
- Temple University, Thoracic Medicine and Surgery, Philadelphia, Pennsylvania, United States
| | - Gerald J Criner
- Temple University Hospital, Pulm & Crit Care Medicine, Philadelphia, Pennsylvania, United States
- Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, United States
| | - Nathaniel Marchetti
- Temple University School of Medicine, Pulmonary and Critical Care Medicine, Philadelphia, Pennsylvania, United States
| | - Durgesh Nandini Das
- The University of Texas Health Science Center at Tyler, Tyler, Texas, United States
| | - Sreerama Shetty
- The University of Texas Health Center at Tyler, Department of Specialty Care Services, Tyler, Texas, United States;
| |
Collapse
|
13
|
Predella C, Lapsley L, Ni K, Murray JW, Liu HY, Motelow JE, Snoeck HW, Glasser SW, Saqi A, Dorrello NV. Engraftment of wild-type alveolar type II epithelial cells in surfactant protein C deficient mice. RESEARCH SQUARE 2024:rs.3.rs-4673915. [PMID: 39315275 PMCID: PMC11419168 DOI: 10.21203/rs.3.rs-4673915/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Childhood interstitial lung disease (chILD) secondary to pulmonary surfactant deficiency is a devastating chronic lung disease in children. Clinical presentation includes mild to severe respiratory failure and fibrosis. There is no specific treatment, except lung transplantation, which is hampered by a severe shortage of donor organs, especially for young patients. Repair of lungs with chILD represents a longstanding therapeutic challenge but cell therapy is a promising strategy. As surfactant is produced by alveolar type II epithelial (ATII) cells, engraftment with normal or gene-corrected ATII cells might provide an avenue to cure. Here we used a chILD disease-like model, Sftpc -/- mice, to provide proof-of-principle for this approach. Sftpc -/- mice developed chronic interstitial lung disease with age and were hypersensitive to bleomycin. We could engraft wild-type ATII cells after low dose bleomycin conditioning. Transplanted ATII cells produced mature SPC and attenuated bleomycin-induced lung injury up to two months post-transplant. This study demonstrates that partial replacement of mutant ATII cells can promote lung repair in a mouse model of chILD-like disease.
Collapse
Affiliation(s)
- Camilla Predella
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Chemistry, Materials and Chemical Engineering “G. Natta”, Politecnico of Milan, Milan, Italy
- Department of Electronics, Information and Bioengineering, Politecnico of Milan, Milan, Italy
| | - Lauren Lapsley
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Keyue Ni
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - John W. Murray
- Columbia Center for Human Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Hsiao-Yun Liu
- Columbia Center for Human Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Joshua E. Motelow
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Morgan Stanley Children’s Hospital, New York, NY, USA
| | - Hans-Willem Snoeck
- Columbia Center for Human Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Stephan W. Glasser
- Medical Sciences Program, Department of Medical Education, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Anjali Saqi
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian, New York, NY, USA
| | - N. Valerio Dorrello
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Morgan Stanley Children’s Hospital, New York, NY, USA
| |
Collapse
|
14
|
Zhang Q, An N, Liu Y, Zhu Y, Pan W, Gu P, Zhao J, Pu Q, Zhu W. Alveolar type 2 cells marker gene SFTPC inhibits epithelial-to-mesenchymal transition by upregulating SOX7 and suppressing WNT/β-catenin pathway in non-small cell lung cancer. Front Oncol 2024; 14:1448379. [PMID: 39346732 PMCID: PMC11427448 DOI: 10.3389/fonc.2024.1448379] [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: 06/13/2024] [Accepted: 08/22/2024] [Indexed: 10/01/2024] Open
Abstract
Introduction Surfactant Protein C gene (SFTPC) is a marker gene of alveolar type 2 cells (AT2), which are the key structures of alveoli. Mutations or deletions in SFTPC cause idiopathic pulmonary fibrosis (IPF). Importantly, IPF is an independent risk factor for non-small cell lung cancer (NSCLC). It suggests that abnormal expression of SFTPC may be relevant to development of NSCLC. However, the function and mechanism of SFTPC in NSCLC are still poor understood until now. Methods The expression of SFTPC and the relationship between SFTPC and prognosis of NSCLC were analyzed in TCGA database and our collected clinical NSCLC tissues. Subsequently, the function and mechanism of SFTPC in NSCLC were explored by RNA-sequence, qRT-PCR, Western blot, Immunohistochemical, Wound-healing, Millicell, Transwell assays and mouse tumor xenograft model. Results SFTPC was dramatically downregulated in NSCLC tissues from TCGA database and 40 out of 46 collected clinical LUAD tissues compared with adjacent non-tumor tissues. Low expression of SFTPC was associated with poor prognosis of LUAD by TCGA database. Importantly, we confirmed that overexpression of SFTPC significantly inhibited Epithelial-to-Mesenchymal Transition (EMT) process of NSCLC cells by upregulating SOX7 and then inactivating WNT/β-catenin pathway in vitro and in vivo. Particularly, we discovered that low expression of SFTPC was associated with EMT process and low expression of SOX7 in NSCLC tissues. Conclusion Our study revealed a novel mechanism of SFTPC in NSCLC development. Meanwhile, it also might provide a new clue for exploring the molecular mechanism about NSCLC development in patients with IPF in the future.
Collapse
Affiliation(s)
- Qiongyin Zhang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ning An
- Cancer Center, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yang Liu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ying Zhu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wuliang Pan
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peiling Gu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinzhu Zhao
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiang Pu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wen Zhu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Jiang H, Zheng B, Hu G, Kuang L, Zhou T, Li S, Chen X, Li C, Zhang D, Zhang J, Yang Z, He J, Jin H. Spatially resolved metabolomics visualizes heterogeneous distribution of metabolites in lung tissue and the anti-pulmonary fibrosis effect of Prismatomeris connate extract. J Pharm Anal 2024; 14:100971. [PMID: 39381647 PMCID: PMC11459407 DOI: 10.1016/j.jpha.2024.100971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/22/2024] [Accepted: 03/25/2024] [Indexed: 10/10/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic progressive end-stage lung disease. However, the mechanisms underlying the progression of this disease remain elusive. Presently, clinically employed drugs are scarce for the treatment of PF. Hence, there is an urgent need for developing novel drugs to address such diseases. Our study found for the first time that a natural source of Prismatomeris connata Y. Z. Ruan (Huang Gen, HG) ethyl acetate extract (HG-2) had a significant anti-PF effect by inhibiting the expression of the transforming growth factor beta 1/suppressor of mothers against decapentaplegic (TGF-β1/Smad) pathway. Network pharmacological analysis suggested that HG-2 had effects on tyrosine kinase phosphorylation, cellular response to reactive oxygen species, and extracellular matrix (ECM) disassembly. Moreover, mass spectrometry imaging (MSI) was used to visualize the heterogeneous distribution of endogenous metabolites in lung tissue and reveal the anti-PF metabolic mechanism of HG-2, which was related to arginine biosynthesis and alanine, asparate and glutamate metabolism, the downregulation of arachidonic acid metabolism, and the upregulation of glycerophospholipid metabolism. In conclusion, we elaborated on the relationship between metabolite distribution and the progression of PF, constructed the regulatory metabolic network of HG-2, and discovered the multi-target therapeutic effect of HG-2, which might be conducive to the development of new drugs for PF.
Collapse
Affiliation(s)
- Haiyan Jiang
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Bowen Zheng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Guang Hu
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Lian Kuang
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Tianyu Zhou
- College of Pharmacy, Shaanxi University of Traditional Chinese Medicine, Xianyang, Shaanxi, 712046, China
| | - Sizheng Li
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Xinyi Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Chuangjun Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Dongming Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jinlan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Zengyan Yang
- Guangxi International Zhuang Medicine Hospital, Nanning, 530201, China
| | - Jiuming He
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Key Laboratory for Safety Research and Evaluation of Innovative Drug, National Medical Products Administration, Beijing, 102206, China
| | - Hongtao Jin
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Key Laboratory for Safety Research and Evaluation of Innovative Drug, National Medical Products Administration, Beijing, 102206, China
- Beijing Union-Genius Pharmaceutical Technology Development Co., Ltd., Beijing, 100176, China
| |
Collapse
|
16
|
Liu Y, Xu C, Gu R, Han R, Li Z, Xu X. Endoplasmic reticulum stress in diseases. MedComm (Beijing) 2024; 5:e701. [PMID: 39188936 PMCID: PMC11345536 DOI: 10.1002/mco2.701] [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: 02/21/2023] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
The endoplasmic reticulum (ER) is a key organelle in eukaryotic cells, responsible for a wide range of vital functions, including the modification, folding, and trafficking of proteins, as well as the biosynthesis of lipids and the maintenance of intracellular calcium homeostasis. A variety of factors can disrupt the function of the ER, leading to the aggregation of unfolded and misfolded proteins within its confines and the induction of ER stress. A conserved cascade of signaling events known as the unfolded protein response (UPR) has evolved to relieve the burden within the ER and restore ER homeostasis. However, these processes can culminate in cell death while ER stress is sustained over an extended period and at elevated levels. This review summarizes the potential role of ER stress and the UPR in determining cell fate and function in various diseases, including cardiovascular diseases, neurodegenerative diseases, metabolic diseases, autoimmune diseases, fibrotic diseases, viral infections, and cancer. It also puts forward that the manipulation of this intricate signaling pathway may represent a novel target for drug discovery and innovative therapeutic strategies in the context of human diseases.
Collapse
Affiliation(s)
- Yingying Liu
- Department of Aviation Clinical Medicine, Air Force Medical CenterPLABeijingChina
| | - Chunling Xu
- School of Pharmaceutical SciencesTsinghua UniversityBeijingChina
| | - Renjun Gu
- School of Chinese MedicineNanjing University of Chinese MedicineNanjingChina
- Department of Gastroenterology and HepatologyJinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Ruiqin Han
- State Key Laboratory of Medical Molecular BiologyDepartment of Biochemistry and Molecular BiologyInstitute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ziyu Li
- School of Acupuncture and TuinaSchool of Regimen and RehabilitationNanjing University of Chinese MedicineNanjingChina
| | - Xianrong Xu
- Department of Aviation Clinical Medicine, Air Force Medical CenterPLABeijingChina
| |
Collapse
|
17
|
Cui X, Zhou Z, Tu H, Wu J, Zhou J, Yi Q, Liu O, Dai X. Mitophagy in fibrotic diseases: molecular mechanisms and therapeutic applications. Front Physiol 2024; 15:1430230. [PMID: 39183973 PMCID: PMC11341310 DOI: 10.3389/fphys.2024.1430230] [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: 05/09/2024] [Accepted: 07/17/2024] [Indexed: 08/27/2024] Open
Abstract
Mitophagy is a highly precise process of selective autophagy, primarily aimed at eliminating excess or damaged mitochondria to maintain the stability of both mitochondrial and cellular homeostasis. In recent years, with in-depth research into the association between mitophagy and fibrotic diseases, it has been discovered that this process may interact with crucial cellular biological processes such as oxidative stress, inflammatory responses, cellular dynamics regulation, and energy metabolism, thereby influencing the occurrence and progression of fibrotic diseases. Consequently, modulating mitophagy holds promise as a therapeutic approach for fibrosis. Currently, various methods have been identified to regulate mitophagy to prevent fibrosis, categorized into three types: natural drug therapy, biological therapy, and physical therapy. This review comprehensively summarizes the current understanding of the mechanisms of mitophagy, delves into its biological roles in fibrotic diseases, and introduces mitophagy modulators effective in fibrosis, aiming to provide new targets and theoretical basis for the investigation of fibrosis-related mechanisms and disease prevention.
Collapse
Affiliation(s)
- Xinyan Cui
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Zekun Zhou
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Hua Tu
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Jianjun Wu
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Jian Zhou
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, China
- Department of VIP Dental Service, School of Stomatology, Capital Medical University, Beijing, China
- Laboratory for Oral and General Health Integration and Translation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qiao Yi
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Ousheng Liu
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Xiaohan Dai
- Hunan Key Laboratory of Oral Health Research, Hunan Clinical Research Center of Oral Major Diseases, Oral Health and Academician Workstation for Oral-maxilofacial, Regenerative Medicine and Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| |
Collapse
|
18
|
Liu Q, Niu Y, Pei Z, Yang Y, Xie Y, Wang M, Wang J, Wu M, Zheng J, Yang P, Hao H, Pang Y, Bao L, Dai Y, Niu Y, Zhang R. Gas6-Axl signal promotes indoor VOCs exposure-induced pulmonary fibrosis via pulmonary microvascular endothelial cells-fibroblasts cross-talk. JOURNAL OF HAZARDOUS MATERIALS 2024; 474:134786. [PMID: 38824778 DOI: 10.1016/j.jhazmat.2024.134786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/14/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
Volatile organic compounds (VOCs) as environmental pollutants were associated with respiratory diseases. Pulmonary fibrosis (PF) was characterized by an increase of extracellular matrix, leading to deterioration of lung function. The adverse effects on lung and the potential mechanism underlying VOCs induced PF had not been elucidated clearly. In this study, the indoor VOCs exposure mouse model along with an ex vivo biosensor assay was established. Based on scRNA-seq analysis, the adverse effects on lung and potential molecular mechanism were studied. Herein, the results showed that VOCs exposure from indoor decoration contributed to decreased lung function and facilitated pulmonary fibrosis in mice. Then, the whole lung cell atlas after VOCs exposure and the heterogeneity of fibroblasts were revealed. We explored the molecular interactions among various pulmonary cells, suggesting that endothelial cells contributed to fibroblasts activation in response to VOCs exposure. Mechanistically, pulmonary microvascular endothelial cells (MPVECs) secreted Gas6 after VOCs-induced PANoptosis phenotype, bound to the Axl in fibroblasts, and then activated fibroblasts. Moreover, Atf3 as the key gene negatively regulated PANoptosis phenotype to ameliorate fibrosis induced by VOCs exposure. These novel findings provided a new perspective about MPVECs could serve as the initiating factor of PF induced by VOCs exposure.
Collapse
Affiliation(s)
- Qingping Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Yong Niu
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Zijie Pei
- Department of Thoracic Surgery, the 2nd Hospital of Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yizhe Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Yujia Xie
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Mengruo Wang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Jingyuan Wang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Mengqi Wu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Jie Zheng
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Peihao Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Haiyan Hao
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Hebei Province Center for Disease Control and Prevention, Shijiazhuang 050021, Hebei, PR China
| | - Yaxian Pang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Lei Bao
- Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Yufei Dai
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Yujie Niu
- Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Rong Zhang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China.
| |
Collapse
|
19
|
Murthy A, Rodriguez LR, Dimopoulos T, Bui S, Iyer S, Chavez K, Tomer Y, Abraham V, Cooper C, Renner DM, Katzen JB, Bentley ID, Ghadiali SN, Englert JA, Weiss SR, Beers MF. Activation of alveolar epithelial ER stress by β-coronavirus infection disrupts surfactant homeostasis in mice: implications for COVID-19 respiratory failure. Am J Physiol Lung Cell Mol Physiol 2024; 327:L232-L249. [PMID: 38860845 PMCID: PMC11444511 DOI: 10.1152/ajplung.00324.2023] [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: 10/19/2023] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024] Open
Abstract
COVID-19 syndrome is characterized by acute lung injury, hypoxemic respiratory failure, and high mortality. Alveolar type 2 (AT2) cells are essential for gas exchange, repair, and regeneration of distal lung epithelium. We have shown that the causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and other members of the β-coronavirus genus induce an endoplasmic reticulum (ER) stress response in vitro; however, the consequences for host AT2 cell function in vivo are less understood. To study this, two murine models of coronavirus infection were used-mouse hepatitis virus-1 (MHV-1) in A/J mice and a mouse-adapted SARS-CoV-2 strain. MHV-1-infected mice exhibited dose-dependent weight loss with histological evidence of distal lung injury accompanied by elevated bronchoalveolar lavage fluid (BALF) cell counts and total protein. AT2 cells showed evidence of both viral infection and increased BIP/GRP78 expression, consistent with activation of the unfolded protein response (UPR). The AT2 UPR included increased inositol-requiring enzyme 1α (IRE1α) signaling and a biphasic response in PKR-like ER kinase (PERK) signaling accompanied by marked reductions in AT2 and BALF surfactant protein (SP-B and SP-C) content, increases in surfactant surface tension, and emergence of a reprogrammed epithelial cell population (Krt8+ and Cldn4+). The loss of a homeostatic AT2 cell state was attenuated by treatment with the IRE1α inhibitor OPK-711. As a proof-of-concept, C57BL6 mice infected with mouse-adapted SARS-CoV-2 demonstrated similar lung injury and evidence of disrupted surfactant homeostasis. We conclude that lung injury from β-coronavirus infection results from an aberrant host response, activating multiple AT2 UPR stress pathways, altering surfactant metabolism/function, and changing AT2 cell state, offering a mechanistic link between SARS-CoV-2 infection, AT2 cell biology, and acute respiratory failure.NEW & NOTEWORTHY COVID-19 syndrome is characterized by hypoxemic respiratory failure and high mortality. In this report, we use two murine models to show that β-coronavirus infection produces acute lung injury, which results from an aberrant host response, activating multiple epithelial endoplasmic reticular stress pathways, disrupting pulmonary surfactant metabolism and function, and forcing emergence of an aberrant epithelial transition state. Our results offer a mechanistic link between SARS-CoV-2 infection, AT2 cell biology, and respiratory failure.
Collapse
Affiliation(s)
- Aditi Murthy
- Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
- PENN-CHOP Lung Biology Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Luis R Rodriguez
- Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
- PENN-CHOP Lung Biology Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Thalia Dimopoulos
- Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Sarah Bui
- Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
- PENN-CHOP Lung Biology Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Swati Iyer
- Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Katrina Chavez
- Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Yaniv Tomer
- Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Valsamma Abraham
- Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Charlotte Cooper
- Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - David M Renner
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Penn Center for Research on Coronaviruses and Emerging Pathogens, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Jeremy B Katzen
- Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
- PENN-CHOP Lung Biology Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Ian D Bentley
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Samir N Ghadiali
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Department of Biomedical Engineering, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Joshua A Englert
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Susan R Weiss
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Penn Center for Research on Coronaviruses and Emerging Pathogens, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Michael F Beers
- Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
- PENN-CHOP Lung Biology Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
20
|
Liang J, Huang G, Liu X, Zhang X, Rabata A, Liu N, Fang K, Taghavifar F, Dai K, Kulur V, Jiang D, Noble PW. Lipid Deficiency Contributes to Impaired Alveolar Progenitor Cell Function in Aging and Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2024; 71:242-253. [PMID: 38657143 PMCID: PMC11299087 DOI: 10.1165/rcmb.2023-0290oc] [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: 08/04/2023] [Accepted: 04/24/2024] [Indexed: 04/26/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an aging-associated interstitial lung disease resulting from repeated epithelial injury and inadequate epithelial repair. Alveolar type II cells (AEC2s) are progenitor cells that maintain epithelial homeostasis and repair the lung after injury. In the current study, we assessed lipid metabolism in AEC2s from human lungs of patients with IPF and healthy donors, as well as AEC2s from bleomycin-injured young and old mice. Through single-cell RNA sequencing, we observed that lipid metabolism-related genes were downregulated in IPF AEC2s and bleomycin-injured mouse AEC2s. Aging aggravated this decrease and hindered recovery of lipid metabolism gene expression in AEC2s after bleomycin injury. Pathway analyses revealed downregulation of genes related to lipid biosynthesis and fatty acid β-oxidation in AEC2s from IPF lungs and bleomycin-injured, old mouse lungs compared with the respective controls. We confirmed decreased cellular lipid content in AEC2s from IPF lungs and bleomycin-injured, old mouse lungs using immunofluorescence staining and flow cytometry. Futhermore, we show that lipid metabolism was associated with AEC2 progenitor function. Lipid supplementation and PPARγ (peroxisome proliferator activated receptor γ) activation promoted progenitor renewal capacity of both human and mouse AEC2s in three-dimensional organoid cultures. Lipid supplementation also increased AEC2 proliferation and expression of SFTPC in AEC2s. In summary, we identified a lipid metabolism deficiency in AEC2s from lungs of patients with IPF and bleomycin-injured old mice. Restoration of lipid metabolism homeostasis in AEC2s might promote AEC2 progenitor function and offer new opportunities for therapeutic approaches to IPF.
Collapse
Affiliation(s)
- Jiurong Liang
- Department of Medicine and Women’s Guild Lung Institute, and
| | - Guanling Huang
- Department of Medicine and Women’s Guild Lung Institute, and
| | - Xue Liu
- Department of Medicine and Women’s Guild Lung Institute, and
| | - Xuexi Zhang
- Department of Medicine and Women’s Guild Lung Institute, and
| | - Anas Rabata
- Department of Medicine and Women’s Guild Lung Institute, and
| | - Ningshan Liu
- Department of Medicine and Women’s Guild Lung Institute, and
| | - Kai Fang
- Department of Medicine and Women’s Guild Lung Institute, and
| | | | - Kristy Dai
- Department of Medicine and Women’s Guild Lung Institute, and
| | - Vrishika Kulur
- Department of Medicine and Women’s Guild Lung Institute, and
| | - Dianhua Jiang
- Department of Medicine and Women’s Guild Lung Institute, and
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Paul W. Noble
- Department of Medicine and Women’s Guild Lung Institute, and
| |
Collapse
|
21
|
He A, He L, Chen T, Li X, Cao C. Biomechanical Properties and Cellular Responses in Pulmonary Fibrosis. Bioengineering (Basel) 2024; 11:747. [PMID: 39199705 PMCID: PMC11351367 DOI: 10.3390/bioengineering11080747] [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: 06/07/2024] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 09/01/2024] Open
Abstract
Pulmonary fibrosis is a fatal lung disease affecting approximately 5 million people worldwide, with a 5-year survival rate of less than 50%. Currently, the only available treatments are palliative care and lung transplantation, as there is no curative drug for this condition. The disease involves the excessive synthesis of the extracellular matrix (ECM) due to alveolar epithelial cell damage, leading to scarring and stiffening of the lung tissue and ultimately causing respiratory failure. Although multiple factors contribute to the disease, the exact causes remain unclear. The mechanical properties of lung tissue, including elasticity, viscoelasticity, and surface tension, are not only affected by fibrosis but also contribute to its progression. This paper reviews the alteration in these mechanical properties as pulmonary fibrosis progresses and how cells in the lung, including alveolar epithelial cells, fibroblasts, and macrophages, respond to these changes, contributing to disease exacerbation. Furthermore, it highlights the importance of developing advanced in vitro models, based on hydrogels and 3D bioprinting, which can accurately replicate the mechanical and structural properties of fibrotic lungs and are conducive to studying the effects of mechanical stimuli on cellular responses. This review aims to summarize the current understanding of the interaction between the progression of pulmonary fibrosis and the alterations in mechanical properties, which could aid in the development of novel therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Andong He
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310028, China
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, 59 Liuting Road, Ningbo 315010, China
- Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
| | - Lizhe He
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, China
| | - Tianwei Chen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xuejin Li
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310028, China
| | - Chao Cao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, 59 Liuting Road, Ningbo 315010, China
| |
Collapse
|
22
|
Tan Q, Wellmerling JH, Song S, Dresler SR, Meridew JA, Choi KM, Li Y, Prakash Y, Tschumperlin DJ. Targeting CEBPA to restore cellular identity and tissue homeostasis in pulmonary fibrosis. JCI Insight 2024; 9:e175290. [PMID: 39012710 PMCID: PMC11343593 DOI: 10.1172/jci.insight.175290] [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: 09/13/2023] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
Fibrosis in the lung is thought to be driven by epithelial cell dysfunction and aberrant cell-cell interactions. Unveiling the molecular mechanisms of cellular plasticity and cell-cell interactions is imperative to elucidating lung regenerative capacity and aberrant repair in pulmonary fibrosis. By mining publicly available RNA-Seq data sets, we identified loss of CCAAT enhancer-binding protein alpha (CEBPA) as a candidate contributor to idiopathic pulmonary fibrosis (IPF). We used conditional KO mice, scRNA-Seq, lung organoids, small-molecule inhibition, and potentially novel gene manipulation methods to investigate the role of CEBPA in lung fibrosis and repair. Long-term (6 months or more) of Cebpa loss in AT2 cells caused spontaneous fibrosis and increased susceptibility to bleomycin-induced fibrosis. Cebpa knockout (KO) in these mice significantly decreased AT2 cell numbers in the lung and reduced expression of surfactant homeostasis genes, while increasing inflammatory cell recruitment as well as upregulating S100a8/a9 in AT2 cells. In vivo treatment with an S100A8/A9 inhibitor alleviated experimental lung fibrosis. Restoring CEBPA expression in lung organoids ex vivo and during experimental lung fibrosis in vivo rescued CEBPA deficiency-mediated phenotypes. Our study establishes a direct mechanistic link between CEBPA repression, impaired AT2 cell identity, disrupted tissue homeostasis, and lung fibrosis.
Collapse
Affiliation(s)
- Qi Tan
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Jack H. Wellmerling
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Shengren Song
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Sara R. Dresler
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Jeffrey A. Meridew
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Kyoung M. Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Yong Li
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Y.S. Prakash
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Daniel J. Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
23
|
Yin Y, Zeng Z, Wei S, Shen Z, Cong Z, Zhu X. Using the sympathetic system, beta blockers and alpha-2 agonists, to address acute respiratory distress syndrome. Int Immunopharmacol 2024; 139:112670. [PMID: 39018694 DOI: 10.1016/j.intimp.2024.112670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Abstract
Acute Respiratory Distress Syndrome (ARDS) manifests as an acute inflammatory lung injury characterized by persistent hypoxemia, featuring a swift onset, high mortality, and predominantly supportive care as the current therapeutic approach, while effective treatments remain an area of active investigation. Adrenergic receptors (AR) play a pivotal role as stress hormone receptors, extensively participating in various inflammatory processes by initiating downstream signaling pathways. Advancements in molecular biology and pharmacology continually unveil the physiological significance of distinct AR subtypes. Interventions targeting these subtypes have the potential to induce specific alterations in cellular and organismal functions, presenting a promising avenue as a therapeutic target for managing ARDS. This article elucidates the pathogenesis of ARDS and the basic structure and function of AR. It also explores the relationship between AR and ARDS from the perspective of different AR subtypes, aiming to provide new insights for the improvement of ARDS.
Collapse
Affiliation(s)
- Yiyuan Yin
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Zhaojin Zeng
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Senhao Wei
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Ziyuan Shen
- Department of Anaesthesiology, Peking University Third Hospital, Beijing, China
| | - Zhukai Cong
- Department of Anaesthesiology, Peking University Third Hospital, Beijing, China.
| | - Xi Zhu
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
24
|
Yang Z, Yang Y, Han X, Hou J. Novel AT2 Cell Subpopulations and Diagnostic Biomarkers in IPF: Integrating Machine Learning with Single-Cell Analysis. Int J Mol Sci 2024; 25:7754. [PMID: 39062997 PMCID: PMC11277372 DOI: 10.3390/ijms25147754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/08/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a long-term condition with an unidentified cause, and currently there are no specific treatment options available. Alveolar epithelial type II cells (AT2) constitute a heterogeneous population crucial for secreting and regenerative functions in the alveolus, essential for maintaining lung homeostasis. However, a comprehensive investigation into their cellular diversity, molecular features, and clinical implications is currently lacking. In this study, we conducted a comprehensive examination of single-cell RNA sequencing data from both normal and fibrotic lung tissues. We analyzed alterations in cellular composition between IPF and normal tissue and investigated differentially expressed genes across each cell population. This analysis revealed the presence of two distinct subpopulations of IPF-related alveolar epithelial type II cells (IR_AT2). Subsequently, three unique gene co-expression modules associated with the IR_AT2 subtype were identified through the use of hdWGCNA. Furthermore, we refined and identified IPF-related AT2-related gene (IARG) signatures using various machine learning algorithms. Our analysis demonstrated a significant association between high IARG scores in IPF patients and shorter survival times (p-value < 0.01). Additionally, we observed a negative correlation between the percent predicted diffusing capacity for lung carbon monoxide (% DLCO) and increased IARG scores (cor = -0.44, p-value < 0.05). The cross-validation findings demonstrated a high level of accuracy (AUC > 0.85, p-value < 0.01) in the prognostication of patients with IPF utilizing the identified IARG signatures. Our study has identified distinct molecular and biological features among AT2 subpopulations, specifically highlighting the unique characteristics of IPF-related AT2 cells. Importantly, our findings underscore the prognostic relevance of specific genes associated with IPF-related AT2 cells, offering valuable insights into the advancement of IPF.
Collapse
Affiliation(s)
| | | | - Xin Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; (Z.Y.); (Y.Y.)
| | - Jiwei Hou
- Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; (Z.Y.); (Y.Y.)
| |
Collapse
|
25
|
Su W, Guo Y, Wang Q, Ma L, Zhang Q, Zhang Y, Geng Y, Jin T, Guo J, Yang R, Niu Z, Ren L, Wang Y, Ning Z, Li W, He W, Sun J, Li T, Li Z, Shan H, Liang H. YAP1 inhibits the senescence of alveolar epithelial cells by targeting Prdx3 to alleviate pulmonary fibrosis. Exp Mol Med 2024; 56:1643-1654. [PMID: 38945958 PMCID: PMC11297023 DOI: 10.1038/s12276-024-01277-0] [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: 08/26/2023] [Revised: 04/02/2024] [Accepted: 04/02/2024] [Indexed: 07/02/2024] Open
Abstract
The senescence of alveolar type II (AT2) cells impedes self-repair of the lung epithelium and contributes to lung injury in the setting of idiopathic pulmonary fibrosis (IPF). Yes-associated protein 1 (YAP1) is essential for cell growth and organ development; however, the role of YAP1 in AT2 cells during pulmonary fibrosis is still unclear. YAP1 expression was found to be downregulated in the AT2 cells of PF patients. Deletion of YAP1 in AT2 cells resulted in lung injury, exacerbated extracellular matrix (ECM) deposition, and worsened lung function. In contrast, overexpression of YAP1 in AT2 cells promoted alveolar regeneration, mitigated pulmonary fibrosis, and improved lung function. In addition, overexpression of YAP1 alleviated bleomycin (BLM) -induced senescence of alveolar epithelial cells both in vivo and in vitro. Moreover, YAP1 promoted the expression of peroxiredoxin 3 (Prdx3) by directly interacting with TEAD1. Forced expression of Prdx3 inhibited senescence and improved mitochondrial dysfunction in BLM-treated MLE-12 cells, whereas depletion of Prdx3 partially abrogated the protective effect of YAP1. Furthermore, overexpression of Prdx3 facilitated self-repair of the injured lung and reduced ECM deposition, while silencing Prdx3 attenuated the antifibrotic effect of YAP1. In conclusion, this study demonstrated that YAP1 alleviates lung injury and pulmonary fibrosis by regulating Prdx3 expression to improve mitochondrial dysfunction and block senescence in AT2 cells, revealing a potential novel therapeutic strategy for pulmonary fibrosis.
Collapse
Affiliation(s)
- Wei Su
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Yingying Guo
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Qianqian Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Lu Ma
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Qing Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Yuhan Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Yiding Geng
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Tongzhu Jin
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Jiayu Guo
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Ruoxuan Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Zhihui Niu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Lingxue Ren
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Yan Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Zhiwei Ning
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Wenyue Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Wenxin He
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, 200433, China
| | - Jian Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
- Zhuhai People's Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, Guangdong, China
| | - Tianyu Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Zhixin Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, 200433, China.
| | - Hongli Shan
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, China.
| | - Haihai Liang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China.
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China.
| |
Collapse
|
26
|
Liang N, Song W, Li J. BPA promotes lung fibrosis in mice by regulating autophagy-dependent ferroptosis in alveolar epithelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 278:116412. [PMID: 38691879 DOI: 10.1016/j.ecoenv.2024.116412] [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: 01/22/2024] [Revised: 04/23/2024] [Accepted: 04/27/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND Bisphenol A (BPA) is an industrial chemical that is commonly found in daily consumer products. BPA is reportedly associated with lung diseases. However, the impact of BPA on pulmonary fibrosis (PF) and its possible mechanisms of action both remain unclear. METHODS A PF mouse model was induced by bleomycin (BLM). Mouse lung fibroblasts (MLG 2908) and mouse alveolar epithelial cells (MLE-12) were treated with BPA to establish a PF cell model. Tissue staining, CCK-8 assays, western blot experiments and relevant indicator kits were used to detect and evaluate the effect of BPA on PF. RESULTS BPA dose-dependently promoted oxidative stress and induced ferroptosis, leading to PF. The ferroptosis inhibitor Fer-1 partly attenuated the effect of BPA. In addition, among the two main cell types associated with the progression of PF, MLE-12 cells are more sensitive to BPA than are MLG 2908 cells, and BPA induces ferroptosis in MLE-12 cells. Furthermore, BPA promoted autophagy-mediated ferroptosis by activating the AMPK/mTOR signaling pathway, thereby exacerbating the progression of PF. The autophagy inhibitor CQ1 partly attenuated the effect of BPA. CONCLUSION BPA promotes the progression of PF by promoting autophagy-dependent ferroptosis in alveolar epithelial cells, which provides a new theoretical basis for understanding BPA-induced PF.
Collapse
Affiliation(s)
- Ni Liang
- Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming, Yunnan 650500, China
| | - Wenyi Song
- Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming, Yunnan 650500, China
| | - Jing Li
- Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming, Yunnan 650500, China.
| |
Collapse
|
27
|
Wang Y, Zhang J, Liu Y, Yue X, Han K, Kong Z, Dong Y, Yang Z, Fu Z, Tang C, Shi C, Zhao X, Han M, Wang Z, Zhang Y, Chen C, Li A, Sun P, Zhu D, Zhao K, Jiang X. Realveolarization with inhalable mucus-penetrating lipid nanoparticles for the treatment of pulmonary fibrosis in mice. SCIENCE ADVANCES 2024; 10:eado4791. [PMID: 38865465 PMCID: PMC11168475 DOI: 10.1126/sciadv.ado4791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/08/2024] [Indexed: 06/14/2024]
Abstract
The stemness loss-associated dysregeneration of impaired alveolar type 2 epithelial (AT2) cells abolishes the reversible therapy of idiopathic pulmonary fibrosis (IPF). We here report an inhalable mucus-penetrating lipid nanoparticle (LNP) for codelivering dual mRNAs, promoting realveolarization via restoring AT2 stemness for IPF treatment. Inhalable LNPs were first formulated with dipalmitoylphosphatidylcholine and our in-house-made ionizable lipids for high-efficiency pulmonary mucus penetration and codelivery of dual messenger RNAs (mRNAs), encoding cytochrome b5 reductase 3 and bone morphogenetic protein 4, respectively. After being inhaled in a bleomycin model, LNPs reverses the mitochondrial dysfunction through ameliorating nicotinamide adenine dinucleotide biosynthesis, which inhibits the accelerated senescence of AT2 cells. Concurrently, pathological epithelial remodeling and fibroblast activation induced by impaired AT2 cells are terminated, ultimately prompting alveolar regeneration. Our data demonstrated that the mRNA-LNP system exhibited high protein expression in lung epithelial cells, which markedly extricated the alveolar collapse and prolonged the survival of fibrosis mice, providing a clinically viable strategy against IPF.
Collapse
Affiliation(s)
- Yan Wang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Jing Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Ying Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Xiao Yue
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Kun Han
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Zhichao Kong
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Yuanmin Dong
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Zhenmei Yang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Zhipeng Fu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Chunwei Tang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Chongdeng Shi
- Department of Emergency, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| | - Xiaotian Zhao
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Maosen Han
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Zhibin Wang
- Lingyi iTECH Manufacturing Co. Ltd., No. 2988, Taidong Road, Xiangcheng District, Suzhou, Jiangsu Province 215000, China
| | - Yulin Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Chen Chen
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College of Shandong University, Jinan, Shandong Province 250012, China
| | - Anning Li
- Department of Radiology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong Province 250012, China
| | - Peng Sun
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Danqing Zhu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, 4572A Academic Building, Clear Water Bay, Kowloon 999077 Hong Kong, China
| | - Kun Zhao
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Xinyi Jiang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| |
Collapse
|
28
|
Seasock MJ, Shafiquzzaman M, Ruiz-Echartea ME, Kanchi RS, Tran BT, Simon LM, Meyer MD, Erice PA, Lotlikar SL, Wenlock SC, Ochsner SA, Enright A, Carisey AF, Romero F, Rosas IO, King KY, McKenna NJ, Coarfa C, Rodriguez A. Let-7 restrains an oncogenic epigenetic circuit in AT2 cells to prevent ectopic formation of fibrogenic transitional cell intermediates and pulmonary fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595205. [PMID: 38826218 PMCID: PMC11142151 DOI: 10.1101/2024.05.22.595205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Analysis of lung alveolar type 2 (AT2) progenitor stem cells has highlighted fundamental mechanisms that direct their differentiation into alveolar type 1 cells (AT1s) in lung repair and disease. However, microRNA (miRNA) mediated post-transcriptional mechanisms which govern this nexus remain understudied. We show here that the let-7 miRNA family serves a homeostatic role in governance of AT2 quiescence, specifically by preventing the uncontrolled accumulation of AT2 transitional cells and by promoting AT1 differentiation to safeguard the lung from spontaneous alveolar destruction and fibrosis. Using mice and organoid models with genetic ablation of let-7a1/let-7f1/let-7d cluster (let-7afd) in AT2 cells, we demonstrate prevents AT1 differentiation and results in aberrant accumulation of AT2 transitional cells in progressive pulmonary fibrosis. Integration of enhanced AGO2 UV-crosslinking and immunoprecipitation sequencing (AGO2-eCLIP) with RNA-sequencing from AT2 cells uncovered the induction of direct targets of let-7 in an oncogene feed-forward regulatory network including BACH1/EZH2 which drives an aberrant fibrotic cascade. Additional analyses by CUT&RUN-sequencing revealed loss of let-7afd hampers AT1 differentiation by eliciting aberrant histone EZH2 methylation which prevents the exit of AT2 transitional cells into terminal AT1s. This study identifies let-7 as a key gatekeeper of post-transcriptional and epigenetic chromatin signals to prevent AT2-driven pulmonary fibrosis.
Collapse
Affiliation(s)
- Matthew J. Seasock
- Immunology & Microbiology Graduate Program, Baylor College of Medicine, Houston, TX, 77030
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
| | - Md Shafiquzzaman
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
| | - Maria E. Ruiz-Echartea
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
| | - Rupa S. Kanchi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine Houston, TX, 77030
| | - Brandon T. Tran
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030
- Department of Pediatrics, Division of Infectious Diseases, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, 77030
| | - Lukas M. Simon
- Therapeutic Innovation Center, Baylor College of Medicine, Houston, TX, 77030
| | | | - Phillip A. Erice
- Immunology & Microbiology Graduate Program, Baylor College of Medicine, Houston, TX, 77030
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
| | - Shivani L. Lotlikar
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
| | | | - Scott A. Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
| | - Anton Enright
- Department of Pathology, University of Cambridge, Cambridge, CB2 1TN, UK
| | - Alex F. Carisey
- William T. Shearer Center for Immunobiology, Texas Children’s Hospital, Houston, TX, 77030
- Current Address: Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Freddy Romero
- Department of Medicine, Section of Pulmonary and Critical Care, Baylor College of Medicine. Houston, TX, 77030
- Current Address: Vertex Pharmaceuticals, 3215 Merryfield Row, San Diego, CA, 92121
| | - Ivan O. Rosas
- Department of Medicine, Section of Pulmonary and Critical Care, Baylor College of Medicine. Houston, TX, 77030
| | - Katherine Y. King
- Department of Pediatrics, Division of Infectious Diseases, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, 77030
| | - Neil J. McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine Houston, TX, 77030
| | - Antony Rodriguez
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine Houston, TX, 77030
- Center for Translational Research on Inflammatory Diseases, Michael E. Debakey, Baylor College of Medicine, Houston, TX, 77030
| |
Collapse
|
29
|
Zhu Y, Meng X, Zhu X, Zhang J, Lv H, Wang F, Wang J, Chen C, Chen M, Wang D, Jin W, Tian R, Wang R. Circular RNA MKLN1 promotes epithelial-mesenchymal transition in pulmonary fibrosis by regulating the miR-26a/b-5p/CDK8 axis in human alveolar epithelial cells and mice models. Arch Toxicol 2024; 98:1399-1413. [PMID: 38460002 PMCID: PMC10965569 DOI: 10.1007/s00204-024-03700-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: 10/13/2023] [Accepted: 01/31/2024] [Indexed: 03/11/2024]
Abstract
Pulmonary fibrosis involves destruction of the lung parenchyma and extracellular matrix deposition. Effective treatments for pulmonary fibrosis are lacking and its pathogenesis is still unclear. Studies have found that epithelial-mesenchymal transition (EMT) of alveolar epithelial cells (AECs) plays an important role in progression of pulmonary fibrosis. Thus, an in-depth exploration of its mechanism might identify new therapeutic targets. In this study, we revealed that a novel circular RNA, MKLN1 (circMKLN1), was significantly elevated in two pulmonary fibrosis models (intraperitoneally with PQ, 50 mg/kg for 7 days, and intratracheally with BLM, 5 mg/kg for 28 days). Additionally, circMKLN1 was positively correlated with the severity of pulmonary fibrosis. Inhibition of circMKLN1 expression significantly reduced collagen deposition and inhibited EMT in AECs. EMT was aggravated after circMKLN1 overexpression in AECs. MiR-26a-5p/miR-26b-5p (miR-26a/b), the targets of circMKLN1, were confirmed by luciferase reporter assays. CircMKLN1 inhibition elevated miR-26a/b expression. Significantly decreased expression of CDK8 (one of the miR-26a/b targets) was observed after inhibition of circMKLN1. EMT was exacerbated again, and CDK8 expression was significantly increased after circMKLN1 inhibition and cotransfection of miR-26a/b inhibitors in AECs. Our research indicated that circMKLN1 promoted CDK8 expression through sponge adsorption of miR-26a/b, which regulates EMT and pulmonary fibrosis. This study provides a theoretical basis for finding new targets or biomarkers in pulmonary fibrosis.
Collapse
Affiliation(s)
- Yong Zhu
- Department of Critical Care Medicine, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, 650 Xinsongjiang Road, Shanghai, 201620, China
| | - Xiaoxiao Meng
- Department of Critical Care Medicine, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, 650 Xinsongjiang Road, Shanghai, 201620, China
| | - Xian Zhu
- Department of Critical Care Medicine, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, 650 Xinsongjiang Road, Shanghai, 201620, China
| | - Jiaxiang Zhang
- Department of Critical Care Medicine, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, 650 Xinsongjiang Road, Shanghai, 201620, China
| | - Hui Lv
- Department of Critical Care Medicine, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, 650 Xinsongjiang Road, Shanghai, 201620, China
| | - Feiyao Wang
- Department of Critical Care Medicine, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, 650 Xinsongjiang Road, Shanghai, 201620, China
| | - Jinfeng Wang
- Department of Critical Care Medicine, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, 650 Xinsongjiang Road, Shanghai, 201620, China
| | - Cheng Chen
- Department of Critical Care Medicine, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, 650 Xinsongjiang Road, Shanghai, 201620, China
| | - Mengting Chen
- Department of Critical Care Medicine, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, 650 Xinsongjiang Road, Shanghai, 201620, China
| | - Dapeng Wang
- Department of Intensive Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, 214021, Jiangsu, China
| | - Wei Jin
- Department of Critical Care Medicine, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, 650 Xinsongjiang Road, Shanghai, 201620, China.
| | - Rui Tian
- Department of Critical Care Medicine, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, 650 Xinsongjiang Road, Shanghai, 201620, China.
| | - Ruilan Wang
- Department of Critical Care Medicine, School of Medicine, Shanghai General Hospital, Shanghai Jiaotong University, 650 Xinsongjiang Road, Shanghai, 201620, China.
| |
Collapse
|
30
|
Chong L, Zou L, Xiang L, Song X, Miao W, Yan X, Xu M, Ling G, El Agha E, Bellusci S, Lou Z, Zhang H, Zhang JS. WSB1, a Hypoxia-Inducible E3 Ligase, Promotes Myofibroblast Accumulation and Attenuates Alveolar Epithelial Regeneration in Mouse Lung Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:656-672. [PMID: 38325552 DOI: 10.1016/j.ajpath.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 12/19/2023] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
Idiopathic pulmonary fibrosis is a progressive interstitial lung disease for which there is no curative therapy available. Repetitive alveolar epithelial injury repair, myofibroblast accumulation, and excessive collagen deposition are key pathologic features of idiopathic pulmonary fibrosis, eventually leading to cellular hypoxia and respiratory failure. The precise mechanism driving this complex maladaptive process remains inadequately understood. WD repeat and suppressor of cytokine signaling box containing 1 (WSB1) is an E3 ubiquitin ligase, the expression of which is associated strongly with hypoxia, and forms a positive feedback loop with hypoxia-inducible factor 1α (HIF-1α) under anoxic condition. This study explored the expression, cellular distribution, and function of WSB1 in bleomycin (BLM)-induced mouse lung injury and fibrosis. WSB1 expression was highly induced by BLM injury and correlated with the progression of lung fibrosis. Significantly, conditional deletion of Wsb1 in adult mice ameliorated BLM-induced pulmonary fibrosis. Phenotypically, Wsb1-deficient mice showed reduced lipofibroblast to myofibroblast transition, but enhanced alveolar type 2 proliferation and differentiation into alveolar type 1 after BLM injury. Proteomic analysis of mouse lung tissues identified caveolin 2 as a potential downstream target of WSB1, contributing to BLM-induced epithelial injury repair and fibrosis. These findings unravel a vital role for WSB1 induction in lung injury repair, thus highlighting it as a potential therapeutic target for pulmonary fibrosis.
Collapse
Affiliation(s)
- Lei Chong
- Department of Pediatric Respiratory Medicine, National Key Clinical Specialty of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lihui Zou
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liyan Xiang
- Department of Pediatric Respiratory Medicine, National Key Clinical Specialty of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinyue Song
- Department of Pediatric Respiratory Medicine, National Key Clinical Specialty of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wanqi Miao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, and Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Wenzhou, China
| | - Xihua Yan
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ming Xu
- Department of Pediatric Respiratory Medicine, National Key Clinical Specialty of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gongxia Ling
- Department of Pediatric Respiratory Medicine, National Key Clinical Specialty of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Elie El Agha
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus-Liebig University Giessen, Giessen, Germany
| | - Saverio Bellusci
- Cardio-Pulmonary Institute, Institute for Lung Health, German Center for Lung Research, Justus-Liebig University Giessen, Giessen, Germany
| | - Zhenkun Lou
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Hailin Zhang
- Department of Pediatric Respiratory Medicine, National Key Clinical Specialty of Pediatric Respiratory Medicine, Institute of Pediatrics, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Jin-San Zhang
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, and Zhejiang Provincial Key Laboratory of Interventional Pulmonology, Wenzhou, China.
| |
Collapse
|
31
|
Wang R, Shi Y, Lv Y, Xie C, Hu Y. The novel insights of epithelial-derived exosomes in various fibrotic diseases. Biomed Pharmacother 2024; 174:116591. [PMID: 38631144 DOI: 10.1016/j.biopha.2024.116591] [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: 01/14/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
The characteristics of fibrosis include the abnormal accumulation of extracellular matrix proteins and abnormal tissue repair caused by injury, infection, and inflammation, leading to a significant increase in organ failure and mortality. Effective and precise treatments are urgently needed to halt and reverse the progression of fibrotic diseases. Exosomes are tiny vesicles derived from endosomes, spanning from 40 to 160 nanometers in diameter, which are expelled into the extracellular matrix environment by various cell types. They play a crucial role in facilitating cell-to-cell communication by transporting a variety of cargoes, including proteins, RNA, and DNA. Epithelial cells serve as the primary barrier against diverse external stimuli that precipitate fibrotic diseases. Numerous research suggests that exosomes from epithelial cells have a significant impact on several fibrotic diseases. An in-depth comprehension of the cellular and molecular mechanisms of epithelial cell-derived exosomes in fibrosis holds promise for advancing the exploration of novel diagnostic biomarkers and clinical drug targets. In this review, we expand upon the pathogenic mechanisms of epithelium-derived exosomes and highlight their role in the fibrotic process by inducing inflammation and activating fibroblasts. In addition, we are particularly interested in the bioactive molecules carried by epithelial-derived exosomes and their potential value in the diagnosis and treatment of fibrosis and delineate the clinical utility of exosomes as an emerging therapeutic modality, highlighting their potential application in addressing various medical conditions.
Collapse
Affiliation(s)
- Rifu Wang
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yuxin Shi
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yonglin Lv
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Changqing Xie
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, China.
| | - Yanjia Hu
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China.
| |
Collapse
|
32
|
Shao M, Cheng H, Li X, Qiu Y, Zhang Y, Chang Y, Fu J, Shen M, Xu X, Feng D, Han Y, Yue S, Zhou Y, Luo Z. Abnormal mitochondrial iron metabolism damages alveolar type II epithelial cells involved in bleomycin-induced pulmonary fibrosis. Theranostics 2024; 14:2687-2705. [PMID: 38773980 PMCID: PMC11103499 DOI: 10.7150/thno.94072] [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: 01/09/2024] [Accepted: 04/08/2024] [Indexed: 05/24/2024] Open
Abstract
Rationale: Pulmonary fibrosis is a chronic progressive lung disease with limited therapeutic options. We previously revealed that there is iron deposition in alveolar epithelial type II cell (AECII) in pulmonary fibrosis, which can be prevented by the iron chelator deferoxamine. However, iron in the cytoplasm and the mitochondria has two relatively independent roles and regulatory systems. In this study, we aimed to investigate the role of mitochondrial iron deposition in AECII injury and pulmonary fibrosis, and to find potential therapeutic strategies. Methods: BLM-treated mice, MLE-12 cells, and primary AECII were employed to establish the mouse pulmonary fibrosis model and epithelial cells injury model, respectively. Mitochondrial transplantation, siRNA and plasmid transfection, western blotting (WB), quantitative real-time polymerase chain reaction (RT-qPCR), polymerase chain reaction (PCR), immunofluorescence, immunoprecipitation (IP), MitoSOX staining, JC-1 staining, oxygen consumption rate (OCR) measurement, and Cell Counting Kit-8 (CCK8) assay were utilized to elucidate the role of mitochondrial iron deposition in cell and lung fibrosis and determine its mechanism. Results: This study showed that prominent mitochondrial iron deposition occurs within AECII in bleomycin (BLM)-induced pulmonary fibrosis mouse model and in BLM-treated MLE-12 epithelial cells. Further, the study revealed that healthy mitochondria rescue BLM-damaged AECII mitochondrial iron deposition and cell damage loss. Mitoferrin-2 (MFRN2) is the main transporter that regulates mitochondrial iron metabolism by transferring cytosolic iron into mitochondria, which is upregulated in BLM-treated MLE-12 epithelial cells. Direct overexpression of MFRN2 causes mitochondrial iron deposition and cell damage. In this study, decreased ubiquitination of the ubiquitin ligase F-box/LRR-repeat protein 5 (FBXL5) degraded iron-reactive element-binding protein 2 (IREB2) and promoted MFRN2 expression as well as mitochondrial iron deposition in damaged AECII. Activation of the prostaglandin E2 receptor EP4 subtype (EP4) receptor signaling pathway counteracted mitochondrial iron deposition by downregulating IREB2-MFRN2 signaling through upregulation of FBXL5. This intervention not only reduced mitochondrial iron content but also preserved mitochondrial function and protected against AECII damage after BLM treatment. Conclusion: Our findings highlight the unexplored roles, mechanisms, and regulatory approaches of abnormal mitochondrial iron metabolism of AECII in pulmonary fibrosis. Therefore, this study deepens the understanding of the mechanisms underlying pulmonary fibrosis and offers a promising strategy for developing effective therapeutic interventions using the EP4 receptor activator.
Collapse
Affiliation(s)
- Min Shao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Haipeng Cheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China
| | - Xiaohong Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China
| | - Yujia Qiu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yunna Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yanfen Chang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Jiafeng Fu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Mengxia Shen
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xinxin Xu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Dandan Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yang Han
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - ShaoJie Yue
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Ziqiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, Hunan, 410013, China
| |
Collapse
|
33
|
Chu L, Zhuo J, Huang H, Chen W, Zhong W, Zhang J, Meng X, Zou F, Cai S, Zou M, Dong H. Tetrandrine alleviates pulmonary fibrosis by inhibiting alveolar epithelial cell senescence through PINK1/Parkin-mediated mitophagy. Eur J Pharmacol 2024; 969:176459. [PMID: 38438063 DOI: 10.1016/j.ejphar.2024.176459] [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: 12/02/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/06/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal and insidious interstitial lung disease. So far, there are no effective drugs for preventing the disease process. Cellular senescence plays a critical role in the development of IPF, with the senescence and insufficient mitophagy of alveolar epithelial cells being implicated in its pathogenesis. Tetrandrine is a natural alkaloid which is now produced synthetically. It was known that the tetrandrine has anti-fibrotic effects, but the efficacy and mechanisms are still not well evaluated. Here, we reveal the roles of tetrandrine on AECs senescence and the antifibrotic effects by using a bleomycin challenged mouse model of pulmonary fibrosis and a bleomycin-stimulated mouse alveolar epithelial cell line (MLE-12). We performed the β-galactosidase staining, immunohistochemistry and fluorescence to assess senescence in MLE-12 cells. The mitophagy levels were detected by co-localization of LC3 and COVIX. Our findings indicate that tetrandrine suppressed bleomycin-induced fibroblast activation and ultimately blocked the increase of collagen deposition in mouse model lung tissue. It has significantly inhibited the bleomycin-induced senescence and senescence-associated secretory phenotype (SASP) in alveolar epithelial cells (AECs). Mechanistically, tetrandrine suppressed the decrease of mitochondrial autophagy-related protein expression to rescue the bleomycin-stimulated impaired mitophagy in MLE-12 cells. We revealed that knockdown the putative kinase 1 (PINK1) gene by a short interfering RNA (siRNA) could abolish the ability of tetrandrine and reverse the MLE-12 cells senescence, which indicated the mitophagy of MLE-12 cells is PINK1 dependent. Our data suggest the tetrandrine could be a novel and effective drug candidate for lung fibrosis and senescence-related fibrotic diseases.
Collapse
Affiliation(s)
- Lanhe Chu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinzhong Zhuo
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haohua Huang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weimou Chen
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenshan Zhong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinming Zhang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaojing Meng
- School of Public Health, Southern Medical University, Guangzhou, China
| | - Fei Zou
- School of Public Health, Southern Medical University, Guangzhou, China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengchen Zou
- Department of Endocrinology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Hangming Dong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
34
|
Shi X, Chen Y, Shi M, Gao F, Huang L, Wang W, Wei D, Shi C, Yu Y, Xia X, Song N, Chen X, Distler JHW, Lu C, Chen J, Wang J. The novel molecular mechanism of pulmonary fibrosis: insight into lipid metabolism from reanalysis of single-cell RNA-seq databases. Lipids Health Dis 2024; 23:98. [PMID: 38570797 PMCID: PMC10988923 DOI: 10.1186/s12944-024-02062-8] [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: 12/26/2023] [Accepted: 02/27/2024] [Indexed: 04/05/2024] Open
Abstract
Pulmonary fibrosis (PF) is a severe pulmonary disease with limited available therapeutic choices. Recent evidence increasingly points to abnormal lipid metabolism as a critical factor in PF pathogenesis. Our latest research identifies the dysregulation of low-density lipoprotein (LDL) is a new risk factor for PF, contributing to alveolar epithelial and endothelial cell damage, and fibroblast activation. In this study, we first integrative summarize the published literature about lipid metabolite changes found in PF, including phospholipids, glycolipids, steroids, fatty acids, triglycerides, and lipoproteins. We then reanalyze two single-cell RNA-sequencing (scRNA-seq) datasets of PF, and the corresponding lipid metabolomic genes responsible for these lipids' biosynthesis, catabolism, transport, and modification processes are uncovered. Intriguingly, we found that macrophage is the most active cell type in lipid metabolism, with almost all lipid metabolic genes being altered in macrophages of PF. In type 2 alveolar epithelial cells, lipid metabolic differentially expressed genes (DEGs) are primarily associated with the cytidine diphosphate diacylglycerol pathway, cholesterol metabolism, and triglyceride synthesis. Endothelial cells are partly responsible for sphingomyelin, phosphatidylcholine, and phosphatidylethanolamines reprogramming as their metabolic genes are dysregulated in PF. Fibroblasts may contribute to abnormal cholesterol, phosphatidylcholine, and phosphatidylethanolamine metabolism in PF. Therefore, the reprogrammed lipid profiles in PF may be attributed to the aberrant expression of lipid metabolic genes in different cell types. Taken together, these insights underscore the potential of targeting lipid metabolism in developing innovative therapeutic strategies, potentially leading to extended overall survival in individuals affected by PF.
Collapse
Affiliation(s)
- Xiangguang Shi
- Department of Dermatology, Huashan Hospital, and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yahui Chen
- Human Phenome Institute, and Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China Fudan University, Shanghai, China
| | - Mengkun Shi
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Fei Gao
- Wuxi Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, China
| | - Lihao Huang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism & Integrative Biology, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Wei Wang
- Wuxi Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, China
- MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Dong Wei
- Wuxi Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, China
| | - Chenyi Shi
- MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yuexin Yu
- Human Phenome Institute, and Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China Fudan University, Shanghai, China
| | - Xueyi Xia
- Human Phenome Institute, and Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China Fudan University, Shanghai, China
| | - Nana Song
- Department of Nephrology, Zhongshan Hospital, Fudan University, Fudan Zhangjiang Institute, Shanghai, People's Republic of China
| | - Xiaofeng Chen
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jörg H W Distler
- Department of Internal Medicine 3 and Institute for Clinical Immunology, University of Erlangen, Nuremberg, Germany
| | - Chenqi Lu
- MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China.
| | - Jingyu Chen
- Wuxi Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, China.
- Center for Lung Transplantation, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jiucun Wang
- Department of Dermatology, Huashan Hospital, and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China.
- Human Phenome Institute, and Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China Fudan University, Shanghai, China.
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Beijing, China.
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
| |
Collapse
|
35
|
Luo L, Zhang W, You S, Cui X, Tu H, Yi Q, Wu J, Liu O. The role of epithelial cells in fibrosis: Mechanisms and treatment. Pharmacol Res 2024; 202:107144. [PMID: 38484858 DOI: 10.1016/j.phrs.2024.107144] [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: 11/02/2023] [Revised: 02/19/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Fibrosis is a pathological process that affects multiple organs and is considered one of the major causes of morbidity and mortality in multiple diseases, resulting in an enormous disease burden. Current studies have focused on fibroblasts and myofibroblasts, which directly lead to imbalance in generation and degradation of extracellular matrix (ECM). In recent years, an increasing number of studies have focused on the role of epithelial cells in fibrosis. In some cases, epithelial cells are first exposed to external physicochemical stimuli that may directly drive collagen accumulation in the mesenchyme. In other cases, the source of stimulation is mainly immune cells and some cytokines, and epithelial cells are similarly altered in the process. In this review, we will focus on the multiple dynamic alterations involved in epithelial cells after injury and during fibrogenesis, discuss the association among them, and summarize some therapies targeting changed epithelial cells. Especially, epithelial mesenchymal transition (EMT) is the key central step, which is closely linked to other biological behaviors. Meanwhile, we think studies on disruption of epithelial barrier, epithelial cell death and altered basal stem cell populations and stemness in fibrosis are not appreciated. We believe that therapies targeted epithelial cells can prevent the progress of fibrosis, but not reverse it. The epithelial cell targeting therapies will provide a wonderful preventive and delaying action.
Collapse
Affiliation(s)
- Liuyi Luo
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Wei Zhang
- Department of Oral Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Siyao You
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Xinyan Cui
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Hua Tu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Qiao Yi
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Jianjun Wu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| | - Ousheng Liu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
36
|
Zheng M, Zhu W, Gao F, Zhuo Y, Zheng M, Wu G, Feng C. Novel inhalation therapy in pulmonary fibrosis: principles, applications and prospects. J Nanobiotechnology 2024; 22:136. [PMID: 38553716 PMCID: PMC10981316 DOI: 10.1186/s12951-024-02407-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 03/18/2024] [Indexed: 04/01/2024] Open
Abstract
Pulmonary fibrosis (PF) threatens millions of people worldwide with its irreversible progression. Although the underlying pathogenesis of PF is not fully understood, there is evidence to suggest that the disease can be blocked at various stages. Inhalation therapy has been applied for lung diseases such as asthma and chronic obstructive pulmonary disease, and its application for treating PF is currently under consideration. New techniques in inhalation therapy, such as the application of microparticles and nanoparticles, traditional Chinese medicine monomers, gene therapy, inhibitors, or agonists of signaling pathways, extracellular vesicle interventions, and other specific drugs, are effective in treating PF. However, the safety and effectiveness of these therapeutic techniques are influenced by the properties of inhaled particles, biological and pathological barriers, and the type of inhalation device used. This review provides a comprehensive overview of the pharmacological, pharmaceutical, technical, preclinical, and clinical experimental aspects of novel inhalation therapy for treating PF and focus on therapeutic methods that significantly improve existing technologies or expand the range of drugs that can be administered via inhalation. Although inhalation therapy for PF has some limitations, the advantages are significant, and further research and innovation about new inhalation techniques and drugs are encouraged.
Collapse
Affiliation(s)
- Meiling Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100010, China
- Peking University People's Hospital, Beijing, 100032, China
| | - Wei Zhu
- Department of Ophthalmology, Changshu No. 2 People's Hospital, Changshu, 215500, China
| | - Fei Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Yu Zhuo
- Department of Medical Oncology Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100010, China
| | - Mo Zheng
- Department of Medical Oncology Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100010, China
| | - Guanghao Wu
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China.
| | - Cuiling Feng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100010, China.
- Peking University People's Hospital, Beijing, 100032, China.
| |
Collapse
|
37
|
Zhang K, Yao E, Aung T, Chuang PT. The alveolus: Our current knowledge of how the gas exchange unit of the lung is constructed and repaired. Curr Top Dev Biol 2024; 159:59-129. [PMID: 38729684 DOI: 10.1016/bs.ctdb.2024.01.002] [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] [Indexed: 05/12/2024]
Abstract
The mammalian lung completes its last step of development, alveologenesis, to generate sufficient surface area for gas exchange. In this process, multiple cell types that include alveolar epithelial cells, endothelial cells, and fibroblasts undergo coordinated cell proliferation, cell migration and/or contraction, cell shape changes, and cell-cell and cell-matrix interactions to produce the gas exchange unit: the alveolus. Full functioning of alveoli also involves immune cells and the lymphatic and autonomic nervous system. With the advent of lineage tracing, conditional gene inactivation, transcriptome analysis, live imaging, and lung organoids, our molecular understanding of alveologenesis has advanced significantly. In this review, we summarize the current knowledge of the constituents of the alveolus and the molecular pathways that control alveolar formation. We also discuss how insight into alveolar formation may inform us of alveolar repair/regeneration mechanisms following lung injury and the pathogenic processes that lead to loss of alveoli or tissue fibrosis.
Collapse
Affiliation(s)
- Kuan Zhang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Erica Yao
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Thin Aung
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Pao-Tien Chuang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States.
| |
Collapse
|
38
|
Zhang L, Wang Y, Tian L, Li L, Chen Z, Ding C, Tian J, Song D, Yao S, Ren W. Thrombospondin-1-mediated crosstalk between autophagy and oxidative stress orchestrates repair of blast lung injury. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167026. [PMID: 38237742 DOI: 10.1016/j.bbadis.2024.167026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
Coal mining carries inherent risks of catastrophic gas explosions capable of inflicting severe lung injury. Using complementary in vivo and in vitro models, we explored mechanisms underlying alveolar epithelial damage and repair following a gas explosion in this study. In a rat model, the gas explosion was demonstrated to trigger inflammation and injury within the alveolar epithelium. The following scRNA-sequencing revealed that alveolar epithelial cells exhibited the most profound transcriptomic changes after gas explosion compared to other pulmonary cell types. In the L2 alveolar epithelial cells, the blast was found to cause autophagic flux by inducing autophagosome formation, LC3 lipidation, and p62 degradation. Transcriptomic profiling of the L2 cells identified PI3K-Akt and p53 pathways as critical modulators governing autophagic and oxidative stress responses to blast damage. Notably, Thrombospondin-1 (Thbs1) was determined for the first time as a pivotal node interconnecting these two pathways. The findings of this study illuminate intricate mechanisms of alveolar epithelial injury and recovery after blast trauma, highlighting autophagic and oxidative stress responses mediated by Thbs1-associated PI3K-Akt and p53 pathways as high-value therapeutic targets, and strategic modulation of these pathways in future studies may mitigate lung damage by reducing oxidative stress while engaging endogenous tissue repair processes like autophagy.
Collapse
Affiliation(s)
- Lin Zhang
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang 453000, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China; Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250001, China; Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Jinan 250001, China.
| | - Yongheng Wang
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Linqiang Tian
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang 453000, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China
| | - Long Li
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang 453000, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China
| | - Zhen Chen
- School of Public Health, Weifang Medical University, Weifang 261000, China
| | - Chunjie Ding
- Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, Henan, China
| | - Jiaqi Tian
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250001, China; Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Jinan 250001, China
| | - Dandan Song
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250001, China; Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Jinan 250001, China
| | - Sanqiao Yao
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang 453000, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China.
| | - Wenjie Ren
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang 453000, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China.
| |
Collapse
|
39
|
Zhang S, Tu D, Liu W, Li R, Jiang M, Yuan X, Luan J, Li H, Lv C, Song X. circELP2 reverse-splicing biogenesis and function as a pro-fibrogenic factor by targeting mitochondrial quality control pathway. J Cell Mol Med 2024; 28:e18098. [PMID: 38159063 PMCID: PMC10844706 DOI: 10.1111/jcmm.18098] [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: 08/08/2023] [Revised: 11/14/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is considered as a chronic, fibrosing interstitial pneumonia with unknown mechanism. The present work aimed to explore the function, biogenesis and regulatory mechanism of circELP2 in pulmonary fibrosis and evaluate the value of blocking circELP2-medicated signal pathway for IPF treatment. The results showed that heterogeneous nuclear ribonucleoprotein L initiated reverse splicing of circELP2 resulting in the increase of circELP2 generation. The biogenetic circELP2 activated the abnormal proliferation and migration of fibroblast and extracellular matrix deposition to promote pulmonary fibrogenesis. Mechanistic studies demonstrated that cytoplasmic circELP2 sponged miR-630 to increase transcriptional co-activators Yes-associated protein 1 (YAP1) and transcriptional co-activator with PDZ-binding motif (TAZ). Then, YAP1/TAZ bound to the promoter regions of their target genes, such as mTOR, Raptor and mLST8, which in turn activated or inhibited the genes expression in mitochondrial quality control pathway. Finally, the overexpressed circELP2 and miR-630 mimic were packaged into adenovirus vector for spraying into the mice lung to evaluate therapeutic effect of blocking circELP2-miR-630-YAP1/TAZ-mitochondrial quality control pathway in vivo. In conclusion, blocking circELP2-medicated pathway can alleviate pulmonary fibrosis, and circELP2 may be a potential target to treat lung fibrosis.
Collapse
Affiliation(s)
- Songzi Zhang
- Department of Respiratory and Critical Care MedicineBinzhou Medical University Hospital, Binzhou Medical UniversityBinzhouChina
- Department of Cellular and Genetic MedicineBinzhou Medical UniversityYantaiChina
| | - Diwei Tu
- Department of Respiratory and Critical Care MedicineBinzhou Medical University Hospital, Binzhou Medical UniversityBinzhouChina
| | - Weili Liu
- Department of Respiratory and Critical Care MedicineBinzhou Medical University Hospital, Binzhou Medical UniversityBinzhouChina
| | - Ruiqiong Li
- Department of Clinical NursingBinzhou Medical University Hospital, Binzhou Medical UniversityBinzhouChina
| | - Mengqi Jiang
- Department of Cellular and Genetic MedicineBinzhou Medical UniversityYantaiChina
| | - Xinglong Yuan
- Department of Respiratory and Critical Care MedicineBinzhou Medical University Hospital, Binzhou Medical UniversityBinzhouChina
| | - Jianlin Luan
- Department of Cellular and Genetic MedicineBinzhou Medical UniversityYantaiChina
| | - Hongbo Li
- Department of Respiratory and Critical Care MedicineBinzhou Medical University Hospital, Binzhou Medical UniversityBinzhouChina
| | - Changjun Lv
- Department of Respiratory and Critical Care MedicineBinzhou Medical University Hospital, Binzhou Medical UniversityBinzhouChina
| | - Xiaodong Song
- Department of Respiratory and Critical Care MedicineBinzhou Medical University Hospital, Binzhou Medical UniversityBinzhouChina
- Department of Cellular and Genetic MedicineBinzhou Medical UniversityYantaiChina
| |
Collapse
|
40
|
Chen Y, Li Z, Ji G, Wang S, Mo C, Ding B. Lung regeneration: diverse cell types and the therapeutic potential. MedComm (Beijing) 2024; 5:e494. [PMID: 38405059 PMCID: PMC10885188 DOI: 10.1002/mco2.494] [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: 07/30/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
Lung tissue has a certain regenerative ability and triggers repair procedures after injury. Under controllable conditions, lung tissue can restore normal structure and function. Disruptions in this process can lead to respiratory system failure and even death, causing substantial medical burden. The main types of respiratory diseases are chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and acute respiratory distress syndrome (ARDS). Multiple cells, such as lung epithelial cells, endothelial cells, fibroblasts, and immune cells, are involved in regulating the repair process after lung injury. Although the mechanism that regulates the process of lung repair has not been fully elucidated, clinical trials targeting different cells and signaling pathways have achieved some therapeutic effects in different respiratory diseases. In this review, we provide an overview of the cell type involved in the process of lung regeneration and repair, research models, and summarize molecular mechanisms involved in the regulation of lung regeneration and fibrosis. Moreover, we discuss the current clinical trials of stem cell therapy and pharmacological strategies for COPD, IPF, and ARDS treatment. This review provides a reference for further research on the molecular and cellular mechanisms of lung regeneration, drug development, and clinical trials.
Collapse
Affiliation(s)
- Yutian Chen
- The Department of Endovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan UniversityChengduChina
| | - Zhen Li
- The Department of Endovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Gaili Ji
- Department of GynecologyThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Shaochi Wang
- Department of Translational MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Chunheng Mo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan UniversityChengduChina
| | - Bi‐Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
41
|
Song C, Xu Z, Liang Q, Mu Y, Liu M, Wu Z, Li X, Li J, Chen H, Wang Y, Gao S, Li A, Yao W, Liu G. OGG1 promoted lung fibrosis by activating fibroblasts via interacting with Snail1. Int Immunopharmacol 2024; 126:111148. [PMID: 37977070 DOI: 10.1016/j.intimp.2023.111148] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/01/2023] [Accepted: 10/18/2023] [Indexed: 11/19/2023]
Abstract
One of abundant DNA lesions induced by reactive oxygen species is 8-oxoguanine (8-oxoG), which compromises genetic instability. 8-oxoG is recognized by the DNA repair protein 8-oxoguanine DNA glycosylase-1 (OGG1) that not only participates in base excision repair but also involves in transcriptional regulation.OGG1 has an important role inIdiopathic Pulmonary Fibrosis (IPF) processing and targeting fibroblasts is a major strategy for the treatment of pulmonary fibrosis, but whether OGG1 activate fibroblast is not clear. In this study, we show that OGG1 expression level is increased at the fibroblast activation stage in mouse lungs induced by bleomycin (BLM) treatment. OGG1 promoted the expression level of fibroblast activation markers (CTGF, fibronectin, and collagen 1) in a pro-fibrotic gene transcriptional regulation pathway via interacting with Snail1, which dependent on 8-oxoG recognition. Global inhibition of OGG1 at the middle stage of lung fibrosis also relieved BLM-induced lung fibrosis in mice. Our results suggest that OGG1 is a target for inhibiting fibroblast activation and a potential therapeutic target for IPF.
Collapse
Affiliation(s)
- Chuge Song
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Department of Respiratory Medicine, Second Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Zhiliang Xu
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Department of Breast Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Dongguan Institute of Respiratory Medicine, Guangdong Medical University, Dongguan 523121, China.
| | - Qingyun Liang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Department of Breast Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Yifan Mu
- Department of Respiratory Medicine, Second Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Manqi Liu
- Department of Cardiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Zijun Wu
- Department of Respiratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Xiaomin Li
- Department of Cardiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Jiali Li
- Department of Respiratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Hongqiao Chen
- Department of Cardiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Yahong Wang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Shenglan Gao
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Ao Li
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Weimin Yao
- Department of Respiratory Medicine, Second Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China.
| | - Gang Liu
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China.
| |
Collapse
|
42
|
Isshiki T, Naiel S, Vierhout M, Otsubo K, Ali P, Tsubouchi K, Yazdanshenas P, Kumaran V, Dvorkin-Gheva A, Kolb MRJ, Ask K. Therapeutic strategies to target connective tissue growth factor in fibrotic lung diseases. Pharmacol Ther 2024; 253:108578. [PMID: 38103794 DOI: 10.1016/j.pharmthera.2023.108578] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
The treatment of interstitial lung diseases, including idiopathic pulmonary fibrosis (IPF), remains challenging as current available antifibrotic agents are not effective in halting disease progression. Connective tissue growth factor (CTGF), also known as cellular communication factor 2 (CCN2), is a member of the CCN family of proteins that regulates cell signaling through cell surface receptors such as integrins, the activity of cytokines/growth factors, and the turnover of extracellular matrix (ECM) proteins. Accumulating evidence indicates that CTGF plays a crucial role in promoting lung fibrosis through multiple processes, including inducing transdifferentiation of fibroblasts to myofibroblasts, epithelial-mesenchymal transition (EMT), and cooperating with other fibrotic mediators such as TGF-β. Increased expression of CTGF has been observed in fibrotic lungs and inhibiting CTGF signaling has been shown to suppress lung fibrosis in several animal models. Thus, the CTGF signaling pathway is emerging as a potential therapeutic target in IPF and other pulmonary fibrotic conditions. This review provides a comprehensive overview of the current evidence on the pathogenic role of CTGF in pulmonary fibrosis and discusses the current therapeutic agents targeting CTGF using a systematic review approach.
Collapse
Affiliation(s)
- Takuma Isshiki
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada; Department of Respiratory Medicine, Toho University School of Medicine, 6-11-1 Omori Nisi, Ota-ku, Tokyo 143-8541, Japan
| | - Safaa Naiel
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Megan Vierhout
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Kohei Otsubo
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Pareesa Ali
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Kazuya Tsubouchi
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Parichehr Yazdanshenas
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Vaishnavi Kumaran
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Anna Dvorkin-Gheva
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Martin R J Kolb
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada
| | - Kjetil Ask
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada.
| |
Collapse
|
43
|
Yan P, Liu J, Li Z, Wang J, Zhu Z, Wang L, Yu G. Glycolysis Reprogramming in Idiopathic Pulmonary Fibrosis: Unveiling the Mystery of Lactate in the Lung. Int J Mol Sci 2023; 25:315. [PMID: 38203486 PMCID: PMC10779333 DOI: 10.3390/ijms25010315] [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: 11/16/2023] [Revised: 12/17/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung disease characterized by excessive deposition of fibrotic connective tissue in the lungs. Emerging evidence suggests that metabolic alterations, particularly glycolysis reprogramming, play a crucial role in the pathogenesis of IPF. Lactate, once considered a metabolic waste product, is now recognized as a signaling molecule involved in various cellular processes. In the context of IPF, lactate has been shown to promote fibroblast activation, myofibroblast differentiation, and extracellular matrix remodeling. Furthermore, lactate can modulate immune responses and contribute to the pro-inflammatory microenvironment observed in IPF. In addition, lactate has been implicated in the crosstalk between different cell types involved in IPF; it can influence cell-cell communication, cytokine production, and the activation of profibrotic signaling pathways. This review aims to summarize the current research progress on the role of glycolytic reprogramming and lactate in IPF and its potential implications to clarify the role of lactate in IPF and to provide a reference and direction for future research. In conclusion, elucidating the intricate interplay between lactate metabolism and fibrotic processes may lead to the development of innovative therapeutic strategies for IPF.
Collapse
Affiliation(s)
| | | | | | | | | | - Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China; (P.Y.); (J.L.); (Z.L.); (J.W.); (Z.Z.)
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang 453007, China; (P.Y.); (J.L.); (Z.L.); (J.W.); (Z.Z.)
| |
Collapse
|
44
|
Rodriguez LR, Tang SY, Roque Barboza W, Murthy A, Tomer Y, Cai TQ, Iyer S, Chavez K, Das US, Ghosh S, Cooper CH, Dimopoulos TT, Babu A, Connelly C, FitzGerald GA, Beers MF. PGF2α signaling drives fibrotic remodeling and fibroblast population dynamics in mice. JCI Insight 2023; 8:e172977. [PMID: 37934604 PMCID: PMC10807712 DOI: 10.1172/jci.insight.172977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/02/2023] [Indexed: 11/09/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic parenchymal lung disease characterized by repetitive alveolar cell injury, myofibroblast proliferation, and excessive extracellular matrix deposition for which unmet need persists for effective therapeutics. The bioactive eicosanoid, prostaglandin F2α, and its cognate receptor FPr (Ptgfr) are implicated as a TGF-β1-independent signaling hub for IPF. To assess this, we leveraged our published murine PF model (IER-SftpcI73T) expressing a disease-associated missense mutation in the surfactant protein C (Sftpc) gene. Tamoxifen-treated IER-SftpcI73T mice developed an early multiphasic alveolitis and transition to spontaneous fibrotic remodeling by 28 days. IER-SftpcI73T mice crossed to a Ptgfr-null (FPr-/-) line showed attenuated weight loss and gene dosage-dependent rescue of mortality compared with FPr+/+ cohorts. IER-SftpcI73T/FPr-/- mice also showed reductions in multiple fibrotic endpoints for which administration of nintedanib was not additive. Single-cell RNA-Seq, pseudotime analysis, and in vitro assays demonstrated Ptgfr expression predominantly within adventitial fibroblasts, which were reprogrammed to an "inflammatory/transitional" cell state in a PGF2α /FPr-dependent manner. Collectively, the findings provide evidence for a role for PGF2α signaling in IPF, mechanistically identify a susceptible fibroblast subpopulation, and establish a benchmark effect size for disruption of this pathway in mitigating fibrotic lung remodeling.
Collapse
Affiliation(s)
- Luis R. Rodriguez
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine
- PENN-CHOP Lung Biology Institute, and
| | - Soon Yew Tang
- Institute for Translational Medicine and Therapeutics, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Willy Roque Barboza
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine
- PENN-CHOP Lung Biology Institute, and
| | - Aditi Murthy
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine
- PENN-CHOP Lung Biology Institute, and
| | - Yaniv Tomer
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine
- PENN-CHOP Lung Biology Institute, and
| | - Tian-Quan Cai
- Calico Life Sciences LLC, South San Francisco, California, USA
| | - Swati Iyer
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine
- PENN-CHOP Lung Biology Institute, and
| | - Katrina Chavez
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine
- PENN-CHOP Lung Biology Institute, and
| | - Ujjalkumar Subhash Das
- Institute for Translational Medicine and Therapeutics, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Soumita Ghosh
- Institute for Translational Medicine and Therapeutics, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Charlotte H. Cooper
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine
- PENN-CHOP Lung Biology Institute, and
| | - Thalia T. Dimopoulos
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine
- PENN-CHOP Lung Biology Institute, and
| | | | | | - Garret A. FitzGerald
- Institute for Translational Medicine and Therapeutics, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael F. Beers
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine
- PENN-CHOP Lung Biology Institute, and
| |
Collapse
|
45
|
Abstract
Pulmonary surfactant is a critical component of lung function in healthy individuals. It functions in part by lowering surface tension in the alveoli, thereby allowing for breathing with minimal effort. The prevailing thinking is that low surface tension is attained by a compression-driven squeeze-out of unsaturated phospholipids during exhalation, forming a film enriched in saturated phospholipids that achieves surface tensions close to zero. A thorough review of past and recent literature suggests that the compression-driven squeeze-out mechanism may be erroneous. Here, we posit that a surfactant film enriched in saturated lipids is formed shortly after birth by an adsorption-driven sorting process and that its composition does not change during normal breathing. We provide biophysical evidence for the rapid formation of an enriched film at high surfactant concentrations, facilitated by adsorption structures containing hydrophobic surfactant proteins. We examine biophysical evidence for and against the compression-driven squeeze-out mechanism and propose a new model for surfactant function. The proposed model is tested against existing physiological and pathophysiological evidence in neonatal and adult lungs, leading to ideas for biophysical research, that should be addressed to establish the physiological relevance of this new perspective on the function of the mighty thin film that surfactant provides.
Collapse
Affiliation(s)
- Fred Possmayer
- Department of Biochemistry, Western University, London, Ontario N6A 3K7, Canada
- Department of Obstetrics/Gynaecology, Western University, London, Ontario N6A 3K7, Canada
| | - Yi Y Zuo
- Department of Mechanical Engineering, University of Hawaii at Manon, Honolulu, Hawaii 96822, United States
- Department of Pediatrics, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96826, United States
| | - Ruud A W Veldhuizen
- Department of Physiology & Pharmacology, Western University, London, Ontario N6A 5C1, Canada
- Department of Medicine, Western University, London, Ontario N6A 3K7, Canada
- Lawson Health Research Institute, London, Ontario N6A 4V2, Canada
| | - Nils O Petersen
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
- Department of Chemistry, Western University, London, Ontario N6A 5B7, Canada
| |
Collapse
|
46
|
Yu F, Yang S, Ni H, Heng D, Wu X, Yang M, Zhang X, Cao Y, Pei Y, An D, Li D, Liu D, Liu L, Pan L, Chen Q, Zhu X, Zhou J. O-GlcNAcylation Regulates Centrosome Behavior and Cell Polarity to Reduce Pulmonary Fibrosis and Maintain the Epithelial Phenotype. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303545. [PMID: 37963851 PMCID: PMC10754140 DOI: 10.1002/advs.202303545] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/11/2023] [Indexed: 11/16/2023]
Abstract
O-GlcNAcylation functions as a cellular nutrient and stress sensor and participates in almost all cellular processes. However, it remains unclear whether O-GlcNAcylation plays a role in the establishment and maintenance of cell polarity, because mice lacking O-GlcNAc transferase (OGT) are embryonically lethal. Here, a mild Ogt knockout mouse model is constructed and the important role of O-GlcNAcylation in establishing and maintaining cell polarity is demonstrated. Ogt knockout leads to severe pulmonary fibrosis and dramatically promotes epithelial-to-mesenchymal transition. Mechanistic studies reveal that OGT interacts with pericentriolar material 1 (PCM1) and centrosomal protein 131 (CEP131), components of centriolar satellites required for anchoring microtubules to the centrosome. These data further show that O-GlcNAcylation of PCM1 and CEP131 promotes their centrosomal localization through phase separation. Decrease in O-GlcNAcylation prevents PCM1 and CEP131 from localizing to the centrosome, instead dispersing these proteins throughout the cell and impairing the microtubule-centrosome interaction to disrupt centrosome positioning and cell polarity. These findings identify a previously unrecognized role for protein O-GlcNAcylation in establishing and maintaining cell polarity with important implications for the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Fan Yu
- State Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemFrontiers Science Center for Cell ResponsesTianjin Key Laboratory of Protein ScienceCollege of Life SciencesNankai UniversityTianjin300071China
- School of Health and Life SciencesUniversity of Health and Rehabilitation SciencesQingdao266071China
| | - Song Yang
- State Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemFrontiers Science Center for Cell ResponsesTianjin Key Laboratory of Protein ScienceCollege of Life SciencesNankai UniversityTianjin300071China
- School of Health and Life SciencesUniversity of Health and Rehabilitation SciencesQingdao266071China
| | - Hua Ni
- State Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemFrontiers Science Center for Cell ResponsesTianjin Key Laboratory of Protein ScienceCollege of Life SciencesNankai UniversityTianjin300071China
| | - Dai Heng
- State Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemFrontiers Science Center for Cell ResponsesTianjin Key Laboratory of Protein ScienceCollege of Life SciencesNankai UniversityTianjin300071China
| | - Xuemei Wu
- State Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemFrontiers Science Center for Cell ResponsesTianjin Key Laboratory of Protein ScienceCollege of Life SciencesNankai UniversityTianjin300071China
| | - Mulin Yang
- State Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemFrontiers Science Center for Cell ResponsesTianjin Key Laboratory of Protein ScienceCollege of Life SciencesNankai UniversityTianjin300071China
| | - Xinming Zhang
- Department of Endodontics and Laboratory of Stem Cells Endocrine ImmunologyTianjin Medical University School of StomatologyTianjin300070China
| | - Yuxin Cao
- Department of Endodontics and Laboratory of Stem Cells Endocrine ImmunologyTianjin Medical University School of StomatologyTianjin300070China
| | - Yandong Pei
- State Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemFrontiers Science Center for Cell ResponsesTianjin Key Laboratory of Protein ScienceCollege of Life SciencesNankai UniversityTianjin300071China
| | - Di An
- State Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemFrontiers Science Center for Cell ResponsesTianjin Key Laboratory of Protein ScienceCollege of Life SciencesNankai UniversityTianjin300071China
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemFrontiers Science Center for Cell ResponsesTianjin Key Laboratory of Protein ScienceCollege of Life SciencesNankai UniversityTianjin300071China
| | - Dayong Liu
- Department of Endodontics and Laboratory of Stem Cells Endocrine ImmunologyTianjin Medical University School of StomatologyTianjin300070China
| | - Lin Liu
- State Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemFrontiers Science Center for Cell ResponsesTianjin Key Laboratory of Protein ScienceCollege of Life SciencesNankai UniversityTianjin300071China
| | - Leiting Pan
- State Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemFrontiers Science Center for Cell ResponsesTianjin Key Laboratory of Protein ScienceCollege of Life SciencesNankai UniversityTianjin300071China
| | - Quan Chen
- State Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemFrontiers Science Center for Cell ResponsesTianjin Key Laboratory of Protein ScienceCollege of Life SciencesNankai UniversityTianjin300071China
| | - Xueliang Zhu
- State Key Laboratory of Cell BiologyCAS Centre for Excellence in Molecular Cell ScienceInstitute of Biochemistry and Cell BiologyShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghai200031China
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemFrontiers Science Center for Cell ResponsesTianjin Key Laboratory of Protein ScienceCollege of Life SciencesNankai UniversityTianjin300071China
- Center for Cell Structure and FunctionShandong Provincial Key Laboratory of Animal Resistance BiologyCollege of Life SciencesShandong Normal UniversityJinan250014China
| |
Collapse
|
47
|
Wang F, Ting C, Riemondy KA, Douglas M, Foster K, Patel N, Kaku N, Linsalata A, Nemzek J, Varisco BM, Cohen E, Wilson JA, Riches DW, Redente EF, Toivola DM, Zhou X, Moore BB, Coulombe PA, Omary MB, Zemans RL. Regulation of epithelial transitional states in murine and human pulmonary fibrosis. J Clin Invest 2023; 133:e165612. [PMID: 37768734 PMCID: PMC10645382 DOI: 10.1172/jci165612] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 09/21/2023] [Indexed: 09/29/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive scarring disease arising from impaired regeneration of the alveolar epithelium after injury. During regeneration, type 2 alveolar epithelial cells (AEC2s) assume a transitional state that upregulates multiple keratins and ultimately differentiate into AEC1s. In IPF, transitional AECs accumulate with ineffectual AEC1 differentiation. However, whether and how transitional cells cause fibrosis, whether keratins regulate transitional cell accumulation and fibrosis, and why transitional AECs and fibrosis resolve in mouse models but accumulate in IPF are unclear. Here, we show that human keratin 8 (KRT8) genetic variants were associated with IPF. Krt8-/- mice were protected from fibrosis and accumulation of the transitional state. Keratin 8 (K8) regulated the expression of macrophage chemokines and macrophage recruitment. Profibrotic macrophages and myofibroblasts promoted the accumulation of transitional AECs, establishing a K8-dependent positive feedback loop driving fibrogenesis. Finally, rare murine transitional AECs were highly senescent and basaloid and may not differentiate into AEC1s, recapitulating the aberrant basaloid state in human IPF. We conclude that transitional AECs induced and were maintained by fibrosis in a K8-dependent manner; in mice, most transitional cells and fibrosis resolved, whereas in human IPF, transitional AECs evolved into an aberrant basaloid state that persisted with progressive fibrosis.
Collapse
Affiliation(s)
- Fa Wang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Ting
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kent A. Riemondy
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Michael Douglas
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Nisha Patel
- College of Literature, Science, and the Arts
| | - Norihito Kaku
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Jean Nemzek
- Unit for Laboratory Animal Medicine, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Brian M. Varisco
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Erez Cohen
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jasmine A. Wilson
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - David W.H. Riches
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Research, Veterans Affairs Eastern Colorado Health Care System, Denver Colorado, USA
| | - Elizabeth F. Redente
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Diana M. Toivola
- Cell Biology, Biosciences, Faculty of Science and Engineering, and InFLAMES Research Flagship Center, Åbo Akademi University, Turku, Finland
| | - Xiaofeng Zhou
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Bethany B. Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Pierre A. Coulombe
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - M. Bishr Omary
- Department of Medicine, Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Rachel L. Zemans
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Program in Cellular and Molecular Biology, School of Medicine, and
| |
Collapse
|
48
|
Luo W, Gu Y, Fu S, Wang J, Zhang J, Wang Y. Emerging opportunities to treat idiopathic pulmonary fibrosis: Design, discovery, and optimizations of small-molecule drugs targeting fibrogenic pathways. Eur J Med Chem 2023; 260:115762. [PMID: 37683364 DOI: 10.1016/j.ejmech.2023.115762] [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: 06/30/2023] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common fibrotic form of idiopathic diffuse lung disease. Due to limited treatment options, IPF patients suffer from poor survival. About ten years ago, Pirfenidone (Shionogi, 2008; InterMune, 2011) and Nintedanib (Boehringer Ingelheim, 2014) were approved, greatly changing the direction of IPF drug design. However, limited efficacy and side effects indicate that neither can reverse the process of IPF. With insights into the occurrence of IPF, novel targets and agents have been proposed, which have fundamentally changed the treatment of IPF. With the next-generation agents, targeting pro-fibrotic pathways in the epithelial-injury model offers a promising approach. Besides, several next-generation IPF drugs have entered phase II/III clinical trials with encouraging results. Due to the rising IPF treatment requirements, there is an urgent need to completely summarize the mechanisms, targets, problems, and drug design strategies over the past ten years. In this review, we summarize known mechanisms, target types, drug design, and novel technologies of IPF drug discovery, aiming to provide insights into the future development and clinical application of next-generation IPF drugs.
Collapse
Affiliation(s)
- Wenxin Luo
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yilin Gu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Siyu Fu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Jifa Zhang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
49
|
Fastrès A, Roels E, Tutunaru AC, Bolen G, Merveille A, Day MJ, Garigliany M, Antoine N, Clercx C. Osteopontin and fibronectin in lung tissue, serum, and bronchoalveolar lavage fluid of dogs with idiopathic pulmonary fibrosis and control dogs. J Vet Intern Med 2023; 37:2468-2477. [PMID: 37853926 PMCID: PMC10658509 DOI: 10.1111/jvim.16870] [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: 05/30/2023] [Accepted: 09/08/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) affects West Highland white terriers (WHWTs). Osteopontin (SPP1) and fibronectin (FN1) are associated with human IPF and are overexpressed by bronchoalveolar lavage fluid (BALF) macrophages in dogs with IPF. OBJECTIVE To investigate the value of these proteins as biomarkers of IPF. ANIMALS West Highland white terriers (WHWTs) with IPF, control WHWTs, and terriers. METHODS Cross-sectional observational study. Immunohistochemistry was used to localize SPP1 and FN1 in lung tissue. Serum and BALF SPP1 and FN1 concentrations were measured using canine ELISA kits and compared between groups. RESULTS Osteopontin stained ciliated epithelial cells, smooth muscular cells, and macrophages of all included dogs, and type-II pneumocytes and extracellular matrix of all 12 diseased WHWTs, 4/6 control WHWTs, and none of the 3 terriers. Osteopontin serum concentration was higher in diseased WHWTs (n = 22; 2.15 ng/mL [0.74-5.30]) compared with control WHWTs (n = 13; 0.63 ng/mL [0.41-1.63]; P = .005) and terriers (n = 15; 0.31 ng/mL [0.19-0.51]; P < .0001), and in control WHWTs compared with terriers (P = .005). Osteopontin BALF concentrations were higher in diseased (0.27 ng/mL [0.14-0.43]) and control WHWTs (0.25 ng/mL [0.14-0.40]), compared with terriers (0.02 ng/mL [0.01-0.08]; P < .0001 and P = .003, respectively). Fibronectin (FN1) serum concentrations were lower in diseased dogs (1.03 ng/mL [0.35-1.48]) and control WHWTs (0.61 ng/mL [0.24-0.65]) compared with terriers (2.72 ng/mL [0.15-5.21]; P < .0001 and P = .0001, respectively). There was no difference in FN1 immunostaining and FN1 BALF concentrations between groups. CONCLUSIONS Results suggest that SPP1 is involved in pathogenesis of IPF and could predispose that breed to the disease. Osteopontin serum concentration could serve as a diagnostic biomarker of IPF.
Collapse
Affiliation(s)
- Aline Fastrès
- Department of Clinical Sciences, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Elodie Roels
- Department of Clinical Sciences, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Alexandru C. Tutunaru
- Department of Clinical Sciences, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Géraldine Bolen
- Department of Clinical Sciences, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Anne‐Christine Merveille
- Department of Clinical Sciences, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Michael J. Day
- School of Veterinary SciencesUniversity of BristolLangfordUnited Kingdom
| | - Mutien‐Marie Garigliany
- Department of Morphology and Pathology, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Nadine Antoine
- Department of Morphology and Pathology, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Cécile Clercx
- Department of Clinical Sciences, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| |
Collapse
|
50
|
Liu X, Niu W, Zhao S, Zhang W, Zhao Y, Li J. Piezo1:the potential new therapeutic target for fibrotic diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 184:42-49. [PMID: 37722629 DOI: 10.1016/j.pbiomolbio.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 09/20/2023]
Abstract
Fibrosis is a pathological process that occurs in various organs, characterized by excessive deposition of extracellular matrix (ECM), leading to structural damage and, in severe cases, organ failure. Within the fibrotic microenvironment, mechanical forces play a crucial role in shaping cell behavior and function, yet the precise molecular mechanisms underlying how cells sense and transmit these mechanical cues, as well as the physical aspects of fibrosis progression, remain less understood. Piezo1, a mechanosensitive ion channel protein, serves as a pivotal mediator, converting mechanical stimuli into electrical or chemical signals. Accumulating evidence suggests that Piezo1 plays a central role in ECM formation and hemodynamics in the mechanical transduction of fibrosis expansion. This review provides an overview of the current understanding of the role of Piezo1 in fibrosis progression, encompassing conditions such as myocardial fibrosis, pulmonary fibrosis, renal fibrosis, and other fibrotic diseases. The main goal is to pave the way for potential clinical applications in the field of fibrotic diseases.
Collapse
Affiliation(s)
- Xin Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Weipin Niu
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuqing Zhao
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenjuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying Zhao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Jing Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|