1
|
Zhang Y, Zhao Y, An C, Guo Y, Ma Y, Shao F, Zhang Y, Sun K, Cheng F, Ren C, Zhang L, Sun B, Zhang Y, Wang H. Material-driven immunomodulation and ECM remodeling reverse pulmonary fibrosis by local delivery of stem cell-laden microcapsules. Biomaterials 2025; 313:122757. [PMID: 39178558 DOI: 10.1016/j.biomaterials.2024.122757] [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: 06/20/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 08/26/2024]
Abstract
Recent progress in stem cell therapy has demonstrated the therapeutic potential of intravenous stem cell infusions for treating the life-threatening lung disease of pulmonary fibrosis (PF). However, it is confronted with limitations, such as a lack of control over cellular function and rapid clearance by the host after implantation. In this study, we developed an innovative PF therapy through tracheal administration of microfluidic-templated stem cell-laden microcapsules, which effectively reversed the progression of inflammation and fibrotic injury. Our findings highlight that hydrogel microencapsulation can enhance the persistence of donor mesenchymal stem cells (MSCs) in the host while driving MSCs to substantially augment their therapeutic functions, including immunoregulation and matrix metalloproteinase (MMP)-mediated extracellular matrix (ECM) remodeling. We revealed that microencapsulation activates the MAPK signaling pathway in MSCs to increase MMP expression, thereby degrading overexpressed collagen accumulated in fibrotic lungs. Our research demonstrates the potential of hydrogel microcapsules to enhance the therapeutic efficacy of MSCs through cell-material interactions, presenting a promising yet straightforward strategy for designing advanced stem cell therapies for fibrotic diseases.
Collapse
Affiliation(s)
- Yujie Zhang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China
| | - Yuan Zhao
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China
| | - Chuanfeng An
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China
| | - Yiyang Guo
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China; School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024, Dalian, PR China
| | - Yubin Ma
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China; School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024, Dalian, PR China
| | - Fei Shao
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China
| | - Yonggang Zhang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China
| | - Kai Sun
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China
| | - Fang Cheng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China
| | - Changle Ren
- Faculty of Medicine, Dalian University of Technology, Dalian, 116023, PR China; Department of Joint Surgery, Dalian Municipal Central Hospital, Dalian, 116044, PR China
| | - Lijun Zhang
- Third People's Hospital of Dalian, Dalian Eye Hospital, Dalian, 116024, PR China
| | - Bingbing Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China; School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024, Dalian, PR China
| | - Yang Zhang
- School of Dentistry, Health Science Center, Shenzhen University, Shenzhen, 518015, PR China
| | - Huanan Wang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China; State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China.
| |
Collapse
|
2
|
Shen N, Shao Z, Xin H, Che F, Cui Y. Exploring TβRI inhibitors from Arenaria kansuensis based on 3D-QSAR, molecular docking and molecular dynamics simulation methods and its anti-pulmonary fibrosis molecular mechanism validation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118788. [PMID: 39245240 DOI: 10.1016/j.jep.2024.118788] [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: 06/24/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pulmonary fibrosis (PF) is a kind of interstitial lung disease that seriously threatens human life and health. Up to now, there is no specifically therapeutic drug. Arenaria kansuensis, a typical Tibetan medicine, has been previously proved to have anti-PF pharmacological activity by our group. However, the specific target and molecular mechanism of pharmacological active ingredients from it are still unknown. AIM OF THE STUDY This study aimed to explore the molecular mechanism and specific target of pharmacological active ingredients from A. kansuensis for treating PF. MATERIALS AND METHODS Virtual screening including 3D-QSAR, molecular docking and molecular dynamics simulation were used to screen TβRI inhibitor. CETSA experiment was used to verify the interaction between GAK (a β-carboline alkaloid isolated from A. kansuensis) and TβRI. Cell and molecular experiments including observation of cell morphology and Western blot were applied to investigate the molecular mechanism of action of GAK for treating PF. Animal experiments including physiological index, immunohistochemistry and ELISA were used to comprehensively evaluate the anti-PF effect of GAK and explore the corresponding mechanism of action. RESULTS Results of 3D-QSAR experiment indicated that GAK is a much stronger potential TβRI inhibitor, molecular mechanism study showed that 30 μM GAK could significantly keep TβRI more stable which indicated that the direct binding interaction between GAK and TβRI, it targetedly inhibited TβRI through forming hydrogen bonds with LYS232, SER280 and ASP351 and the binding energies is -56.05 kcal/mol. In vitro experiment showed GAK could suppress downstream signal pathways of TβRI including MAPK, PI3K/AKT and NF-κB pathways during EMT process. In vivo experiment showed that GAK could improve the survival rate and body weight of PF mice, alleviate the symptoms of histopathological severity, inflammatory cell infiltration and collagen deposition in lung tissue of PF mice through inhibiting EMT process of PF. CONCLUSIONS This work not only provided evidence to support GAK as a novel TβRI inhibitor for treating PF through multiple pathways, but also reveal the specific target and molecular mechanism of β-carboline alkaloids from A. kansuensis for treating PF.
Collapse
Affiliation(s)
- Na Shen
- School of Medicine, Linyi University, Linyi, 276000, Shandong, China
| | - Ziyao Shao
- School of Medicine, Linyi University, Linyi, 276000, Shandong, China
| | - Huawei Xin
- School of Medicine, Linyi University, Linyi, 276000, Shandong, China
| | - Fengyuan Che
- Linyi People's Hospital, Linyi, 276000, Shandong, China.
| | - Yulei Cui
- School of Medicine, Linyi University, Linyi, 276000, Shandong, China; Linyi People's Hospital, Linyi, 276000, Shandong, China.
| |
Collapse
|
3
|
Zheng H, Li J, Leung SSY. Inhalable polysorbates stabilized nintedanib nanocrystals to facilitate pulmonary nebulization and alveolar macrophage evasion. BIOMATERIALS ADVANCES 2025; 166:214084. [PMID: 39471574 DOI: 10.1016/j.bioadv.2024.214084] [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: 06/28/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024]
Abstract
Pulmonary delivery of nintedanib has noticeable advantages over the current oral administration in managing idiopathic pulmonary fibrosis (IPF). However, it remains a challenge to construct an efficient lung delivery system for insoluble nintedanib to resist nebulization instabilities and alveolar macrophage clearance. Herein, we attempted to develop nintedanib as inhalable nanocrystals stabilized with polysorbates. Different types of polysorbates (polysorbate 20, 40, 60, 80) and various drug-surfactant molar ratios (DSR = 10, 30, 60) were screened to determine the optimal nintedanib nanocrystal formulation. Most formulations (except those stabilized by polysorbate 40) could tailor nintedanib nanocrystals with sizes around 600 nm, and the nebulization-caused drug loss could be significantly reduced when DSR increased to 60. Meanwhile, all nanocrystals boosted the in vitro drug dissolution rate and improved the aerosol performance of nintedanib. Although nebulization-caused particle aggregation was found in most formulations, the nanocrystal stabilized with polysorbate 80 at DSR 60 presented no apparent size change after nebulization. This formulation exhibited superior alveolar macrophage evasion, enhanced fibroblast cluster infiltration, and improved fibroblast cluster inhibition compared with other formulations, indicating its significant potential for pulmonary nintedanib delivery. Overall, this study explored the potential of polysorbates in stabilizing nintedanib nanocrystals for nebulization and proposed practical solutions to transfer nintedanib from oral to lung delivery.
Collapse
Affiliation(s)
- Huangliang Zheng
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jiaqi Li
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | | |
Collapse
|
4
|
Liu JQ, Zhou HB, Bai WF, Wang J, Li Q, Fan LY, Chang H, Shi SL. Assessment of progression of pulmonary fibrosis based on metabonomics and analysis of intestinal microbiota. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:201-217. [PMID: 38488151 DOI: 10.1080/21691401.2024.2326616] [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: 06/26/2023] [Accepted: 02/27/2024] [Indexed: 03/19/2024]
Abstract
The main purpose of this study was to explore the changes of biomarkers in different developmental stages of bleomycin-induced pulmonary fibrosis (PF) in rats via comprehensive pathophysiology, UPLC-QTOF/MS metabonomic technology, and 16S rRNA gene sequencing of intestinal microbiota. The rats were randomly divided into normal control and 1-, 2- and 4-week model group. The rat model of PF was established by one-time intratracheal instillation of bleomycin. The levels of inflammatory and fibrosis-related factors such as hydroxyproline (HYP), type III procollagen (COL-III), type IV collagen (COL-IV), hyaluronidase (HA), laminin (LN), interleukin (IL)-1β, IL-6, malondialdehyde (MDA) increased and superoxide dismutase (SOD) decreased as the PF cycle progressed. In the 1-, 2- and 4-week model group, 2, 19 and 18 potential metabolic biomarkers and 3, 16 and 12 potential microbial biomarkers were detected, respectively, which were significantly correlated. Glycerophospholipid metabolism pathway was observed to be an important pathway affecting PF at 1, 2 and 4 weeks; arginine and proline metabolism pathways significantly affected PF at 2 weeks. Linoleic acid metabolism pathway exhibited clear metabolic abnormalities at 2 and 4 weeks of PF, and alpha-linolenic acid metabolism pathway significantly affected PF at 4 weeks.
Collapse
Affiliation(s)
- Jia-Qi Liu
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
| | - Hong-Bing Zhou
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
- Institute of Bioactive Substance and Function of Mongolian Medicine and Chinese Materia Medica, Baotou Medical College, Baotou, PR China
| | - Wan-Fu Bai
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
| | - Jia Wang
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
| | - Qian Li
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
| | - Li-Ya Fan
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
| | - Hong Chang
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
| | - Song-Li Shi
- Department of Pharmacy, Baotou Medical College, Baotou, PR China
- Institute of Bioactive Substance and Function of Mongolian Medicine and Chinese Materia Medica, Baotou Medical College, Baotou, PR China
| |
Collapse
|
5
|
Morikka J, Federico A, Möbus L, Inkala S, Pavel A, Sani S, Vaani M, Peltola S, Serra A, Greco D. Toxicogenomic assessment of in vitro macrophages exposed to profibrotic challenge reveals a sustained transcriptomic immune signature. Comput Struct Biotechnol J 2024; 25:194-204. [PMID: 39430886 PMCID: PMC11490883 DOI: 10.1016/j.csbj.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/27/2024] [Accepted: 10/05/2024] [Indexed: 10/22/2024] Open
Abstract
Immune signalling is a crucial component in the progression of fibrosis. However, approaches for the safety assessment of potentially profibrotic substances, that provide information on mechanistic immune responses, are underdeveloped. This study aimed to develop a novel framework for assessing the immunotoxicity of fibrotic compounds. We exposed macrophages in vitro to multiple sublethal concentrations of the profibrotic agent bleomycin, over multiple timepoints, and generated RNA sequencing data. Using a toxicogenomic approach, we performed dose-dependent analysis to discover genes dysregulated by bleomycin exposure in a dose-responsive manner. A subset of immune genes displayed a sustained dose-dependent and differential expression response to profibrotic challenge. An immunoassay revealed cytokines and proteinases responding to bleomycin exposure that closely correlated to transcriptomic alterations, underscoring the integration between transcriptional immune response and external immune signalling activity. This study not only increases our understanding of the immunological mechanisms of fibrosis, but also offers an innovative framework for the toxicological evaluation of substances with potential fibrogenic effects on macrophage signalling. Our work brings a new immunotoxicogenomic direction for hazard assessment of fibrotic compounds, through the implementation of a time and resource efficient in vitro methodology.
Collapse
Affiliation(s)
- Jack Morikka
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
| | - Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Lena Möbus
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Simo Inkala
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Alisa Pavel
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Copenhagen, Denmark
| | - Saara Sani
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Maaret Vaani
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sanna Peltola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
6
|
Akca MN, Kasavi C. Identifying new molecular signatures and potential therapeutics for idiopathic pulmonary fibrosis: a network medicine approach. Mamm Genome 2024; 35:734-748. [PMID: 39254743 DOI: 10.1007/s00335-024-10069-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/31/2024] [Indexed: 09/11/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease characterized by excessive collagen deposition and fibrosis of the lung parenchyma, leading to respiratory failure. The molecular mechanisms underlying IPF pathogenesis remain incompletely understood, hindering the development of effective therapeutic strategies. We have used a network medicine approach to comprehensively analyze molecular interactions and identify novel molecular signatures and potential therapeutics associated with IPF progression. Our integrative analysis revealed dysregulated molecular networks that are central to IPF pathophysiology. We have highlighted key molecular players and signaling pathways that are implicated in aberrant fibrotic processes. This systems-level understanding enables the identification of new biomarkers and therapeutic targets for IPF, providing potential avenues for precision medicine. Drug repurposing analysis revealed several drug candidates with anti-fibrotic, anti-inflammatory, and anti-cancer activities that could potentially slow fibrotic progression and improve patient outcomes. This study offers new insights into the molecular underpinnings of IPF and highlights network medicine approaches in uncovering complex disease mechanisms. The molecular signatures and therapeutic targets identified hold promise for developing precision therapies tailored to individual patients, ultimately advancing the management of this debilitating lung disease.
Collapse
Affiliation(s)
- Mecbure Nur Akca
- Department of Bioengineering, Faculty of Engineering, Marmara University, İstanbul, Türkiye
| | - Ceyda Kasavi
- Department of Bioengineering, Faculty of Engineering, Marmara University, İstanbul, Türkiye.
| |
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
|
Reininger D, Fundel-Clemens K, Mayr CH, Wollin L, Laemmle B, Quast K, Nickolaus P, Herrmann FE. PDE4B inhibition by nerandomilast: Effects on lung fibrosis and transcriptome in fibrotic rats and on biomarkers in human lung epithelial cells. Br J Pharmacol 2024; 181:4766-4781. [PMID: 39183442 DOI: 10.1111/bph.17303] [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: 02/22/2024] [Revised: 06/17/2024] [Accepted: 07/02/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND AND PURPOSE The PDE4 family is considered a prime target for therapeutic intervention in several fibro-inflammatory diseases. We have investigated the molecular mechanisms of nerandomilast (BI 1015550), a preferential PDE4B inhibitor. EXPERIMENTAL APPROACH In addition to clinically relevant parameters of idiopathic pulmonary fibrosis (IPF; lung function measurement/high-resolution computed tomography scan/AI-Ashcroft score), whole-lung homogenates from a therapeutic male Wistar rat model of pulmonary fibrosis were analysed by next-generation sequencing (NGS). Data were matched with public domain data derived from human IPF samples to investigate how well the rat model reflected human IPF. We scored the top counter-regulated genes following treatment with nerandomilast in human single cells and validated disease markers discovered in the rat model using a human disease-relevant in vitro assay of IPF. KEY RESULTS Nerandomilast improved the decline of lung function parameters in bleomycin-treated animals. In the NGS study, most transcripts deregulated by bleomycin treatment were normalised by nerandomilast treatment. Most notably, a significant number of deregulated transcripts that were identified in human IPF disease were also found in the animal model and reversed by nerandomilast. Mapping to single-cell data revealed the strongest effects on mesenchymal, epithelial and endothelial cell populations. In a primary human epithelial cell culture system, several disease-related (bio)markers were inhibited by nerandomilast in a concentration-dependent manner. CONCLUSIONS AND IMPLICATIONS This study further supports the available knowledge about the anti-inflammatory/antifibrotic mechanisms of nerandomilast and provides novel insights into the mode of action and signalling pathways influenced by nerandomilast treatment of lung fibrosis.
Collapse
Affiliation(s)
- Dennis Reininger
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Katrin Fundel-Clemens
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Christoph H Mayr
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Lutz Wollin
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Baerbel Laemmle
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Karsten Quast
- Global Clinical Development & Operations, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Peter Nickolaus
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Franziska Elena Herrmann
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
9
|
Ghanem M, Justet A, Jaillet M, Vasarmidi E, Boghanim T, Hachem M, Vadel A, Joannes A, Mordant P, Balayev A, Adams T, Mal H, Cazes A, Poté N, Mailleux A, Crestani B. Identification of FGFR4 as a regulator of myofibroblast differentiation in pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2024; 327:L818-L830. [PMID: 39350729 DOI: 10.1152/ajplung.00184.2023] [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: 06/13/2023] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 11/13/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease with limited therapeutic options. Fibroblast growth factor receptor-4 (FGFR4) is a known receptor for several paracrine fibroblast growth factors (FGFs). FGFR4 is also the main receptor for FGF19, an endocrine FGF that was demonstrated by our group to have antifibrotic properties in the lung. We aimed to determine whether FGFR4 could modulate pulmonary fibrogenesis. We assessed FGFR4 mRNA and protein levels in IPF and control lungs. In vitro, we determined the effect of transforming growth factor-β (TGF-β), endothelin-1, and platelet-derived growth factor (PDGF) on FGFR4 expression in human lung fibroblasts. We determined the effect of FGFR4 inhibition, using a specific pharmacological inhibitor (FGF401), or genetic deletion in murine embryonic fibroblasts (MEFs) on TGF-β-induced myofibroblastic differentiation. In vivo, we evaluated the development of bleomycin-induced lung fibrosis in Fgfr4-deficient (Fgfr4-/-) mice compared with wild-type littermates (WT) and after FGF401 treatment in WT mice compared with a control group receiving the solvent only. FGFR4 was decreased in IPF lungs, as compared with control lungs, at mRNA and protein levels. In vitro, FGFR4 was downregulated after treatment with TGF-β, endothelin-1, and PDGF. In vitro, FGFR4 inhibition by FGF401 prevented TGF-β1-induced collagen and ACTA2 increase in lung fibroblasts. Similar results were observed in Fgfr4-/- MEFs. In vivo, FGFR4 genetic deficiency or FGFR4 pharmacological inhibition did not modulate bleomycin-induced pulmonary fibrosis. Our data suggest that FGFR4 exerts profibrotic properties by enhancing TGF-β signaling in vitro. However, the inhibition of FGFR4 is not sufficient to prevent the development of pulmonary fibrosis in vivo.NEW & NOTEWORTHY FGFR4 has been reported to have antifibrotic effects in the liver. We aimed to determine the involvement of FGFR4 during IPF. Our data suggest that FGFR4 exerts profibrotic properties by enhancing TGF-β signaling in vitro. However, the inhibition of FGFR4 is not sufficient to prevent the development of pulmonary fibrosis in vivo. To our knowledge, this is the first study to assess the profibrotic action of FGFR4 during pulmonary fibrosis.
Collapse
Affiliation(s)
- Mada Ghanem
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Faculté de médecine Xavier Bichat, Paris, France
| | - Aurélien Justet
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Faculté de médecine Xavier Bichat, Paris, France
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, United States
| | - Madeleine Jaillet
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Faculté de médecine Xavier Bichat, Paris, France
| | - Eirini Vasarmidi
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Faculté de médecine Xavier Bichat, Paris, France
| | - Tiara Boghanim
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Faculté de médecine Xavier Bichat, Paris, France
| | - Mouna Hachem
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Faculté de médecine Xavier Bichat, Paris, France
| | - Aurélie Vadel
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Faculté de médecine Xavier Bichat, Paris, France
| | - Audrey Joannes
- INSERM U1085, IRSET Institut de Recherche sur la Santé, l'Environnement et le Travail, Université de Rennes-1, Rennes, France
| | - Pierre Mordant
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Faculté de médecine Xavier Bichat, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Service de Chirurgie Thoracique et vasculaire, Paris, France
| | - Agshin Balayev
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, United States
| | - Taylor Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, United States
| | - Hervé Mal
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Faculté de médecine Xavier Bichat, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Service de Pneumologie et Transplantation, Paris, France
| | - Aurélie Cazes
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Faculté de médecine Xavier Bichat, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Bichat, Département d'Anatomopathologie, Paris, France
| | - Nicolas Poté
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Faculté de médecine Xavier Bichat, Paris, France
- Assistance Publique des Hôpitaux de Paris, Hôpital Bichat, Département d'Anatomopathologie, Paris, France
| | - Arnaud Mailleux
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Faculté de médecine Xavier Bichat, Paris, France
| | - Bruno Crestani
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Faculté de médecine Xavier Bichat, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Service de Pneumologie A, Centre de Référence des Maladies Pulmonaires Rares, Paris, France
| |
Collapse
|
10
|
Mohamed BME, Abdelrahim MEA. Timing impact on the initiation of pirfenidone therapy on idiopathic pulmonary fibrosis disease progression. World J Clin Cases 2024; 12:6538-6542. [DOI: 10.12998/wjcc.v12.i32.6538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 09/24/2024] Open
Abstract
In this editorial, we comment on the article by Lei et al, with a specific focus on the timing of the initiation of the antifibrotic agent pirfenidone (PFD) in the management of idiopathic pulmonary fibrosis (IPF) and its impact on lung function of IPF patients. PFD is an antifibrotic agent that is widely used in the management of IPF in both early and advanced stages. It inhibits various pathways and has antifibrotic, anti-inflammatory, and antioxidant properties. Despite dosage lowering, PFD slowed IPF progression and maintained functional capacity. The 6-min walk distance test indicated that patients tolerated adverse events well, and PFD significantly reduced the incidence of progression episodes and death. Even when a single disease-progression event occurred, continuing PFD treatment had benefits.
Collapse
Affiliation(s)
- Basma M E Mohamed
- Department of Clinical Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 343433, Egypt
| | - Mohamed E A Abdelrahim
- Department of Clinical Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 343433, Egypt
| |
Collapse
|
11
|
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. [PMID: 39546755 DOI: 10.1021/acsnano.4c10328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
12
|
Jiang MY, Zhang C, Huang QH, Feng LL, Yang YY, Zhou Q, Luo HB, Wu Y. Discovery of Selective PDE1 Inhibitors with Anti-pulmonary Fibrosis Effects by Targeting the Metal Pocket. J Med Chem 2024. [PMID: 39546471 DOI: 10.1021/acs.jmedchem.4c01533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease with no ideal drugs. Our previous research demonstrated that phosphodiesterase 1 (PDE1) could be a promising target for the treatment of IPF. However, only a few selective PDE1 inhibitors are available, and the mechanism of recognition between inhibitors and the PDE1 protein is not fully understood. This study carried out a step-by-step optimization of a dihydropyrimidine hit Z94555858. By targeting the metal pocket of PDE1, a lead compound 3f was obtained, exhibiting an IC50 value of 11 nM against PDE1, moderate selectivity over other PDEs, and significant anti-fibrotic effects in bleomycin-induced pulmonary fibrosis rats. The structure-activity relationship study aided by molecular docking revealed that forming halogen bonds with water in the metal pocket greatly enhanced the PDE1 inhibition, providing a novel strategy for further rational design of PDE1 inhibitors.
Collapse
Affiliation(s)
- Mei-Yan Jiang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Chen Zhang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Qing-Hua Huang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Ling-Ling Feng
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yi-Yi Yang
- Key Laboratory of Tropical Biological Resources of Ministry of Education and Hainan Engineering Research Center for Drug Screening and Evaluation, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, Hainan, China
| | - Qian Zhou
- Key Laboratory of Tropical Biological Resources of Ministry of Education and Hainan Engineering Research Center for Drug Screening and Evaluation, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, Hainan, China
| | - Hai-Bin Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education and Hainan Engineering Research Center for Drug Screening and Evaluation, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, Hainan, China
- Song Li' Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Yazhou Bay, Sanya 572000, China
| | - Yinuo Wu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
13
|
Peng M, Deng J, Li X. Clinical advances and challenges in targeting FGF/FGFR signaling in lung cancer. Mol Cancer 2024; 23:256. [PMID: 39543657 PMCID: PMC11566285 DOI: 10.1186/s12943-024-02167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024] Open
Abstract
Fibroblast growth factors (FGFs) and their receptors regulate numerous cellular processes, such as metabolism and signal transduction, but can also drive tumorigenesis. Specifically, in lung cancer, the overexpression of FGFs, as well as the amplification, mutation and fusion of FGFR genes, are closely linked to the initiation, progression and resistance of the disease, suggesting that targeting FGF/FGFR is an attractive therapeutic strategy for lung cancer treatment. Nintedanib, a multitarget tyrosine kinase inhibitor (TKI) used in combination with docetaxel, has shown some success as a second-line therapy for lung cancer. However, clinical trials evaluating other FGFR inhibitors have yielded mixed results, indicating substantial complexity in targeting aberrant FGF/FGFR signaling. In this review, we describe the aberrations in FGF/FGFR signaling in lung cancer and summarize the clinical efficacy of FGFR inhibitors, such as multitarget TKIs, selective FGFR-TKIs and biological agents. We also discuss various challenges associated with FGFR targeting in lung cancer, including precision patient selection, toxicity and resistance. Finally, we provide perspectives on future directions, namely, developing novel FGFR-targeting drugs, such as FGFR degraders and more specific FGFR-TKIs, adopting combination therapy and targeting FGFs.
Collapse
Affiliation(s)
- Mei Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China.
| | - Jun Deng
- Department of Pharmacy, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, 410000, P. R. China
| | - Xiangping Li
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China.
| |
Collapse
|
14
|
Li XN, Lin YP, Han MM, Fang YF, Xing L, Jeong JH, Jiang HL. Modulating Fibrotic Mechanical Microenvironment for Idiopathic Pulmonary Fibrosis Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2407661. [PMID: 39529565 DOI: 10.1002/adma.202407661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is exacerbated by injurious mechanical forces that destabilize the pulmonary mechanical microenvironment homeostasis, leading to alveolar dysfunction and exacerbating disease severity. However, given the inherent mechanosensitivity of fibrotic lungs, where type II alveolar epithelial cells (AEC IIs) are subjected to persistent stretching and overactivated myofibroblasts experience malignant interactions during mechanotransduction, it becomes imperative to develop effective strategies to modulate the pulmonary mechanical microenvironment. Herein, cyclo (RGDfC) peptide-decorated zeolitic imidazolate framework-8 nanoparticles (named ZDFPR NPs) are constructed to target and repair the aberrant mechanical force levels in pathological lungs. Specifically, reduces mechanical tension in AEC IIs by pH-responsive ZDFPR NPs that release zinc ions and 7, 8-dihydroxyflavone to promote alveolar repair and differentiation. Meanwhile, malignant interactions between myofibroblast contractility and extracellular matrix stiffness during mechanotransduction are disrupted by the fasudil inhibition ROCK signaling pathway. The results show that ZDFPR NPs successfully restored pulmonary mechanical homeostasis and terminated the fibrosis process in bleomycin-induced fibrotic mice. This study not only presents a promising strategy for modulating pulmonary mechanical microenvironment but also pioneers a novel avenue for IPF treatment.
Collapse
Affiliation(s)
- Xue-Na Li
- College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Ya-Ping Lin
- State Key Laboratory of Natural Medicines, Department of Pharmaceuticals, China Pharmaceutical University, Nanjing, 210009, China
| | - Meng-Meng Han
- State Key Laboratory of Natural Medicines, Department of Pharmaceuticals, China Pharmaceutical University, Nanjing, 210009, China
| | - Yue-Fei Fang
- State Key Laboratory of Natural Medicines, Department of Pharmaceuticals, China Pharmaceutical University, Nanjing, 210009, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, Department of Pharmaceuticals, China Pharmaceutical University, Nanjing, 210009, China
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, 210009, China
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Hu-Lin Jiang
- College of Pharmacy, Yanbian University, Yanji, 133002, China
- State Key Laboratory of Natural Medicines, Department of Pharmaceuticals, China Pharmaceutical University, Nanjing, 210009, China
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, 210009, China
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, 16419, South Korea
| |
Collapse
|
15
|
Lee J, Kim KJ, Nam JH, Choi JY, Rhee CK, Jo YS. Prevalence, Clinical Features, and Outcomes of Young Patients with Idiopathic Pulmonary Fibrosis. Respiration 2024:1-12. [PMID: 39510056 DOI: 10.1159/000541692] [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: 06/26/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024] Open
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) can occur at any age; however, studies on younger IPF patients are scarce because it primarily affects the elderly. This study aimed to investigate the clinical features and outcomes of younger IPF patients. METHODS We analyzed the National Korean Health Insurance Review and Assessment Service (HIRA) database from 2015 to 2021. Patients with IPF were identified using the International Classification of Diseases 10th Revision (ICD-10) codes and the Rare Intractable Diseases codes and were categorized into three age groups: <50, ≥50 and <65, and ≥65 years. The risk of acute exacerbation (AE) and mortality was analyzed. RESULTS Among 4,243 patients with IPF, 91 were under 50. These younger patients, who were predominantly female, exhibited less comorbidities and received more systemic steroids, whereas older group received more pirfenidone. Although AE risk increased with age, it was not statistically significant. Mortality and lung transplantation risks increased notably with age from the <50 group to the ≥50 and <65 group (hazard ratio [HR]: 1.52, 95% confidence interval [CI]: 0.93-2.49) and the ≥65 group (HR: 2.44, 95% CI: 1.51-3.93). These risks were influenced by factors such as age, comorbidities, previous AEs, and steroid use. Conversely, pirfenidone treatment reduced the risk. CONCLUSION While younger IPF patients had a lower risk of mortality and lung transplantation, with no significant differences in the risk of AEs, they were less likely to receive antifibrotic therapy and more often treated with steroids, which may affect outcomes. Early, targeted treatment strategies, including antifibrotic use, are crucial for improving their prognosis.
Collapse
Affiliation(s)
- Jeewon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea,
| | - Kyung Joo Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Hyun Nam
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joon Young Choi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chin Kook Rhee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yong Suk Jo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
16
|
Kambhampati V, Eedara A, Andugulapati SB. Yohimbine treatment improves pulmonary fibrosis by attenuating the inflammation and oxidative stress via modulating the MAPK pathway. Biochem Pharmacol 2024; 230:116613. [PMID: 39515589 DOI: 10.1016/j.bcp.2024.116613] [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: 07/17/2024] [Revised: 10/16/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating interstitial lung disorder characterized by the accumulation of extracellular matrix and collagen, resulting in significant parenchymal scarring and respiratory failure that leads to mortality. Yohimbine (YBH) is an α-2 adrenergic receptor antagonist with anti-oxidant and anti-inflammatory properties. In the current study, we aimed to investigate the anti-inflammatory, anti-oxidant and anti-fibrotic activity of YBH against LPS/TGF-β-induced differentiation in BEAS-2B/LL29 cells and bleomycin (BLMN) induced pulmonary fibrosis model in rats. Network pharmacology, gene expression, Western-blot analysis, immune-cytochemistry/immunohistochemistry, lung functional analysis, and histology techniques were used to assess the fibrotic marker expression/levels in cells or rat lung tissues. YBH treatment significantly attenuated the LPS-induced pro-inflammatory (identified through a network-pharmacology approach) and oxidative stress markers expression in lung epithelial cells. TGF-β stimulation significantly elevated the fibrotic cascade of markers and treatment with YBH attenuated these markers' expression/levels. Intra-tracheal administration of BLMN caused a significant elevation of various inflammatory/oxidative stress and fibrotic markers expression in lung tissues and treatment with YBH significantly mitigated the same. Ashcroft score analysis revealed that BLMN exhibited severe distortion of the lungs, elevation of thickness of the alveolar walls and accumulation of collagen in tissues, further treatment with YBH significantly suppressed these events and improved the lung architecture. Lung functional parameters demonstrated that BLMN-induced stiffness and resistance were reduced considerably upon YBH treatment and restored lung function dose-dependently. Overall, this study reveals that YBH treatment significantly attenuated the BLMN-induced fibrosis by regulating the MAPK pathway and provided insightful information for progressing towards translational outcomes.
Collapse
Affiliation(s)
- Vaishnavi Kambhampati
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500007, India
| | - Abhisheik Eedara
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500007, India; Academy of Science and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Sai Balaji Andugulapati
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad 500007, India; Academy of Science and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India.
| |
Collapse
|
17
|
Piecyk M, Ferraro-Peyret C, Laville D, Perros F, Chaveroux C. Novel insights into the GCN2 pathway and its targeting. Therapeutic value in cancer and lessons from lung fibrosis development. FEBS J 2024; 291:4867-4889. [PMID: 38879870 DOI: 10.1111/febs.17203] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/19/2024] [Accepted: 06/06/2024] [Indexed: 11/14/2024]
Abstract
Defining the mechanisms that allow cells to adapt to environmental stress is critical for understanding the progression of chronic diseases and identifying relevant drug targets. Among these, activation of the pathway controlled by the eIF2-alpha kinase GCN2 is critical for translational and metabolic reprogramming of the cell in response to various metabolic, proteotoxic, and ribosomal stressors. However, its role has frequently been investigated through the lens of a stress pathway signaling via the eIF2α-activating transcription factor 4 (ATF4) downstream axis, while recent advances in the field have revealed that the GCN2 pathway is more complex than previously thought. Indeed, this kinase can be activated through a variety of mechanisms, phosphorylate substrates other than eIF2α, and regulate cell proliferation in a steady state. This review presents recent findings regarding the fundamental mechanisms underlying GCN2 signaling and function, as well as the development of drugs that modulate its activity. Furthermore, by comparing the literature on GCN2's antagonistic roles in two challenging pathologies, cancer and pulmonary diseases, the benefits, and drawbacks of GCN2 targeting, particularly inhibition, are discussed.
Collapse
Affiliation(s)
- Marie Piecyk
- Department of Biochemistry and Molecular Biology, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
- Center for Innovation in Cancerology of Lyon (CICLY) EA 3738, Faculty of Medicine and Maieutic Lyon Sud, University Lyon I, Oullins, France
| | - Carole Ferraro-Peyret
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, France
- Hospices Civils de Lyon, Plateforme AURAGEN, France
| | - David Laville
- Department of Pathology, Hospices Civils de Lyon, East Hospital Group, Bron, France
| | - Frédéric Perros
- Laboratoire CarMeN, UMR INSERM U1060/INRA U1397, University of Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Cedric Chaveroux
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, France
| |
Collapse
|
18
|
Li H, Dai X, Zhou J, Wang Y, Zhang S, Guo J, Shen L, Yan H, Jiang H. Mitochondrial dynamics in pulmonary disease: Implications for the potential therapeutics. J Cell Physiol 2024; 239:e31370. [PMID: 38988059 DOI: 10.1002/jcp.31370] [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: 02/26/2024] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Mitochondria are dynamic organelles that continuously undergo fusion/fission to maintain normal cell physiological activities and energy metabolism. When mitochondrial dynamics is unbalanced, mitochondrial homeostasis is broken, thus damaging mitochondrial function. Accumulating evidence demonstrates that impairment in mitochondrial dynamics leads to lung tissue injury and pulmonary disease progression in a variety of disease models, including inflammatory responses, apoptosis, and barrier breakdown, and that the role of mitochondrial dynamics varies among pulmonary diseases. These findings suggest that modulation of mitochondrial dynamics may be considered as a valid therapeutic strategy in pulmonary diseases. In this review, we discuss the current evidence on the role of mitochondrial dynamics in pulmonary diseases, with a particular focus on its underlying mechanisms in the development of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD), asthma, pulmonary fibrosis (PF), pulmonary arterial hypertension (PAH), lung cancer and bronchopulmonary dysplasia (BPD), and outline effective drugs targeting mitochondrial dynamics-related proteins, highlighting the great potential of targeting mitochondrial dynamics in the treatment of pulmonary disease.
Collapse
Affiliation(s)
- Hui Li
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xinyan Dai
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Junfu Zhou
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yujuan Wang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Shiying Zhang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jiacheng Guo
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lidu Shen
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hengxiu Yan
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Huiling Jiang
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
19
|
Xia Y, Wang H, Shao M, Liu X, Sun F. MAP3K19 Promotes the Progression of Tuberculosis-Induced Pulmonary Fibrosis Through Activation of the TGF-β/Smad2 Signaling Pathway. Mol Biotechnol 2024; 66:3300-3310. [PMID: 37906388 DOI: 10.1007/s12033-023-00941-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/09/2023] [Indexed: 11/02/2023]
Abstract
Tuberculosis-induced pulmonary fibrosis (PF) is a chronic, irreversible interstitial lung disease, which severely affects lung ventilation and air exchange, leading to respiratory distress, impaired lung function, and ultimately death. As previously reported, epithelial-mesenchymal transition (EMT) and fibrosis in type II alveolar epithelial cells (AEC II) are two critical processes that contributes to the initiation and progression of tuberculosis-related PF, but the underlying pathological mechanisms remain unclear. In this study, through performing Real-Time quantitative PCR (RT-qPCR), Western blot, immunohistochemistry, and immunofluorescence staining assay, we confirmed that the expression levels of EMT and fibrosis-related biomarkers were significantly increased in lung tissues with tuberculosis-associated PF in vivo and Mycobacterium bovis Bacillus Calmette-Guérin (BCG) strain-infected AEC II cells in vitro. Besides, we noticed that the mitogen-activated protein kinase 19 (MAP3K19) was aberrantly overexpressed in PF models, and silencing of MAP3K19 significantly reduced the expression levels of fibronectin, collagen type I, and alpha-smooth muscle actin to decrease fibrosis, and upregulated E-cadherin and downregulated vimentin to suppress EMT in BCG-treated AEC II cells. Then, we uncovered the underlying mechanisms and found that BCG synergized with MAP3K19 to activate the pro-inflammatory transforming growth factor-beta (TGF-β)/Smad2 signal pathway in AEC II cells, and BCG-induced EMT process and fibrosis in AEC II cells were all abrogated by co-treating cells with TGF-β/Smad2 signal pathway inhibitor LY2109761. In summary, our results uncovered the underlying mechanisms by which the MAP3K19/TGF-β/Smad2 signaling pathway regulated EMT and fibrotic phenotypes of AEC II cells to facilitate the development of tuberculosis-associated PF, and these findings will provide new ideas and biomarkers to ameliorate tuberculosis-induced PF in clinic.
Collapse
Affiliation(s)
- Yu Xia
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China.
| | - Haiyue Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China
| | - Meihua Shao
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China
| | - Xuemei Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China
| | - Feng Sun
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyu Shan Road, Urumqi, 830054, China
| |
Collapse
|
20
|
Nguyen TH, Nguyen HHN, Nguyen TD, Tran VTH, Nguyen HA, Pham DV. NLRP3 inflammasome activation contributes to the development of the pro-fibrotic phenotype of lung fibroblasts. Biochem Pharmacol 2024; 229:116496. [PMID: 39159876 DOI: 10.1016/j.bcp.2024.116496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/03/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an irreversible progressive interstitial lung disease of unknown cause. The poorly understood pathophysiology of IPF poses substantial challenges to the development of effective anti-lung fibrotic drugs. The NLRP3 inflammasome, a key component of the innate immune system, has recently been linked to the pathogenesis of lung fibrosis. However, the specific contributions of NLRP3 inflammasomes to determination of the pro-fibrotic phenotype of lung fibroblasts, which play a central role in the production of extracellular matrix protein, remain to be investigated. Therefore, the present study was performed to elucidate the involvement of NLRP3 inflammasome signalling pathways in modulation of lung fibroblast proliferation and differentiation. We found that activation of NLRP3 inflammasomes increased in lung fibroblasts derived from individuals with pulmonary fibrosis and in normal lung fibroblasts stimulated with transforming growth factor β and platelet-derived growth factor. Importantly, blockage of NLRP3 inflammasome signalling, either by gene silencing of NLRP3 or using pharmacological inhibitors of NLRP3, caspase-1, or IL-1 receptor, inhibited the proliferation, differentiation, and extracellular matrix protein synthesis of activated lung fibroblasts. Moreover, induction of the reactive oxygen species/thioredoxin-interacting protein axis, an upstream signalling pathway of NLRP3 inflammasomes, was essential for maintenance of the pro-fibrotic phenotype of lung fibroblasts. Interestingly, treatments with pharmacological inhibitors of NLRP3 inflammasomes prevented the progression of bleomycin-induced pulmonary fibrosis in mice. Collectively, these findings suggest that aberrant activation of NLRP3 inflammasomes is a critical event in the pathogenesis of IPF and that targeting NLRP3 inflammasomes may serve as a therapeutic strategy for IPF.
Collapse
Affiliation(s)
- Thu-Hang Nguyen
- Department of Pharmacology, Hanoi University of Pharmacy, Hanoi, Vietnam
| | | | - Thuy-Duong Nguyen
- Department of Pharmacology, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Van Thi-Hong Tran
- Department of Pharmacology and Biochemistry, Vietnam National Institute of Medicinal Materials, Hanoi, Vietnam
| | - Hoang-Anh Nguyen
- Department of Pharmacology, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Duc-Vinh Pham
- Department of Pharmacology, Hanoi University of Pharmacy, Hanoi, Vietnam.
| |
Collapse
|
21
|
Cruwys S, Hein P, Humphries B, Black D. Drug discovery and development in idiopathic pulmonary fibrosis: the changing landscape. Drug Discov Today 2024; 29:104207. [PMID: 39396672 DOI: 10.1016/j.drudis.2024.104207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 08/28/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an area of high unmet clinical need and high research activity in the pharmaceutical and biotech industries. The two approved therapies, nintedanib and pirfenidone, have issues with efficacy and tolerability. Despite a considerable number of development programs reaching late-stage Phase 2b or 3 clinical trials, no drug other than nintedanib and pirfenidone has successfully demonstrated a benefit for patients. An analysis of these failures, and consideration of the trajectories of some of the current development projects, may offer novel paradigms for choosing modes-of-action and for the development of successful drugs.
Collapse
Affiliation(s)
- Simon Cruwys
- TherapeutAix UG, Juttastrasse 18, 52066 Aachen, Germany
| | - Peter Hein
- TherapeutAix UG, Juttastrasse 18, 52066 Aachen, Germany
| | - Bob Humphries
- TherapeutAix UG, Juttastrasse 18, 52066 Aachen, Germany
| | - Darcey Black
- TherapeutAix UG, Juttastrasse 18, 52066 Aachen, Germany.
| |
Collapse
|
22
|
Hao W, Yu TT, Li W, Wang GG, Hu HX, Zhou PP. Hemin attenuates bleomycin-induced lung fibrosis in mice by regulating the TGF-β1/MAPK and AMPK/SIRT1/PGC-1α/HO-1/NF-κB pathways. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:559-568. [PMID: 39467719 PMCID: PMC11519717 DOI: 10.4196/kjpp.2024.28.6.559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 10/30/2024]
Abstract
The objective of this study was to investigate the protective effect and potential mechanism of action of hemin on bleomycin-induced pulmonary fibrosis in mice. Male C57BL/6 mice were randomly divided into control, bleomycin and bleomycin + hemin groups. Mice in the bleomycin and bleomycin + hemin groups were injected intratracheally with bleomycin to establish the pulmonary fibrosis model. The bleomycin + hemin group mice were injected intraperitoneally with hemin starting 7 days before modeling until the end of Day 21 after modeling. Pathological changes in lung tissue were assessed by HE and Masson staining. Malondialdehyde (MDA), superoxide dismutase (SOD) and catalase (CAT) levels were determined in lung tissue. Immunohistochemistry was performed to assess the expression of α-SMA and collagen I. The serum levels of IL-6 and TNF-α were measured via ELISA. Western blotting was used to determine the expression of TGF-β1, SIRT1, PGC-1α and HO-1 and the phosphorylation levels of p38, ERK1/2, JNK, AMPK and NF-κB p65 in lung tissue. Hemin significantly reduced lung indices, increased terminal body weight. It also significantly increased SOD and CAT activities; decreased MDA, IL-6 and TNF-α levels; reduced the levels of α-SMA and collagen I-positive cells; upregulated SIRT1, PGC-1α and HO-1 expression; promoted AMPK phosphorylation; and downregulated TGF-β1 expression and p38, ERK1/2, JNK and NF-κB p65 phosphorylation. Hemin might attenuate oxidative damage and inflammatory responses and reduces extracellular matrix deposition by regulating the expression and phosphorylation of proteins associated with the TGF-β1/MAPK and AMPK/SIRT1/PGC-1α/HO-1/NF-κB pathways, thereby alleviating bleomycin-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Wei Hao
- Department of Functional Experimental Training Center, Wu Hu 241002, China
| | - Ting-ting Yu
- Department of Functional Experimental Training Center, Wu Hu 241002, China
| | - Wei Li
- Department of Pathophysiology, Basic Medical College, Wannan Medical College, Wu Hu 241002, China
| | - Guo-guang Wang
- Department of Pathophysiology, Basic Medical College, Wannan Medical College, Wu Hu 241002, China
| | - Hui-xian Hu
- Department of Medical Imageology, Wannan Medical College, Wu Hu 241002, China
| | - Ping-ping Zhou
- Department of Physiology, Basic Medical College, Wannan Medical College, Wu Hu 241002, China
| |
Collapse
|
23
|
John J, Clark AR, Kumar H, Burrowes KS, Vandal AC, Wilsher ML, Milne DG, Bartholmai BJ, Levin DL, Tawhai MH. Quantitative Analysis of Lung Shape in Idiopathic Pulmonary Fibrosis: Insights Into Disease- and Age-Associated Patterns. Acad Radiol 2024; 31:4650-4660. [PMID: 38679527 DOI: 10.1016/j.acra.2024.04.026] [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: 12/12/2023] [Revised: 04/03/2024] [Accepted: 04/14/2024] [Indexed: 05/01/2024]
Abstract
RATIONALE AND OBJECTIVES Fibrotic scarring in idiopathic pulmonary fibrosis (IPF) typically develops first in the posterior-basal lung tissue before advancing to involve more of the lung. The complexity of lung shape in the costo-diaphragmatic region has been proposed as a potential factor in this regional development. Intrinsic and disease-related shape could therefore be important for understanding IPF risk and its staging. We hypothesized that lung and lobe shape in IPF would have important differences from controls. MATERIALS AND METHODS A principal component (PC) analysis was used to derive a statistical shape model (SSM) of the lung for a control cohort aged > 50 years (N = 39), using segmented lung and fissure surface data from CT imaging. Individual patient shape models derived for baseline (N = 18) and follow-up (N = 16) CT scans in patients with IPF were projected to the SSM to describe shape as the sum of the SSM average and weighted PC modes. Associations between the first four PC shape modes, lung function, percentage of fibrosis (fibrosis%) and pulmonary vessel-related structures (PVRS%), and other tissue metrics were assessed and compared between the two cohorts. RESULTS Shape was different between IPF and controls (P < 0.05 for all shape modes), with IPF shape forming a distinct shape cluster. Shape had a negative relationship with age in controls (P = 0.013), but a positive relationship with age in IPF (P = 0.026). Some features of shape changed on follow-up. Shape in IPF was associated with fibrosis% (P < 0.05) and PVRS% (P < 0.05). CONCLUSION Quantitative comparison of lung and lobe shape in IPF with controls of a similar age reveals shape differences that are strongly associated with age and percent fibrosis. The clustering of IPF cohort shape suggests that it could be an important feature to describe disease.
Collapse
Affiliation(s)
- Joyce John
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Alys R Clark
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Haribalan Kumar
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Kelly S Burrowes
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Alain C Vandal
- Department of Statistics, University of Auckland, Auckland, New Zealand
| | | | - David G Milne
- Radiology, Auckland City Hospital, Auckland, New Zealand
| | | | | | - Merryn H Tawhai
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
24
|
Wang S, Li J, Wu C, Lei Z, Wang T, Huang X, Zhang S, Liu Y, Bi X, Zheng F, Zhu X, Huang Z, Yi X. Single-Cell RNA Sequencing Reveals Monocyte-Derived Interstitial Macrophages with a Pro-Fibrotic Phenotype in Bleomycin-Induced Pulmonary Fibrosis. Int J Mol Sci 2024; 25:11669. [PMID: 39519222 PMCID: PMC11545836 DOI: 10.3390/ijms252111669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive lung disease with limited effective therapies. Interstitial macrophages (IMs), especially those derived from monocytes, play an unknown role in IPF pathogenesis. By using single-cell RNA sequencing (scRNA-seq), bleomycin (BLM)-induced pulmonary fibrosis mouse lungs were analyzed to characterize the cellular landscape and heterogeneity of macrophages in this model. scRNA-seq was used to identify distinct interstitial macrophage subpopulations in fibrotic lungs, with monocyte-derived macrophages exhibiting a pro-fibrotic gene expression profile enriched in wound healing, extracellular matrix (ECM) remodeling, and pro-fibrotic cytokine production functions. A pseudotime analysis revealed that IMs originated from monocytes and differentiated along a specific trajectory. A cell-cell communication analysis demonstrated strong interactions between monocyte-derived interstitial macrophages (Mo-IMs) and fibroblasts through the transforming growth factor beta (TGFβ), secreted phosphoprotein 1 (SPP1), and platelet-derived growth factor (PDGF) signaling pathways. Flow cytometry validated the presence and expansion of Mo-IMs subpopulations in BLM-treated mice. This study reveals the cellular heterogeneity and developmental trajectory of lung macrophages in early BLM-induced pulmonary fibrosis, highlighting the crucial role of Mo-IMs with a pro-fibrotic phenotype in IPF pathogenesis via interactions with fibroblasts. Targeting these specific macrophage subpopulations and associated signaling pathways may provide novel therapeutic strategies for IPF.
Collapse
Affiliation(s)
- Shunli Wang
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jie Li
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Caixia Wu
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Zhengyao Lei
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Tong Wang
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xinxin Huang
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Suxia Zhang
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yuting Liu
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xiaohan Bi
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Fanshuo Zheng
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xuyou Zhu
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Ziling Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xianghua Yi
- Department of Pathology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| |
Collapse
|
25
|
Zhuo J, Liu D, Yu Q, Hu M, Huang H, Chen Y, Li Y, Gao Y, Chen W, Meng X, Zou F, Zhang J, Cai S, Dong H. Indole-3-acetic acid attenuates pulmonary fibrosis by modulating lung microbiota, inhibiting fibroblast activation, and alleviating alveolar epithelial cell senescence. Life Sci 2024; 359:123191. [PMID: 39481838 DOI: 10.1016/j.lfs.2024.123191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/15/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
AIM Pulmonary fibrosis (PF) is a relentlessly progressive disorder characterized by high mortality and limited effective therapeutic options. Indole-3-acetic acid (IAA), originally recognized as a plant hormone, is also identified as a tryptophan-derived metabolite catabolized from microbiota in mammals. IAA has exhibited antioxidative, anti-inflammatory, and anti-tumor effects in various disorders, yet its role in PF remains elusive. MAIN METHODS Bleomycin (BLM) was employed to induce PF in a mouse model. TGF-β1 was utilized in primary mouse lung fibroblasts (pMLFs) to establish a pro-fibrotic in vitro cellular model, and in A549 cells to create an in vitro cellular senescence model. The therapeutic effects of IAA on PF were evaluated using hematoxylin-eosin staining, immunofluorescence staining, western blotting, SA-β-gal assay, and network pharmacology analysis. Additionally, the effect of IAA on lung microbiota of PF was investigated using 16S rRNA gene sequencing analysis. KEY FINDINGS we observed a significant reduction in IAA levels in both PF patients and mouse models. Moreover, we demonstrated the therapeutic potential of IAA in alleviating PF in BLM-induced mouse models, showing a dose-dependent response. Mechanistically, we delineated three perspectives. Firstly, IAA promoted autophagic flux by inhibiting the PI3K/AKT/mTOR pathway, thereby suppressing lung fibroblast differentiation and extracellular matrix (ECM) deposition. Secondly, IAA attenuated alveolar epithelial cell senescence by modulating the PI3K/AKT and HIF-1 pathways. Lastly, IAA displayed the ability to mitigate PF by modulating the structure and composition of lung microbiota. SIGNIFICANCE Our study demonstrates that IAA alleviates PF through multiple pathways, highlighting its potential as a therapeutic agent.
Collapse
Affiliation(s)
- Jinzhong Zhuo
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Dongyu Liu
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qi Yu
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Minxuan Hu
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Haohua Huang
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yixin Chen
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanqun Li
- Ganzhou People's Hospital, Ganzhou 341000, China
| | - Yimei Gao
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Weimou Chen
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaojin Meng
- School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Fei Zou
- School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Jinming Zhang
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Shaoxi Cai
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Hangming Dong
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
26
|
Wang H, Sun K, Peng H, Wang Y, Zhang L. Emerging roles of noncoding RNAs in idiopathic pulmonary fibrosis. Cell Death Discov 2024; 10:443. [PMID: 39433746 PMCID: PMC11494106 DOI: 10.1038/s41420-024-02170-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/24/2024] [Accepted: 08/28/2024] [Indexed: 10/23/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrotic lung disease with limited treatment options and efficacy. Evidence suggests that IPF arises from genetic, environmental, and aging-related factors. The pathogenic mechanisms of IPF primarily involve dysregulated repeated microinjuries to epithelial cells, abnormal fibroblast/myofibroblast activation, and extracellular matrix (ECM) deposition, but thus far, the exact etiology remains unclear. Noncoding RNAs (ncRNAs) play regulatory roles in various biological processes and have been implicated in the pathophysiology of multiple fibrotic diseases, including IPF. This review summarizes the roles of ncRNAs in the pathogenesis of IPF and their potential as diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Kai Sun
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Hao Peng
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Yi Wang
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
| | - Lei Zhang
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
- Xianning Medical College, Hubei University of Science & Technology, Xianning, 437000, Hubei, China.
| |
Collapse
|
27
|
Kong J, Chen L. Gene expression profile analysis of severe influenza-based modulation of idiopathic pulmonary fibrosis. Eur J Med Res 2024; 29:501. [PMID: 39420432 PMCID: PMC11488079 DOI: 10.1186/s40001-024-02107-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/12/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND It is known severe influenza infections and idiopathic pulmonary fibrosis (IPF) disease might stimulate each other. Till now, no associated mechanism has been reported. METHOD We collected the genetic pattern of expression of severe influenza (GSE111368) and IPF (GSE70866) from the Gene Expression Omnibus (GEO) database. Common differentially expressed genes (C-DEGs) were identified from the two datasets, and using this data, we conducted three forms of analyses, functional annotation, protein-protein interaction (PPI) network and module construction, and hub gene identification and co-expression analysis. RESULTS In all, 174 C-DEGs were selected for additional analyses. Based on our functional analysis, these C-DEGs mediated inflammatory response and cell differentiation. Furthermore, using cytoHubba, we identified 15 genes, namely, MELK, HJURP, BIRC5, TPX2, TK1, CDT1, UBE2C, UHRF1, CCNA2, TYMS, CDCA5, CDCA8, RAD54L, CCNB2, and ITGAM, which served as hub genes to possibly contribute to severe influenza patients with IPF disease as comorbidity. The hub gene expressions were further confirmed using two stand-alone datasets (GSE101702 for severe influenza and GSE10667 for IPF). CONCLUSION Herein, we demonstrated the significance of common pathways and critical genes in severe influenza and IPF etiologies. The identified pathways and genes may be employed as possible therapeutic targets for future therapy against severe influenza patients with IPF.
Collapse
Affiliation(s)
- Jianping Kong
- Department of Nephrology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, 211200, China
| | - Liang Chen
- Department of Infectious Diseases, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical College of Nanjing University, NO 188 Lingshan North Road, Qixia District, Nanjing, 210046, China.
| |
Collapse
|
28
|
Kortekaas RK, Geillinger-Kästle KE, Fuentes-Mateos R, Schönleber A, van der Koog L, Elferink RAB, Al-Alyan N, Burgess JK, Gosens R. The soluble factor milieu in idiopathic pulmonary fibrosis dysregulates epithelial differentiation. FASEB J 2024; 38:e70077. [PMID: 39370849 DOI: 10.1096/fj.202302405rr] [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: 11/24/2023] [Revised: 08/23/2024] [Accepted: 09/16/2024] [Indexed: 10/08/2024]
Abstract
In idiopathic pulmonary fibrosis (IPF), epithelial abnormalities are present including bronchiolization and alveolar cell dysfunction. We hypothesized that the IPF microenvironment disrupts normal epithelial growth and differentiation. We mimicked the soluble factors within an IPF microenvironment using an IPF cocktail (IPFc), composed of nine factors which are increased in IPF lungs (CCL2, IL-1β, IL-4, IL-8, IL-13, IL-33, TGF-β, TNFα, and TSLP). Using IPFc, we asked whether the soluble factor milieu in IPF affects epithelial growth and differentiation and how IPFc compares to TGF-β alone. Epithelial growth and differentiation were studied using mouse lung organoids (primary Epcam+ epithelial cells co-cultured with CCL206 fibroblasts). Organoids exposed to IPFc and TGF-β were re-sorted into epithelial and fibroblast fractions and subjected to RNA sequencing. IPFc did not affect the number of organoids formed. However, pro-surfactant protein C expression was decreased. On these parameters, TGF-β alone had similar effects. However, RNA sequencing of re-sorted organoids revealed that IPFc and TGF-β had distinct effects on both epithelial cells and fibroblasts. IPFc upregulated goblet cell markers, whereas these were inhibited by TGF-β. Although both IPFc and TGF-β increased extracellular matrix gene expression, only TGF-β increased myofibroblast markers. VEGF-C and Wnt signaling were among the most differentially regulated signaling pathways by IPFc versus TGF-β. Interestingly, Wnt pathway activation rescued Sftpc downregulation induced by IPFc. In conclusion, IPFc alters epithelial differentiation in a way that is distinct from TGF-β. Alterations in Wnt signaling contribute to these effects. IPFc may be a more comprehensive representation of the soluble factor microenvironment in IPF.
Collapse
Affiliation(s)
- Rosa K Kortekaas
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kerstin E Geillinger-Kästle
- Department of Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Rocío Fuentes-Mateos
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anita Schönleber
- Department of Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Luke van der Koog
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Robin A B Elferink
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Nakaa Al-Alyan
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Janette K Burgess
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
29
|
Chen W, Gao Y, Liu Y, Luo Y, Xue X, Xiao C, Wei K. Tanshinone IIA Loaded Inhaled Polymer Nanoparticles Alleviate Established Pulmonary Fibrosis. ACS Biomater Sci Eng 2024; 10:6250-6262. [PMID: 39288315 DOI: 10.1021/acsbiomaterials.4c00532] [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: 09/19/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal respiratory disease characterized by chronic, progressive scarring of the lung parenchyma, leading to an irreversible decline in lung function. Apart from supportive care, there is currently no specific treatment available to reverse the disease. Based on the fact that tanshinone IIA (TAN) had an effect on protecting against TGF-β1-induced fibrosis through the inhibition of Smad and non-Smad signal pathways to avoid myofibroblasts activation, this study reported the development of the inhalable tanshinone IIA-loaded chitosan-oligosaccharides-coated poly(lactic-co-glycolic acid) (PLGA) nanoparticles (CPN@TAN) for enhancing the pulmonary delivery of tanshinone IIA to treat pulmonary fibrosis. The CPN@TAN with a size of 206.5 nm exhibited excellent in vitro aerosol delivery characteristics, featuring a mass median aerodynamic diameter (MMAD) of 3.967 ± 0.025 μm and a fine particle fraction (FPF) of 70.516 ± 0.929%. Moreover, the nanoparticles showed good stability during atomization and enhanced the mucosal penetration capabilities. The results of confocal spectroscopy confirmed the potential of the nanoparticles as carriers that facilitated the uptake of drugs by NIH3T3, A549, and MH-S cells. Additionally, the nanoparticles demonstrated good in vitro biocompatibility. In a mouse model of bleomycin-induced pulmonary fibrosis, noninvasive inhalation of aerosol CPN@TAN greatly suppressed collagen formation and facilitated re-epithelialization of the destroyed alveolar epithelium without causing systemic toxicity compared with intravenous administration. Consequently, our noninvasive inhalation drug delivery technology based on polymers may represent a promising paradigm and open the door to overcoming the difficulties associated with managing pulmonary fibrosis.
Collapse
Affiliation(s)
- Wenyu Chen
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yuanyuan Gao
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yuanqi Liu
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yujia Luo
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Xinrui Xue
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Chujie Xiao
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Kun Wei
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| |
Collapse
|
30
|
Wu L, Liu Y, Zhang Y, Xu R, Bi K, Li J, Wang J, Liu Y, Guo W, Wang Q, Chen Z. Identification of PANoptosis-related genes for idiopathic pulmonary fibrosis by machine learning and molecular subtype analysis. Sci Rep 2024; 14:24068. [PMID: 39402203 PMCID: PMC11473738 DOI: 10.1038/s41598-024-76263-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/11/2024] [Indexed: 10/17/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a severe interstitial lung disease characterized by a grim prognosis, in which various forms of cell death are significant contributors to its development. The objective of this study is to explore diagnostic biomarkers and molecular subtypes associated with PANoptosis in IPF, and to develop reliable diagnostic models based on PANoptosis-related mechanisms. The peripheral blood transcriptomic data of IPF from the Gene Expression Omnibus (GEO) database and PANoptosis-related genes from the GeneCards database were utilized to conduct differential gene expression analysis and weighted gene co-expression network analysis (WGCNA), identifying PANoptosis-related differentially expressed genes (PDEGs). We yielded 9 PDEGs and employed machine learning algorithms to identify 3 key diagnostic biomarkers for PANoptosis in IPF: MMP9, FCMR, NIBAN3. Consensus clustering algorithm was applied to recognize two PANoptosis-related subtypes. Cluster 1 exhibited higher abundance of adaptive immune response cells and significant enrichment in DNA damage and repair-related pathways. Cluster 2 exhibited greater prevalence of innate immune response cells and predominant enhancement in pathways related to lipid cholesterol metabolism and vascular remodeling. Diagnostic models were developed with the aid of clinical decision-making and a novel approach to the diagnosis and treatment for IPF.
Collapse
Affiliation(s)
- Li Wu
- Department of Anesthesiology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yang Liu
- School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Yifan Zhang
- The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Rui Xu
- Department of Anesthesiology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Kaixin Bi
- The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jing Li
- Department of Anesthesiology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Jia Wang
- Department of Anesthesiology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Yabing Liu
- Department of Anesthesiology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Wanjin Guo
- Department of Respiratory and Critical Care Medicine, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China.
| | - Qi Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China.
- School of Management, Shanxi Medical University, Taiyuan, China.
| | - Zhiqiang Chen
- Department of Galactophore, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, 030000, Taiyuan, China.
| |
Collapse
|
31
|
Dong Z, Wang X, Wang P, Bai M, Wang T, Chu Y, Qin Y. Idiopathic Pulmonary Fibrosis Caused by Damaged Mitochondria and Imbalanced Protein Homeostasis in Alveolar Epithelial Type II Cell. Adv Biol (Weinh) 2024:e2400297. [PMID: 39390651 DOI: 10.1002/adbi.202400297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/21/2024] [Indexed: 10/12/2024]
Abstract
Alveolar epithelial Type II (ATII) cells are closely associated with early events of Idiopathic pulmonary fibrosis (IPF). Proteostasis dysfunction, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction are known causes of decreased proliferation of alveolar epithelial cells and the secretion of pro-fibrotic mediators. Here, a large body of evidence is systematized and a cascade relationship between protein homeostasis, endoplasmic reticulum stress, mitochondrial dysfunction, and fibrotropic cytokines is proposed, providing a theoretical basis for ATII cells dysfunction as a possible pathophysiological initiating event for idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Zhaoxiong Dong
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
- Institute of Biophysics, Chinese Academy of Sciences 15 Datun Road, Chaoyang District, Beijing, 100101, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Xiaolong Wang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
| | - Peiwen Wang
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Mingjian Bai
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100101, China
| | - Tianyu Wang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100101, China
| | - Yanhui Chu
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Yan Qin
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
- Institute of Biophysics, Chinese Academy of Sciences 15 Datun Road, Chaoyang District, Beijing, 100101, China
| |
Collapse
|
32
|
Pei Z, Fan J, Tang M, Li Y. Ferroptosis: A New Strategy for the Treatment of Fibrotic Diseases. Adv Biol (Weinh) 2024:e2400383. [PMID: 39377183 DOI: 10.1002/adbi.202400383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/07/2024] [Indexed: 10/09/2024]
Abstract
Ferroptosis is a new type of cell death characterized by iron dependence and the excessive accumulation of lipid reactive oxygen species (lipid ROS) that has gradually become better characterized. There is sufficient evidence indicating that ferroptosis is associated with a variety of human life activities and diseases, such as tumor suppression, ischemic organ injury, and degenerative disorders. Notably, ferroptosis is also involved in the initiation and development of fibrosis in various organs, including liver fibrosis, pulmonary fibrosis, renal fibrosis, and cardiac fibrosis, which is usually irreversible and refractory. Although a large number of patients with fibrosis urgently need to be treated, the current treatment options are still limited and unsatisfactory. Organ fibrosis involves a series of complex and orderly processes, such as parenchymal cell damage, recruitment of inflammatory cells and activation of fibroblasts, which ultimately leads to the accumulation of extracellular matrix (ECM) and the formation of fibrosis. An increasing number of studies have confirmed the close association between these pathological processes and ferroptosis. This review summarizes the role and function of ferroptosis in fibrosis and proposes several potential therapeutic strategies and pathways based on ferroptosis.
Collapse
Affiliation(s)
- Zhuo Pei
- Air Force Hospital of the Central Theater Command of PLA, Datong, 037006, China
| | - Jing Fan
- Air Force Hospital of the Northern Theater Command of the People's Liberation Army of China, Shenyang, 110044, China
| | - Maolin Tang
- Air Force Hospital of the Central Theater Command of PLA, Datong, 037006, China
| | - Yuhong Li
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
| |
Collapse
|
33
|
Sun N, Ogulur I, Mitamura Y, Yazici D, Pat Y, Bu X, Li M, Zhu X, Babayev H, Ardicli S, Ardicli O, D'Avino P, Kiykim A, Sokolowska M, van de Veen W, Weidmann L, Akdis D, Ozdemir BG, Brüggen MC, Biedermann L, Straumann A, Kreienbühl A, Guttman-Yassky E, Santos AF, Del Giacco S, Traidl-Hoffmann C, Jackson DJ, Wang DY, Lauerma A, Breiteneder H, Zhang L, O'Mahony L, Pfaar O, O'Hehir R, Eiwegger T, Fokkens WJ, Cabanillas B, Ozdemir C, Walter K, Bayik M, Nadeau KC, Torres MJ, Akdis M, Jutel M, Agache I, Akdis CA. The epithelial barrier theory and its associated diseases. Allergy 2024. [PMID: 39370939 DOI: 10.1111/all.16318] [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: 05/17/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024]
Abstract
The prevalence of many chronic noncommunicable diseases has been steadily rising over the past six decades. During this time, over 350,000 new chemical substances have been introduced to the lives of humans. In recent years, the epithelial barrier theory came to light explaining the growing prevalence and exacerbations of these diseases worldwide. It attributes their onset to a functionally impaired epithelial barrier triggered by the toxicity of the exposed substances, associated with microbial dysbiosis, immune system activation, and inflammation. Diseases encompassed by the epithelial barrier theory share common features such as an increased prevalence after the 1960s or 2000s that cannot (solely) be accounted for by the emergence of improved diagnostic methods. Other common traits include epithelial barrier defects, microbial dysbiosis with loss of commensals and colonization of opportunistic pathogens, and circulating inflammatory cells and cytokines. In addition, practically unrelated diseases that fulfill these criteria have started to emerge as multimorbidities during the last decades. Here, we provide a comprehensive overview of diseases encompassed by the epithelial barrier theory and discuss evidence and similarities for their epidemiology, genetic susceptibility, epithelial barrier dysfunction, microbial dysbiosis, and tissue inflammation.
Collapse
Affiliation(s)
- Na Sun
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Xiangting Bu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Xueyi Zhu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Division of Food Processing, Milk and Dairy Products Technology Program, Karacabey Vocational School, Bursa Uludag University, Bursa, Turkey
| | - Paolo D'Avino
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ayca Kiykim
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Cerrahpasa School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Lukas Weidmann
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Deniz Akdis
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | | | - Marie Charlotte Brüggen
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Luc Biedermann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Alex Straumann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Andrea Kreienbühl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Emma Guttman-Yassky
- Department of Dermatology, and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandra F Santos
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Children's Allergy Service, Evelina London Children's Hospital, Guy's and St. Thomas' Hospital, London, UK
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Stefano Del Giacco
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | - David J Jackson
- Guy's Severe Asthma Centre, Guy's Hospital, Guy's & St Thomas' NHS Trust, London, UK
- School of Immunology & Microbial Sciences, King's College London, London, UK
| | - De-Yun Wang
- Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore City, Singapore
| | - Antti Lauerma
- Department of Dermatology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Heimo Breiteneder
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Liam O'Mahony
- Department of Medicine and School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Robyn O'Hehir
- Allergy, Asthma & Clinical Immunology, The Alfred Hospital, Melbourne, Victoria, Australia
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Thomas Eiwegger
- Translational Medicine Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
- Department of Pediatric and Adolescent Medicine, University Hospital St. Pölten, St. Pölten, Austria
| | - Wytske J Fokkens
- Department of Otorhinolaryngology & Head and Neck Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Beatriz Cabanillas
- Department of Allergy, Instituto de Investigación Biosanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Cevdet Ozdemir
- Department of Pediatric Basic Sciences, Institute of Child Health, Istanbul University, Istanbul, Turkey
- Istanbul Faculty of Medicine, Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul University, Istanbul, Turkey
| | - Kistler Walter
- Department of Sports Medicine, Davos Hospital, Davos, Switzerland
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Medical Committee International Ice Hockey Federation (IIHF), Zurich, Switzerland
| | - Mahmut Bayik
- Department of Internal Medicine and Hematology, Marmara University, Istanbul, Turkey
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Maria J Torres
- Allergy Unit, IBIMA-Hospital Regional Universitario de Málaga-ARADyAL, UMA, Málaga, Spain
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Marek Jutel
- Department of Clinical Immunology, Wrocław Medical University, Wroclaw, Poland
| | - Ioana Agache
- Faculty of Medicine, Department of Allergy and Clinical Immunology, Transylvania University, Brasov, Romania
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
34
|
Wang S, Gong X, Yuan J, Huang J, Zhao R, Ji J, Wang M, Shi X, Xin W, Zhong Y, Zheng Y, Jiang Q. Iron-doped diesel exhaust early-in-life inhalation-induced cardiopulmonary toxicity in chicken embryo: Roles of ferroptosis and acyl hydrocarbon signaling. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125085. [PMID: 39374763 DOI: 10.1016/j.envpol.2024.125085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/14/2024] [Accepted: 10/04/2024] [Indexed: 10/09/2024]
Abstract
Diesel exhaust (DE) is a major contributor to air pollution. Iron-doping could improve diesel burning efficacy and decrease emission, however, it will also change the composition of DE, potentially enhancing the toxicities. This study is aimed to assess iron-doped DE-induced cardiopulmonary toxicity in an established in ovo early-in-life inhalation exposure chicken embryo model, and to explore potential mechanisms. Ferrocene (205, 410, 820,1640 mg/L, equivalent to 75, 150, 300, 600 ppm iron mass) was added to diesel fuel, DE was collected from a diesel generator, and then exposed to embryonic day 18-19 chicken embryo via in ovo inhalation. Hatched chickens were kept for 0, 1, or 3 months, and then sacrificed. Histopathology, electrocardiography along with biochemical methods were used to assess cardiopulmonary toxicities. For mechanistic investigation, inhibitor for ferroptosis (ferrostatin-1) or Acyl hydrocarbon receptor (PDM2) were administered before DE (with or without iron-doping), and the cardiopulmonary toxicities were compared. Characterization of DE particles indicated that addition of ferrocene significantly elevated iron content. Additionally, the contents of major toxic polycyclic aromatic hydrocarbons decreased following addition of 820 mg/L ferrocene, but increased at other doses. Remarkable cardiopulmonary toxicities, in the manifestation of elevated heart rates, cardiac remodeling and cardiac/pulmonary fibrosis were observed in animals exposed to iron-doped DEs, in which the addition of ferrocene significantly enhanced the toxicities. Both ferrostatin-1 and PDM2 pretreatment could effectively alleviate the observed effects in animals exposed to iron-doped DE. Inhibition of AhR signaling seems to be capable of alleviating changes to ferroptosis related molecules following exposure to iron-doped DE, while inhibition of ferroptosis does not seem to affect AhR signaling molecules. In summary, iron-doping with ferrocene to diesel enhanced DE-induced cardiopulmonary toxicities in chicken embryos. Ferroptosis and AhR signaling both seem to participate in this process, in which AhR signaling seems to affect ferroptosis.
Collapse
Affiliation(s)
- Siyi Wang
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Xinxian Gong
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Junhua Yuan
- Department of Special Medicine, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Jing Huang
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Rui Zhao
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Jing Ji
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Meinan Wang
- Qingdao Product Quality Testing Research Institute, 77 Keyuanweisi Road, Qingdao, China
| | - Xiaoyu Shi
- Qingdao Product Quality Testing Research Institute, 77 Keyuanweisi Road, Qingdao, China
| | - Wenya Xin
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China
| | - Yuxu Zhong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, China
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China.
| | - Qixiao Jiang
- Department of Toxicology, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao, China.
| |
Collapse
|
35
|
Wang Q, Li A, Li Q, Li J, Wang Q, Wu S, Meng J, Liu C, Wang D, Chen Y. Carbon monoxide attenuates cellular senescence-mediated pulmonary fibrosis via modulating p53/PAI-1 pathway. Eur J Pharmacol 2024; 980:176843. [PMID: 39068977 DOI: 10.1016/j.ejphar.2024.176843] [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: 01/22/2024] [Revised: 07/10/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE Idiopathic pulmonary fibrosis (IPF) is a fatal progressive condition often requiring lung transplantation. Accelerated senescence of type II alveolar epithelial cells (AECII) plays a crucial role in pulmonary fibrosis progression through the secretion of the senescence-associated secretory phenotype (SASP). Low-dose carbon monoxide (CO) possesses anti-inflammatory, anti-oxidative, and anti-aging properties. This study aims to explore the preventive effects of CO-releasing molecule 2 (CORM2) in a bleomycin-induced pulmonary fibrosis model. METHODS We established an pulmonary fibrosis model in C57BL/6J mice and evaluated the impact of CORM2 on fibrosis pathology using Masson's trichrome staining, fluorescence staining, and pulmonary function tests. Fibrogenic marker expression and SASP secretion in tissues and AECII cells were analyzed using qRT-PCR, Western blot, and ELISA assays both in vivo and in vitro. Additionally, we investigated DNA damage and cellular senescence through immunofluorescence and SA-β-gal staining. RESULTS CORM2 showed a preventive effect on bleomycin-induced lung fibrosis by improving pulmonary function and reducing the expression of fibrosis-related genes, such as TGF-β, α-SMA, Collagen I/III. CORM2 decreased the DNA damage response by inhibiting γ-H2AX, p53, and p21. We identified PAI-1 as a new target gene that was downregulated by CORM2, and which was associated with cellular senescence and fibrosis. CORM2 effectively inhibited cellular senescence and delayed EMT occurrence in AECII cells. CONCLUSION Our study highlights the potential of CORM2 in preventing DNA damage-induced cellular senescence in bleomycin-induced pulmonary fibrosis through modulation of the p53/PAI-1 signaling pathway. These findings underscore the promising prospects of CORM2 in targeting cellular senescence and the p53/PAI-1 pathway as a potential preventive strategy for IPF.
Collapse
Affiliation(s)
- Qianqian Wang
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China; Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622, Liaoning, China
| | - Aohan Li
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China
| | - Qian Li
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China
| | - Jiaxin Li
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China; Case Statistics Office, The Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Harbin, 150011, China
| | - Qi Wang
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China
| | - Siyuan Wu
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China
| | - Jiaojiao Meng
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China
| | - Changpeng Liu
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China
| | - Dan Wang
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China; Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622, Liaoning, China.
| | - Yingqing Chen
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622, Liaoning, China; Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622, Liaoning, China.
| |
Collapse
|
36
|
Yue B, Xiong D, Chen J, Yang X, Zhao J, Shao J, Wei D, Gao F, Huang M, Chen J. SPP1 induces idiopathic pulmonary fibrosis and NSCLC progression via the PI3K/Akt/mTOR pathway. Respir Res 2024; 25:362. [PMID: 39369217 PMCID: PMC11456247 DOI: 10.1186/s12931-024-02989-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND The prevalence of non-small cell lung cancer (NSCLC) is notably elevated in individuals diagnosed with idiopathic pulmonary fibrosis (IPF). Secreted phosphoprotein 1 (SPP1), known for its involvement in diverse physiological processes, including oncogenesis and organ fibrosis, has an ambiguous role at the intersection of IPF and NSCLC. Our study sought to elucidate the function of SPP1 within the pathogenesis of IPF and its subsequent impact on NSCLC progression. METHODS Four GEO datasets was analyzed for common differential genes and TCGA database was used to analyze the prognosis. The immune infiltration was analyzed by TIMER database. SPP1 expression was examined in human lung tissues, the IPF fibroblasts and the BLM-induced mouse lung fibrosis model. Combined with SPP1 gene gain- and loss-of-function, qRT-PCR, Western blot, EdU and CCK-8 experiments were performed to evaluate the effects and mechanisms of SPP1 in IPF progression. Effect of SPP1 on NSCLC was detected by co-cultured IPF fibroblasts and NSCLC cells. RESULTS Through bioinformatics analysis, we observed a significant overexpression of SPP1 in both IPF and NSCLC patient datasets, correlating with enhanced immune infiltration of cancer-associated fibroblasts in NSCLC. Elevated levels of SPP1 were detected in lung tissue samples from IPF patients and bleomycin-induced mouse models, with partial colocalization observed with α-smooth muscle actin. Knockdown of SPP1 inhibits TGF-β1-induced differentiation of fibroblasts to myofibroblasts and the proliferation of IPF fibroblasts. Conversely, SPP1 overexpression promoted IPF fibroblast proliferation via PI3K/Akt/mTOR pathway. Furthermore, IPF fibroblasts promoted NSCLC cell proliferation and activated the PI3K/Akt/mTOR pathway; these effects were attenuated by SPP1 knockdown in IPF fibroblasts. CONCLUSIONS Our findings suggest that SPP1 functions as a molecule promoting both fibrosis and tumorigenesis, positioning it as a prospective therapeutic target for managing the co-occurrence of IPF and NSCLC.
Collapse
Affiliation(s)
- Bingqing Yue
- Department of lung transplantation, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Dian Xiong
- Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214000, China
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330000, China
| | - Juan Chen
- Department of General Intensive Care Unit, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Xiucheng Yang
- Department of lung transplantation, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Jin Zhao
- Department of lung transplantation, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Jingbo Shao
- Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Dong Wei
- Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Fei Gao
- Department of Emergency, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Man Huang
- Department of lung transplantation, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
- Department of General Intensive Care Unit, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
- Key Laboratory of Multiple Organ Failure, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingyu Chen
- Department of lung transplantation, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
- Lung Transplant Center, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214000, China.
| |
Collapse
|
37
|
Chen X, Yang Y, Sun S, Liu Q, Yang Y, Jiang L. CX3C chemokine: Hallmarks of fibrosis and ageing. Pharmacol Res 2024; 208:107348. [PMID: 39134186 DOI: 10.1016/j.phrs.2024.107348] [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: 04/19/2024] [Revised: 07/03/2024] [Accepted: 08/07/2024] [Indexed: 08/18/2024]
Abstract
Fibrosis refers to the progressive tissue lesion process characterized by excessive secretion and deposition of extracellular matrix (ECM). Abnormal fibrous tissue deposition distorts tissue architecture and leads to the progressive loss of organ function. Notably, fibrosis is one of the primary pathological appearances of many end stage illnesses, and is considered as a lethal threat to human health, especially in the elderly with ageing-related diseases. CX3C ligand 1 (CX3CL1) is the only member of chemokine CX3C and binds specifically to CX3C receptor 1 (CX3CR1). Different from other chemokines, CX3CL1 possesses both chemotactic and adhesive activity. CX3CL1/CX3CR1 axis involves in various physiological and pathological processes, and exerts a critical role in cells from the immune system, vascular system, and nervous system etc. Notably, increasing evidence has demonstrated that CX3CL1/CX3CR1 signaling pathway is closely related to the pathological process of fibrosis in multiple tissue and organs. We reviewed the crucial role of CX3CL1/CX3CR1 axis in fibrosis and ageing and systematically summarized the underlying mechanism, which offers prospective strategies of targeting CX3C for the therapy of fibrosis and ageing-related diseases.
Collapse
Affiliation(s)
- Xuanning Chen
- School of Medicine, Shanghai Jiao Tong University, 227 Chongqing South Road, Shanghai 200011, China
| | - Yiling Yang
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-maxillofacial Science, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| | - Siyuan Sun
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-maxillofacial Science, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China
| | - Qiong Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| | - Lingyong Jiang
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-maxillofacial Science, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai 200011, China.
| |
Collapse
|
38
|
Guan J, Yin L, Huang Q, Chen J, Liu H, Li J. m 6A methyltransferase ZC3H13 improves pulmonary fibrosis in mice through regulating Bax expression. Exp Cell Res 2024; 442:114255. [PMID: 39307407 DOI: 10.1016/j.yexcr.2024.114255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 07/07/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease. N6-methyladenosine (m6A) is a reversible RNA modification that was shown to be associated with IPF development. The present study aimed to explore the function and potential mechanism of the m6A methylation enzyme zinc finger CCCH-type containing 13 (ZC3H13) in IPF. In the study, bioinformatic screening yielded a differentially expressed m6A gene, ZC3H13, which was down-regulated in GEO microarrays, BLM-induced mouse models, and cellular models. Overexpression of ZC3H13 reduced histopathological damage of lung tissues in mice, mitigated fibrosis (including reduced α-SMA, collagen Ⅰ, and Vimentin levels, and elevated E-cadherin levels), decreased lung/body weight ratio and lung hydroxyproline levels, reduced oxidative stress (increased SOD activity and GSH-Px activity and decreased MDA levels), suppressed apoptosis within lung tissues and MLE-12 cells, promoted Bcl-2 expression, and inhibited Bax expression. Bax expression was found to be negatively correlated with ZC3H13 expression by correlation analysis. ZC3H13 could bind Bax mRNA and promote its m6A methylation through reading protein YTHDC1, thereby inhibiting its stability. Bax inhibition ameliorated BLM-induced MLE-12 cell dysfunction and partially abrogated the inhibition of MLE-12 cell function by ZC3H13 downregulation. In conclusion, m6A methyltransferase ZC3H13 impedes lung epithelial cell apoptosis and thus improves pulmonary fibrosis by promoting Bax mRNA m6A methylation and down-regulating Bax expression through reading protein YTHDC1.
Collapse
Affiliation(s)
- Jing Guan
- Department of Science and Education, The First Hospital of Changsha, Changsha, 410005, Hunan, China
| | - Lengyun Yin
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China
| | - Qi Huang
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China
| | - Jiamei Chen
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China
| | - Hui Liu
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China
| | - Jianmin Li
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China.
| |
Collapse
|
39
|
Ma J, Wang L, Yang D, Luo J, Gao J, Wang J, Guo H, Li J, Wang F, Wu J, Hu R. Chronic stress causes ovarian fibrosis to impair female fertility in mice. Cell Signal 2024; 122:111334. [PMID: 39102927 DOI: 10.1016/j.cellsig.2024.111334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 08/07/2024]
Abstract
OBJECTIVE Chronic psychological stress is associated with impaired follicular development and ovarian dysfunction. Many aspects of this dysfunction and the underlying mechanisms remain unclear. Using a chronic unpredictable mild stress (CUMS) mouse model, we investigate the influence of chronic stress on ovarian function and explore potential mechanisms. METHODS A CUMS mouse model was constructed over eight months, covering the period from sexual maturity to the onset of declining fertility in mice. At the end of the 2nd, 4th, 6th, and 8th months of exposure to CUMS, behavioral and physiological assays, including the sucrose preference test, tail suspension test, and serum corticosterone levels, were conducted to validate the effectiveness of the stress model. Fertility and ovarian function were assessed by analyzing the estrous cycle, number of offspring, sex hormone levels, follicle counts, granulosa cell proliferation and apoptosis, and the expression levels of fibrosis markers. Furthermore, proteomic analyses were performed on the ovaries to investigate the molecular mechanisms of ovarian fibrosis induced by CUMS. RESULTS With continued CUMS exposure, there was a gradual decline in both the ovary-to-body weight ratio and the number of offspring. Moreover, the percentage of atretic follicles was notably higher in the CUMS-exposed groups compared to the control groups. It is noticeable that CUMS triggered granulosa cell apoptosis and halted proliferation. Additionally, increased expression of α-SMA and Collagen I in the ovaries of CUMS-exposed mice indicated that CUMS could induce ovarian fibrosis. Proteomic analysis provided insights into the activation of specific biological processes and molecules associated with fibrosis induced by chronic stress. CONCLUSIONS Our results strongly suggest that exposure to CUMS induces ovarian fibrosis, which influences follicular development and ultimately contributes to fertility decline. These findings offer novel perspectives on the impact of chronic stress on ovarian dysfunction.
Collapse
Affiliation(s)
- Jie Ma
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750004, China; General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Lu Wang
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Danyu Yang
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Jia Luo
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750004, China; General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Jinmei Gao
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Jinfang Wang
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Hua Guo
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Jialing Li
- Reproductive Medicine Center, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750004, China; General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Feimiao Wang
- Reproductive Medicine Center, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750004, China; General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Ji Wu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Rong Hu
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750004, China; Reproductive Medicine Center, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, Ningxia 750004, China; General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| |
Collapse
|
40
|
Huang P, Wang X, Cao Y, Yang J, Yao R, Liang L, Cheng G, Yang L. Research progress on the use of Salvia miltiorrhiza Bunge extracts in the treatment of pulmonary diseases. Biomed Pharmacother 2024; 179:117282. [PMID: 39146764 DOI: 10.1016/j.biopha.2024.117282] [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: 06/12/2024] [Revised: 08/01/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024] Open
Abstract
Salvia miltiorrhiza Bunge extracts, known for their diverse biological activities, often have remarkable efficacy in treating pulmonary diseases overlooked due to their specific cardiovascular actions. With the recent outbreak of COVID-19, research into pulmonary-related diseases has garnered significant attention. Salvia miltiorrhiza Bunge extracts can be broadly categorized into lipophilic and hydrophilic components; however, a comprehensive summary of their mechanisms in treating pulmonary diseases is lacking. Therefore, this review aims to systematically summarize the therapeutic mechanisms of 10 major Salvia miltiorrhiza Bunge extracts in treating pulmonary fibrosis, lung cancer, acute lung injury, and chronic obstructive pulmonary disease, with the goal of identifying promising options for efficacious therapies.
Collapse
Affiliation(s)
- Peifeng Huang
- School of Integrative medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xuezhen Wang
- School of Integrative medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yingyi Cao
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Jiaming Yang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rongmei Yao
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Leiqin Liang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518000, China.
| | - Long Yang
- School of Integrative medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
41
|
Ren C, Wang Q, Fan S, Mi T, Zhang Z, He D. Toll-Like Receptor 9 Aggravates Pulmonary Fibrosis by Promoting NLRP3-Mediated Pyroptosis of Alveolar Epithelial Cells. Inflammation 2024; 47:1744-1761. [PMID: 38498270 DOI: 10.1007/s10753-024-02006-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024]
Abstract
The apoptosis-prone property of alveolar epithelial cells plays a crucial role in pulmonary fibrosis(PF), but the role of pyroptosis in it is still unclear. Toll-like receptor 9(TLR9) has been reported to play a vital role in the pathogenesis of many diseases. However, the effect of TLR9 on alveolar epithelial cells in PF has not been fully elucidated. Gene expression microarray related to Idiopathic pulmonary fibrosis(IPF) was obtained from the Gene Expression Omnibus(GEO) database. In the mouse model of bleomycin-induced PF, adeno-associated virus(AAV6) was used to interfere with TLR9 to construct TLR9 knockdown mice to study the role of TLR9 in PF, and the specific mechanism was studied by intratracheal instillation of NLR family pyrin domain containing 3(NLRP3) activator. In vitro experiments were performed using A549 cells. Bleomycin-induced pyroptosis in the lung tissue of PF mice increased, and TLR9 protein levels also increased, especially in alveolar epithelial cells. The levels of fibrosis and pyroptosis in lung tissue of TLR9 knockdown mice were improved. We found that TLR9 can bind to the NLRP3, thereby increasing the activation of the NLRP3/caspase-1 inflammasome pathway. When we use the NLRP3 activator, the levels of fibrosis and pyroptosis in lung tissue of TLR9 knockout mice can be counteracted. Pyroptosis of alveolar epithelial cells plays a vital role in PF, and TLR9 can promote NLRP3-mediated pyroptosis of alveolar epithelial cells to aggravate the progression of PF and may become a feasible target for the prevention and treatment of PF.
Collapse
Affiliation(s)
- Chunnian Ren
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child development and Disorders, China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
- Department of Cardiothoracic Surgery, Children's Hospital of Chongqing Medical University , National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Quan Wang
- Department of Cardiothoracic Surgery, Children's Hospital of Chongqing Medical University , National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Shulei Fan
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Mi
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child development and Disorders, China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Zhaoxia Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child development and Disorders, China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China
| | - Dawei He
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child development and Disorders, China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, People's Republic of China.
- Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
42
|
Han X, Zhang A, Meng Z, Wang Q, Liu S, Wang Y, Tan J, Guo L, Li F. Bioinformatics analysis based on extracted ingredients combined with network pharmacology, molecular docking and molecular dynamics simulation to explore the mechanism of Jinbei oral liquid in the therapy of idiopathic pulmonary fibrosis. Heliyon 2024; 10:e38173. [PMID: 39364246 PMCID: PMC11447332 DOI: 10.1016/j.heliyon.2024.e38173] [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: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024] Open
Abstract
Objective Jinbei oral liquid (JBOL), which is derived from a traditional hospital preparation, is frequently utilized to treat idiopathic pulmonary fibrosis (IPF) and has shown efficacy in clinical therapy. However, there are now several obstacles facing the mechanism inquiry, including target proteins, active components, and the binding affinity between crucial compounds and target proteins. To gain additional insight into the mechanisms underlying JBOL in anti-IPF, this study used bioinformation technologies, including network pharmacology, molecular docking, and molecular dynamic simulation, with a substantial amount of data based on realistic constituents. Methods Using network pharmacology, we loaded 118 realistic compounds into the SwissTargetPrediction and SwissADME databases and screened the active compounds and target proteins. IPF-related targets were collected from the OMIM, DisGeNET, and GeneCards databases, and the network of IPF-active constituents was built with Cytoscape 3.10.1. The GO and KEGG pathway enrichment analyses were carried out using Metascape, and the protein-protein interaction (PPI) network was constructed to screen the key targets with the STRING database. Finally, the reciprocal affinity between the active molecules and the crucial targets was assessed through the use of molecular docking and molecular dynamics simulation. Results A total of 122 targets and 34 tested active compounds were summarized in this investigation. Among these, kaempferol, apigenin, baicalein were present in high degree. PPI networks topological analysis identified eight key target proteins. AGE-RAGE, EGFR, and PI3K-Akt signaling pathways were found to be regulated during the phases of cell senescence, inflammatory response, autophagy, and immunological response in anti-IPF of JBOL. It was verified by molecular docking and molecular dynamics simulation that the combining way and binding energy between active ingredients and selected targets. Conclusions This work forecasts the prospective core ingredients, targets, and signal pathways of JBOL in anti-IPF, which has confirmed the multiple targets and pathways of JBOL in anti-IPF and provided the first comprehensive assessment with bioinformatic approaches. With empirical backing and an innovative approach to the molecular mechanism, JBOL is being considered as a potential new medication.
Collapse
Affiliation(s)
- Xinru Han
- Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Pharmacy, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Aijun Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, China
- Institute of Chinese Materia Medica, Shandong Hongji-tang Pharmaceutical Group Co., Ltd., Jinan, China
| | - Zhaoqing Meng
- Institute of Chinese Materia Medica, Shandong Hongji-tang Pharmaceutical Group Co., Ltd., Jinan, China
| | - Qian Wang
- Department of Pharmacy, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Song Liu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunjia Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiaxin Tan
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lubo Guo
- Department of Pharmacy, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Feng Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
43
|
Viswanathan VK, Ghoshal AG, Mohan A, Patil K, Bhargave C, Choudhari S, Mehta S. Patient Profile-Based Management with Nintedanib in Patients with Idiopathic Pulmonary Fibrosis. Pulm Ther 2024:10.1007/s41030-024-00271-1. [PMID: 39340742 DOI: 10.1007/s41030-024-00271-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
A severe and progressive interstitial lung disease (ILD) known as idiopathic pulmonary fibrosis (IPF) has an unknown etiology with poorly defined mechanisms of development. Among the currently prescribed pharmacological interventions for IPF, nintedanib demonstrates the ability to decelerate the deterioration of lung function and yield positive clinical outcomes. Multiple randomized placebo-controlled trials have confirmed the efficacy and acceptable safety profile of nintedanib. Real-world evidence studies also support the use of nintedanib in IPF, being an efficient and well-tolerated treatment option. It has the potential to stabilize the disease progression in patients with ILD. Patients with IPF frequently have comorbidities like diabetes and hypertension, which can exacerbate the course of disease, reduce quality of life, and decrease treatment adherence. For well-informed decision-making, it is important for healthcare professionals to recognize the position of nintedanib therapy in IPF with comorbidities. The gastrointestinal adverse effects, notably diarrhea, dominate the nintedanib safety profile. These can be effectively controlled by closely monitoring side effects, administering anti-diarrheal and anti-emetic drugs, reducing the nintedanib dose, and discontinuing it in case of severe symptoms with an option to reintroduce the treatment after side effects subside. Symptomatic interventions and monitoring of liver enzymes may reduce the occurrence of permanent treatment discontinuations.
Collapse
Affiliation(s)
| | - Aloke G Ghoshal
- National Allergy Asthma Bronchitis Institute, Kolkata, West Bengal, India
| | - Anant Mohan
- Department of Pulmonary, Critical Care & Sleep Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Ketaki Patil
- Medical Affairs & Clinical Research, Sun Pharma Laboratories Limited, Mumbai, India.
| | - Chaitanya Bhargave
- Medical Affairs & Clinical Research, Sun Pharma Laboratories Limited, Mumbai, India
| | - Sanjay Choudhari
- Medical Affairs & Clinical Research, Sun Pharma Laboratories Limited, Mumbai, India
| | - Suyog Mehta
- Medical Affairs & Clinical Research, Sun Pharma Laboratories Limited, Mumbai, India
| |
Collapse
|
44
|
Liu T, Ning J, Fan X, Wei H, Shi G, Fu QB. Identification of immune patterns in idiopathic pulmonary fibrosis patients driven by PLA2G7-positive macrophages using an integrated machine learning survival framework. Sci Rep 2024; 14:22369. [PMID: 39333367 PMCID: PMC11437001 DOI: 10.1038/s41598-024-73625-z] [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: 02/13/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024] Open
Abstract
Patients with advanced idiopathic pulmonary fibrosis (IPF), a complex and incurable lung disease with an elusive pathology, are nearly exclusive candidates for lung transplantation. Improved identification of patient subtypes can enhance early diagnosis and intervention, ultimately leading to better prognostic outcomes for patients. The goal of this study is to identify new immune patterns and biomarkers in patients. Immune subtypes in IPF patients were identified using single-sample gene set enrichment analysis, and immune subtype-related genes were explored using the weighted correlation network analysis algorithm. A machine learning integration framework was used to establish the optimal prognostic model, known as the immune-related risk score (IRS). Single-cell sequencing was conducted to investigate the major role of macrophage-derived PLA2G7 in the immune microenvironment. We assessed the stability of celecoxib in targeting PLA2G7 through molecular docking and surface plasmon resonance. IPF patients present two distinct immune subtypes, one characterized by immune activation and inflammation, and the other by immune suppression. IRS can predict the immune status and prognosis of IPF patients. Furthermore, multi-cohort analysis and single-cell sequencing analysis demonstrated the diagnostic and prognostic value of PLA2G7 derived from macrophages and its role in shaping the inflammatory immune microenvironment in IPF patients. Celecoxib could effectively and stably bind with PLA2G7. PLA2G7, as identified through IRS, demonstrates marked stability in diagnosing and predicting the prognosis of IPF patients as well as predicting their immune status. It can serve as a novel biomarker for IPF patients.
Collapse
Affiliation(s)
- Tianxi Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jingyuan Ning
- Department of Immunology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xiaoqing Fan
- Institute of Microbiological Testing and Inspection, Tianjin Centre for Disease Control and Prevention, Tianjin , People's Republic of China
| | - Huan Wei
- Department of Neurology, The Affiliated Yan'an Hospital of Kunming Medical University, Kunming, People's Republic of China
| | - Guangsen Shi
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, People's Republic of China.
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Qingshan Bill Fu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, People's Republic of China.
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
45
|
Curioni AV, Borie R, Crestani B, Helou DG. Updates on the controversial roles of regulatory lymphoid cells in idiopathic pulmonary fibrosis. Front Immunol 2024; 15:1466901. [PMID: 39386201 PMCID: PMC11461235 DOI: 10.3389/fimmu.2024.1466901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common and severe form of pulmonary fibrosis, characterized by scar formation in the lung interstitium. Transforming growth factor beta (TGF-β) is known as a key mediator in the fibrotic process, acting on fibroblasts and mediating their proliferation and differentiation into myofibroblasts. Although the immune system is not considered responsible for the initiation of IPF, markers of tolerogenic immunity define the pro-fibrotic microenvironment in the lungs. In homeostatic conditions, regulatory T cells (Tregs) constitute the main lymphoid population responsible for maintaining peripheral tolerance. Similar to Tregs, regulatory B cells (Bregs) represent a recently described subset of B lymphocytes with immunosuppressive functions. In the context of IPF, numerous studies have suggested a role for Tregs in enhancing fibrosis, mainly via the secretion of TGF-β. In humans, most studies show increased percentages of Tregs associated with the severity of IPF, although their exact role remains unclear. In mice, the most commonly used model involves triggering acute lung inflammation with bleomycin, leading to a subsequent fibrotic process. Consequently, data are still conflicting, as Tregs may play a protective role during the inflammatory phase and a deleterious role during the fibrotic phase. Bregs have been less studied in the context of IPF, but their role appears to be protective in experimental models of lung fibrosis. This review presents the latest updates on studies exploring the implication of regulatory lymphoid cells in IPF and compares the different approaches to better understand the origins of conflicting findings.
Collapse
Affiliation(s)
- Anna V. Curioni
- Université Paris Cité, Institut national de la santé et de la recherche médicale (INSERM), Physiopathologie et épidémiologie des maladies respiratoires (PHERE), Paris, France
| | - Raphaël Borie
- Université Paris Cité, Institut national de la santé et de la recherche médicale (INSERM), Physiopathologie et épidémiologie des maladies respiratoires (PHERE), Paris, France
- Service Pneumologie A, Assistance publique – Hôpitaux de Paris (AP-HP), Hôpital Bichat, Paris, France
| | - Bruno Crestani
- Université Paris Cité, Institut national de la santé et de la recherche médicale (INSERM), Physiopathologie et épidémiologie des maladies respiratoires (PHERE), Paris, France
- Service Pneumologie A, Assistance publique – Hôpitaux de Paris (AP-HP), Hôpital Bichat, Paris, France
| | - Doumet Georges Helou
- Université Paris Cité, Institut national de la santé et de la recherche médicale (INSERM), Physiopathologie et épidémiologie des maladies respiratoires (PHERE), Paris, France
| |
Collapse
|
46
|
Jin L, Bao B, Huang XT, Tao JH, Duan JX, Zhong WJ, Zhang CY, Liu YB, Chen H, Yang NSY, Guan CX, Zhou Y. MEOX1 triggers myofibroblast apoptosis resistance, contributing to pulmonary fibrosis in mice. J Cell Physiol 2024:e31442. [PMID: 39319990 DOI: 10.1002/jcp.31442] [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: 06/06/2024] [Revised: 08/22/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Abstract
The apoptosis resistance of myofibroblasts is a hallmark in the irreversible progression of pulmonary fibrosis (PF). While the underlying molecular mechanism remains elusive. In this study, we unveiled a previously unrecognized mechanism underlying myofibroblast apoptosis resistance during PF. Our investigation revealed heightened expression of mesenchyme homeobox 1 (MEOX1) in the lungs of idiopathic pulmonary fibrosis (IPF) patients and bleomycin-induced PF mice. Silencing MEOX1 significantly attenuated PF progression in mice. In vitro, we found a notable increase in MEOX1 expression in transforming growth factor-β1 (TGF-β1)-induced myofibroblasts. Silencing MEOX1 enhanced apoptosis of myofibroblasts. Mechanistically, we identified G-protein signaling pathway regulatory factor 4 (RGS4) as a critical downstream target of MEOX1, as predicted by bioinformatics analysis. MEOX1 enhanced apoptosis resistance by upregulating RGS4 expression in myofibroblasts. In conclusion, our study highlights MEOX1 as a promising therapeutic target for protecting against PF by modulating myofibroblast apoptosis resistance.
Collapse
Affiliation(s)
- Ling Jin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Bo Bao
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, China
| | - Jia-Hao Tao
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Jia-Xi Duan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Wen-Jin Zhong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Yu-Biao Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Hui Chen
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Nan-Shi-Yu Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, China
- National Experimental Teaching Demonstration Center for Medical Function, Changsha, China
| |
Collapse
|
47
|
Wang S, Yu H, Liu S, Liu Y, Gu X. Regulation of idiopathic pulmonary fibrosis: a cross-talk between TGF- β signaling and MicroRNAs. Front Med (Lausanne) 2024; 11:1415278. [PMID: 39386739 PMCID: PMC11461268 DOI: 10.3389/fmed.2024.1415278] [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: 04/10/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Pulmonary fibrosis (PF) is a highly complex and challenging disease affecting the respiratory system. Patients with PF usually have an abbreviated survival period and a consequential high mortality rate after the diagnosis is confirmed, posing serious threats to human health. In clinical practice, PF is typically treated by antifibrotic agents, such as Pirfenidone and Nintedanib. However, these agents have been reported to correlate with substantial adverse effects, escalating costs, and insufficient efficacy. Moreover, it remains unclarified about the multifactorial pathology of PF. Therefore, there is an urgent demand for elucidating these underlying mechanisms and identifying safe, efficient, and targeted therapeutic strategies for PF treatment. The crucial role of the transforming growth factor-β (TGF-β) signaling pathway in PF development has been explored in many studies. MicroRNAs (miRNAs), which function as post-transcriptional regulators of gene expression, can significantly affect the development of PF by modulating TGF-β signaling. In turn, TGF-β signaling can regulate the expression and biogenesis of miRNAs, thereby substantially affecting the progression of PF. Hence, the therapeutic strategies that focus on the drug-targeted regulation of miRNAs, either by augmenting down-regulated miRNAs or inhibiting overexpressed miRNAs, may hinder the pathways related to TGF-β signaling. These strategies may contribute to the prevention and suppression of PF progression and may provide novel insights into the treatment of this disease.
Collapse
Affiliation(s)
| | | | | | | | - Xiu Gu
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
48
|
Wei S, Liu Y, Ran C, Li Y, Tang B, Lu M, Wang H. Calpain-1 Up-Regulation Promotes Bleomycin-Induced Pulmonary Fibrosis by Activating Ferroptosis. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00356-0. [PMID: 39326733 DOI: 10.1016/j.ajpath.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/30/2024] [Accepted: 09/03/2024] [Indexed: 09/28/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and fatal disease. Calpain-1 was shown to be an effective therapeutic target for vascular endothelial dysfunction and pulmonary hypertension. However, the role of calpain-1 in bleomycin (BLM)-induced IPF has not been defined. The aim of this study was to assess the targeting of calpain-1 by activating ferroptosis in BLM-treated knockout mice and murine lung epithelial-12 cells. In this study, the role of calpain-1 in the regulation of IPF was investigated using a BLM-induced IPF mouse model. The results of our study showed that increased expression of calpain-1 was accompanied by increased fibrosis, lipid peroxidation, iron ion accumulation, and YAP levels and decreased levels of p-AMPK in BLM-induced IPF. MDL-28170 (calpain-1 inhibition) treatment and calpain-1 knockdown alleviated ferroptosis and IPF induced by BLM. Overexpression of calpain-1 in murine lung epithelial-12 cells further exacerbated iron accumulation and IPF. Mechanistically, lentivirus-mediated up-regulation of calpain-1 inhibited AMPK activity and promoted the nuclear translocation of YAP, leading to high levels of acyl-CoA synthetase long-chain family 4 -and transferrin receptor protein 1 and triggering a ferroptosis response that ultimately exacerbated BLM-induced lung fibrosis. Calpain-1 inhibition reversed these results and ameliorated BLM-induced IPF. In conclusion, these findings suggest that the calpain-1-acyl-CoA synthetase long-chain family 4-transferrin receptor protein 1-ferroptosis-positive regulatory axis contributes to BLM-induced IPF, which indicates that calpain-1 has potential therapeutic value for the treatment of IPF.
Collapse
Affiliation(s)
- Silin Wei
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Yu Liu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Chenyang Ran
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Yunhan Li
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Bailin Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Meili Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China.
| | - Hongxin Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
49
|
Atzeni F, Alciati A, Gozza F, Masala IF, Siragusano C, Pipitone N. Interstitial lung disease in rheumatic diseases: an update of the 2018 review. Expert Rev Clin Immunol 2024:1-18. [PMID: 39302018 DOI: 10.1080/1744666x.2024.2407536] [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: 07/20/2023] [Revised: 07/25/2024] [Accepted: 09/18/2024] [Indexed: 09/22/2024]
Abstract
INTRODUCTION Interstitial lung disease (ILD) is a potential severe complication of various rheumatic diseases, typically connective tissue diseases (CTD), associated with significant morbidity and mortality. ILD may occur during the course of the disease but may also be its first manifestation. Several cell types are involved in ILD's pathogenesis, and if not controlled, pulmonary inflammation may lead to pulmonary fibrosis. AREAS COVERED We searched PubMed, Medline, and the Cochrane Library for papers published between 1995 and February 2017 in the first version, and between 2017 and April 2023 using combinations of words. The most frequent systemic rheumatic diseases associated with ILD are systemic sclerosis (SSc), rheumatoid arthritis (RA), and idiopathic inflammatory myositis. Treatment and monitoring guidelines are still lacking, and current treatment strategies have been extrapolated from the literature on SSc and established treatments for non-pulmonary systemic rheumatic manifestations. EXPERT OPINION Given the complexity of diagnosis and the paucity of treatment trials, managing CTD patients with ILD is challenging. It requires the skills of multidisciplinary CTD-ILD clinics including at least rheumatologists and lung specialists.
Collapse
Affiliation(s)
- Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Messina, Italy
| | - Alessandra Alciati
- Department of Clinical Neurosciences, Villa S. Benedetto Menni, Albese, Como, Italy
- Humanitas Clinical and Research Center, Rozzano, Italy
| | - Francesco Gozza
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Messina, Italy
| | | | - Cesare Siragusano
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Messina, Italy
| | - Nicolò Pipitone
- Rheumatology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
50
|
Wang Q, Ning Y, Zhang J, Du X, Xu Z, Hu Y, Gao F, Chen Y. Rapamycin and Hyperoside-Co-loaded Macrophage Delivery System Enhanced Pulmonary Fibrosis Therapy by Autophagy Upregulation and Epithelial-to-Mesenchymal Transition Inhibition. ACS APPLIED MATERIALS & INTERFACES 2024; 16:48993-49002. [PMID: 39225760 DOI: 10.1021/acsami.4c07604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Pulmonary fibrosis is a lethal interstitial lung disease, for which current treatments are inadequate in halting its progression. A significant factor contributing to the development of fibrosis is insufficient autophagy, which leads to increased fibroblast proliferation and collagen deposition. However, treatments aimed at upregulating autophagy often cause further lung pathology due to the disruption of epithelial cell balance. In response, we have developed a novel macrophage delivery system loaded with an epithelial-to-mesenchymal transition inhibitor, hyperoside (HYP), and an autophagy inducer, rapamycin (RAP). This system targets the fibrotic areas of the lung through chemotaxis, releases liposomes via macrophage extracellular traps, and effectively inhibits fibroblast proliferation while restoring the alveolar structure through the combined effects of RAP and HYP, ultimately reducing lung pathology without causing systemic toxicity. Our findings not only highlight a promising method to enhance autophagy-based treatments for pulmonary fibrosis but also demonstrate the potential of macrophages as effective nanocarriers for drug delivery.
Collapse
Affiliation(s)
- Qi Wang
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yuanmeng Ning
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jinru Zhang
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xu Du
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zihan Xu
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yongcheng Hu
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Feng Gao
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yanzuo Chen
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|