1
|
Li D, Qian L, Du Y, Liu L, Sun Z, Han Y, Guo X, Shen C, Zhang Z, Liu X. METTL14-mediated m 6A modification of DDIT4 promotes its mRNA stability in aging-related idiopathic pulmonary fibrosis. Epigenetics 2025; 20:2462898. [PMID: 39916577 PMCID: PMC11810098 DOI: 10.1080/15592294.2025.2462898] [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: 06/23/2024] [Revised: 01/01/2025] [Accepted: 01/29/2025] [Indexed: 02/12/2025] Open
Abstract
Although N6-methyladenosine (m6A) may be related to the pathogenesis of fibrotic process, the mechanism of m6A modification in aging-related idiopathic pulmonary fibrosis (IPF) remains unclear. Three-milliliter venous blood was collected from IPF patients and healthy controls. MeRIP-seq and RNA-seq were utilized to investigate differential m6A modification. The expressions of identified m6A regulator and target gene were validated using MeRIP-qPCR and real-time PCR. Moreover, we established an animal model and a senescent model of A549 cells to explore the associated molecular mechanism. Our study provided a panorama of m6A methylation in IPF. Increased peaks (3756) and decreased peaks (4712) were observed in the IPF group. The association analysis showed that 749 DEGs were affected by m6A methylation in IPF. Among the m6A regulators, the expression of METTL14 decreased in IPF. The m6A level of our interested gene DDIT4 decreased significantly, but the mRNA level of DDIT4 was higher in IPF. This was further verified in bleomycin-induced pulmonary fibrosis. At the cellular level, it was further confirmed that METTL14 and DDIT4 might participate in the senescence of alveolar epithelial cells. The downregulation of METTL14 might inhibit the decay of DDIT4 mRNA by reducing the m6A modification level of DDIT4 mRNA, leading to high expression of DDIT4 mRNA and protein. Our study provided a panorama of m6A alterations in IPF and discovered METTL14 as a potential intervention target for epigenetic modification in IPF. These results pave the way for future investigations regarding m6A modifications in aging-related IPF.
Collapse
Affiliation(s)
- Dan Li
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of Geriatrics, the First Hospital of Shanxi Medical University, Taiyuan, China
| | - Li Qian
- Department of Geriatrics, the First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yufeng Du
- Department of Geriatrics, the First Hospital of Shanxi Medical University, Taiyuan, China
| | - Lifang Liu
- Department of Geriatrics, the First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ziyue Sun
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yongkang Han
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Xiangrui Guo
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Chao Shen
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zheng Zhang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Xuejun Liu
- Department of Geriatrics, the First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
2
|
Bhardwaj P, Maruthi M. Hyperglycaemia-induced fibrotic and inflammatory gene expression alterations in lung epithelial cells: Implications for pulmonary fibrosis development. Gene 2025; 959:149520. [PMID: 40254080 DOI: 10.1016/j.gene.2025.149520] [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: 03/17/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/22/2025]
Abstract
Hyperglycaemia has a significant long-term impact on multiple organ systems, including renal, cardiovascular, central nervous, hepatic and ocular systems, leading to the gradual loss of their functional abilities. Numerous studies have elucidated the pathophysiology, etiology, and consequences of hyperglycaemia on these organs. The pulmonary system is also considered as a target of hyperglycaemia, several factors cause lung injury which leads to the development of pulmonary fibrosis, a chronic fibrotic disease with usual interstitial pneumonia patterns. Nevertheless, the effects of hyperglycaemia on the development of pulmonary fibrosis remain poorly understood. We intend to understand the cellular and morphological changes, and the progression of fibrosis in lung epithelial cells subjected to hyperglycaemia. Our experimental data indicate that hyperglycaemia induces fibrotic and inflammatory alterations in cultured lung epithelial cells. These alterations are facilitated by the upregulation of genes related to fibrosis and inflammation, promoting cell proliferation and migration. Further research is required to comprehensively elucidate the impact of hyperglycaemia during lung injury progression of fibrosis, these findings may reveal novel mechanisms that may help in the assessment and treatment of lung ailments in people with hyperglycaemia.
Collapse
Affiliation(s)
- Priya Bhardwaj
- Department of Biochemistry, School of Interdisciplinary and Applied Sciences, Central University of Haryana
| | - Mulaka Maruthi
- Department of Biochemistry, School of Interdisciplinary and Applied Sciences, Central University of Haryana.
| |
Collapse
|
3
|
Wilson TM, Bolt M, Stahly A, Lee JS, Bang TJ, Sachs PB, Deane KD, Humphries SM, Solomon JJ, Demoruelle MK. Transforming growth factor-beta is increased in sputum from individuals with rheumatoid arthritis-associated pulmonary fibrosis. Rheumatology (Oxford) 2025; 64:3989-3995. [PMID: 39693122 PMCID: PMC12107045 DOI: 10.1093/rheumatology/keae697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/04/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Interstitial lung disease (ILD) develops in 5-10% of patients with RA and contributes significantly to morbidity and mortality, particularly in those with a fibrotic phenotype. Yet, biomarkers to reliably identify RA patients with underlying pulmonary fibrosis are inadequate. Herein, we used sputum to identify lung-based biomarkers that distinguish RA patients with underlying pulmonary fibrosis and may better inform underlying pathogenesis in RA-ILD. METHODS We included 37 RA patients with pulmonary fibrosis (RA-PF) and 30 RA patients without ILD (RA-no-ILD). Induced sputum and serum were tested for TGF-β levels by immunoassay. DNA was extracted to determine presence of the MUC5B ILD-risk allele ('T'). High-resolution CT (HRCT) and pulmonary function tests (PFTs) were completed within 3 months of sputum collection and quantified to determine lung disease severity. RESULTS Sputum TGF-β was significantly elevated in individuals with RA-PF compared with RA-no-ILD (P < 0.001) and correlated with more fibrosis on HRCT (P = 0.005) and lower forced vital capacity (P = 0.006) and diffusion capacity of carbon monoxide (P = 0.044) on PFTs. Within RA-PF patients, sputum TGF-β was higher in those with the MUC5B ILD-risk genotype (GT/TT) (P = 0.038). There were no differences in serum levels of TGF-β between groups. CONCLUSION We demonstrate that sputum levels of TGF-β are significantly elevated in individuals with RA-PF, correlate with lung disease severity, and are elevated in those with the MUC5B ILD-risk polymorphism. These findings could identify novel approaches to ILD screening in RA and potential targeted therapeutic strategies for RA-ILD.
Collapse
Affiliation(s)
- Timothy M Wilson
- Division of Rheumatology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Matthew Bolt
- Department of Biostatistics, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Andrew Stahly
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Joyce S Lee
- Division of Pulmonary and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Tami J Bang
- Department of Radiology, National Jewish Health, Denver, CO, USA
| | - Peter B Sachs
- Department of Radiology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Kevin D Deane
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | | | - Joshua J Solomon
- Center for Interstitial Lung Disease, National Jewish Health, Denver, CO, USA
| | - M Kristen Demoruelle
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| |
Collapse
|
4
|
Quan J, Ma C, Zhao X, Guo Y, Qu W, Zhou X, Ma E, Xu Y. Discovery of novel selective HDAC6 inhibitors via a scaffold hopping approach for the treatment of idiopathic pulmonary fibrosis (IPF) in vitro and in vivo. Bioorg Chem 2025; 159:108360. [PMID: 40112668 DOI: 10.1016/j.bioorg.2025.108360] [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: 01/22/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/22/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, irreversible, and fatal pulmonary disease. Owing to its complex pathogenesis and lack of effective treatment, patients have a short survival time after diagnosis. Although pirfenidone and nintedanib can mitigate declines in lung function, neither has stopped the progression of IPF nor significantly improved long-term survival in patients. HDAC6 inhibitors have been reported to inhibit TGF-β1-induced collagen expression to protect mice from pulmonary fibrosis, and this pharmacological mechanism has been supported by immunohistochemical studies of HDAC6 overexpression in IPF lung tissue. In this study, a series of novel derivatives were obtained based on the reported active compounds through the ring closure strategy in scaffold hopping theory. Compound W28 was selected from in vitro screening for better HDAC6 selectivity, and it was used for in-depth pharmacokinetic and pharmacodynamic studies. Detailed molecular docking studies, molecular dynamics (MD) simulations and the structure-activity relationship (SAR) discussion will contribute to guiding the design of new molecules. In further studies, the ability of W28 to inhibit the IPF phenotype was confirmed, and the corresponding pharmacological mechanism was also demonstrated. Moreover, the pharmacokinetic characteristics of W28 were also tested to guide pharmacodynamic studies in vivo, and the therapeutic effect of W28 on bleomycin-induced pulmonary fibrosis in mice was found to be satisfactory. The results reported in this paper may provide a reference for promoting the discovery of new selective HDAC6 inhibitors as drug molecules for the treatment of IPF.
Collapse
Affiliation(s)
- Jishun Quan
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Chao Ma
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Xianchen Zhao
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Yuxi Guo
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Wenhui Qu
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Xinru Zhou
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Enlong Ma
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China.
| | - Yongnan Xu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China.
| |
Collapse
|
5
|
Shen J, Jiang Y, Bu W, Yu M, Huang R, Tang C, Yang Z, Gao H, Su L, Cheng D, Zhao X. Protein Ubiquitination Modification in Pulmonary Fibrosis. Compr Physiol 2025; 15:e70013. [PMID: 40312137 DOI: 10.1002/cph4.70013] [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: 02/05/2025] [Revised: 03/31/2025] [Accepted: 04/22/2025] [Indexed: 05/03/2025]
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive fibrotic interstitial lung disease characterized by a high incidence and mortality rate, which encompasses features, such as diffuse alveolar inflammation, invasive fibroblast activation, and uncontrolled extracellular matrix (ECM) deposition. Beyond the local pathological processes, PF can be better understood in light of interorgan communication networks that are involved in its progression. Notably, pulmonary inflammation can affect cardiovascular, renal, hepatic, and neural functions, highlighting the importance of understanding these systemic interactions. Posttranslational modifications play a crucial role in regulating protein function, localization, stability, and activity. Specifically, protein ubiquitination modifications are involved in PF induced by various stimuli, involving a range of ubiquitin-modifying enzymes and substrates. In this review, we provide an overview of how E3 ubiquitin ligases and deubiquitinating enzymes (DUBs) modulate PF through several signaling pathways, such as TGF-β, Wnt, metabolic activity, aging, ferroptosis, endoplasmic reticulum stress, and inflammatory responses. This perspective includes the role of ubiquitin-proteasome systems in interorgan communication, affecting the progression of PF and related systemic conditions. Additionally, we also summarize the currently available therapeutic compounds targeting protein ubiquitination-related enzymes or ubiquitination substrates for the treatment of PF. Understanding the interplay between ubiquitination and interorgan communication may pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Jinping Shen
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
- Nantong Center for Disease Control and Prevention, Nantong, China
| | - Yuling Jiang
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Wenxia Bu
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Mengjiao Yu
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Ruiyao Huang
- Department of Clinical Medicine, Nantong University Xinglin College, Nantong, China
| | - Can Tang
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Zeyun Yang
- Nantong Center for Disease Control and Prevention, Nantong, China
| | - Haiping Gao
- Nantong Center for Disease Control and Prevention, Nantong, China
| | - Liling Su
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, China
| | - Demin Cheng
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Xinyuan Zhao
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| |
Collapse
|
6
|
Zhou M, Liu S, Yuan F, Li J, Zhou M, Huang J, Zhang Y, Liang Q. Isomeric 2-isobutylmalate derivatives with anti-pulmonary fibrosis effects from the leaves of Bletilla striata via LC-MS/MS-based molecular networking. Bioorg Chem 2025; 159:108351. [PMID: 40117752 DOI: 10.1016/j.bioorg.2025.108351] [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: 12/09/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/23/2025]
Abstract
Based on the LC-MS/MS molecular networking strategy, nine undescribed 2-isobutylmalate derivatives, namely bletistrosides M-U (compounds 1-7, 9, and 11), together with two known analogues (compounds 8 and 10), were isolated and identified from the leaves of Bletilla striata. Their structures with absolute configurations were deduced from spectroscopic data, acidic hydrolysis, and comparison with reported compounds. Compounds 1/2, 3/4, 5/6, and 7/8 represented four pairs of Z/E isomers regarding cinnamoyl groups, and each pair underwent interconversion under UV radiation at 254 nm. Biologically, compounds 1, 2, and 10 exhibited anti-pulmonary fibrosis effects against bleomycin-stimulated cell injury in A549 cells. Further investigations demonstrated that the anti-pulmonary fibrosis potential of 2 was related to the inhibition of apoptosis and epithelial-mesenchymal transition by blocking the Bax/Bcl-2, TGF-β1/Smad2/3, and PI3K/AKT signaling pathways, while concurrently enhancing the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Ming Zhou
- Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, People's Republic of China
| | - Si Liu
- Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, People's Republic of China
| | - Fang Yuan
- Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, People's Republic of China
| | - Jun Li
- College of Pharmacy, South-Central Minzu University, Wuhan 430074, People's Republic of China
| | - Mengchen Zhou
- National Demonstration Center for Experimental Basic Medical Education, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Junfeng Huang
- Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, People's Republic of China
| | - Yanjun Zhang
- Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, People's Republic of China
| | - Qiong Liang
- Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, People's Republic of China.
| |
Collapse
|
7
|
Liao CY, Hundscheid JH, Crawford J, ten Dijke P, Coornaert B, Danen EH. Novel high throughput 3D ECM remodeling assay identifies MEK as key driver of fibrotic fibroblast activity. Mater Today Bio 2025; 32:101800. [PMID: 40343164 PMCID: PMC12059351 DOI: 10.1016/j.mtbio.2025.101800] [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: 07/30/2024] [Revised: 01/21/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
In fibrotic tissues, activated fibroblasts remodel the collagen-rich extracellular matrix (ECM). Intervening with this process represents a candidate therapeutic strategy to attenuate disease progression. Models that generate quantitative data on 3D fibroblast-mediated ECM remodeling with the reproducibility and throughput needed for drug testing are lacking. Here, we develop a model that fits this purpose and produces combined quantitative information on drug efficacy and cytotoxicity. We use microinjection robotics to design patterns of fibrillar collagen-embedded fibroblast clusters and apply automated microscopy and image analysis to quantify ECM remodeling between-, and cell viability within clusters of TGFβ-activated primary human skin or lung fibroblasts. We apply this assay to compound screening and reveal actionable targets to suppress fibrotic ECM remodeling. Strikingly, we find that after an initial phase of fibroblast activation by TGFβ, canonical TGFβ signaling is dispensable and, instead, non-canonical activation of MEK-ERK signaling drives ECM remodeling. Moreover, we reveal that higher concentrations of two TGFβ receptor inhibitors while blocking canonical TGFβ signaling, in fact stimulate this MEK-mediated profibrotic ECM remodeling activity.
Collapse
Affiliation(s)
- Chen-Yi Liao
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | | | | | - Peter ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Erik H.J. Danen
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| |
Collapse
|
8
|
Elshal M, Hamdi A, Khalil IT, Arafa ESA, Althobaiti MM, Arab HH, Haikal A. 3,4-Dimethoxycinnamic acid from coffee silverskin biowaste ameliorates bleomycin-induced pulmonary fibrosis via modulating caveolin-1-dependent activation of NF-κB, TGF-β1/Smad3, and ERK1/2 signaling pathways. Toxicol Appl Pharmacol 2025; 501:117414. [PMID: 40425068 DOI: 10.1016/j.taap.2025.117414] [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: 02/22/2025] [Revised: 05/02/2025] [Accepted: 05/23/2025] [Indexed: 05/29/2025]
Abstract
The aim of the current study was to explore the potential beneficial effect of 3,4-dimethoxycinnamic acid (DMCA), obtained from coffee silverskin (CS) biowaste, on bleomycin-induced lung fibrosis in rats and elucidate its possible underlying mechanisms. Lung fibrosis was induced in rats by a single intratracheal administration of bleomycin. DMCA (25 and 50 mg/kg) and pirfenidone (50 mg/kg) were orally administered. Our results showed that DMCA markedly reduced lactate dehydrogenase activity, total protein content, and total and differential inflammatory cell counts in the bronchoalveolar lavage fluid. DMCA also inhibited oxidative stress and nuclear factor kappa B (NF-κB)-mediated tumor necrosis factor and interleukin-1β production. Furthermore, DMCA markedly downregulated the pulmonary levels of transforming growth factor beta 1 (TGF-β1), SMAD family member 3 (Smad3), and extracellular signal-regulated kinase 1/2 (ERK1/2), concurrently with a marked reduction in collagen-1 expression in the lung. Contrarily, DMCA upregulated lung caveolin-1 expression. These findings were concomitantly accompanied by remarkable improvement in the lung histopathology. In conclusion, DMCA effectively mitigates bleomycin-induced lung fibrosis in rats, which is comparable to pirfenidone, through antioxidant, anti-inflammatory, and anti-fibrotic effects. These beneficial effects are mainly mediated through boosting caveolin-1 expression that halts NF-κB, TGF-β1/smad3, and ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Mahmoud Elshal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Abdelrahman Hamdi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Ibrahim T Khalil
- Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - El-Shaimaa A Arafa
- College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates.
| | - Musaad M Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia..
| | - Hany H Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia..
| | - Abdullah Haikal
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
9
|
Wan HQ, Xie LF, Li HL, Ma Y, Li QH, Dai MQ, Fu YD, Li WJ, Zhou JP, Qian MY, Shen X. GPR40 activation alleviates pulmonary fibrosis by repressing M2 macrophage polarization through the PKD1/CD36/TGF-β1 pathway. Acta Pharmacol Sin 2025:10.1038/s41401-025-01558-y. [PMID: 40369224 DOI: 10.1038/s41401-025-01558-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 04/01/2025] [Indexed: 05/16/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive interstitial lung disease characterized by complex aetiologies involving the accumulation of inflammatory cells, such as macrophages, in the alveoli. This process is driven by uncontrolled extracellular matrix (ECM) deposition and the development of fibrous connective tissues. Here, we observed that the mRNA expression of Ffar1, the gene encoding G protein-coupled receptor 40 (GPR40), is repressed, while Cd36 is increased in the bronchoalveolar lavage fluid (BALF), which is predominantly composed of alveolar macrophages, of IPF patients. Furthermore, the GPR40 protein was found to be largely adhered to macrophages and was pathologically downregulated in the lungs of bleomycin (BLM)-induced PF model mice (PF mice) compared with those of control mice. Specific knockdown of GPR40 in pulmonary macrophages by adeno-associated virus 9-F4/80-shGPR40 (AAV9-shGPR40) exacerbated the fibrotic phenotype in the PF mice, and activation of GPR40 by its determined agonist compound SC (1,3-dihydroxy-8-methoxy-9H-xanthen-9-one) effectively protected the PF mice from pathological exacerbation. Moreover, Ffar1 or Cd36 gene knockout mouse-based assays were performed to explore the mechanism underlying the regulation of GPR40 activation in pulmonary macrophages with compound SC as a probe. We found that compound SC mitigated pulmonary fibrosis progression by preventing M2 macrophage polarization from exerting profibrotic effects through the GPR40/PKD1/CD36 axis. Our results strongly support the therapeutic potential of targeting intrinsic GPR40 activation in pulmonary macrophages for IPF and highlight the potential of compound SC in treating this disease.
Collapse
Affiliation(s)
- Hui-Qi Wan
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ling-Feng Xie
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Hong-Lin Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yan Ma
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qiu-Hui Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Meng-Qing Dai
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuan-Dong Fu
- Pulmonary Disease Department, Nanjing Pukou District Central Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wen-Jun Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jin-Pei Zhou
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, China.
| | - Min-Yi Qian
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xu Shen
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing, China.
| |
Collapse
|
10
|
Nguyen H, Juang U, Gwon S, Jung W, Huang Q, Lee S, Lee B, Kwon SH, Kim SH, Park J. Effect of CTMP1 gene on pulmonary fibrosis. Toxicol Res 2025; 41:235-244. [PMID: 40291111 PMCID: PMC12021751 DOI: 10.1007/s43188-024-00269-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 10/09/2024] [Accepted: 11/13/2024] [Indexed: 04/30/2025] Open
Abstract
Protein kinase B (PKB/AKT) is a very important member of the protein kinase family, playing significant roles in various crucial processes including insulin-signaling, cell survival, growth, and metabolism. The carboxyl-terminal modulator protein 1 (CTMP1) inhibits PKB, primarily by attenuating its phosphorylation. Idiopathic pulmonary fibrosis (IPF) is an irreversible, chronic, progressive pulmonary disorder; the clinical treatment options are limited. Of the various experimental models, bleomycin-induced lung fibrosis is the most extensively studied. It closely resembles human lung fibrosis. We explored the impact of CTMP1 on bleomycin-induced fibrosis. In vitro experiments involved knockdown of CTMP1 in A549 cells (human alveolar epithelial cells), followed by bleomycin treatment. In vivo, lung fibrosis was induced in mice with ablated CTMP1 via intratracheal bleomycin administration at 2 mg/kg. CTMP1 deletion reduced pulmonary fibrosis and the epithelial-to-mesenchymal transition by inhibiting PKB phosphorylation. These findings suggest that CTMP1 plays a pivotal role in the regulation of lung fibrosis, offering new insights into potential therapeutic approaches for IPF patients. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-024-00269-6.
Collapse
Affiliation(s)
- Huonggiang Nguyen
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015 Republic of Korea
| | - Uijin Juang
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015 Republic of Korea
| | - Suhwan Gwon
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015 Republic of Korea
| | - Woohyeong Jung
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015 Republic of Korea
| | - Quingzhi Huang
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015 Republic of Korea
| | - Soohyeon Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015 Republic of Korea
| | - Beomwoo Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015 Republic of Korea
| | - So Hee Kwon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, 21983 Republic of Korea
| | - Seon-Hwan Kim
- Department of Neurosurgery, Institute for Cancer Research, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015 Republic of Korea
| | - Jongsun Park
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, 35015 Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015 Republic of Korea
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon, Republic of Korea
| |
Collapse
|
11
|
Tondi F, Cirsmaru RA, Conti C, Follenzi A, Gresele P, Olgasi C, Bury L. Hermansky-Pudlak Syndrome: From Molecular Pathogenesis to Targeted Therapies. IUBMB Life 2025; 77:e70025. [PMID: 40387003 PMCID: PMC12086961 DOI: 10.1002/iub.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/21/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025]
Abstract
Hermansky-Pudlak syndrome (HPS) is a rare inherited disorder caused by defects in lysosome-related organelles (LROs) in various tissues, including platelets, melanocytes, and endothelial cells. Key features of HPS include oculocutaneous albinism, bleeding tendency, and, in some cases, pulmonary fibrosis, granulomatous colitis, and immunodeficiency. The condition is linked to mutations in 11 genes involved in the formation of LROs. Currently, treatment options for HPS are limited and often ineffective. Though cell and gene therapies have been explored for melanosomes and epithelial cells, there is limited knowledge about their application to platelets and endothelial cells. Understanding the detailed mechanisms of HPS pathogenesis is crucial, and using induced pluripotent stem cell (iPSC) models may provide valuable insights into the disease's molecular processes, aiding the development of new treatments. In this review, we will focus on the genetics and molecular mechanisms of HPS, on its clinical manifestations and current therapeutic approaches, highlighting the need for further research into the disease mechanisms and potential innovative therapies.
Collapse
Affiliation(s)
- Francesca Tondi
- Department of Medicine and Surgery, Section of Internal and Cardiovascular MedicineUniversity of PerugiaPerugiaItaly
| | | | - Chiara Conti
- Department of Medicine and Surgery, Section of Internal and Cardiovascular MedicineUniversity of PerugiaPerugiaItaly
| | - Antonia Follenzi
- Department of Health Sciences, School of MedicineUniversity of Piemonte OrientaleNovaraItaly
- Dipartimento Attività Integrate Ricerca InnovazioneAzienda Ospedaliero‐Universitaria SS. Antonio e Biagio e C. ArrigoAlessandriaItaly
| | - Paolo Gresele
- Department of Medicine and Surgery, Section of Internal and Cardiovascular MedicineUniversity of PerugiaPerugiaItaly
| | - Cristina Olgasi
- Department of Translational Medicine, School of MedicineUniversity of Piemonte OrientaleNovaraItaly
| | - Loredana Bury
- Department of Medicine and Surgery, Section of Internal and Cardiovascular MedicineUniversity of PerugiaPerugiaItaly
| |
Collapse
|
12
|
Liu H, Tan S, Zhao J, Lin X. Changes in Serum PDGF-C and TGF-β1 Levels After PCI in Premature Coronary Artery Disease: Combined Predictive Value for MACCE. Int J Gen Med 2025; 18:2367-2377. [PMID: 40321937 PMCID: PMC12050022 DOI: 10.2147/ijgm.s510456] [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: 12/06/2024] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
Objective This study evaluates dynamic changes in platelet derived growth factor C (PDGF-C) and transforming growth factor β1 (TGF-β1) levels after percutaneous coronary intervention (PCI) in patients with premature coronary artery disease (PCAD) and their combined predictive value for major adverse cardiac and cerebrovascular events (MACCE). Methods A total of 100 PCAD patients admitted to the hospital from July 2021 to July 2023 who had completed 2 years of follow-up were retrospectively selected as the research objects. The patients were divided into MACCE group and non-MACCE group according to the occurrence of MACCE. The changes of serum PDGF-C and TGF-β1 levels were compared before operation, 1 year after operation and 2 years after operation. Cox regression was used to test the influencing factors. Receiver operating characteristic (ROC) curve was used to predict the predictive value. The decision curve was used to analyze the predicting value of serum PDGF-C and TGF-β1. Results Compared with that before operation, serum PDGF-C levels increased, while TGF-β1 levels decreased at 1 year and 2 years post-PCI (P<0.05). The levels of hs-CRP, HDL-C, MPV and PDGF-C in the MACCE group were higher than those in the non-MACCE group, and the level of TGF-β1 was lower than that in the non-MACCE group (P<0.05). The hs-CRP, MPV and PDGF-C were identified as independent risk factors for MACCE (HR>1, P<0.05), and TGF-β1 was identified as a protective factor (HR<1, P<0.05). The AUC of PDGF-C levels and TGF-β1 levels n in predicting MACCE after PCI in PCAD patients were 0.796 and 0.837, respectively. Combined prediction has higher sensitivity and specificity than individual markers. The decision curve showed that within the threshold range of 0.141-0.202 and 0.216-0.998, the net return rate of the combination of PDGF-C and TGF-β1 levels in predicting MACCE after PCI in PCAD patients was better than that of either alone. Conclusion hs-CRP, MPV, PDGF-C and TGF-β1 were the influencing factors of MACCE in PCAD patients after PCI. Combined detection of PDGF-C and TGF- β1 enhanced predictive accuracy for MACCE, offering potential value for risk stratification in PCAD patients post-PCI.
Collapse
Affiliation(s)
- Haide Liu
- Department of Cardiology, Guigang City People’s Hospital, Guigang, Guangxi, 537100, People’s Republic of China
| | - Shanglang Tan
- Department of Cardiology, Guigang City People’s Hospital, Guigang, Guangxi, 537100, People’s Republic of China
| | - Jiaxin Zhao
- Department of Ultrasound, Guigang City People’s Hospital, Guigang, Guangxi, 537100, People’s Republic of China
| | - Xuejuan Lin
- Department of Surgery, Guigang Maternal and Child Health Care Hospital, Guigang, Guangxi, 537100, People’s Republic of China
| |
Collapse
|
13
|
Liao W, Ang Y, Kee ACL, Lim V, Lim AYH, Chai CLL, Wong WSF. Calcaratarin D, A Labdane Diterpenoid, Attenuates Bleomycin-Induced Pulmonary Fibrosis by Blocking Wnt/β-Catenin Signaling Pathway. Pharmacol Res 2025; 216:107756. [PMID: 40311955 DOI: 10.1016/j.phrs.2025.107756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is one of the most common interstitial lung diseases with a high mortality rate. Calcaratarin D (CalD), a labdane diterpenoid, has been shown to possess anti-inflammatory properties. The present study evaluated the therapeutic potential of CalD in pulmonary fibrosis. A single dose of bleomycin (BLM, 2.5mg/kg) was instilled intratracheally in mice for up to 21 days to develop lung fibrosis. Oral CalD (50mg/kg) reduced BLM-induced inflammatory cell infiltration, especially pro-fibrotic Arg1-expressing interstitial macrophages in the bronchoalveolar lavage fluid. During the late fibrotic phase, CalD decreased BLM-induced mortality and body weight loss. In addition, CalD ameliorated lung histopathology, reduced collagen deposition and mucus hypersecretion, and improved lung functions in BLM-exposed mice. Furthermore, CalD modulated the levels of pro-inflammatory cytokines, chemokines, and growth factors in BAL fluid and lung tissues. In mouse lungs, BLM selectively upregulated Wnt10A level and promoted β-catenin nuclear translocation. CalD not only blocked Wnt10A/β-catenin signaling pathway but also reduced pro-fibrotic markers such as collagens, α-SMA and FHL2. In normal human lung fibroblasts, CalD inhibited TGF-β1-stimulated pro-fibrotic markers and Wnt/β-catenin signaling pathway by reducing Wnt10A production, upregulating endogenous Wnt antagonist DKK1 level, dephosphorylating Wnt ligand co-receptor LRP6, and preventing β-catenin and YAP/TAZ nuclear translocation. The antifibrotic action of CalD was shown to be dependent on its α,β-unsaturated γ-butyrolactone structure that is essential for CalD to form covalent interaction with cellular protein targets. Our results imply that CalD could be a novel antifibrotic agent for IPF, acting through blockade of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore
| | - Yuet Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore
| | - Adrian C L Kee
- Division of Respiratory and Critical Care Medicine, Department of Medicine, National University Health System, Singapore
| | - Valencia Lim
- Division of Respiratory and Critical Care Medicine, Department of Medicine, National University Health System, Singapore
| | - Albert Y H Lim
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Christina L L Chai
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore; Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore; Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, Singapore.
| |
Collapse
|
14
|
Qian R, Xu Y, Zhang L, Wang L, Chen X, Wang M, Bao Q, Yao Y, Xie L. Haizao Yuhu decoction ameliorates silica-induced lung injury by inhibiting transforming growth factor-beta1/Smad pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119669. [PMID: 40122314 DOI: 10.1016/j.jep.2025.119669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Haizao Yuhu Decoction (HYD) is a traditional Chinese herbal formula known for regulating Qi and dispersing stasis. AIM OF THE STUDY This study investigates the effects of HYD on silica-induced lung injury and the underlying mechanisms. MATERIALS AND METHODS The main constituents of HYD were identified using ultra-performance liquid chromatography Q-Exactive mass spectrometry (UPLC-QE-MS). Network pharmacology was employed to predict the targets and pathways through which HYD ameliorates silicosis, which were validated in a silica-induced lung injury mouse model and a TGF-β1-induced alveolar epithelial cell model. Pathological evaluation was conducted using hematoxylin-eosin (H&E) and Masson staining, while inflammatory cytokines and fibrosis were assessed via enzyme-linked immunosorbent assay (ELISA) and hydroxyproline quantification. Western blotting (WB) was performed to analyse protein expression levels of targeted markers. Proliferation and migration capabilities of MLE12 cells treated with HYD and its bioactive constituents (glycitein, diosmetin, and limonin) were assessed using cell counting kit-8 (CCK-8) and wound healing assays. RESULTS HYD significantly alleviated silica-induced lung injury, reducing inflammatory response and collagen deposition. A total of 176 constituents were identified in HYD, with 111 being pharmacologically active and linked to 1397 potential therapeutic targets, 107 associated with silicosis. Enrichment analyses highlighted the TGF-β1/Smad pathway and epithelial-mesenchymal transition (EMT) in HYD's anti-silicosis effects, which was validated by the restoration of TGF-β1, p-Smad2/Smad2, p-Smad3/Smad3, E-cadherin, and Vimentin following HYD treatment. Additionally, glycitein, diosmetin, and limonin inhibited the proliferation and migration of TGF-β1-induced MLE12 cells and suppressed the activation of TGF-β1/Smad pathway and EMT. CONCLUSIONS HYD effectively alleviates silica-induced lung injury by specifically inhibiting the TGF-β1/Smad pathway and EMT process.
Collapse
Affiliation(s)
- Rui Qian
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Yunyi Xu
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Luoning Zhang
- Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Liqun Wang
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Xuxi Chen
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Mengzhu Wang
- Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Qixue Bao
- Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Yuqin Yao
- State Key Laboratory of Biotherapy, West China School of Clinical Medicine (West China Hospital) Sichuan University, Chengdu, China; Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Linshen Xie
- Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
15
|
Meshkovska Y, Dzhuraeva B, Godugu C, Pooladanda V, Thatikonda S. Deciphering the interplay: circulating cell-free DNA, signaling pathways, and disease progression in idiopathic pulmonary fibrosis. 3 Biotech 2025; 15:102. [PMID: 40165930 PMCID: PMC11954786 DOI: 10.1007/s13205-025-04272-y] [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: 06/12/2024] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lung disease with an unknown etiology and a short survival rate. There is no accurate method of early diagnosis, and it involves computed tomography (CT) or lung biopsy. Since diagnostic methods are not accurate due to their similarity to other lung pathologies, discovering new biomarkers is a key issue for diagnosticians. Currently, the use of ccf-DNA (circulating cell-free deoxyribonucleic acid) is an important focus due to its association with IPF-induced alterations in metabolic pathways, such as amino acid metabolism, energy metabolism, and lipid metabolism pathways. Other biomarkers associated with metabolic changes have been found, and they are related to changes in type II/type I alveolar epithelial cells (AECs I/II), changes in extracellular matrix (ECM), and inflammatory processes. Currently, IPF pathogenetic treatment remains unknown, and the mortality rates are increasing, and the patients are diagnosed at a late stage. Signaling pathways and metabolic dysfunction have a significant role in the disease occurrence, particularly the transforming growth factor-β (TGF-β) signaling pathway, which plays an essential role. TGF-β, Wnt, Hedgehog (Hh), and integrin signaling are the main drivers of fibrosis. These pathways activate the transformation of fibroblasts into myofibroblasts, extracellular matrix (ECM) deposition, and tissue remodeling fibrosis. Therapy targeting diverse signaling pathways to slow disease progression is crucial in the treatment of IPF. Two antifibrotic medications, including pirfenidone and nintedanib, are Food and Drug Administration (FDA)-approved for treatment. ccf-DNA could become a new biomarker for IPF diagnosis to detect the disease at the early stage, while FDA-approved therapies could help to prevent late conditions from forming and decrease mortality rates.
Collapse
Affiliation(s)
- Yeva Meshkovska
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL 33612 USA
| | - Barchinai Dzhuraeva
- Department of Hospital Pediatrics, Moffitt Cancer Center, Tampa, FL 33612 USA
- Department of Hospital Pediatrics with a Course of Neonatology, National Center of Maternal and Child Health, Bishkek, 720017 Kyrgyzstan
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037 India
| | - Venkatesh Pooladanda
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, 60 Blossom Street, Thier 9, Boston, MA 02114 USA
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA 02115 USA
| | - Sowjanya Thatikonda
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL 33612 USA
| |
Collapse
|
16
|
Farouk H, Moustafa PE, Khattab MS, El-Marasy SA. Diacerein ameliorates amiodarone-induced pulmonary fibrosis via targeting the TGFβ1/α-SMA/Smad3 pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4111-4122. [PMID: 39417843 PMCID: PMC11978552 DOI: 10.1007/s00210-024-03450-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024]
Abstract
This study is aimed at investigating the possible protective effect of diacerein (DIA) against AMD-induced pulmonary fibrosis in rats. Rats were classified into 4 groups: a normal group that received distilled water, control group that received AMD (100 mg/kg, p.o.) for 21 days to induce pulmonary fibrosis, and 2 treatment groups that received diacerein, in 2 dose levels (50 and 100 mg/kg, p.o., respectively) in addition to AMD (100 mg/kg, p.o.), for 21 days. Lung function test was assessed using a spirometer; serum and tissue were collected. Biochemical, real-time PCR, histopathological, and immunohistopathological analyses were carried out. AMD reduced tidal volume (TV), peripheral expiratory rate (PER), forced vital capacity (FVC), serum reduced glutathione (GSH) levels, Beclin, and LCII, while it elevated transform growth factor (TGF-β1) gene expression, serum malondialdehyde (MDA) level, alpha-smooth muscle actin (α-SMA), Smad3, phosphorylated signal transducer and activator of transcription (p-STAT3), and p62 lung content. Also, AMD elevated tumor necrosis factor-alpha (TNF-α) and caspase-3 protein expression. DIA elevated TV, PER, FVC, serum GSH level, Beclin, and LCII, while it reduced TGF-β1 gene expression, serum MDA level, α-SMA, Smad3, p-STAT-3, and p62 lung content. Moreover, DIA reduced TNF-α and caspase-3 protein expression. DIA attenuated AMD-induced pulmonary fibrosis via alleviating the TGF1/α-SMA/Smad3 pathway, reducing STAT-3 activation, and combating oxidative stress and inflammation in addition to promoting autophagy and abrogating apoptosis.
Collapse
Affiliation(s)
- Hadir Farouk
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Passant E Moustafa
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Marwa S Khattab
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Salma A El-Marasy
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt.
| |
Collapse
|
17
|
St Pierre L, Berhan A, Sung EK, Alvarez JR, Wang H, Ji Y, Liu Y, Yu H, Meier A, Afshar K, Golts EM, Lin GY, Castaldi A, Calvert BA, Ryan A, Zhou B, Offringa IA, Marconett CN, Borok Z. Integrated multiomic analysis identifies TRIP13 as a mediator of alveolar epithelial type II cell dysfunction in idiopathic pulmonary fibrosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167572. [PMID: 39547519 PMCID: PMC11951472 DOI: 10.1016/j.bbadis.2024.167572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 10/14/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal progressive lung disease urgently needing new therapies. Current treatments only delay disease progression, leaving lung transplant as the sole remaining option. Recent studies support a model whereby IPF arises because alveolar epithelial type II (AT2) cells, which normally mediate distal lung regeneration, acquire airway and/or mesenchymal characteristics, preventing proper repair. Mechanisms driving this abnormal differentiation remain unclear. We performed integrated transcriptomic and epigenomic analysis of purified AT2 cells which revealed genome-wide alterations in IPF lungs. The most prominent epigenetic alteration was activation of an enhancer in thyroid receptor interactor 13 (TRIP13), although TRIP13 was not the most significantly transcriptionally upregulated gene. TRIP13 is broadly implicated in epithelial-mesenchymal plasticity. In cultured human AT2 cells and lung slices, small molecule TRIP13 inhibitor DCZ0415 prevented acquisition of the mesenchymal gene signature characteristic of IPF, suggesting TRIP13 inhibition as a potential therapeutic approach to fibrotic disease.
Collapse
Affiliation(s)
- Laurence St Pierre
- Department of Surgery, University of Southern California, Los Angeles, CA 90089, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Asres Berhan
- Department of Medicine, University of California San Diego, CA 92037, USA
| | - Eun K Sung
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Department of Integrative Translational Sciences, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Juan R Alvarez
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Hongjun Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Yanbin Ji
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Yixin Liu
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Haoze Yu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Angela Meier
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA 92037, USA
| | - Kamyar Afshar
- Department of Medicine, University of California San Diego, CA 92037, USA
| | - Eugene M Golts
- Department of Surgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - Grace Y Lin
- Department of Pathology, University of California, San Diego, La Jolla, CA 92037, USA
| | | | - Ben A Calvert
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Amy Ryan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Beiyun Zhou
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Ite A Offringa
- Department of Surgery, University of Southern California, Los Angeles, CA 90089, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| | - Crystal N Marconett
- Department of Surgery, University of Southern California, Los Angeles, CA 90089, USA; Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Department of Integrative Translational Sciences, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA.
| | - Zea Borok
- Department of Medicine, University of California San Diego, CA 92037, USA.
| |
Collapse
|
18
|
Ratnasingham M, Bradding P, Roach KM. The role of TRP channels in lung fibrosis: Mechanisms and therapeutic potential. Int J Biochem Cell Biol 2025; 180:106728. [PMID: 39672503 DOI: 10.1016/j.biocel.2024.106728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a severe lung disease affecting around 5 million people globally, with a median survival of 3-4 years. Characterized by excessive scarring of lung tissue, IPF results from the accumulation of myofibroblasts that deposit extracellular matrix (ECM), causing fibrosis. Current treatments, pirfenidone and nintedanib, slow the disease but do not stop its progression. IPF pathogenesis involves repeated alveolar injury, leading to pro-fibrotic mediators like TGFβ1, which trigger fibroblast-to-myofibroblast transitions and ECM deposition. Recent research suggests that transient receptor potential (TRP) channels, such as TRPV4, TRPC6, and TRPA1, play a key role in regulating calcium signalling and mechanical stress, crucial in myofibroblast activation. Targeting TRP channels may disrupt fibrosis and offer new therapeutic strategies. Preclinical studies indicate that inhibiting TRP channels could reduce fibrosis, warranting further trials to explore their efficacy and safety in treating IPF and related fibrotic conditions.
Collapse
Affiliation(s)
- M Ratnasingham
- NIHR Respiratory BRC, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - P Bradding
- NIHR Respiratory BRC, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - K M Roach
- NIHR Respiratory BRC, Department of Respiratory Sciences, University of Leicester, Leicester, UK.
| |
Collapse
|
19
|
Xu W, Jieda X, Wu Y, Du F, Ma L, Luo L, Liu D, Guo L, Liu J, Dong W. Safety, Efficacy and Bio-Distribution Analysis of Exosomes Derived From Human Umbilical Cord Mesenchymal Stem Cells for Effective Treatment of Bronchopulmonary Dysplasia by Intranasal Administration in Mice Model. Int J Nanomedicine 2025; 20:2521-2553. [PMID: 40034220 PMCID: PMC11874997 DOI: 10.2147/ijn.s501843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
Purpose Exosomes (Exos) derived from human umbilical cord mesenchymal stem cells (hUC-MSCs) hold great potential for treating bronchopulmonary dysplasia (BPD); however, safety concerns and effects of intranasal administration remain unexplored. This study aimed to explore the safety of hUC-MSCs and Exos and to investigate the efficacy and bio-distribution of repeated intranasal Exos administration in neonatal BPD models. Methods Characteristics of hUC-MSCs and Exos were analyzed. A subcutaneous tumor formation assay using a single dose of hUC-MSCs or Exos was conducted in Crl:NU-Foxn1nu mice. Vital signs, biochemical indices, pathological alterations, and 18F-FDG microPET/CT analysis were examined. Pulmonary pathology, three-dimensional reconstructions, ultrastructural structures, in vivo and ex vivo bio-distribution imaging analyses, enzyme-linked immunoassay assays, and reverse transcription-quantitative polymerase chain reaction analyses of lung tissues were all documented following intranasal Exos administration. Results Characteristics of hUC-MSCs and Exos satisfied specifications. Crl:NU-Foxn1nu mice did not exhibit overt toxicity or carcinogenicity following a single dose of hUC-MSCs or Exos after 60 days of observation. Repeated intranasal Exos administration effectively alleviated pathological injuries, restored pulmonary ventilation in three-dimensional reconstruction, and recovered endothelial cell layer integrity in ultrastructural analysis. Exos steadily accumulated in lung tissues from postnatal day 1 to 14. Exos also interrupted the epithelial-mesenchymal transition and inflammation reactions in BPD models. Conclusion As a nanoscale, non-cellular therapy, intranasal administration of Exos was an effective, noninvasive treatment for BPD. This approach was free from toxic, tumorigenic risks and repaired alveolar damage while interrupting epithelial-mesenchymal transition and inflammation in neonatal mice with BPD.
Collapse
Affiliation(s)
- Wanting Xu
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Xiaolin Jieda
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Yue Wu
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Fengling Du
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Lu Ma
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Lijuan Luo
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Dong Liu
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Ling Guo
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Jing Liu
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Wenbin Dong
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| |
Collapse
|
20
|
Tan C, Wang J, Ye X, Kasimu K, Li Y, Luo F, Yi H, Luo Y. Genome-wide CRISPR/Cas9 screening identifies key profibrotic regulators of TGF-β1-induced epithelial-mesenchymal transformation and pulmonary fibrosis. Front Mol Biosci 2025; 12:1507163. [PMID: 40034336 PMCID: PMC11872725 DOI: 10.3389/fmolb.2025.1507163] [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: 10/07/2024] [Accepted: 01/09/2025] [Indexed: 03/05/2025] Open
Abstract
Background The idiopathic pulmonary fibrosis (IPF) is a progressive and lethal interstitial lung disease with high morbidity and mortality. IPF is characterized by excessive extracellular matrix accumulation (ECM) and epithelial-mesenchymal transformation (EMT). To date, few anti-fibrotic therapeutics are available to reverse the progression of pulmonary fibrosis, and it is important to explore new profibrotic molecular regulators mediating EMT and pulmonary fibrosis. Methods Based on our model of TGF-β1-induced EMT in BEAS-2B cells, we performed the genome-wide CRISPR/Cas9 knockout (GeCKO) screening technique, pathway and functional enrichment analysis, loss-of-function experiment, as well as other experimental techniques to comprehensively investigate profibrotic regulators contributing to EMT and the pathogenesis of pulmonary fibrosis. Results Utilizing the GeCKO library screening, we identified 76 top molecular regulators. Ten candidate genes were subsequently confirmed by integrating the high-throughput data with findings from pathway and functional enrichment analysis. Among the candidate genes, knockout of COL20A1 and COL27A1 led to decreased mRNA expression of ECM components (Fibronectin and Collagen-I), as well as an increased rate of cell apoptosis. The mRNA expression of Collagen-I, together with the cell viability and migration, were inhibited when knocking out the WNT11. In addition, a decrease in the protein deposition of ECM components was observed by suppressing the expression of COL20A1, COL27A1, and WNT11. Conclusion Our study demonstrates that the COL20A1, COL27A1, and WNT11 serve as key profibrotic regulators of EMT. Gaining understanding and insights into these key profibrotic regulators of EMT paves the way for the discovery of new therapeutic targets against the onset and progression of IPF.
Collapse
Affiliation(s)
- Chunjiang Tan
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Respiratory Diseases, Sun Yat-sen University, Guangzhou, China
| | - Juan Wang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Respiratory Diseases, Sun Yat-sen University, Guangzhou, China
| | - Xiangrong Ye
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Respiratory Diseases, Sun Yat-sen University, Guangzhou, China
| | - Kaidirina Kasimu
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Respiratory Diseases, Sun Yat-sen University, Guangzhou, China
| | - Ye Li
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Respiratory Diseases, Sun Yat-sen University, Guangzhou, China
| | - Feng Luo
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Respiratory Diseases, Sun Yat-sen University, Guangzhou, China
| | - Hui Yi
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Respiratory Diseases, Sun Yat-sen University, Guangzhou, China
| | - Yifeng Luo
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Respiratory Diseases, Sun Yat-sen University, Guangzhou, China
- Department of Emergency Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
21
|
Wang Y, Ma Z, Peng W, Yu Q, Liang W, Cao L, Wang Z. 3,5,6,7,8,3',4'- Heptamethoxyflavonoid inhibits TGF-β1-induced epithelial-mesenchymal transition by regulating oxidative stress and autophagy through MEK/ERK/PI3K/AKT/mTOR signaling pathway. Sci Rep 2025; 15:4567. [PMID: 39915543 PMCID: PMC11802913 DOI: 10.1038/s41598-025-88869-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 01/31/2025] [Indexed: 02/09/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a crucial pathological process in the pathogenesis of fibrosis. 3,5,6,7,8,3',4'-hepmethoxyflavone (HMF), the main active ingredient extracted from the Chinese herb Breynia fruticosa (L.) Hook. f., has been shown to have beneficial effects on regulating apoptosis and inhibiting collagen deposition. However, it remains unclear whether and how HMF alleviates transforming growth factor-β1 (TGF-β1)-induced EMT. The objective of this study was to investigate the impact of HMF on TGF-β1-induced EMT in human alveolar Type II epithelial cells (A549) and its underlying mechanism. In vitro culture of TGF-β1-induced EMT in A549 cells revealed that HMF reduced cell viability and migration, inhibited collagen deposition, decreased expression levels of mesenchymal cell markers and fibrosis markers α-SMA, MMP2, TIMP1, β-catenin, and Snail. Meanwhile, the expression level of E-cadherin increased as an epithelial cell marker. Additionally, we discussed the effects of HMF on oxidative stress and autophagy. Various experiments confirmed that HMF regulated the expression levels of Nrf2, keap-1, HO-1, ROS, MDA, SOD, GSH, and played a role in reducing oxidative stress. At the same time, HMF significantly activated autophagy by increasing expressions of Beclin-1 and LC3B as well as enhancing autophagosome content. The addition 3-MA, an autophagy inhibitor attenuated these beneficial effects. Furthermore, HMF significantly inhibited phosphorylation levels of MEK, ERK, PI3K, AKT, and mTOR through various pathways. In conclusion, HMF effectively inhibits TGF-β1-induced EMT in A549 cells by targeting the MEK/ERK/PI3K/AKT/mTOR signaling pathway. Moreover, it exhibits a close correlation with the suppression of oxidative stress and induction of autophagy.
Collapse
Affiliation(s)
- Yiting Wang
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Zhiheng Ma
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Weiwen Peng
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Qinglian Yu
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Wenjie Liang
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Liu Cao
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China
| | - Zhuqiang Wang
- Department of Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, Guangdong, China.
- National Engineering Research Center for Modernization of Traditional Chinese Medicine, Hospital Preparation Transformation Branch, Zhongshan, China.
| |
Collapse
|
22
|
Korytina GF, Markelov VA, Gibadullin IA, Zulkarneev SR, Nasibullin TR, Zulkarneev RH, Avzaletdinov AM, Avdeev SN, Zagidullin NS. The Relationship Between Differential Expression of Non-coding RNAs (TP53TG1, LINC00342, MALAT1, DNM3OS, miR-126-3p, miR-200a-3p, miR-18a-5p) and Protein-Coding Genes (PTEN, FOXO3) and Risk of Idiopathic Pulmonary Fibrosis. Biochem Genet 2025:10.1007/s10528-024-11012-z. [PMID: 39881079 DOI: 10.1007/s10528-024-11012-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a rapidly progressive interstitial lung disease of unknown pathogenesis with no effective treatment currently available. Given the regulatory roles of lncRNAs (TP53TG1, LINC00342, H19, MALAT1, DNM3OS, MEG3), miRNAs (miR-218-5p, miR-126-3p, miR-200a-3p, miR-18a-5p, miR-29a-3p), and their target protein-coding genes (PTEN, TGFB2, FOXO3, KEAP1) in the TGF-β/SMAD3, Wnt/β-catenin, focal adhesion, and PI3K/AKT signaling pathways, we investigated the expression levels of selected genes in peripheral blood mononuclear cells (PBMCs) and lung tissue from patients with IPF. Lung tissue and blood samples were collected from 33 newly diagnosed, treatment-naive patients and 70 healthy controls. Gene expression levels were analyzed by RT-qPCR. TaqMan assays and TaqMan MicroRNA assay were employed to quantify the expression of target lncRNAs, mRNAs, and miRNAs. Our study identified significant differential expression in PBMCs from IPF patients compared to healthy controls, including lncRNAs MALAT1 (Fold Change = 3.809, P = 0.0001), TP53TG1 (Fold Change = 0.4261, P = 0.0021), and LINC00342 (Fold Change = 1.837, P = 0.0448); miRNAs miR-126-3p (Fold Change = 0.102, P = 0.0028), miR-200a-3p (Fold Change = 0.442, P = 0.0055), and miR-18a-5p (Fold Change = 0.154, P = 0.0034); and mRNAs FOXO3 (Fold Change = 4.604, P = 0.0032) and PTEN (Fold Change = 2.22, P = 0.0011). In lung tissue from IPF patients, significant expression changes were observed in TP53TG1 (Fold Change = 0.2091, P = 0.0305) and DNM3OS (Fold Change = 4.759, P = 0.05). Combined analysis of PBMCs expression levels for TP53TG1, MALAT1, miRNA miR-126-3p, and PTEN distinguished IPF patients from healthy controls with an AUC = 0.971, sensitivity = 0.80, and specificity = 0.955 (P = 6 × 10-8). These findings suggest a potential involvement of the identified ncRNAs and mRNAs in IPF pathogenesis. However, additional functional validation studies are needed to elucidate the precise molecular mechanisms by which these lncRNAs, miRNAs, and their targets contribute to PF.
Collapse
Affiliation(s)
- Gulnaz F Korytina
- Institute of Biochemistry and Genetics-Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences (IBG UFRC RAS), Pr. Oktyabrya, 71, Ufa, 450054, Russian Federation.
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation.
| | - Vitaly A Markelov
- Institute of Biochemistry and Genetics-Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences (IBG UFRC RAS), Pr. Oktyabrya, 71, Ufa, 450054, Russian Federation
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| | - Irshat A Gibadullin
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| | - Shamil R Zulkarneev
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| | - Timur R Nasibullin
- Institute of Biochemistry and Genetics-Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences (IBG UFRC RAS), Pr. Oktyabrya, 71, Ufa, 450054, Russian Federation
| | - Rustem H Zulkarneev
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| | | | - Sergey N Avdeev
- Sechenov First Moscow State Medical University (Sechenov University), 8-2, Trubetskaya Str., Moscow, 119991, Russian Federation
| | - Naufal Sh Zagidullin
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| |
Collapse
|
23
|
Wang Y, Liu C, Wang N, Weng D, Zhao Y, Yang H, Wang H, Xu S, Gao J, Lang C, Fan Z, Yu L, He Z. hAMSCs regulate EMT in the progression of experimental pulmonary fibrosis through delivering miR-181a-5p targeting TGFBR1. Stem Cell Res Ther 2025; 16:2. [PMID: 39757225 DOI: 10.1186/s13287-024-04095-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a common and multidimensional devastating interstitial lung disease. The development of novel and more effective interventions for PF is an urgent clinical need. A previous study has found that miR-181a-5p plays an important role in the development of PF, and human amniotic mesenchymal stem cells (hAMSCs) exert potent therapeutic potential on PF. However, whether hAMSCs act on PF by delivering miR-181a-5p and its detailed mechanism still remain unknown. Thus, this study was designed to investigate the underlying possible mechanism of hAMSCs on PF in bleomycin (BLM)-induced mouse PF model, and a co-culture system of hAMSCs and A549 cells epithelial mesenchymal transition (EMT) model, focusing on its effects on collagen deposition, EMT, and epithelial cell cycle regulation. METHODS hAMSCs with different miR-181a-5p expression levels were constructed. BLM (4 mg/kg) was used to create a PF model, while TGF-β1 was used to induce A549 cells to construct an EMT model. Furthermore, the effects of different miR-181a-5p expression in hAMSCs on collagen deposition and EMT during lung fibrosis were assessed in vivo and in vitro. RESULTS We found that hAMSCs exerted anti-fibrotic effect in BLM-induced mouse PF model. Moreover, hAMSCs also exerted protective effect on TGFβ1-induced A549 cell EMT model. Furthermore, hAMSCs ameliorated PF by promoting epithelial cell proliferation, reducing epithelial cell apoptosis, and attenuating EMT of epithelial cells through paracrine effects. hAMSCs regulated EMT in PF through delivering miR-181a-5p targeting TGFBR1. CONCLUSIONS Our findings reveal for the first time that hAMSCs inhibit PF by promoting epithelial cell proliferation, reducing epithelial cell apoptosis, and attenuating EMT. Mechanistically, the therapeutic effect of hMASCs on PF is achieved through delivering miR-181a-5p targeting TGFBR1.
Collapse
Affiliation(s)
- Yanyang Wang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Chan Liu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Nuoxin Wang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Dong Weng
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Yan Zhao
- Department of Prevention Healthcare, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, 400038, China
| | - Hongyu Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Haoyuan Wang
- Department of Cardiothoracic Surgery, Liuzhou People's Hospital, Liuzhou, 545001, Guangxi, China
| | - Shangfu Xu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Jianmei Gao
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi, 563000, Guizhou, China
| | - Changhui Lang
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Zhenhai Fan
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Limei Yu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Zhixu He
- Center of Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, 550025, Guizhou, China.
- Department of Pediatric Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
24
|
Sumanaweera D, Suo C, Cujba AM, Muraro D, Dann E, Polanski K, Steemers AS, Lee W, Oliver AJ, Park JE, Meyer KB, Dumitrascu B, Teichmann SA. Gene-level alignment of single-cell trajectories. Nat Methods 2025; 22:68-81. [PMID: 39300283 PMCID: PMC11725504 DOI: 10.1038/s41592-024-02378-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 07/12/2024] [Indexed: 09/22/2024]
Abstract
Single-cell data analysis can infer dynamic changes in cell populations, for example across time, space or in response to perturbation, thus deriving pseudotime trajectories. Current approaches comparing trajectories often use dynamic programming but are limited by assumptions such as the existence of a definitive match. Here we describe Genes2Genes, a Bayesian information-theoretic dynamic programming framework for aligning single-cell trajectories. It is able to capture sequential matches and mismatches of individual genes between a reference and query trajectory, highlighting distinct clusters of alignment patterns. Across both real world and simulated datasets, it accurately inferred alignments and demonstrated its utility in disease cell-state trajectory analysis. In a proof-of-concept application, Genes2Genes revealed that T cells differentiated in vitro match an immature in vivo state while lacking expression of genes associated with TNF signaling. This demonstrates that precise trajectory alignment can pinpoint divergence from the in vivo system, thus guiding the optimization of in vitro culture conditions.
Collapse
Affiliation(s)
- Dinithi Sumanaweera
- Wellcome Sanger Institute; Wellcome Genome Campus, Hinxton, Cambridge, UK
- Theory of Condensed Matter, Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Chenqu Suo
- Wellcome Sanger Institute; Wellcome Genome Campus, Hinxton, Cambridge, UK
- Department of Paediatrics, Cambridge University Hospitals; Hills Road, Cambridge, UK
| | - Ana-Maria Cujba
- Wellcome Sanger Institute; Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Daniele Muraro
- Wellcome Sanger Institute; Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Emma Dann
- Wellcome Sanger Institute; Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Krzysztof Polanski
- Wellcome Sanger Institute; Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Alexander S Steemers
- Wellcome Sanger Institute; Wellcome Genome Campus, Hinxton, Cambridge, UK
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Woochan Lee
- Wellcome Sanger Institute; Wellcome Genome Campus, Hinxton, Cambridge, UK
- Department of Biomedical Sciences, Seoul National University, Seoul, Korea
| | - Amanda J Oliver
- Wellcome Sanger Institute; Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Jong-Eun Park
- Wellcome Sanger Institute; Wellcome Genome Campus, Hinxton, Cambridge, UK
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Kerstin B Meyer
- Wellcome Sanger Institute; Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Bianca Dumitrascu
- Department of Statistics, Columbia University, New York, NY, USA
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
| | - Sarah A Teichmann
- Wellcome Sanger Institute; Wellcome Genome Campus, Hinxton, Cambridge, UK.
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Co-director of CIFAR Macmillan Research Program, Toronto, Ontario, Canada.
| |
Collapse
|
25
|
Du C, Ma C, Geng R, Wang X, Wang X, Yang J, Hu J. Bruceine A inhibits TGF-β1/Smad pathway in pulmonary fibrosis by blocking gal3/TGF-β1 interaction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156267. [PMID: 39615217 DOI: 10.1016/j.phymed.2024.156267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 10/13/2024] [Accepted: 11/14/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Bruceine A(BA) has many pharmacological activities and significantly inhibits fibrosis in keloid fibroblasts. However, the underlying mechanisms have not yet been fully elucidated. OBJECTIVE This study aimed to investigate the effects of BA on pulmonary fibrosis(PF) and explore its underlying mechanisms. METHODS PF models were constructed by BLM-induced C57BL/6 J mice, TGF-β1- induced MRC-5 and HFL-1 cells. Cell proliferation, MMP, apoptosis, and ROS levels were analyzed in vitro. In vivo, experiments were performed to evaluate the therapeutic effect of BA on PF by detecting respiratory function, histopathology, and collagen level. Fibro-associated, ECM, and EMT key proteins were used to assess the degree of PF. To predict the target of BA by molecular docking technology, and verified by DARTS, CETSA, MST,and SPR. Then overexpression gal3-lentivirus, GB1107 gal3 inhibitor, and BA addition were used to verify the TGF-β1/Smad pathway key protein by western blot. RESULTS We found that BA inhibited PF both in vitro and in vivo. The predicted and validated results showed that gal3 was the target of BA, and the binding site was Arg144, His158, and Trp181. Mechanistically, BA disrupts the interaction between gal3 and TGF-β1. BA reduced Smad2/3 and p-Smad2/3 protein content and inhibited TGF-β1/Smad pathway in the overexpressing gal3 HFL-1 cells. After adding GB1107, the inhibitory effect of BA on TGF-β1/Smad pathway disappeared. CONCLUSION This study is the first to demonstrate that BA can target gal3, interfere with the interaction between gal3 and TGF-β1 protein, inhibit the downstream TGF-β1/Smad pathway, and act as a "brake" to reverse the PF process. These findings provide a solid scientific basis for the clinical application of BA in the prevention and treatment of PF.
Collapse
Affiliation(s)
- Chao Du
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, China.
| | - Chong Ma
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, China.
| | - Ruoyu Geng
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, China.
| | - Xiaomei Wang
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China; Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, China; Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, China.
| | - Xinling Wang
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China; Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, China; Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, China.
| | - Jianhua Yang
- Department of Pharmacy, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China; Xinjiang Key Laboratory of Clinical Drug Research, China.
| | - Junping Hu
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China; Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, China; Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, China.
| |
Collapse
|
26
|
Zhao T, Zhou ZR, Wan HQ, Feng T, Hu XH, Li XQ, Zhao SM, Li HL, Hou JW, Li W, Lu DY, Qian MY, Shen X. Otilonium bromide ameliorates pulmonary fibrosis in mice through activating phosphatase PPM1A. Acta Pharmacol Sin 2025; 46:107-121. [PMID: 39160244 PMCID: PMC11695943 DOI: 10.1038/s41401-024-01368-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/24/2024] [Indexed: 08/21/2024]
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive and irreversible interstitial lung disease characterized by unremitting pulmonary myofibroblasts activation, extracellular matrix (ECM) deposition and inflammatory recruitment. PF has no curable medication yet. In this study we investigated the molecular pathogenesis and potential therapeutic targets of PF and discovered drug lead compounds for PF therapy. A murine PF model was established in mice by intratracheal instillation of bleomycin (BLM, 5 mg/kg). We showed that the protein level of pulmonary protein phosphatase magnesium-dependent 1A (PPM1A, also known as PP2Cα) was significantly downregulated in PF patients and BLM-induced PF mice. We demonstrated that TRIM47 promoted ubiquitination and decreased PPM1A protein in PF progression. By screening the lab in-house compound library, we discovered otilonium bromide (OB, clinically used for treating irritable bowel syndrome) as a PPM1A enzymatic activator with an EC50 value of 4.23 μM. Treatment with OB (2.5, 5 mg·kg-1·d-1, i.p., for 20 days) significantly ameliorated PF-like pathology in mice. We constructed PF mice with PPM1A-specific knockdown in the lung tissues, and determined that by targeting PPM1A, OB treatment suppressed ECM deposition through TGF-β/SMAD3 pathway in fibroblasts, repressed inflammatory responses through NF-κB/NLRP3 pathway in alveolar epithelial cells, and blunted the crosstalk between inflammation in alveolar epithelial cells and ECM deposition in fibroblasts. Together, our results demonstrate that pulmonary PPM1A activation is a promising therapeutic strategy for PF and highlighted the potential of OB in the treatment of the disease.
Collapse
Affiliation(s)
- Tong Zhao
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhi-Ruo Zhou
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Hui-Qi Wan
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tian Feng
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xu-Hui Hu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiao-Qian Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shi-Mei Zhao
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hong-Lin Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ji-Wei Hou
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, China
| | - Wei Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Da-Yun Lu
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Min-Yi Qian
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xu Shen
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
27
|
Colin Waldo MD, Quintero-Millán X, Negrete-García MC, Ruiz V, Sommer B, Romero-Rodríguez DP, Montes-Martínez E. Circulating MicroRNAs in Idiopathic Pulmonary Fibrosis: A Narrative Review. Curr Issues Mol Biol 2024; 46:13746-13766. [PMID: 39727949 DOI: 10.3390/cimb46120821] [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: 10/14/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, deathly disease with no recognized effective cure as yet. Furthermore, its diagnosis and differentiation from other diffuse interstitial diseases remain a challenge. Circulating miRNAs have been measured in IPF and have proven to be an adequate option as biomarkers for this disease. These miRNAs, released into the circulation outside the cell through exosomes and proteins, play a crucial role in the pathogenic pathways and mechanisms involved in IPF development. This review focuses on the serum/plasma miRNAs reported in IPF that have been validated by real-time PCR and the published evidence regarding the fibrotic process. First, we describe the mechanisms by which miRNAs travel through the circulation (contained in exosomes and bound to proteins), as well as the mechanism by which miRNAs perform their function within the cell. Subsequently, we summarize the evidence concerning miRNAs reported in serum/plasma, where we find contradictory functions in some miRNAs (dual functions in IPF) when comparing the findings in vitro vs. in vivo. The most relevant finding, for instance, the levels of miRNAs let-7d and miR-21 reported in the serum/plasma in IPF, correspond to those found in studies in lung fibroblasts and the murine bleomycin model, reinforcing the usefulness of these miRNAs as future biomarkers in IPF.
Collapse
Affiliation(s)
- Marisa Denisse Colin Waldo
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Xochipilzihuitl Quintero-Millán
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Maria Cristina Negrete-García
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Víctor Ruiz
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Bettina Sommer
- Bronchial Hyperreactivity Research Department, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Dámaris P Romero-Rodríguez
- Conahcyt National Laboratory for Research and Diagnosis by Immunocytofluorometry (LANCIDI), National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Eduardo Montes-Martínez
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| |
Collapse
|
28
|
Vissers G, Giacomozzi M, Verdurmen W, Peek R, Nap A. The role of fibrosis in endometriosis: a systematic review. Hum Reprod Update 2024; 30:706-750. [PMID: 39067455 PMCID: PMC11532625 DOI: 10.1093/humupd/dmae023] [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: 03/15/2024] [Revised: 06/04/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Fibrosis is an important pathological feature of endometriotic lesions of all subtypes. Fibrosis is present in and around endometriotic lesions, and a central role in its development is played by myofibroblasts, which are cells derived mainly after epithelial-to-mesenchymal transition (EMT) and fibroblast-to-myofibroblast transdifferentiation (FMT). Transforming growth factor-β (TGF-β) has a key role in this myofibroblastic differentiation. Myofibroblasts deposit extracellular matrix (ECM) and have contracting abilities, leading to a stiff micro-environment. These aspects are hypothesized to be involved in the origin of endometriosis-associated pain. Additionally, similarities between endometriosis-related fibrosis and other fibrotic diseases, such as systemic sclerosis or lung fibrosis, indicate that targeting fibrosis could be a potential therapeutic strategy for non-hormonal therapy for endometriosis. OBJECTIVE AND RATIONALE This review aims to summarize the current knowledge and to highlight the knowledge gaps about the role of fibrosis in endometriosis. A comprehensive literature overview about the role of fibrosis in endometriosis can improve the efficiency of fibrosis-oriented research in endometriosis. SEARCH METHODS A systematic literature search was performed in three biomedical databases using search terms for 'endometriosis', 'fibrosis', 'myofibroblasts', 'collagen', and 'α-smooth muscle actin'. Original studies were included if they reported about fibrosis and endometriosis. Both preclinical in vitro and animal studies, as well as research concerning human subjects were included. OUTCOMES Our search yielded 3441 results, of which 142 studies were included in this review. Most studies scored a high to moderate risk of bias according to the bias assessment tools. The studies were divided in three categories: human observational studies, experimental studies with human-derived material, and animal studies. The observational studies showed details about the histologic appearance of fibrosis in endometriosis and the co-occurrence of nerves and immune cells in lesions. The in vitro studies identified several pro-fibrotic pathways in relation to endometriosis. The animal studies mainly assessed the effect of potential therapeutic strategies to halt or regress fibrosis, for example targeting platelets or mast cells. WIDER IMPLICATIONS This review shows the central role of fibrosis and its main cellular driver, the myofibroblast, in endometriosis. Platelets and TGF-β have a pivotal role in pro-fibrotic signaling. The presence of nerves and neuropeptides is closely associated with fibrosis in endometriotic lesions, and is likely a cause of endometriosis-associated pain. The process of fibrotic development after EMT and FMT shares characteristics with other fibrotic diseases, so exploring similarities in endometriosis with known processes in diseases like systemic sclerosis, idiopathic pulmonary fibrosis or liver cirrhosis is relevant and a promising direction to explore new treatment strategies. The close relationship with nerves appears rather unique for endometriosis-related fibrosis and is not observed in other fibrotic diseases. REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Guus Vissers
- Department of Obstetrics & Gynaecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maddalena Giacomozzi
- Department of Obstetrics & Gynaecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wouter Verdurmen
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ron Peek
- Department of Obstetrics & Gynaecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Annemiek Nap
- Department of Obstetrics & Gynaecology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
29
|
Mu Z, Li B, Chen M, Liang C, Gu W, Su J. Endoplasmic reticulum stress induces renal fibrosis in high‑fat diet mice via the TGF‑β/SMAD pathway. Mol Med Rep 2024; 30:235. [PMID: 39422027 PMCID: PMC11544397 DOI: 10.3892/mmr.2024.13360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
The aim of the present study was to investigate the role and mechanism of endoplasmic reticulum stress (ERS) in kidney injury caused by high‑fat diet (HFD). An obese mouse model was established via HFD feeding and intervention was performed by intraperitoneal injection of the ERS inhibitor salubrinal (Sal). Changes in the body and kidney weight and serum biochemical indices of the mice were determined. Hematoxylin and eosin and Masson staining were used to observe the pathological changes of renal tissues. Reverse transcription‑quantitative PCR and western blotting were used to observe the expression of ERS‑related proteins and TGF‑β/SMAD pathway‑related proteins. Immunohistochemistry was employed to explore the distribution of these proteins. Compared with those in the control group, the weight gain, lipid metabolism disorders and deterioration of renal function in the model group were greater. Malondialdehyde was elevated and superoxide dismutase was decreased in renal tissues. The mRNA and protein levels of TGF‑β1, SMAD2/3, α‑smooth muscle actin, collagen I, glucose‑regulated protein 78 and C/EBP‑homologous protein were markedly elevated, whereas SMAD7 was markedly decreased. Sal markedly inhibited the aforementioned effects. This investigation revealed a link between ERS and renal injury caused by HFD. ERS in HFD‑fed mice triggers renal fibrosis through the TGF‑β/SMAD pathway.
Collapse
Affiliation(s)
- Zhidan Mu
- Department of Physiology and Pathophysiology, College of Basic Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Bin Li
- Department of Physiology and Pathophysiology, College of Basic Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Mingyang Chen
- Department of Physiology and Pathophysiology, College of Basic Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Chen Liang
- Department of Physiology and Pathophysiology, College of Basic Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| | - Wei Gu
- Department of Infection Disease, First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Juan Su
- Department of Physiology and Pathophysiology, College of Basic Medicine, Dali University, Dali, Yunnan 671000, P.R. China
| |
Collapse
|
30
|
Yao Y, Ritzmann F, Miethe S, Kattler-Lackes K, Colakoglu B, Herr C, Kamyschnikow A, Brand M, Garn H, Yildiz D, Langer F, Bals R, Beisswenger C. Co-culture of human AT2 cells with fibroblasts reveals a MUC5B phenotype: insights from an organoid model. Mol Med 2024; 30:227. [PMID: 39578767 PMCID: PMC11585087 DOI: 10.1186/s10020-024-00990-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 11/05/2024] [Indexed: 11/24/2024] Open
Abstract
Impaired interaction of fibroblasts with pneumocytes contributes to the progression of chronic lung disease such as idiopathic pulmonary fibrosis (IPF). Mucin 5B (MUC5B) is associated with IPF. Here we analyzed the interaction of primary fibroblasts and alveolar type 2 (AT2) pneumocytes in the organoid model. Single-cell analysis, histology, and qRT-PCR revealed that fibroblasts expressing high levels of fibrosis markers regulate STAT3 signaling in AT2 cells, which is accompanied by cystic organoid growth and MUC5B expression. Cystic growth and MUC5B expression were also caused by the cytokine IL-6. The PI3K-Akt signaling pathway was activated in fibroblasts. The drug dasatinib prevented the formation of MUC5B-expressing cystic organoids. MUC5B associated with AT2 cells in samples obtained from IPF patients. Our model shows that fibrotic primary fibroblasts induce impaired differentiation of AT2 cells via STAT3 signaling pathways, as observed in IPF patients. It can be used for mechanistic studies and drug development.
Collapse
Affiliation(s)
- Yiwen Yao
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
- Department of Clinical Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, 200065, Shanghai, China
| | - Felix Ritzmann
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Sarah Miethe
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, Philipps University of Marburg, D-35043, Marburg, Germany
| | | | - Betül Colakoglu
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Christian Herr
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Andreas Kamyschnikow
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Michelle Brand
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Holger Garn
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, Philipps University of Marburg, D-35043, Marburg, Germany
| | - Daniela Yildiz
- Experimental and Clinical Pharmacology and Toxicology, PZMS, Saarland University, 66421, Homburg, Germany
| | - Frank Langer
- Department of Thoracic- and Cardiovascular Surgery, Saarland University Hospital, Homburg/Saar, Germany
| | - Robert Bals
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
- Department of Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany.
| |
Collapse
|
31
|
Santos-Álvarez JC, Velázquez-Enríquez JM, Reyes-Jiménez E, Ramírez-Hernández AA, Iñiguez-Palomares R, Rodríguez-Beas C, Canseco SP, Aguilar-Ruiz SR, Castro-Sánchez L, Vásquez-Garzón VR, Baltiérrez-Hoyos R. Allium sativum nanovesicles exhibit anti-inflammatory and antifibrotic activity in a bleomycin-induced lung fibrosis model. Mol Biol Rep 2024; 51:1166. [PMID: 39560703 DOI: 10.1007/s11033-024-10104-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic and highly fatal disease characterized by excessive accumulation of extracellular matrix (ECM), foci of myofibroblasts, and a usual pattern of interstitial pneumonia. As suggested by international guidelines, the treatment for this disease involves supportive therapies, as there is currently no effective treatment. Plant-derived nanovesicles have emerged as a new treatment for various diseases and have been tested in cellular and murine models. METHODS AND RESULTS This research aimed to test the use of Allium sativum nanovesicles (AS-NV) in a murine model of IPF induced by bleomycin. AS-NV reduced the amount of collagen and restored lung architecture in the mouse model. AS-NV was tested on human lung fibroblasts, which do not affect the viability of healthy cells. AS-NV treatment decreases the mRNA levels of genes related to fibrosis, inflammation, and ECM deposition (Mmp2,Timp-2,Vegf,Pcna,Col1a1,Tgf-β,α-Sma,IL-1β,and Hif1a) in bleomycin-induced idiopathic pulmonary fibrosis. CONCLUSIONS This research highlights the anti-inflammatory and antifibrotic activity of AS-NV, which contributes to plant nanovesicle mechanisms in IPF; however, more AS-NV studies are needed to identify alternative treatments for idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Jovito Cesar Santos-Álvarez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez, 68120, Mexico
| | - Juan Manuel Velázquez-Enríquez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez, 68120, Mexico
| | - Edilburga Reyes-Jiménez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez, 68120, Mexico
| | - Alma Aurora Ramírez-Hernández
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez, 68120, Mexico
| | - Ramon Iñiguez-Palomares
- Departamento de Física, Universidad de Sonora, Rosales y Luis Encinas, Hermosillo, 83000, Mexico
| | - César Rodríguez-Beas
- Departamento de Física, Universidad de Sonora, Rosales y Luis Encinas, Hermosillo, 83000, Mexico
| | - Socorro Pina Canseco
- Centro de Investigación Facultad de Medicina, UNAM-UABJO, Oaxaca de Juárez, 68120, Mexico
| | - Sergio Roberto Aguilar-Ruiz
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez, Oaxaca, 68120, Mexico
| | - Luis Castro-Sánchez
- CONAHCYT-Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, 28045, Mexico
| | - Verónica Rocío Vásquez-Garzón
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez, 68120, Mexico
- CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez, Oaxaca, 68120, Mexico
| | - Rafael Baltiérrez-Hoyos
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez, 68120, Mexico.
- CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez, Oaxaca, 68120, Mexico.
| |
Collapse
|
32
|
Pommerolle L, Beltramo G, Biziorek L, Truchi M, Dias AMM, Dondaine L, Tanguy J, Pernet N, Goncalves V, Bouchard A, Monterrat M, Savary G, Pottier N, Ask K, Kolb MRJ, Mari B, Garrido C, Collin B, Bonniaud P, Burgy O, Goirand F, Bellaye PS. CD206 + macrophages are relevant non-invasive imaging biomarkers and therapeutic targets in experimental lung fibrosis. Thorax 2024; 79:1124-1135. [PMID: 39033028 PMCID: PMC11672011 DOI: 10.1136/thorax-2023-221168] [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: 11/06/2023] [Accepted: 06/25/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Interstitial lung diseases (ILDs) include a large number of diseases associated with progressive pulmonary fibrosis (PPF), including idiopathic pulmonary fibrosis (IPF). Despite the rarity of each of the fibrotic ILDs individually, they cumulatively affect a considerable number of patients. PPF is characterised by an excessive collagen deposition leading to functional decline. OBJECTIVES Therapeutic options are limited to nintedanib and pirfenidone which are only able to reduce fibrosis progression. CD206-expressing M2 macrophages are involved in fibrosis progression, and whether they may be relevant therapeutic targets or biomarkers remains an open question. RESULTS In our study, CD206+ lung macrophages were monitored in bleomycin-induced lung fibrosis in mice by combining flow cytometry, scRNAseq and in vivo molecular imaging using a single photon emission computed tomography (SPECT) radiopharmaceutical, 99mTc-tilmanocept. The antifibrotic effect of the inhibition of M2 macrophage polarisation with a JAK inhibitor, tofacitinib, was assessed in vivo. We demonstrate that CD206-targeted in vivo SPECT imaging with 99mTc-tilmanocept was able to accurately detect and quantify the increase in CD206+ macrophages from early to advanced stages of experimental fibrosis and ex vivo in lung biopsies from patients with IPF. CD206-targeted imaging also specifically detected a decrease in CD206+ lung macrophages on nintedanib and tofacitinib treatment. Importantly, early in vivo imaging of CD206+ macrophages allowed the prediction of experimental lung fibrosis progression as well as nintedanib and tofacitinib efficacy. CONCLUSIONS These findings indicate that M2 macrophages may be relevant theranostic targets for personalised medicine for patients with PPF.
Collapse
Affiliation(s)
- Lenny Pommerolle
- HSP-pathies Team, INSERM U1231 CTM Labex LIPSTIC and Label of Excellence from la Ligue National Contre le Cancer, Dijon, France
| | - Guillaume Beltramo
- HSP-pathies Team, INSERM U1231 CTM Labex LIPSTIC and Label of Excellence from la Ligue National Contre le Cancer, Dijon, France
- Pneumology and Respiratory Intensive Care, CHU Dijon, Dijon, France
- Reference Center for Rare Pulmonary Diseases, OrphaLung Network, RespiFil, CHU Dijon Bourgogne, Dijon, France, Valbonne, France
- Faculty of Medicine and Pharmacy, University of Burgundy, Dijon, France, Dijon, France
| | - Leo Biziorek
- HSP-pathies Team, INSERM U1231 CTM Labex LIPSTIC and Label of Excellence from la Ligue National Contre le Cancer, Dijon, France
| | - Marin Truchi
- Université Côte d'Azur, IPMC, UMR CNRS 7275 Inserm 1323, IHU RespiERA, Valbonne, France
| | | | - Lucile Dondaine
- HSP-pathies Team, INSERM U1231 CTM Labex LIPSTIC and Label of Excellence from la Ligue National Contre le Cancer, Dijon, France
- Reference Center for Rare Pulmonary Diseases, OrphaLung Network, RespiFil, CHU Dijon Bourgogne, Dijon, France, Valbonne, France
| | - Julie Tanguy
- HSP-pathies Team, INSERM U1231 CTM Labex LIPSTIC and Label of Excellence from la Ligue National Contre le Cancer, Dijon, France
| | - Nicolas Pernet
- Faculty of Medicine and Pharmacy, University of Burgundy, Dijon, France, Dijon, France
| | - Victor Goncalves
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université de Bourgogne, Dijon, France
| | | | - Marie Monterrat
- Team IMATHERA, Centre Georges François Leclerc, Dijon, France
| | - Grégoire Savary
- FHU-OncoAge, CNRS, IPMC, Université Côte d'Azur, Valbonne, France
| | - Nicolas Pottier
- FHU-OncoAge, CNRS, IPMC, Université Côte d'Azur, Valbonne, France
| | - Kjetil Ask
- Department of Medicine, Pathology, and Molecular Medicine, McMaster University, Dijon, Ontario, Canada
| | - Martin R J Kolb
- Department of Medicine, Pathology, and Molecular Medicine, McMaster University, Dijon, Ontario, Canada
| | - Bernard Mari
- Université Côte d'Azur, IPMC, UMR CNRS 7275 Inserm 1323, IHU RespiERA, Valbonne, France
| | - Carmen Garrido
- HSP-pathies Team, INSERM U1231 CTM Labex LIPSTIC and Label of Excellence from la Ligue National Contre le Cancer, Dijon, France
- Reference Center for Rare Pulmonary Diseases, OrphaLung Network, RespiFil, CHU Dijon Bourgogne, Dijon, France, Valbonne, France
- Team IMATHERA, Centre Georges François Leclerc, Dijon, France
| | - Bertrand Collin
- Team IMATHERA, Centre Georges François Leclerc, Dijon, France
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR 6302, CNRS, Université de Bourgogne, Dijon, France
| | - Philippe Bonniaud
- HSP-pathies Team, INSERM U1231 CTM Labex LIPSTIC and Label of Excellence from la Ligue National Contre le Cancer, Dijon, France
- Pneumology and Respiratory Intensive Care, CHU Dijon, Dijon, France
- Reference Center for Rare Pulmonary Diseases, OrphaLung Network, RespiFil, CHU Dijon Bourgogne, Dijon, France, Valbonne, France
- Faculty of Medicine and Pharmacy, University of Burgundy, Dijon, France, Dijon, France
| | - Olivier Burgy
- HSP-pathies Team, INSERM U1231 CTM Labex LIPSTIC and Label of Excellence from la Ligue National Contre le Cancer, Dijon, France
- Reference Center for Rare Pulmonary Diseases, OrphaLung Network, RespiFil, CHU Dijon Bourgogne, Dijon, France, Valbonne, France
- Faculty of Medicine and Pharmacy, University of Burgundy, Dijon, France, Dijon, France
| | - Françoise Goirand
- HSP-pathies Team, INSERM U1231 CTM Labex LIPSTIC and Label of Excellence from la Ligue National Contre le Cancer, Dijon, France
- Reference Center for Rare Pulmonary Diseases, OrphaLung Network, RespiFil, CHU Dijon Bourgogne, Dijon, France, Valbonne, France
- Faculty of Medicine and Pharmacy, University of Burgundy, Dijon, France, Dijon, France
| | - Pierre-Simon Bellaye
- HSP-pathies Team, INSERM U1231 CTM Labex LIPSTIC and Label of Excellence from la Ligue National Contre le Cancer, Dijon, France
- Reference Center for Rare Pulmonary Diseases, OrphaLung Network, RespiFil, CHU Dijon Bourgogne, Dijon, France, Valbonne, France
- Team IMATHERA, Centre Georges François Leclerc, Dijon, France
| |
Collapse
|
33
|
Li H, Wang J, Li Z, Wu Z, Zhang Y, Kong L, Yang Q, Wang D, Shi H, Shen G, Zou S, Zhu W, Fan K, Xu Z. Quantitative proteomics reveals the mechanism of endoplasmic reticulum stress-mediated pulmonary fibrosis in mice. Heliyon 2024; 10:e39150. [PMID: 39640640 PMCID: PMC11620036 DOI: 10.1016/j.heliyon.2024.e39150] [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: 07/11/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
Pulmonary fibrosis is a progressive disease that can lead to respiratory failure. Many types of cells are involved in the progression of pulmonary fibrosis. This study utilized quantitative proteomics to investigate the mechanism of TGF-β-induced fibrosis-like changes in mouse epithelial cells. Our findings revealed that TGF-β significantly impacted biological processes related to the endoplasmic reticulum, mitochondrion, and ribonucleoprotein complex. Pull-down assay coupled with proteomics identified 114 proteins that may directly interact with TGF-β, and their functions were related to mitochondria, translation, ubiquitin ligase conjugation, mRNA processing, and actin binding. Among them, 17 molecules were also found in different expression proteins (DEPs) of quantitative proteomic, such as H1F0, MED21, SDF2L1, DAD1, and TMX1. Additionally, TGF-β decreased the folded structure and the number of ribosomes in the endoplasmic reticulum and increased the expression of key proteins in the unfolded protein response, including HRD1, PERK, and ERN1. Overall, our study suggested that TGF-β induced fibrotic changes in mouse lung epithelial cells by ER stress and initiated the unfolded protein response through the PRKCSH/IRE1 and PERK/GADD34/CHOP signaling pathways.
Collapse
Affiliation(s)
- Heng Li
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Jin Wang
- Department of Clinical Laboratory, Tianjin Third Central Hospital, Tianjin, 300170, People's Republic of China
| | - Ziling Li
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Zhidong Wu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Yan Zhang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Lingjia Kong
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Qingqing Yang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Dong Wang
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - He Shi
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Guozheng Shen
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Shuang Zou
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Wenqing Zhu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Kaiyuan Fan
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| | - Zhongwei Xu
- Central Laboratory, Logistics University of Chinese People's Armed Police Force, Tianjin, 300309, People's Republic of China
- Tianjin key laboratory for prevention and control of occupational and environmental hazards, 300309, People's Republic of China
| |
Collapse
|
34
|
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
|
35
|
Shi Z, Zhou M, Zhai J, Sun J, Wang X. Novel therapeutic strategies and drugs for idiopathic pulmonary fibrosis. Arch Pharm (Weinheim) 2024; 357:e2400192. [PMID: 38961537 DOI: 10.1002/ardp.202400192] [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: 03/14/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 07/05/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease of unknown etiology. Currently, drugs used to treat IPF in clinical practice exhibit severe side effects and limitations. To address these issues, this paper discusses the therapeutic effects of preclinical targeted drugs (such as STAT3 and TGF-β/Smad pathway inhibitors, chitinase inhibitors, PI3K and phosphodiesterase inhibitors, etc.) and natural products on IPF. Through a summary of current research progress, it is found that natural products possess multitarget effects, stable therapeutic efficacy, low side effects, and nondrug dependence. Furthermore, we discuss the significant prospects of natural product molecules in combating fibrosis by influencing the immune system, expecting that current analytical data will aid in the development of new drugs or the investigation of active ingredients in natural products for potential IPF treatments in the future.
Collapse
Affiliation(s)
- Zezhou Shi
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| | - Min Zhou
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| | - Jingfang Zhai
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| | - Jie Sun
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| | - Xiaojing Wang
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| |
Collapse
|
36
|
Li Y, Du X, Hu Y, Wang D, Duan L, Zhang H, Zhang R, Xu Y, Zhou R, Zhang X, Zhang M, Liu J, Lv Z, Chen Y, Wang W, Sun Y, Cui Y. Iron-laden macrophage-mediated paracrine profibrotic signaling induces lung fibroblast activation. Am J Physiol Cell Physiol 2024; 327:C979-C993. [PMID: 39183565 DOI: 10.1152/ajpcell.00675.2023] [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: 12/06/2023] [Revised: 07/22/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating condition characterized by progressive lung scarring and uncontrolled fibroblast proliferation, inevitably leading to organ dysfunction and mortality. Although elevated iron levels have been observed in patients and animal models of lung fibrosis, the mechanisms linking iron dysregulation to lung fibrosis pathogenesis, particularly the role of macrophages in orchestrating this process, remain poorly elucidated. Here we evaluate iron metabolism in macrophages during pulmonary fibrosis using both in vivo and in vitro approaches. In murine bleomycin- and amiodarone-induced pulmonary fibrosis models, we observed significant iron deposition and lipid peroxidation in pulmonary macrophages. Intriguingly, the ferroptosis regulator glutathione peroxidase 4 (GPX4) was upregulated in pulmonary macrophages following bleomycin instillation, a finding corroborated by single-cell RNA sequencing analysis. Moreover, macrophages isolated from fibrotic mouse lungs exhibited increased transforming growth factor (TGF)-β1 expression that correlated with lipid peroxidation. In vitro, iron overload in bone marrow-derived macrophages triggered lipid peroxidation and TGF-β1 upregulation, which was effectively suppressed by ferroptosis inhibitors. When cocultured with iron-overloaded macrophages, lung fibroblasts exhibited heightened activation, evidenced by increased α-smooth muscle actin and fibronectin expression. Importantly, this profibrotic effect was attenuated by treating macrophages with a ferroptosis inhibitor or blocking TGF-β receptor signaling in fibroblasts. Collectively, our study elucidates a novel mechanistic paradigm in which the accumulation of iron within macrophages initiates lipid peroxidation, thereby amplifying TGF-β1 production, subsequently instigating fibroblast activation through paracrine signaling. Thus, inhibiting iron overload and lipid peroxidation warrants further exploration as a strategy to suppress fibrotic stimulation by disease-associated macrophages. NEW & NOTEWORTHY This study investigates the role of iron in pulmonary fibrosis, specifically focusing on macrophage-mediated mechanisms. Iron accumulation in fibrotic lung macrophages triggers lipid peroxidation and an upregulation of transforming growth factor (TGF)-β1 expression. Coculturing iron-laden macrophages activates lung fibroblasts in a TGF-β1-dependent manner, which can be mitigated by ferroptosis inhibitors. These findings underscore the potential of targeting iron overload and lipid peroxidation as a promising strategy to alleviate fibrotic stimulation provoked by disease-associated macrophages.
Collapse
Affiliation(s)
- Yunqi Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Xinqian Du
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yue Hu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Dan Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Luo Duan
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Hanxiao Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ruoyang Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People's Republic of China
- National Center for Respiratory Medicine, Beijing, People's Republic of China
| | - Yingjie Xu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ruonan Zhou
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Xinyu Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Muzhi Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Jie Liu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yan Chen
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ying Sun
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
37
|
Glatt J, Chandar P. The future of chemical pleurodesis: A review of novel and investigational sclerosant agents. Am J Med Sci 2024; 368:175-181. [PMID: 38670530 DOI: 10.1016/j.amjms.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024]
Abstract
Chemical pleurodesis is a technique in which an injurious and inflammatory substance is instilled into the pleural cavity to cause adhesion and fibrosis. It is commonly used in the management of recurrent malignant pleural effusions. Historically, many different types of sclerosants have been described, though only a few, including talc, the tetracycline derivatives, and anti-septic compounds such as silver nitrate and iodopovidone, have found their way into mainstream use. However, over the past several years, there has been increasing interest in alternative pleurodesis agents. In this review, we will explore future directions in the field, with an eye toward novel and investigational agents.
Collapse
Affiliation(s)
- Jonathan Glatt
- Thomas Jefferson University Hospital, Department of Pulmonary, Allergy, and Critical Care Medicine, 834 Walnut Street, Suite 650, Philadelphia, PA 19107, USA.
| | - Prarthna Chandar
- Thomas Jefferson University Hospital, Department of Pulmonary, Allergy, and Critical Care Medicine, 834 Walnut Street, Suite 650, Philadelphia, PA 19107, USA
| |
Collapse
|
38
|
Kim MJ, Hwang HS, Choi JH, Yoo ES, Jang MI, Lee J, Oh SM. Development of a multi-analysis model using an epithelial-fibroblast co-culture system as an alternative to animal testing. Environ Anal Health Toxicol 2024; 39:e2024024-0. [PMID: 39536704 PMCID: PMC11560297 DOI: 10.5620/eaht.2024024] [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: 07/08/2024] [Accepted: 09/13/2024] [Indexed: 11/16/2024] Open
Abstract
The evaluation of respiratory chemical substances has been mostly performed in animal tests (OECD TG 403, TG 412, TG 413, etc.). However, there have been ongoing discussions about the limited use of these inhalation toxicity tests due to differences in the anatomical structure of the respiratory tract, difficulty in exposure, laborious processes, and ethical reasons. Alternative animal testing methods that mimic in vivo testing are required. Therefore, in this study, we established a co-culture system composed of differentiated epithelial cells under an air-liquid interface (ALI) system in the apical part and fibroblasts in the basal part. This system was designed to mimic the wound-healing mechanism in the respiratory system. In addition, we developed a multi-analysis system that simultaneously performs toxicological and functional evaluations. Several individual assays were used sequentially in a multi-analysis model for pulmonary toxicity. Briefly, cytokine analysis, histology, and cilia motility were measured in the apical part, and cell migration and gel contraction assay were performed by exposing MRC-5 cells to the basal culture. First, human airway epithelial cells from bronchial (hAECB) were cultured under air-liquid interface (ALI) system conditions and validated pseudostratified epithelium by detecting differentiation-related epithelial markers using Transepithelial Electrical Resistance (TEER) measurement, Hematoxylin and Eosin (H&E) staining, and immunocytochemistry (ICC) staining. Afterward, the co-culture cells exposed to Transforming growth factor-beta 1 (TGF-β1), a key mediator of pulmonary fibrosis, induced significant toxicological responses such as cytotoxicity, cell migration, and gel contraction, which are wound-healing markers. In addition, cilia motility in epithelial cells was significantly decreased compared to control. Therefore, the multi-analysis model with a 3D epithelial-fibroblast co-culture system is expected to be useful in predicting pulmonary toxicity as a simple and efficient high-throughput screening method and as an alternative to animal testing.
Collapse
Affiliation(s)
- Min-Ju Kim
- Department of Bio-application toxicity, Hoseo University, Asan, Republic of Korea
| | - Hee-Sung Hwang
- Department of Bio-application toxicity, Hoseo University, Asan, Republic of Korea
| | - Jee Hoon Choi
- Department of Bio-application toxicity, Hoseo University, Asan, Republic of Korea
| | - Eun-Seon Yoo
- Department of Bio-application toxicity, Hoseo University, Asan, Republic of Korea
| | - Mi-Im Jang
- Department of Bio-application toxicity, Hoseo University, Asan, Republic of Korea
| | - Juhee Lee
- Department of ICT Automotive Engineering, Hoseo University, Asan, Republic of Korea
| | - Seung Min Oh
- Department of Bio-application toxicity, Hoseo University, Asan, Republic of Korea
- Department of Animal Health and Welfare, Hoseo University, Asan, Republic of Korea
| |
Collapse
|
39
|
Enzel D, Kriventsov M, Sataieva T, Malygina V. Cellular and Molecular Genetic Mechanisms of Lung Fibrosis Development and the Role of Vitamin D: A Review. Int J Mol Sci 2024; 25:8946. [PMID: 39201632 PMCID: PMC11355055 DOI: 10.3390/ijms25168946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Idiopathic pulmonary fibrosis remains a relevant problem of the healthcare system with an unfavorable prognosis for patients due to progressive fibrous remodeling of the pulmonary parenchyma. Starting with the damage of the epithelial lining of alveoli, pulmonary fibrosis is implemented through a cascade of complex mechanisms, the crucial of which is the TGF-β/SMAD-mediated pathway, involving various cell populations. Considering that a number of the available drugs (pirfenidone and nintedanib) have only limited effectiveness in slowing the progression of fibrosis, the search and justification of new approaches aimed at regulating the immune response, cellular aging processes, programmed cell death, and transdifferentiation of cell populations remains relevant. This literature review presents the key modern concepts concerning molecular genetics and cellular mechanisms of lung fibrosis development, based mainly on in vitro and in vivo studies in experimental models of bleomycin-induced pulmonary fibrosis, as well as the latest data on metabolic features, potential targets, and effects of vitamin D and its metabolites.
Collapse
Affiliation(s)
| | | | - Tatiana Sataieva
- Medical Institute Named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University, Lenina Boulevard 5/7, 295051 Simferopol, Russia; (D.E.); (M.K.); (V.M.)
| | | |
Collapse
|
40
|
Franzén L, Olsson Lindvall M, Hühn M, Ptasinski V, Setyo L, Keith BP, Collin A, Oag S, Volckaert T, Borde A, Lundeberg J, Lindgren J, Belfield G, Jackson S, Ollerstam A, Stamou M, Ståhl PL, Hornberg JJ. Mapping spatially resolved transcriptomes in human and mouse pulmonary fibrosis. Nat Genet 2024; 56:1725-1736. [PMID: 38951642 PMCID: PMC11319205 DOI: 10.1038/s41588-024-01819-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/30/2024] [Indexed: 07/03/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with poor prognosis and limited treatment options. Efforts to identify effective treatments are thwarted by limited understanding of IPF pathogenesis and poor translatability of available preclinical models. Here we generated spatially resolved transcriptome maps of human IPF (n = 4) and bleomycin-induced mouse pulmonary fibrosis (n = 6) to address these limitations. We uncovered distinct fibrotic niches in the IPF lung, characterized by aberrant alveolar epithelial cells in a microenvironment dominated by transforming growth factor beta signaling alongside predicted regulators, such as TP53 and APOE. We also identified a clear divergence between the arrested alveolar regeneration in the IPF fibrotic niches and the active tissue repair in the acutely fibrotic mouse lung. Our study offers in-depth insights into the IPF transcriptional landscape and proposes alveolar regeneration as a promising therapeutic strategy for IPF.
Collapse
Affiliation(s)
- Lovisa Franzén
- Safety Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
| | - Martina Olsson Lindvall
- Safety Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Michael Hühn
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Victoria Ptasinski
- Safety Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Laura Setyo
- Pathology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Benjamin P Keith
- Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Astrid Collin
- Animal Science and Technology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Steven Oag
- Animal Science and Technology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Thomas Volckaert
- Bioscience In Vivo, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Annika Borde
- Bioscience In Vivo, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Joakim Lundeberg
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
| | - Julia Lindgren
- Translational Genomics, Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Graham Belfield
- Translational Genomics, Centre for Genomics Research, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Sonya Jackson
- Late-Stage Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Ollerstam
- Safety Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Marianna Stamou
- Safety Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| | - Patrik L Ståhl
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden.
| | - Jorrit J Hornberg
- Safety Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
41
|
Sampsonas F, Bosgana P, Bravou V, Tzouvelekis A, Dimitrakopoulos FI, Kokkotou E. Interstitial Lung Diseases and Non-Small Cell Lung Cancer: Particularities in Pathogenesis and Expression of Driver Mutations. Genes (Basel) 2024; 15:934. [PMID: 39062713 PMCID: PMC11276289 DOI: 10.3390/genes15070934] [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: 06/15/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
INTRODUCTION Interstitial lung diseases are a varied group of diseases associated with chronic inflammation and fibrosis. With the emerging and current treatment options, survival rates have vastly improved. Having in mind that the most common type is idiopathic pulmonary fibrosis and that a significant proportion of these patients will develop lung cancer as the disease progresses, prompt diagnosis and personalized treatment of these patients are fundamental. SCOPE AND METHODS The scope of this review is to identify and characterize molecular and pathogenetic pathways that can interconnect Interstitial Lung Diseases and lung cancer, especially driver mutations in patients with NSCLC, and to highlight new and emerging treatment options in that view. RESULTS Common pathogenetic pathways have been identified in sites of chronic inflammation in patients with interstitial lung diseases and lung cancer. Of note, the expression of driver mutations in EGFR, BRAF, and KRAS G12C in patients with NSCLC with concurrent interstitial lung disease is vastly different compared to those patients with NSCLC without Interstitial Lung Disease. CONCLUSIONS NSCLC in patients with Interstitial Lung Disease is a challenging diagnostic and clinical entity, and a personalized medicine approach is fundamental to improving survival and quality of life. Newer anti-fibrotic medications have improved survival in IPF/ILD patients; thus, the incidence of lung cancer is going to vastly increase in the next 5-10 years.
Collapse
Affiliation(s)
- Fotios Sampsonas
- Department of Respiratory Medicine, Medical School, University of Patras, 26504 Patras, Greece;
| | - Pinelopi Bosgana
- Department of Pathology, Medical School, University of Patras, 26504 Patras, Greece;
| | - Vasiliki Bravou
- Department of Anatomy, Embryology and Histology, Medical School, University of Patras, 26504 Patras, Greece;
| | - Argyrios Tzouvelekis
- Department of Respiratory Medicine, Medical School, University of Patras, 26504 Patras, Greece;
| | | | - Eleni Kokkotou
- Oncology Unit, The Third Department of Medicine, Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| |
Collapse
|
42
|
Shaikh TB, Chandra Y, Andugulapati SB, Sistla R. Vistusertib improves pulmonary inflammation and fibrosis by modulating inflammatory/oxidative stress mediators via suppressing the mTOR signalling. Inflamm Res 2024; 73:1223-1237. [PMID: 38789791 DOI: 10.1007/s00011-024-01894-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
INTRODUCTION Inflammation and oxidative stress are key factors in the development of pulmonary fibrosis (PF) by promoting the differentiation of fibroblasts through modulating various pathways including Wnt/β-catenin, TGF-β and mTOR signalling. OBJECTIVE AND METHODS This study aimed to evaluate the effects and elucidate the mechanisms of vistusertib (VSB) in treating pulmonary inflammation/fibrosis, specifically by targeting the mTOR pathway using various in vitro and in vivo models. RESULTS Lipopolysaccharide (LPS)-induced inflammation model in macrophages (RAW 264.7), epithelial (BEAS-2B) and endothelial (HMVEC-L) cells revealed that treatment with VSB significantly reduced the IL-6, TNF-α, CCL2, and CCL7 expression. TGF-β induced differentiation was also significantly reduced upon VSB treatment in fibrotic cells (LL29 and DHLF). Further, bleomycin-induced inflammation and fibrosis models demonstrated that treatment with VSB significantly ameliorated the severe inflammation, and lung architectural distortion, by reducing the inflammatory markers expression/levels, inflammatory cells and oxidative stress indicators. Further, fibrosis model results exhibited that, VSB treatment significantly reduced the α-SMA, collagen and TGF-β expressions, improved the lung architecture and restored lung functions. CONCLUSION Overall, this study uncovers the anti-inflammatory/anti-fibrotic effects of VSB by modulating the mTOR activation. Although VSB was tested for lung fibrosis, it can be tested for other fibrotic disorders to improve the patient's survival and quality of life.
Collapse
Affiliation(s)
- Taslim B Shaikh
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India
| | - Yogesh Chandra
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India
| | - Sai Balaji Andugulapati
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India.
| | - Ramakrishna Sistla
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India.
| |
Collapse
|
43
|
Ke HL, Li RJ, Yu CC, Wang XP, Wu CY, Zhang YW. Network pharmacology and experimental verification to decode the action of Qing Fei Hua Xian Decotion against pulmonary fibrosis. PLoS One 2024; 19:e0305903. [PMID: 38913698 PMCID: PMC11195996 DOI: 10.1371/journal.pone.0305903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 06/06/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a common interstitial pneumonia disease, also occurred in post-COVID-19 survivors. The mechanism underlying the anti-PF effect of Qing Fei Hua Xian Decotion (QFHXD), a traditional Chinese medicine formula applied for treating PF in COVID-19 survivors, is unclear. This study aimed to uncover the mechanisms related to the anti-PF effect of QFHXD through analysis of network pharmacology and experimental verification. METHODS The candidate chemical compounds of QFHXD and its putative targets for treating PF were achieved from public databases, thereby we established the corresponding "herb-compound-target" network of QFHXD. The protein-protein interaction network of potential targets was also constructed to screen the core targets. Furthermore, Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were used to predict targets, and pathways, then validated by in vivo experiments. RESULTS A total of 188 active compounds in QFHXD and 50 target genes were identified from databases. The key therapeutic targets of QFHXD, such as PI3K/Akt, IL-6, TNF, IL-1β, STAT3, MMP-9, and TGF-β1 were identified by KEGG and GO analysis. Anti-PF effects of QFHXD (in a dose-dependent manner) and prednisone were confirmed by HE, Masson staining, and Sirius red staining as well as in vivo Micro-CT and immunohistochemical analysis in a rat model of bleomycin-induced PF. Besides, QFXHD remarkably inhibits the activity of PI3K/Akt/NF-κB and TGF-β1/Smad2/3. CONCLUSIONS QFXHD significantly attenuated bleomycin-induced PF via inhibiting inflammation and epithelial-mesenchymal transition. PI3K/Akt/NF-κB and TGF-β1/Smad2/3 pathways might be the potential therapeutic effects of QFHXD for treating PF.
Collapse
Affiliation(s)
- Hao-Liang Ke
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Rui-Jie Li
- School of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Chao-Chao Yu
- Department of Rehabilitation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiu-Ping Wang
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chao-Yan Wu
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ying-Wen Zhang
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
44
|
Taherian M, Bayati P, Mojtabavi N. Stem cell-based therapy for fibrotic diseases: mechanisms and pathways. Stem Cell Res Ther 2024; 15:170. [PMID: 38886859 PMCID: PMC11184790 DOI: 10.1186/s13287-024-03782-5] [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: 01/29/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Fibrosis is a pathological process, that could result in permanent scarring and impairment of the physiological function of the affected organ; this condition which is categorized under the term organ failure could affect various organs in different situations. The involvement of the major organs, such as the lungs, liver, kidney, heart, and skin, is associated with a high rate of morbidity and mortality across the world. Fibrotic disorders encompass a broad range of complications and could be traced to various illnesses and impairments; these could range from simple skin scars with beauty issues to severe rheumatologic or inflammatory disorders such as systemic sclerosis as well as idiopathic pulmonary fibrosis. Besides, the overactivation of immune responses during any inflammatory condition causing tissue damage could contribute to the pathogenic fibrotic events accompanying the healing response; for instance, the inflammation resulting from tissue engraftment could cause the formation of fibrotic scars in the grafted tissue, even in cases where the immune system deals with hard to clear infections, fibrotic scars could follow and cause severe adverse effects. A good example of such a complication is post-Covid19 lung fibrosis which could impair the life of the affected individuals with extensive lung involvement. However, effective therapies that halt or slow down the progression of fibrosis are missing in the current clinical settings. Considering the immunomodulatory and regenerative potential of distinct stem cell types, their application as an anti-fibrotic agent, capable of attenuating tissue fibrosis has been investigated by many researchers. Although the majority of the studies addressing the anti-fibrotic effects of stem cells indicated their potent capabilities, the underlying mechanisms, and pathways by which these cells could impact fibrotic processes remain poorly understood. Here, we first, review the properties of various stem cell types utilized so far as anti-fibrotic treatments and discuss the challenges and limitations associated with their applications in clinical settings; then, we will summarize the general and organ-specific mechanisms and pathways contributing to tissue fibrosis; finally, we will describe the mechanisms and pathways considered to be employed by distinct stem cell types for exerting anti-fibrotic events.
Collapse
Affiliation(s)
- Marjan Taherian
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Paria Bayati
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Nazanin Mojtabavi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
Zhou Y, Ling T, Shi W. Current state of signaling pathways associated with the pathogenesis of idiopathic pulmonary fibrosis. Respir Res 2024; 25:245. [PMID: 38886743 PMCID: PMC11184855 DOI: 10.1186/s12931-024-02878-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) represents a chronic and progressive pulmonary disorder distinguished by a notable mortality rate. Despite the elusive nature of the pathogenic mechanisms, several signaling pathways have been elucidated for their pivotal roles in the progression of this ailment. This manuscript aims to comprehensively review the existing literature on the signaling pathways linked to the pathogenesis of IPF, both within national and international contexts. The objective is to enhance the comprehension of the pathogenic mechanisms underlying IPF and offer a scholarly foundation for the advancement of more efficacious therapeutic strategies, thereby fostering research and clinical practices within this domain.
Collapse
Affiliation(s)
- Yang Zhou
- School of Medicine, Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, 224005, China
| | - Tingting Ling
- School of Medicine, Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, 224005, China
| | - Weihong Shi
- School of Medicine, Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, 224005, China.
| |
Collapse
|
46
|
Min Y, Wu L, Xu C, Han W, Yin Z, Pan X, Sun L, Zhang J, Wan G, Zhou T. Macrophages and pulmonary fibrosis: a bibliometric and visual analysis of publications from 1990 to 2023. Front Med (Lausanne) 2024; 11:1374177. [PMID: 38952862 PMCID: PMC11215139 DOI: 10.3389/fmed.2024.1374177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/03/2024] [Indexed: 07/03/2024] Open
Abstract
Background The role of macrophages in the symptomatic and structural progression of pulmonary fibrosis (PF) has garnered significant scholarly attention in recent years. This study employs a bibliometric approach to examine the present research status and areas of focus regarding the correlation between macrophages and PF, aiming to provide a comprehensive understanding of their relationship. Methodology The present study employed VOSviewer, CiteSpace, and Microsoft Excel software to visualize and analyze various aspects such as countries, institutions, authors, journals, co-cited literature, keywords, related genes, and diseases. These analyses were conducted using the Web of Science core collection database. Results A comprehensive collection of 3,479 records pertaining to macrophages and PF from the period of 1990 to 2023 was obtained. Over the years, there has been a consistent increase in research literature on this topic. Notably, the United States and China exhibited the highest level of collaboration in this field. Through careful analysis, the institutions, authors, and prominent journals that hold significant influence within this particular field have been identified as having the highest publication output. The pertinent research primarily concentrates on the domains of Biology and Medicine. The prevailing keywords encompass pulmonary fibrosis, acute lung injury, idiopathic pulmonary fibrosis, and others. Notably, TGFβ1, TNF, and CXCL8 emerge as the most frequently studied targets, primarily associated with signaling pathways such as cytokine-cytokine receptor interaction. Additionally, cluster analysis of related diseases reveals their interconnectedness with ailments such as cancer. Conclusion The present study employed bibliometric methods to investigate the knowledge structure and developmental trends in the realm of macrophage and PF research. The findings shed light on the introduction and research hotspots that facilitate a more comprehensive understanding of macrophages and PF.
Collapse
Affiliation(s)
- Yi Min
- Yixing Traditional Chinese Medicine Hospital, Wuxi, Jiangsu, China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Lifei Wu
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Physical Education, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Cheng Xu
- Yixing Traditional Chinese Medicine Hospital, Wuxi, Jiangsu, China
| | - Wen Han
- Yixing Traditional Chinese Medicine Hospital, Wuxi, Jiangsu, China
| | - Zhi Yin
- Yixing Traditional Chinese Medicine Hospital, Wuxi, Jiangsu, China
| | - Xu Pan
- Yixing Traditional Chinese Medicine Hospital, Wuxi, Jiangsu, China
| | - Luyao Sun
- Yixing Traditional Chinese Medicine Hospital, Wuxi, Jiangsu, China
| | - Jinrong Zhang
- Fifth People’s Hospital of Suzhou, Suzhou, Jiangsu, China
| | - Guoqiang Wan
- Yixing Traditional Chinese Medicine Hospital, Wuxi, Jiangsu, China
| | - Tongxin Zhou
- Yixing Traditional Chinese Medicine Hospital, Wuxi, Jiangsu, China
| |
Collapse
|
47
|
Wu Z, Shi R, Yan S, Zhang S, Lu B, Huang Z, Ji L. Integrating network pharmacology, experimental validation and molecular docking to reveal the alleviation of Yinhuang granule on idiopathic pulmonary fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155368. [PMID: 38498951 DOI: 10.1016/j.phymed.2024.155368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/21/2023] [Accepted: 01/15/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic disease characterized by the abnormal proliferation of fibroblast and excessive deposition of extracellular matrix (ECM), accompanied by inflammation and ultimately respiratory failure. Yinhuang granule (YHG), with clinical properties of clearing heat, detoxifying and anti-inflammation, is commonly used to heal upper respiratory diseases in China for decades. PURPOSE To explore the improvement of YHG on bleomycin (BLM)-induced IPF in mice and its possible engaged mechanism. METHODS The mortality rate was recorded, lung function was determined and hematoxylin-eosin (H&E) staining was carried out to explore the alleviation of YHG on BLM-caused IPF in mice. Hydroxyproline, collagen I and collagen III contents were detected, and Sirius red and Masson staining were conducted to evaluate YHG's alleviation on lung fibrosis. The underlying mechanism was predicted by network pharmacology, and confirmed by Real-time polymerase chain reaction (RT-PCR), Western-blot (WB) and enzyme linked immunosorbent assay (ELISA). The binding affinity between related key proteins and active compounds in YHG was calculated by using molecular docking, and further validated by cellular thermal shift assay (CESTA). RESULTS YHG (400, 800 mg/kg) weakened lung damage and pulmonary fibrosis in mice induced by BLM. Network pharmacology and experimental validation displayed that inflammation and angiogenesis participated in the YHG-provided improvement on IPF, and key involved molecules included tumor necrosis factor-α (TNFα), vascular endothelial growth factor-A (VEGFA), interleukine-6 (IL-6), etc. The data of molecular docking presented that some main active compounds from YHG had a high binding affinity with TNFR1 or VEGFR2, and some of them were further validated by CESTA. CONCLUSION YHG effectively improved the BLM-induced IPF in mice via reducing inflammation and angiogenesis.
Collapse
Affiliation(s)
- Zeqi Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ruijia Shi
- School of Basic Medical Science of Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shihao Yan
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai,200123, China
| | - Shaobo Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bin Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhenlin Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
48
|
Ran B, Ren X, Lin X, Teng Y, Xin F, Ma W, Zhao X, Li M, Wang J, Wang C, Sun L, Zhang J. Glycyrrhetinic acid loaded in milk-derived extracellular vesicles for inhalation therapy of idiopathic pulmonary fibrosis. J Control Release 2024; 370:811-820. [PMID: 38754632 DOI: 10.1016/j.jconrel.2024.05.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/27/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and life-threatening lung disease for which treatment options are limited. Glycyrrhetinic acid (GA) is a triterpenoid with multiple biological effects, such as anti-inflammatory and anti-fibrotic properties. Herein, inhalable milk-derived extracellular vesicles (mEVs) encapsulating GA (mEVs@GA) were screened and evaluated for IPF treatment. The results indicated that the loading efficiency of GA in mEVs@GA was 8.65%. Therapeutic effects of inhalable mEVs@GA were investigated in vitro and in vivo. The mEVs@GA demonstrated superior anti-inflammatory effects on LPS-stimulated MHS cells. Furthermore, repeated noninvasive inhalation delivery of mEVs@GA in bleomycin-induced IPF mice could decrease the levels of transforming growth factors β1 (TGF-β1), Smad3 and inflammatory cytokines IL-6, IL-1β and TNF-α. The mEVs@GA effectively diminished the development of fibrosis and improved pulmonary function in the IPF mice model at a quarter of the dose compared with the pirfenidone oral administration group. Additionally, compared to pirfenidone-loaded mEVs, mEVs@GA demonstrated superior efficacy at the same drug concentration in the pharmacodynamic study. Overall, inhaled mEVs@GA have the potential to serve as an effective therapeutic option in the treatment of IPF.
Collapse
Affiliation(s)
- Bo Ran
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Xiaohong Ren
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Xueyuan Lin
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Yupu Teng
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Fangyuan Xin
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Wuzhen Ma
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiangyu Zhao
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Mingwei Li
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Jinghuang Wang
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Caifen Wang
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Lixin Sun
- Shenyang Pharmaceutiacal University, Shenyang 110016, China.
| | - Jiwen Zhang
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China.
| |
Collapse
|
49
|
Howes AM, Dea NC, Ghosh D, Krishna K, Wang Y, Li Y, Morrison B, Toussaint KC, Dawson MR. Fibroblast senescence-associated extracellular matrix promotes heterogeneous lung niche. APL Bioeng 2024; 8:026119. [PMID: 38855444 PMCID: PMC11161856 DOI: 10.1063/5.0204393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/24/2024] [Indexed: 06/11/2024] Open
Abstract
Senescent cell accumulation in the pulmonary niche is associated with heightened susceptibility to age-related disease, tissue alterations, and ultimately a decline in lung function. Our current knowledge of senescent cell-extracellular matrix (ECM) dynamics is limited, and our understanding of how senescent cells influence spatial ECM architecture changes over time is incomplete. Herein is the design of an in vitro model of senescence-associated extracellular matrix (SA-ECM) remodeling using a senescent lung fibroblast-derived matrix that captures the spatiotemporal dynamics of an evolving senescent ECM architecture. Multiphoton second-harmonic generation microscopy was utilized to examine the spatial and temporal dynamics of fibroblast SA-ECM remodeling, which revealed a biphasic process that established a disordered and heterogeneous architecture. Additionally, we observed that inhibition of transforming growth factor-β signaling during SA-ECM remodeling led to improved local collagen fiber organization. Finally, we examined patient samples diagnosed with pulmonary fibrosis to further tie our results of the in vitro model to clinical outcomes. Moreover, we observed that the senescence marker p16 is correlated with local collagen fiber disorder. By elucidating the temporal dynamics of SA-ECM remodeling, we provide further insight on the role of senescent cells and their contributions to pathological ECM remodeling.
Collapse
Affiliation(s)
| | - Nova C. Dea
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 029012, USA
| | - Deepraj Ghosh
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 029012, USA
| | - Krishangi Krishna
- School of Engineering, Brown University, Providence, Rhode Island 02912, USA
| | - Yihong Wang
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912, USA
| | - Yanxi Li
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 029012, USA
| | - Braxton Morrison
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 029012, USA
| | - Kimani C. Toussaint
- School of Engineering, Brown University, Providence, Rhode Island 02912, USA
| | - Michelle R. Dawson
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 029012, USA
| |
Collapse
|
50
|
Govorova IA, Nikitochkina SY, Vorotelyak EA. Influence of intersignaling crosstalk on the intracellular localization of YAP/TAZ in lung cells. Cell Commun Signal 2024; 22:289. [PMID: 38802925 PMCID: PMC11129370 DOI: 10.1186/s12964-024-01662-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/11/2024] [Indexed: 05/29/2024] Open
Abstract
A cell is a dynamic system in which various processes occur simultaneously. In particular, intra- and intercellular signaling pathway crosstalk has a significant impact on a cell's life cycle, differentiation, proliferation, growth, regeneration, and, consequently, on the normal functioning of an entire organ. Hippo signaling and YAP/TAZ nucleocytoplasmic shuttling play a pivotal role in normal development, homeostasis, and tissue regeneration, particularly in lung cells. Intersignaling communication has a significant impact on the core components of the Hippo pathway and on YAP/TAZ localization. This review describes the crosstalk between Hippo signaling and key lung signaling pathways (WNT, SHH, TGFβ, Notch, Rho, and mTOR) using lung cells as an example and highlights the remaining unanswered questions.
Collapse
Affiliation(s)
- I A Govorova
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilov str, 26, Moscow, 119334, Russia.
| | - S Y Nikitochkina
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilov str, 26, Moscow, 119334, Russia
| | - E A Vorotelyak
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilov str, 26, Moscow, 119334, Russia
| |
Collapse
|