1
|
Yu S, Liu K, Zhou F, Yang X, Zhang J, Yu K, Zhu Y, Qin L, Peng T, Zhang C, He Y. Nobiletin from Citrus reticulata Blanco alleviates pulmonary fibrosis through inhibiting the PI3K/AKT pathway and epithelial-mesenchymal transition. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119965. [PMID: 40354836 DOI: 10.1016/j.jep.2025.119965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 05/07/2025] [Accepted: 05/10/2025] [Indexed: 05/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Citrus reticulata Blanco, a traditional Chinese herb, has been utilized for bronchitis-related conditions. Nobiletin (NOB), the primary bioactive compound in Citrus reticulata Blanco, exhibits anti-inflammatory, antioxidant, and antifibrotic properties. However, the efficacy of NOB in pulmonary fibrosis (PF) and the underlying mechanisms remain ambiguous. AIM OF THE STUDY This study is aimed to assess the effectiveness of NOB in the management of pulmonary fibrosis and to delineate the fundamental molecular mechanisms responsible for its action. MATERIALS AND METHODS Bleomycin (BLM)-induced PF in C57BL/6 mice and transforming growth factor-β1 (TGF-β1)-induced molecular alterations in NIH3T3 and A549 cells, a number of techniques were used to study the efficacy and mechanism of NOB in PF, including Sirius Red collagen assays, Western blot (WB), immunofluorescence (IF), Micro-CT Imaging, pulmonary function assay, histopathological examination, proteomic analysis, wound healing, transwell, and immunocytochemistry (IHC) were utilized to analyze the efficacy and mechanism of NOB in PF. RESULTS Our study demonstrated that NOB exhibited antifibrotic effects in TGF-β1-induced NIH3T3 cells, which was further corroborated in BLM-induced C57BL/6 mice. Utilizing proteomics analysis, we determined that the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signaling cascade and cellular adhesion processes were of significant importance. When A549 cells were induced with TGF-β1, our observations revealed that NOB could inhibit epithelial-mesenchymal transition (EMT) and display antifibrotic effects by modulating the PI3K/AKT pathway. CONCLUSIONS NOB could alleviate PF through inhibiting the PI3K/AKT pathway and EMT, suggesting it as a beneficial adjunct therapy for PF patients.
Collapse
Affiliation(s)
- Shishuang Yu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Ke Liu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Fengjiao Zhou
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Xiuli Yang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Junru Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Kexin Yu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yi Zhu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Ling Qin
- Anti-infective Agent Creation Engineering Research Centre of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Ting Peng
- Anti-infective Agent Creation Engineering Research Centre of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China.
| | - Chuantao Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Yujiao He
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China; Anti-infective Agent Creation Engineering Research Centre of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China; Chengdu Institute of Food Inspection, Chengdu, 611130, China.
| |
Collapse
|
2
|
Miao X, Liu P, Liu Y, Zhang W, Li C, Wang X. Epigenetic targets and their inhibitors in the treatment of idiopathic pulmonary fibrosis. Eur J Med Chem 2025; 289:117463. [PMID: 40048798 DOI: 10.1016/j.ejmech.2025.117463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a deadly lung disease characterized by fibroblast proliferation, excessive extracellular matrix buildup, inflammation, and tissue damage, resulting in respiratory failure and death. Recent studies suggest that impaired interactions among epithelial, mesenchymal, immune, and endothelial cells play a key role in IPF development. Advances in bioinformatics have also linked epigenetics, which bridges gene expression and environmental factors, to IPF. Despite the incomplete understanding of the pathogenic mechanisms underlying IPF, recent preclinical studies have identified several novel epigenetic therapeutic targets, including DNMT, EZH2, G9a/GLP, PRMT1/7, KDM6B, HDAC, CBP/p300, BRD4, METTL3, FTO, and ALKBH5, along with potential small-molecule inhibitors relevant for its treatment. This review explores the pathogenesis of IPF, emphasizing epigenetic therapeutic targets and potential small molecule drugs. It also analyzes the structure-activity relationships of these epigenetic drugs and summarizes their biological activities. The objective is to advance the development of innovative epigenetic therapies for IPF.
Collapse
Affiliation(s)
- Xiaohui Miao
- Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Pan Liu
- Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Yangyang Liu
- Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Wenying Zhang
- Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Chunxin Li
- Department of Clinical Laboratory Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Xiujiang Wang
- Department of Pulmonary Diseases, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China.
| |
Collapse
|
3
|
Pan T, Wu J, Qiu X, Zhu D, Wang J, Li T, Wang Z, Feng F, Xu Y, Zhou X. Identification of potential mechanisms of Schisandrin B in the treatment of idiopathic pulmonary fibrosis by integrating network pharmacology and experimental validation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5389-5403. [PMID: 39549058 DOI: 10.1007/s00210-024-03605-7] [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: 08/13/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a worsening fibrotic condition characterized by a short survival rate and limited treatment options. This study evaluates the potential anti-fibrotic properties of Schisandrin B (Sch B) through network pharmacology and experimental validation. A mouse model of bleomycin-induced pulmonary fibrosis was established, and the modeled mice were treated with Sch B at three doses (20 mg/kg/day, 40 mg/kg/day, and 80 mg/kg/day). A fibrotic model was developed in NIH/3T3 cells by treating them with TGF-β (10 ng/mL) and administering Sch B at various concentrations (10, 20, and 40 µM). The results revealed that Sch B treatment delayed the development of bleomycin-induced pulmonary fibrosis and substantially decreased the transcription levels of collagen I and α-SMA in TGF-β-induced fibroblasts. Core targets were screened with protein-protein interaction network analysis, molecular complex detection (MCODE), and CytoHubba plugin. The application of Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and molecular docking highlighted the significance of the HIF-1α signaling pathway in the potential mechanism of Sch B in IPF therapy. Western blot, PCR, and immunofluorescence were performed to validate the effects of Sch B on HIF-1α. In vivo and in vitro, Sch B administration reduced HIF-1α expression. These outcomes provide valuable insights into the potential mechanism by which Sch B delays IPF development, with HIF-1α potentially serving as a key target. However, further investigation is warranted to assess the safety and efficacy of Sch B in clinical settings.
Collapse
Affiliation(s)
- Tingyu Pan
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jieyu Wu
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xirui Qiu
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Dongwei Zhu
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jing Wang
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Tingyuan Li
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Zhichao Wang
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Fanchao Feng
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yong Xu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Xianmei Zhou
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
4
|
Dev S, Dong Y, Hamilton JP. Hepatic microtubule destabilization facilitates liver fibrosis in the mouse model of Wilson disease. J Mol Med (Berl) 2025; 103:531-545. [PMID: 40140071 PMCID: PMC12078373 DOI: 10.1007/s00109-025-02535-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/24/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025]
Abstract
Wilson disease (WD) is a potentially fatal metabolic disorder caused by the inactivation of the copper (Cu) transporter ATP7B, resulting in systemic Cu overload and fibroinflammatory liver disease. The molecular mechanism and effects of elevated Cu on cytoskeletal dynamics in liver fibrogenesis are not clear. Here, we tested the regulation of hepatic cytoskeleton and fibrogenesis with respect to Cu overload in WD. Atp7b-/- (knockout) mice with established liver disease, hepatocyte-specific Atp7b△Hep knockout mice without fibroinflammatory disease, and the age-and sex-matched controls were compared using Western blotting, real-time quantitative reverse transcription PCR (qRT-PCR), immunohistochemical (IHC) staining and transcriptomics (RNA-sequencing) analysis. In Atp7b-/- mice with developed liver disease, there is a significant increase in cytoskeletal protein expression with a reduction in α-tubulin acetylation. In these mice before the onset of liver pathology, no significant changes in cytoskeletal nor hepatic stellate cell activation are observed. As hepatic copper levels rise, an increase in cytoskeletal proteins with a decrease in acetylated-α-tubulin/α-tubulin ratio occurs. RNA-sequencing, qRT-PCR, and immunostaining confirm that the tubulin is upregulated at the transcriptional level and hepatocytes are the primary source of early tubulin increases before fibrosis. An increase in α-tubulin with a decrease in α-tubulin acetylation via Hdac6 and Sirt2 induction facilitates fibrosis as reflected by concomitant increases in desmin and α-SMA immunostaining in Atp7b-/- mice at 20 weeks. Moreover, strongly positive correlations between α-tubulin and α-tubulin deacetylase with the expression of liver fibrosis markers are observed in animal and human WD. Hepatocyte-specific Atp7b△Hep mice lack significant changes in tubulin as well as fibrosis despite hepatic steatosis. This study provides evidence that microtubule destabilization causes cytoskeletal rearrangement and facilitates hepatic stellate cell (HSC) activation and fibrosis in the murine model of WD. KEY MESSAGES: Hepatic cytoskeleton system is induced in Wilson disease. Hepatic microtubules acetylation is dysregulated in murine Wilson disease. Microtubules destabilization is positively associated with liver fibrosis in Wilson disease. Microtubules destabilization concomitant with fibrogenesis exacerbates WD progression.
Collapse
Affiliation(s)
- Som Dev
- Department of Biochemistry, All India Institute of Medical Sciences, Kalyani, West Bengal, 741245, India.
- Department of Physiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| | - Yixuan Dong
- Department of Physiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - James P Hamilton
- Department of Physiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Division of Gastroenterology and Hepatology, Johns Hopkins University, School of Medicine, 725 North Wolfe St, Baltimore, MD, 21205, USA
| |
Collapse
|
5
|
Aabis M, Tiwari P, Kumar V, Singh G, Panghal A, Jena G. Pentadecanoic acid attenuates thioacetamide-induced liver fibrosis by modulating oxidative stress, inflammation, and ferroptosis pathways in rat. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04143-6. [PMID: 40310526 DOI: 10.1007/s00210-025-04143-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 04/03/2025] [Indexed: 05/02/2025]
Abstract
Pentadecanoic acid (PDA) has been reported as a histone deacetylase 6 inhibitor. Numerous studies have shown that Histone deacetylases (HDACs) are significantly involved in the development of fibrosis. The present study focused on assessing the anti-fibrotic properties of PDA in ameliorating hepatic fibrosis induced by thioacetamide (TAA) in Wistar rats. PDA was administered orally at the doses of 10, 20 and 40 mg/kg daily, whereas TAA was administered intraperitoneally at a dose of 200 mg/kg twice weekly, for a period of 9 weeks. Administration of TAA significantly increased the relative and absolute liver weight, alanine transaminase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), gamma glutamyl transferase (γ-GT), myeloperoxidase (MPO), malondialdehyde (MDA) and reduced the glutathione (GSH) levels and PDA intervention restored the same. PDA treatment ameliorated TAA-induced collagen deposition and infiltration of inflammatory cells as revealed by Sirius red and H&E staining. Additionally, histopathological analysis revealed lymphocyte infiltration, collagen build up, development of bridging fibrosis, degeneration of the portal triad, iron accumulation, and necrosis in TAA-treated rats. The intervention with PDA significantly mitigated these pathological changes. PDA treatment significantly downregulated the expressions of TGF-β1, α-SMA, NLRP3, NF-κB and HDAC6 against TAA-induced liver damage. The present study clearly demonstrated that PDA treatment significantly alleviated TAA-induced hepatic fibrosis by ameliorating the inflammatory markers.
Collapse
Affiliation(s)
- Mohammad Aabis
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Punjab, 160062, India
| | - Priyanka Tiwari
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Punjab, 160062, India
| | - Vinod Kumar
- High resolution Transmission electron microscopy Facility, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali (near to Chandigarh), Punjab, 160062, India
| | - Gurpreet Singh
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Punjab, 160062, India
| | - Archana Panghal
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Punjab, 160062, India
| | - Gopabandhu Jena
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Punjab, 160062, India.
| |
Collapse
|
6
|
Li Y, Zheng J, Liu F, Tan X, Jiang H, Wang Y. Discussion of the material basis for prevention and treatment of pulmonary fibrosis using naturally medicinal and edible homologous herbs based on the dynamic process of Nrf2, NF-κB and TGF-β in PF. Biomed Pharmacother 2025; 185:117911. [PMID: 40090283 DOI: 10.1016/j.biopha.2025.117911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/02/2025] [Accepted: 02/07/2025] [Indexed: 03/18/2025] Open
Abstract
Pulmonary fibrosis (PF) is a progressive chronic lung disease with a high incidence and poor prognosis. Despite extensive research into the mechanisms that initiate and drive the progression of pulmonary fibrosis, developing effective treatments remains challenging due to the multiple etiologies, pathogenic links, and signaling pathways involved in PF. Indeed, nuclear factor erythroid 2-related factor 2 (Nrf2), nuclear factor kappa-B (NF-κB), and transforming growth factor-beta (TGF-β) are central players in the pathogenesis of pulmonary fibrosis, and each of these factors influences distinct yet interconnected processes that collectively contribute to disease progression: Nrf2 upregulates antioxidants to mitigate oxidative stress, NF-κB modulates inflammatory responses, and TGF-β promotes fibroblast activation and extracellular matrix (ECM) deposition, leading to fibrosis. Targeting these pathways may offer therapeutic strategies, uncover new insights and provide potential therapeutic targets for PF. Absolutely, the interactions between Nrf2, NF-κB, and TGF-β pathways are complex and can significantly influence the progression of PF, which indicated that targeting a single pathway may show poor efficacy in managing the condition. Moreover, few therapies that effectively intervene in these pathways have been approved. This review focused on the molecular mechanisms of Nrf2, NF-κB, and TGF-β involving in PF and the material basis of the naturally medicinal and edible homologous herbs, which provides a solid foundation for understanding the disease's pathogenesis, and supports the development of therapeutic drugs or treatments for addressing the complex nature of PF.
Collapse
Affiliation(s)
- Yan Li
- Chongqing Academy of Chinese Materia Medica, Chongqing University of Chinese Medicine, Chongqing 400065, PR China; Chongqing Key Laboratory of Chinese Medicine & Health Science, Chongqing 400065, PR China.
| | - Jia Zheng
- Chongqing University of Chinese Medicine, Chongqing 402760, PR China.
| | - Fei Liu
- Chongqing Academy of Chinese Materia Medica, Chongqing University of Chinese Medicine, Chongqing 400065, PR China.
| | - Xianfeng Tan
- Chongqing Baijiahuan Health Technology Co., Ltd, Chongqing 400065, China.
| | - Huiping Jiang
- Chongqing Baijiahuan Health Technology Co., Ltd, Chongqing 400065, China.
| | - Yongde Wang
- Chongqing Academy of Chinese Materia Medica, Chongqing University of Chinese Medicine, Chongqing 400065, PR China; Chongqing Key Laboratory of Chinese Medicine & Health Science, Chongqing 400065, PR China.
| |
Collapse
|
7
|
Tan S, Fu G, Xie Y, Xie X, Yan J, Jin L. HDAC6 deficiency aggravates ductular reactions through aggresome-mediated hepatocyte apoptosis. Biochem Biophys Res Commun 2025; 753:151511. [PMID: 39986090 DOI: 10.1016/j.bbrc.2025.151511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
Ductular reactions (DRs) contribute significantly to the occurrence and development of liver disease. While histone deacetylase 6 (HDAC6) is known to regulate injury repair in multiple tissues, its exact role in DRs remains unclear. This study examined the role and underlying mechanism of HDAC6 in DRs using an HDAC6 knockout (HDAC6-/y) male mouse model. Wild type and HDAC6-deficient male mice were administered 3,5 diethoxicarbonyl-1,4 dihydrocollidine (DDC) to induce DRs. The impact of HDAC6 inhibition on aggresome formation was assessed in vitro using AML-12 hepatocytes exposed to H2O2 and treated with tubastatin A (TSA), a selective HDAC6 inhibitor. Fluorescence immunohistochemistry and quantitative real-time polymerase chain reaction (qRT-PCR) were employed to quantify protein and gene expression levels, respectively. Immunohistochemical and qRT-PCR analyses revealed that HDAC6 deficiency exacerbated DRs and fibrosis, accompanied by increased expression of transforming growth factor β (TGF-β) and activation of the Notch signaling pathway. Additionally, genetic knockout or pharmacological inhibition of HDAC6 promoted hepatocyte apoptosis in vivo and in vitro, as evidenced by elevated caspase3, caspase9, and p53 expression. Furthermore, TSA treatment induced the formation of aggresomes in H2O2-exposed AML-12 hepatocytes, which were encased by vimentin filaments. These findings demonstrate that HDAC6 deficiency promotes DRs and liver fibrosis through the formation of intracellular aggregates, ultimately leading to hepatocyte apoptosis.
Collapse
Affiliation(s)
- Shanshan Tan
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, 312000, China
| | - Guoquan Fu
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, 312000, China; Hangzhou Hongwang Medical Laboratory Co. Ltd., Hangzhou, Zhejiang, 310000, China
| | - Yixia Xie
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, 312000, China
| | - Xueying Xie
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, 312000, China
| | - Junyan Yan
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, 312000, China.
| | - Lifang Jin
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang, 312000, China; Hangzhou Hongwang Medical Laboratory Co. Ltd., Hangzhou, Zhejiang, 310000, China.
| |
Collapse
|
8
|
Song B, Hu J, Chen S, Zhang Y. The Mechanisms and Therapeutic Implications of PI3K Signaling in Airway Inflammation and Remodeling in Asthma. Biologics 2025; 19:73-86. [PMID: 40070559 PMCID: PMC11895685 DOI: 10.2147/btt.s497622] [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: 10/10/2024] [Accepted: 02/15/2025] [Indexed: 03/14/2025]
Abstract
Bronchial asthma is a complex and heterogeneous disease with ongoing airway inflammation and increased airway responsiveness. Key characteristics of the disease include persistent airway inflammation, airway hyperresponsiveness, and airway remodeling. Asthma's chronic and recurrent characteristics contribute to airway remodeling and inflammation, which can exacerbate lung damage. Presently, inflammation is predominantly managed with corticosteroids, yet there is a notable absence of treatments specifically addressing airway remodeling. The phosphoinositide 3-kinase (PI3K) signaling pathway is integral to the processes of inflammation, airway remodeling, and immune responses. Pharmacological agents targeting this pathway are currently undergoing clinical evaluation. This review elucidates the role of PI3K in the immune responses, airway inflammation, and remodeling associated with asthma, examining its underlying mechanisms. Furthermore, we synthesize the existing literature on the therapeutic potential of PI3K inhibitors for asthma management, emphasizing immune modulation, airway inflammation, and remodeling, including drug development and ongoing clinical trials. Lastly, we explore how various PI3K-targeted therapies may enhance efficacy and improve tolerance.
Collapse
Affiliation(s)
- Bangguo Song
- School of Clinical Chinese Medicine, Gansu University of Traditional Chinese Medicine, Gansu, People’s Republic of China
| | - Jihong Hu
- Teaching Experimental Training Center, Gansu University of Traditional Chinese Medicine, Gansu, People’s Republic of China
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, People’s Republic of China
| | - Shupeng Chen
- School of Clinical Medicine, Jiangxi University of Traditional Chinese Medicine, Jiangxi, People’s Republic of China
| | - Yang Zhang
- Key Laboratory of Dunhuang Medicine and Transformation, Ministry of Education, Gansu, People’s Republic of China
- Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| |
Collapse
|
9
|
Fontana A, Bergantini L, Carullo G, Scalvini L, D’Alessandro M, Papulino C, Cameli P, Gangi S, Vincenzi F, Varani K, Contri C, Pasquini S, Pistocchi A, Pezzotta A, Carbone S, Saponara S, Gemma S, Altucci L, Benedetti R, Lodola A, Mor M, Bargagli E, Butini S, Campiani G. Spirotetrahydroisoquinoline-Based Histone Deacetylase Inhibitors as New Antifibrotic Agents: Biological Evaluation in Human Fibroblasts from Bronchoalveolar Lavages of Idiopathic Pulmonary Fibrosis Patients. ACS Pharmacol Transl Sci 2025; 8:380-393. [PMID: 39974640 PMCID: PMC11833724 DOI: 10.1021/acsptsci.4c00456] [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: 07/26/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 02/21/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a rare interstitial lung disease typified by a progressive fibrosing phenotype. IPF has been associated with aberrant HDAC activity, particularly HDAC6. Combining synthetic and modeling studies, a new family of spirotetrahydroisoquinoline-capped histone deacetylase inhibitors 5a-o was developed. These analogues were prepared via the three-component Castagnoli-Cushman reaction (CCR) as the key step. Structure-activity relationship (SAR) studies identified 5n (fibrostat) as a preferential HDAC6 inhibitor with a suitable degree of selectivity compared to HDAC1, HDAC3, HDAC5, HDAC8, HDAC10, and HDAC11. 5n was able to negatively modulate the expression of fibrotic markers, fibronectin and collagen 1, in fibroblasts derived from bronchoalveolar lavages of IPF patients. In another ex vivo IPF human model, 5n (fibrostat) reduced the expression of fibronectin and negatively affected the expression of collagen 1 and vimentin, the latter being associated with invasiveness. Finally, fibrostat did not show toxicity in rat-perfused heart and zebrafish larvae.
Collapse
Affiliation(s)
- Anna Fontana
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Laura Bergantini
- Department
of Medical Science, Surgery and Neuroscience, Policlinico Santa Maria
alle Scotte, University of Siena, Viale Mario Bracci 16, 53100 Siena, Italy
| | - Gabriele Carullo
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Laura Scalvini
- Department
of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Miriana D’Alessandro
- Department
of Medical Science, Surgery and Neuroscience, Policlinico Santa Maria
alle Scotte, University of Siena, Viale Mario Bracci 16, 53100 Siena, Italy
| | - Chiara Papulino
- Department
of Precision Medicine, University of Campania
Luigi Vanvitelli, 80138 Naples, Italy
| | - Paolo Cameli
- Department
of Medical Science, Surgery and Neuroscience, Policlinico Santa Maria
alle Scotte, University of Siena, Viale Mario Bracci 16, 53100 Siena, Italy
| | - Sara Gangi
- Department
of Medical Science, Surgery and Neuroscience, Policlinico Santa Maria
alle Scotte, University of Siena, Viale Mario Bracci 16, 53100 Siena, Italy
| | - Fabrizio Vincenzi
- Department
of Translational Medicine, University of
Ferrara, 44121 Ferrara, Italy
| | - Katia Varani
- Department
of Translational Medicine, University of
Ferrara, 44121 Ferrara, Italy
| | - Chiara Contri
- Department
of Translational Medicine, University of
Ferrara, 44121 Ferrara, Italy
| | - Silvia Pasquini
- Department
of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Anna Pistocchi
- Department
of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi 93 - L.I.T.A., 20054 Segrate (MI), Italy
| | - Alex Pezzotta
- Department
of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi 93 - L.I.T.A., 20054 Segrate (MI), Italy
| | - Sabrina Carbone
- Department
of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi 93 - L.I.T.A., 20054 Segrate (MI), Italy
| | - Simona Saponara
- Department
of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Sandra Gemma
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Lucia Altucci
- Department
of Precision Medicine, University of Campania
Luigi Vanvitelli, 80138 Naples, Italy
- Biogem
Institute of Molecular and Genetic Biology, 83031 Ariano Irpino, Avellino, Italy
- Program of Medical
Epigenetics, Vanvitelli
Hospital, 80138 Naples, Italy
| | - Rosaria Benedetti
- Department
of Precision Medicine, University of Campania
Luigi Vanvitelli, 80138 Naples, Italy
- Program of Medical
Epigenetics, Vanvitelli
Hospital, 80138 Naples, Italy
| | - Alessio Lodola
- Department
of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Marco Mor
- Department
of Food and Drug, University of Parma, 43124 Parma, Italy
- Microbiome
Research Hub, University of Parma, Parma 43124, Italy
| | - Elena Bargagli
- Department
of Medical Science, Surgery and Neuroscience, Policlinico Santa Maria
alle Scotte, University of Siena, Viale Mario Bracci 16, 53100 Siena, Italy
| | - Stefania Butini
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Giuseppe Campiani
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| |
Collapse
|
10
|
Majidpour M, Azizi SG, Davodabadi F, Sabeti Akbar-Abad M, Abdollahi Z, Sargazi S, Shahriari H. Recent advances in TGF-β signaling pathway in COVID-19 pathogenesis: A review. Microb Pathog 2025; 199:107236. [PMID: 39701478 DOI: 10.1016/j.micpath.2024.107236] [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: 09/02/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/21/2024]
Abstract
The coronavirus disease 2019 (COVID-19) has resulted in approximately 7.0 million fatalities between 2019 and 2022, underscoring a pressing need for comprehensive research into its underlying mechanisms and therapeutic avenues. A distinctive feature of severe COVID-19 is the dysregulated immune response characterized by excessive activation of immune cells and the consequent cytokine storms. Recent advancements in our understanding of cellular signaling pathways have illuminated the role of Transforming Growth Factor Beta (TGF-β) as a pivotal signaling molecule with significant implications for the pathogenesis of infectious diseases, including COVID-19. Emerging evidence reveals that TGF-β signaling, when activated by viral components or secondary pathways, adversely affects diverse cell types, particularly immune cells, and lung tissue, leading to complications such as pulmonary fibrosis. In our review article, we critically evaluate recent literature on the involvement of TGF-β signaling in the progression of COVID-19. We discuss a range of pharmacological interventions, including nintedanib, pirfenidone, corticosteroids, proton pump inhibitors, and histone deacetylase inhibitors, and their potential to modulate the TGF-β pathway in the context of COVID-19 treatment. Additionally, we explore ongoing clinical trials involving mesenchymal stem cells, low-dose radiation therapy, and artemisinin derivatives to assess their impact on TGF-β levels and subsequent clinical outcomes in COVID-19 patients. This review is particularly relevant at this juncture as the global health community continues to grapple with the ramifications of the COVID-19 pandemic, highlighting the urgent need for targeted therapeutic strategies aimed at TGF-β modulation to mitigate disease severity and improve patient outcomes.
Collapse
Affiliation(s)
- Mahdi Majidpour
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Seyed Ghader Azizi
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Fatemeh Davodabadi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahboobeh Sabeti Akbar-Abad
- Department of Clinical Biochemistry, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Zahra Abdollahi
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran.
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Hossein Shahriari
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| |
Collapse
|
11
|
Agrawal A, Clayton EL, Cavazos CL, Clayton BA, Rodney GG. Histone deacetylase 6 inhibition promotes microtubule acetylation and facilitates autophagosome-lysosome fusion in dystrophin-deficient mdx mice. Acta Physiol (Oxf) 2025; 241:e14243. [PMID: 39422111 DOI: 10.1111/apha.14243] [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: 11/29/2023] [Revised: 08/23/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024]
Abstract
AIM Duchenne muscular dystrophy is a progressive muscle-wasting disease caused by mutations in the dystrophin gene. Despite progress in dystrophin-targeted gene therapies, it is still a fatal disease requiring novel therapeutics that can be used synergistically or alternatively to emerging gene therapy. Defective autophagy and disorganized microtubule networks contribute to dystrophic pathogenesis, yet the mechanisms by which microtubule alterations regulate autophagy remain elusive. The present study was designed to uncover possible mechanisms underpinning the role of microtubules in regulating autophagy in dystrophic mice. METHODS Mdx mice were also supplemented with Tubastatin A, a pharmacological inhibitor of histone deacetylase 6, and pathophysiology was assessed. Mdx mice with a genetic deletion of the Nox-2 scaffolding subunit p47phox were used to assess redox dependence on tubulin acetylation. RESULTS Our data show decreased acetylation of α-tubulin with enhanced histone deacetylase 6 expression. Tubastatin A increases tubulin acetylation and Q-SNARE complex formation but does not alter microtubule organization or density, indicating improved autophagosome-lysosome fusion. Tubastatin A increases the acetylation of peroxiredoxin and protects it from hyper-oxidation, hence modulating intracellular redox status in mdx mice. Tubastatin A reduces muscle damage and enhances force production. Genetic down regulation of Nox2 activity in the mdx mice promotes autophagosome maturation but not autolysosome formation. CONCLUSION Our data highlight that autophagy is differentially regulated by redox and acetylation in mdx mice. By improving autophagy through promoting tubulin acetylation, Tubastatin A decreases the dystrophic phenotype and improves muscle function, suggesting a great potential for clinical translation and treating dystrophic patients.
Collapse
Affiliation(s)
- Akanksha Agrawal
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Erin L Clayton
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Courtney L Cavazos
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Benjamin A Clayton
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - George G Rodney
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
12
|
Byeon HJ, Choi SH, Kikkawa DO, Ko J, Yoon JS. Therapeutic role of histone deacetylase inhibition in an in vitro model of Graves' orbitopathy. Mol Med Rep 2024; 30:218. [PMID: 39329199 PMCID: PMC11465418 DOI: 10.3892/mmr.2024.13342] [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: 03/02/2024] [Accepted: 08/20/2024] [Indexed: 09/28/2024] Open
Abstract
Graves' orbitopathy (GO), a manifestation of Graves' disease, is characterized by orbital fibroblast‑induced inflammation, leading to fibrosis or adipogenesis. Histone deacetylase (HDAC) serves a central role in autoimmune diseases and fibrosis. The present study investigated HDAC inhibition in orbital fibroblasts from patients with GO to evaluate its potential as a therapeutic agent. Primary cultured orbital fibroblasts were treated with an HDAC inhibitor, panobinostat, under the stimulation of IL‑1β, TGF‑β or adipogenic medium. Inflammatory cytokines, and fibrosis‑ and adipogenesis‑related proteins were analyzed using western blotting. The effects of panobinostat on HDAC mRNA expression were measured in GO orbital fibroblasts, and specific HDACs were inhibited using small interfering RNA transfection. Panobinostat significantly reduced the IL‑1β‑induced production of inflammatory cytokines and TGF‑β‑induced production of fibrosis‑related proteins. It also suppressed adipocyte differentiation and adipogenic transcription factor production. Furthermore, it significantly attenuated HDAC7 mRNA expression in GO orbital fibroblasts. In addition, the silencing of HDAC7 led to anti‑inflammatory and anti‑fibrotic effects. In conclusion, by inhibiting HDAC7 gene expression, panobinostat may suppress the production of inflammatory cytokines, profibrotic proteins and adipogenesis in GO orbital fibroblasts. The present in vitro study suggested that HDAC7 could be a potential therapeutic target for inhibiting the inflammatory, adipogenic and fibrotic mechanisms of GO.
Collapse
Affiliation(s)
- Hyeong Ju Byeon
- Department of Ophthalmology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Soo Hyun Choi
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Don O. Kikkawa
- Division of Oculofacial Plastic and Reconstructive Surgery, Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92037, USA
| | - Jaesang Ko
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jin Sook Yoon
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
13
|
Guan Y, Li J, Sun B, Xu K, Zhang Y, Ben H, Feng Y, Liu M, Wang S, Gao Y, Duan Z, Zhang Y, Chen D, Wang Y. HBx-induced upregulation of MAP1S drives hepatocellular carcinoma proliferation and migration via MAP1S/Smad/TGF-β1 loop. Int J Biol Macromol 2024; 281:136327. [PMID: 39374711 DOI: 10.1016/j.ijbiomac.2024.136327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 09/12/2024] [Accepted: 10/03/2024] [Indexed: 10/09/2024]
Abstract
Hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC), has a significantly higher risk of recurrence. However, the exact mechanism by which HBV prompts HCC recurrence remains largely unknown. In this study liver microarray test revealed significant upregulation of microtubule associated protein 1S (MAP1S) in metastatic HCC compared to control. MAP1S knockdown suppressed growth of HCCLM3 cells in vitro and in vivo. Mechanistically, HBV-encoded X protein (HBx) upregulates MAP1S, which enhances microtubule (MT) acetylation by promoting the degradation of histone deacetylase 6 (HDAC6), and facilitates the nuclear translocation of Smad complex, and thereby enhancing downstream TGF-β signaling. Smad complex, in turn, increases MAP1S, establishing a feedback loop of MAP1S/Smad/TGF-β1. Finally, survival analysis of 150 HBV-associated HCC patients demonstrated both increased MAP1S and decreased HDAC6 were significantly associated with shorter relapse-free survival. Collectively, this study reveals a unique mechanism whereby HBx-induced upregulation of MAP1S drives HBV-related HCC proliferation and migration through the MAP1S/Smad/TGF-β1 feedback loop. TEASER: MAP1S is a key link between HBV infection and a higher risk of metastatic recurrence of HCC.
Collapse
Affiliation(s)
- Yuanyue Guan
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Tsinghua Changgung Hospital, School Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Jiaxi Li
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Bin Sun
- Clinical Center for Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Intervention Therapy Center of Tumor and Liver Diseases, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Kaikun Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yonghong Zhang
- Clinical Center for Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Intervention Therapy Center of Tumor and Liver Diseases, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Haijing Ben
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Yingmei Feng
- Department of Science and Development, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Mengcheng Liu
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Shanshan Wang
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Yuxue Gao
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Zhongping Duan
- Clinical Center for Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Artificial Liver Center, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | - Yang Zhang
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China.
| | - Dexi Chen
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China.
| | - Yanjun Wang
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing You An Hospital, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
14
|
Lin L, Lin Y, Han Z, Wang K, Zhou S, Wang Z, Wang S, Chen H. Understanding the molecular regulatory mechanisms of autophagy in lung disease pathogenesis. Front Immunol 2024; 15:1460023. [PMID: 39544928 PMCID: PMC11560454 DOI: 10.3389/fimmu.2024.1460023] [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: 07/05/2024] [Accepted: 10/07/2024] [Indexed: 11/17/2024] Open
Abstract
Lung disease development involves multiple cellular processes, including inflammation, cell death, and proliferation. Research increasingly indicates that autophagy and its regulatory proteins can influence inflammation, programmed cell death, cell proliferation, and innate immune responses. Autophagy plays a vital role in the maintenance of homeostasis and the adaptation of eukaryotic cells to stress by enabling the chelation, transport, and degradation of subcellular components, including proteins and organelles. This process is essential for sustaining cellular balance and ensuring the health of the mitochondrial population. Recent studies have begun to explore the connection between autophagy and the development of different lung diseases. This article reviews the latest findings on the molecular regulatory mechanisms of autophagy in lung diseases, with an emphasis on potential targeted therapies for autophagy.
Collapse
Affiliation(s)
- Lin Lin
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhongyu Han
- School of Medicine, Southeast University, Nanjing, China
- Science Education Department, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Ke Wang
- Department of Science and Education, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| | - Shuwei Zhou
- Department of Radiology, Zhongda Hospital, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, School of Medicine, Southeast University, Nanjing, China
| | - Zhanzhan Wang
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Lianyungang, Lianyungang, China
| | - Siyu Wang
- Department of Preventive Medicine, Kunshan Hospital of Chinese Medicine, Kunshan, China
| | - Haoran Chen
- Science Education Department, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| |
Collapse
|
15
|
Zhao X, Yang L. Pharmacological targets and validation of remdesivir for the treatment of COVID-19-associated pulmonary fibrosis: A network-based pharmacology and bioinformatics study. Medicine (Baltimore) 2024; 103:e39062. [PMID: 39331891 PMCID: PMC11441881 DOI: 10.1097/md.0000000000039062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 07/03/2024] [Indexed: 09/29/2024] Open
Abstract
The objective of this study was to employ bioinformatics and network pharmacology methodologies to investigate the targets and molecular mechanisms of remdesivir in the treatment of coronavirus disease 2019 (COVID-19)-associated pulmonary fibrosis (PF). Several open-source databases were utilized to confirm the shared targets of remdesivir, COVID-19, and PF. Following this, a comprehensive analysis incorporating function enrichment, protein-protein interaction (PPI), transcription factor (TF), and molecular docking was conducted to investigate the potential mechanisms underlying the effectiveness of remdesivir in the treatment of COVID-19-associated PF. The initial validation of these findings was performed using publicly available histological and single-cell sequencing databases. The functional enrichment analysis revealed a strong association between remdesivir and viral defense, inflammatory response, and immune response. The key pathways identified in the study were transforming growth factor (TGF-β), PI3K-Akt, mTOR, MAPK, epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor resistance, HIF-1, and Toll-like receptor signaling pathways. Additionally, the PPI analysis demonstrated the network relationships of 13 important targets, while the TF analysis provided valuable insights into the regulatory networks of these targets. Among the identified TFs, RELA was found to be the most significant. To validate our findings, we utilized publicly available histological and single-cell sequencing databases, successfully confirming the involvement of 8 key targets, including AKT1, EGFR, RHOA, MAPK1, PIK3R1, MAPK8, MAPK14, and MTOR. Furthermore, molecular docking studies were conducted to assess the interaction between remdesivir and the identified key targets, thus confirming its effective targeting effects. Remdesivir has the potential to exert antiviral, anti-inflammatory, and immunomodulatory effects in the context of COVID-19-associated PF.
Collapse
Affiliation(s)
- Xueping Zhao
- Department of Pharmacy, The First People’s Hospital of Hangzhou Lin’an District, Hangzhou, China
| | - Liping Yang
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory of Drug Clinical Risk and Personalized Medication Evaluation, Beijing, China
| |
Collapse
|
16
|
Liu B, Li N, Liu Y, Zhang Y, Qu L, Cai H, Li Y, Wu X, Geng Q. BRD3308 suppresses macrophage oxidative stress and pyroptosis via upregulating acetylation of H3K27 in sepsis-induced acute lung injury. BURNS & TRAUMA 2024; 12:tkae033. [PMID: 39224841 PMCID: PMC11367671 DOI: 10.1093/burnst/tkae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/04/2024] [Indexed: 09/04/2024]
Abstract
Background Sepsis-induced acute lung injury (ALI) leads to severe hypoxemia and respiratory failure, contributing to poor prognosis in septic patients. Endotoxin dissemination triggers oxidative stress and the release of inflammatory cytokines in macrophages, initiating diffuse alveolar damage. The role of epigenetic histone modifications in organ injury is increasingly recognized. The present study aimed to investigate the use of a histone modification inhibitor to alleviate sepsis-induced ALI, revealing a new strategy for improving sepsis patient survival. Methods In vivo models of ALI were established through the intraperitoneal injection of lipopolysaccharide and cecal ligation and puncture surgery. Furthermore, the disease process was simulated in vitro by stimulating Tamm-Horsfall protein-1 (THP-1) cells with lipopolysaccharide. Hematoxylin and eosin staining, blood gas analysis and pulmonary function tests were utilized to assess the extent of lung tissue damage. Western blot analysis, real-time polymerase chain reaction, enzyme-linked immunosorbent assay and immunofluorescence were used to measure the levels and distribution of the indicated indicators within cells and tissues. Reactive oxygen species and autophagic flux alterations were detected using specific probes. Results BRD3308, which is a inhibitor of histone deacetylase 3, improved lung tissue damage, inflammatory infiltration and edema in ALI by inhibiting Nod-like receptor protein3-mediated pyroptosis in macrophages. By upregulating autophagy, BRD3308 improved the disruption of redox balance in macrophages and reduced the accumulation of reactive oxygen species. Mechanistically, BRD3308 inhibited histone deacetylase 3 activity by binding to it and altering its conformation. Following histone deacetylase 3 inhibition, acetylation of H3K27 was significantly increased. Moreover, the increase in H3K27Ac led to the upregulation of autophagy-related gene 5, a key component of autophagosomes, thereby activating autophagy. Conclusions BRD3308 inhibits oxidative stress and pyroptosis in macrophages by modulating histone acetylation, thereby preventing sepsis-induced ALI. The present study provides a potential strategy and theoretical basis for the clinical treatment of sepsis-induced ALI.
Collapse
Affiliation(s)
- Bohao Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin, 130021, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, China
| | - Yan Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin, 130021, China
| | - Limei Qu
- Department of Pathology, The First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin, 130021, China
| | - Hongfei Cai
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin, 130021, China
| | - Yang Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin, 130021, China
- Organ Transplantation Center, The First Hospital of Jilin University, 71 Xinmin Street, Chaoyang District, Changchun, Jilin, 130021, China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, China
| |
Collapse
|
17
|
Liu F, Xu J, Li F, Ni W, Chen Z, Hou S, Ke S, Wang B. Resveratrol reinforces the therapeutic effect of mesenchymal stem cell (MSC)-derived exosomes against renal ischemia‒reperfusion injury (RIRI)-associated fibrosis by suppressing TGF-β-induced epithelial-mesenchymal transition. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2024; 22:200242. [PMID: 39280777 PMCID: PMC11401501 DOI: 10.1016/j.ijcrp.2024.200242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 09/18/2024]
Abstract
Resveratrol (RSV) has been shown to prevent epithelial-mesenchymal transition (EMT) in different diseases by modulating several signaling pathways, and RSV can prevent EMT by modulating the signaling of the TGF-β/Smad axis. In the development of renal ischemia‒reperfusion injury (RIRI), RSV and MSC-derived exosomes could ameliorate RIRI via different signaling pathways. In this study, we aimed to investigate the effect of RSV plus MSC-derived exosomes on the prognosis of RIRI. Quantitative real-time polymerase chain reaction (PCR) was performed to measure the expression of E-CAD, SMA, COL10A1, VMT and MMP-7 mRNA in TCMK-1 cells and mice under various conditions. HE and Masson staining were used to evaluate kidney injury and fibrosis in mice under various conditions. RSV effectively maintained the TGF-β- and AA-induced upregulation of E-CAD, SMA, COL10A1, VMT and MMP-7 mRNA expression in TCMK-1 cells. Moreover, MSC-derived exosomes effectively reinforced the effect of RSV on reducing the TGF-β- and AA-induced upregulation of E-CAD, SMA, COL10A1, VMT and MMP-7 mRNA expression in TCMK-1 cells. Furthermore, MSC-derived exosomes enhanced the capability of RSV to maintain the RIRI-induced increases in Cr and BUN, as well as the upregulation of E-CAD, SMA, COL10A1, VMT and MMP-7 mRNA expression in mice. In addition, MSC-derived exosomes enhanced the capability of RSV to decrease RIRI-induced kidney injury and fibrosis in mice. Our findings showed that the administration of MSC-derived exosomes and RSV could suppress the TGF-β-induced epithelial-mesenchymal transition. This suppressive effect was promoted by the coadministration of MSC-derived exosomes and RSV.
Collapse
Affiliation(s)
- Fuhe Liu
- Pharmaceutical Department, Zhejiang Pharmaceutical College, Ningbo, Zhejiang, 315100, China
| | - Jinlong Xu
- Ningbo Yinzhou No.2 Hospital, Ningbo, Zhejiang, 315100, China
| | - Fen Li
- Huzhou Institute for Food and Drug Control, Huzhou, Zhejiang, 313000, China
| | - Wenjuan Ni
- Pharmaceutical Department, Zhejiang Pharmaceutical College, Ningbo, Zhejiang, 315100, China
| | - Ziwei Chen
- Pharmaceutical Department, Zhejiang Pharmaceutical College, Ningbo, Zhejiang, 315100, China
| | - Shanshan Hou
- Pharmaceutical Department, Zhejiang Pharmaceutical College, Ningbo, Zhejiang, 315100, China
| | - Shasha Ke
- Municipal Hospital Affiliated to Taizhou University, Taizhou, Zhejiang, 318000, China
| | - Binhui Wang
- Municipal Hospital Affiliated to Taizhou University, Taizhou, Zhejiang, 318000, China
| |
Collapse
|
18
|
Zhang F, Cui Y, Zhang T, Yin W. Epigenetic regulation of macrophage activation in chronic obstructive pulmonary disease. Front Immunol 2024; 15:1445372. [PMID: 39206196 PMCID: PMC11349576 DOI: 10.3389/fimmu.2024.1445372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Macrophages in the innate immune system play a vital role in various lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), acute lung injury and pulmonary fibrosis. Macrophages involved in the process of immunity need to go through a process of activation, including changes in gene expression and cell metabolism. Epigenetic modifications are key factors of macrophage activation including DNA methylation, histone modification and non-coding RNA regulation. Understanding the role and mechanisms of epigenetic regulation of macrophage activation can provide insights into the function of macrophages in lung diseases and help identification of potential therapeutic targets. This review summarizes the latest progress in the epigenetic changes and regulation of macrophages in their development process and in normal physiological states, and the epigenetic regulation of macrophages in COPD as well as the influence of macrophage activation on COPD development.
Collapse
Affiliation(s)
- Feng Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Yachao Cui
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Tiejun Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University Affiliated Qingyuan Hospital, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenguang Yin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University (GMU) - Guangzhou Institutes of Biomedicine and Health (GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Cheng HP, Jiang SH, Cai J, Luo ZQ, Li XH, Feng DD. Histone deacetylases: potential therapeutic targets for idiopathic pulmonary fibrosis. Front Cell Dev Biol 2024; 12:1426508. [PMID: 39193364 PMCID: PMC11347278 DOI: 10.3389/fcell.2024.1426508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/03/2024] [Indexed: 08/29/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive disease of unknown origin and the most common interstitial lung disease. However, therapeutic options for IPF are limited, and novel therapies are urgently needed. Histone deacetylases (HDACs) are enzymes that participate in balancing histone acetylation activity for chromatin remodeling and gene transcription regulation. Increasing evidence suggests that the HDAC family is linked to the development and progression of chronic fibrotic diseases, including IPF. This review aims to summarize available information on HDACs and related inhibitors and their potential applications in treating IPF. In the future, HDACs may serve as novel targets, which can aid in understanding the etiology of PF, and selective inhibition of single HDACs or disruption of HDAC genes may serve as a strategy for treating PF.
Collapse
Affiliation(s)
- Hai-peng Cheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China
| | - Shi-he Jiang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China
| | - Jin Cai
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China
| | - Zi-qiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, Hunan, China
| | - Xiao-hong Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China
| | - Dan-dan Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
20
|
Ma R, Huang X, Sun D, Wang J, Xue C, Ye Q. Tetrandrine Alleviates Silica-induced Pulmonary Fibrosis Through PI3K/AKT Pathway: Network Pharmacology Investigation and Experimental Validation. Inflammation 2024; 47:1109-1126. [PMID: 38265677 DOI: 10.1007/s10753-023-01964-6] [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: 11/16/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/25/2024]
Abstract
Tetrandrine (TET) is a bisbenzylisoquinoline alkaloid derived from Stephania tetrandra S. Moor, known for its potential use in attenuating the progression of silicosis. However, the precise effects and underlying mechanisms of TET remain controversial. In this study, we aimed to elucidate the pharmacological mechanism of TET using a network pharmacology approach, while also evaluating its effect on silica-induced lung fibrosis in mice and TGF-β1-stimulated pulmonary fibroblasts in vitro. We employed network pharmacology to unravel the biological mechanisms through which TET may exert its therapeutic effects on pulmonary fibrosis and silicosis. In a silica-induced mouse model of lung fibrosis, TET was administered orally either during the early or late stage of fibrotic progression. Additionally, we examined the effects of TET on fibroblasts stimulated by TGF-β1 in vitro. Through the analysis, we identified a total of 101 targets of TET, 7,851 genes associated with pulmonary fibrosis, and 80 overlapping genes. These genes were primarily associated with key pathways such as epidermal growth factor receptor tyrosine kinase inhibitor resistance, the vascular endothelial growth factor signaling pathway, and the phosphatidylinositol 3 kinase (PI3K)-protein kinase B (PKB or AKT) signaling pathway. Furthermore, molecular docking analysis revealed the binding of TET to AKT1, the catalytic subunit of phosphatidylinositol-3 kinase, and KDR. In vivo experiments demonstrated that TET significantly alleviated silica-induced pulmonary fibrosis and reduced the expression of fibrotic markers. Moreover, TET exhibited inhibitory effects on the migration, proliferation, and differentiation of TGF-β1-induced lung fibroblasts in vitro. Notably, TET mitigated silica-induced pulmonary fibrosis by suppressing the PI3K/AKT pathway. In conclusion, our findings suggest that TET possesses the ability to suppress silica-induced pulmonary fibrosis by targeting the PI3K/AKT signaling pathway. These results provide valuable insights into the therapeutic potential of TET in the treatment of pulmonary fibrosis and silicosis.
Collapse
Affiliation(s)
- Ruimin Ma
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chao-Yang District, Beijing, China
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaoxi Huang
- Medical Research Center, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Di Sun
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chao-Yang District, Beijing, China
| | - Jingwei Wang
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chao-Yang District, Beijing, China
| | - Changjiang Xue
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chao-Yang District, Beijing, China
| | - Qiao Ye
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chao-Yang District, Beijing, China.
| |
Collapse
|
21
|
Pan D, Di X, Yan B, Su X. Advances in the Study of Non-Coding RNA in the Signaling Pathway of Pulmonary Fibrosis. Int J Gen Med 2024; 17:1419-1431. [PMID: 38617054 PMCID: PMC11016256 DOI: 10.2147/ijgm.s455707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/24/2024] [Indexed: 04/16/2024] Open
Abstract
Pulmonary fibrosis is a group of chronic, progressive, and irreversible interstitial lung diseases, which are common to most end-stage lung diseases and are one of the most difficult diseases of the respiratory system. In recent years, due to the frequent occurrence of air pollution and smog, the incidence of pulmonary fibrosis in China has increased year by year, the morbidity and mortality rates of pulmonary fibrosis have gradually increased and the age of the disease tends to be younger. However, the pathogenesis of pulmonary fibrosis is not yet fully understood and is needed to further explore new drug targets. Studies have shown that non-coding RNAs play an important role in regulating the process of pulmonary fibrosis, non-coding RNAs and their specifically expressed can promote or inhibit the process. Here, we review the role of some in the regulation of pulmonary fibrosis signaling pathways and provide new ideas for the clinical diagnosis and treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Dengyun Pan
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| | - Xin Di
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| | - Bingdi Yan
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| | - Xiaomin Su
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
22
|
Guan D, Men Y, Bartlett A, Hernández MAS, Xu J, Yi X, Li HS, Kong D, Mazitschek R, Ozcan U. Central inhibition of HDAC6 re-sensitizes leptin signaling during obesity to induce profound weight loss. Cell Metab 2024; 36:857-876.e10. [PMID: 38569472 DOI: 10.1016/j.cmet.2024.02.007] [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: 06/22/2023] [Revised: 11/02/2023] [Accepted: 02/13/2024] [Indexed: 04/05/2024]
Abstract
Leptin resistance during excess weight gain significantly contributes to the recidivism of obesity to leptin-based pharmacological therapies. The mechanisms underlying the inhibition of leptin receptor (LepR) signaling during obesity are still elusive. Here, we report that histone deacetylase 6 (HDAC6) interacts with LepR, reducing the latter's activity, and that pharmacological inhibition of HDAC6 activity disrupts this interaction and augments leptin signaling. Treatment of diet-induced obese mice with blood-brain barrier (BBB)-permeable HDAC6 inhibitors profoundly reduces food intake and leads to potent weight loss without affecting the muscle mass. Genetic depletion of Hdac6 in Agouti-related protein (AgRP)-expressing neurons or administration with BBB-impermeable HDAC6 inhibitors results in a lack of such anti-obesity effect. Together, these findings represent the first report describing a mechanistically validated and pharmaceutically tractable therapeutic approach to directly increase LepR activity as well as identifying centrally but not peripherally acting HDAC6 inhibitors as potent leptin sensitizers and anti-obesity agents.
Collapse
Affiliation(s)
- Dongxian Guan
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yuqin Men
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander Bartlett
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Jie Xu
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xinchi Yi
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hu-Song Li
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dong Kong
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ralph Mazitschek
- Massachusetts General Hospital, Center for Systems Biology, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Umut Ozcan
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
Luo L, Zhang W, You S, Cui X, Tu H, Yi Q, Wu J, Liu O. The role of epithelial cells in fibrosis: Mechanisms and treatment. Pharmacol Res 2024; 202:107144. [PMID: 38484858 DOI: 10.1016/j.phrs.2024.107144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/19/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Fibrosis is a pathological process that affects multiple organs and is considered one of the major causes of morbidity and mortality in multiple diseases, resulting in an enormous disease burden. Current studies have focused on fibroblasts and myofibroblasts, which directly lead to imbalance in generation and degradation of extracellular matrix (ECM). In recent years, an increasing number of studies have focused on the role of epithelial cells in fibrosis. In some cases, epithelial cells are first exposed to external physicochemical stimuli that may directly drive collagen accumulation in the mesenchyme. In other cases, the source of stimulation is mainly immune cells and some cytokines, and epithelial cells are similarly altered in the process. In this review, we will focus on the multiple dynamic alterations involved in epithelial cells after injury and during fibrogenesis, discuss the association among them, and summarize some therapies targeting changed epithelial cells. Especially, epithelial mesenchymal transition (EMT) is the key central step, which is closely linked to other biological behaviors. Meanwhile, we think studies on disruption of epithelial barrier, epithelial cell death and altered basal stem cell populations and stemness in fibrosis are not appreciated. We believe that therapies targeted epithelial cells can prevent the progress of fibrosis, but not reverse it. The epithelial cell targeting therapies will provide a wonderful preventive and delaying action.
Collapse
Affiliation(s)
- Liuyi Luo
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Wei Zhang
- Department of Oral Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Siyao You
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Xinyan Cui
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Hua Tu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Qiao Yi
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Jianjun Wu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| | - Ousheng Liu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
24
|
Yu H, Liu S, Wang S, Gu X. A narrative review of the role of HDAC6 in idiopathic pulmonary fibrosis. J Thorac Dis 2024; 16:688-695. [PMID: 38410580 PMCID: PMC10894383 DOI: 10.21037/jtd-23-1183] [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: 07/29/2023] [Accepted: 11/17/2023] [Indexed: 02/28/2024]
Abstract
Background and Objective Idiopathic pulmonary fibrosis (IPF) is a progressive and irreversible condition characterized by the deposition of extracellular matrix resulting from repetitive damage to the alveolar epithelium. These injuries, along with dysregulated wound repair and fibroblast dysfunction, lead to continuous tissue remodeling and fibrosis, eventually resulting in end-stage pulmonary fibrosis. Currently, there is no specific pharmacological treatment available for IPF. The role of inflammation in the development of IPF is still a topic of debate, and it is sometimes considered incidental to fibrosis. Over the past decade, macrophages have emerged as significant contributors to the pathogenesis of fibrosis. M1 macrophages are responsible for wound healing following alveolar epithelial injury, while M2 macrophages are involved in resolving wound repair and terminating the inflammatory response in the lungs. Various studies provide evidence that M2-like macrophages contribute to the abnormal fibrogenesis. In recent years, there has been growing interest in understanding macrophage polarization and its role in the development of pulmonary fibrosis. Histone deacetylase 6 (HDAC6), a member of the HDAC family with two functional deacetylase structural domains and a ubiquitin-binding zinc finger structural domain (ZnF-BUZ), plays a crucial role in pulmonary fibrosis. This article explores the role of HDAC6 in pulmonary fibrosis and evaluates its potential as a treatment approach for IPF. Methods PubMed, Cochrane Library, China National Knowledge Infrastructure (CNKI), Wanfang, China Biomedical Literature Service System (CBMdisc) and Web of Science were searched to obtain researches, published in English and Chinese, until July 2023. The search was performed using specific keywords such as Histone deacetylase 6, HDAC6, Idiopathic pulmonary fibrosis, IPF, fibrosis. Key Content and Findings HDAC6 has diverse effects on physiological processes, including the NLRP3 inflammasome, epithelial-mesenchymal transition, the TGFβ-PI3K-AKT pathway, macrophage polarization and TGF-β-Smad signaling pathway, due to its unique structure. HDAC6 has been found to enhance the inflammatory response and fibrosis of lung tissues, contributing to the development of IPF. Conclusions In the future, HDAC6 inhibitors are expected to play a crucial role in the treatment of fibrotic disorders and should be studied further deserves to pursue in future research.
Collapse
Affiliation(s)
- Hanming Yu
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shi Liu
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuo Wang
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiu Gu
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
25
|
Yang PY, Fang CY, Cho SC, Lee SP, Liao HY, Liao YW, Yu CC, Huang PH. Targeting histone deacetylase 9 represses fibrogenic phenotypes in buccal mucosal fibroblasts with arecoline stimulation. J Dent Sci 2024; 19:79-85. [PMID: 38303807 PMCID: PMC10829613 DOI: 10.1016/j.jds.2023.05.029] [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: 04/17/2023] [Revised: 05/22/2023] [Indexed: 02/03/2024] Open
Abstract
Background/purpose Oral submucosal fibrosis (OSF) is a premalignant disorder positively associated with betel nut chewing. Recent studies supported the promising benefits of histone deacetylase (HDAC) inhibitors for fibrosis treatment. Here we aim to clarify the pro-fibrogenic role of HDAC9 in regulating OSF. Materials and methods Healthy and OSF specimens were collected to investigate the clinical significance of HDAC9. Chronic arecoline treatment process was used to induce arecoline-mediated myofibroblasts-related activation of primary buccal mucosa fibroblasts (BMFs). Functional analysis of collagen gel contraction, transwell migration, and wound-healing assays were performed to assess the change in pro-fibrogenic properties of BMFs and fibrotic BMFs (fBMFs). Lentiviral-mediated HDAC9 knockdown was used to verify the role of HDAC9 in the pro-fibrogenic process. Results We found that arecoline significantly increased the mRNA and protein expression of HDAC9 of BMFs in a dose-dependent manner. Knockdown of HDAC9 in BMFs reversed the strengthened effects of arecoline on collagen gel contraction, cell migration, and wound-healing ability. We further demonstrated that knockdown of HDAC9 in fBMFs significantly attenuated its inherent pro-fibrogenic properties. Furthermore, we confirmed a significantly increased expression of HDAC9 mRNA in OSF compared to normal tissues, which suggested a positive correlation between the up-regulation of HDAC9 and OSF. Conclusion We demonstrated that silencing of HDAC9 inhibited arecoline-induced activation and inherent pro-fibrogenic properties, suggesting potential therapeutics by targeting HDAC9 in the OSF treatment.
Collapse
Affiliation(s)
- Po-Yu Yang
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chih-Yuan Fang
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Chi Cho
- Department of Medical Research and Education, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Shiao-Pieng Lee
- School of Dentistry, Department of Dentistry of Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Heng-Yi Liao
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Wen Liao
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Cheng-Chia Yu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Pao-Hsien Huang
- Department of Dentistry, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| |
Collapse
|
26
|
Park SJ, Lee N, Jeong CH. ACY-241, a histone deacetylase 6 inhibitor, suppresses the epithelial-mesenchymal transition in lung cancer cells by downregulating hypoxia-inducible factor-1 alpha. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:83-91. [PMID: 38154967 PMCID: PMC10762487 DOI: 10.4196/kjpp.2024.28.1.83] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/30/2023]
Abstract
Hypoxia-inducible factor-1 alpha (HIF-1α) is a transcription factor activated under hypoxic conditions, and it plays a crucial role in cellular stress regulation. While HIF-1α activity is essential in normal tissues, its presence in the tumor microenvironment represents a significant risk factor as it can induce angiogenesis and confer resistance to anti-cancer drugs, thereby contributing to poor prognoses. Typically, HIF-1α undergoes rapid degradation in normoxic conditions via oxygen-dependent degradation mechanisms. However, certain cancer cells can express HIF-1α even under normoxia. In this study, we observed an inclination toward increased normoxic HIF-1α expression in cancer cell lines exhibiting increased HDAC6 expression, which prompted the hypothesis that HDAC6 may modulate HIF-1α stability in normoxic conditions. To prove this hypothesis, several cancer cells with relatively higher HIF-1α levels under normoxic conditions were treated with ACY-241, a selective HDAC6 inhibitor, and small interfering RNAs for HDAC6 knockdown. Our data revealed a significant reduction in HIF-1α expression upon HDAC6 inhibition. Moreover, the downregulation of HIF-1α under normoxic conditions decreased zinc finger E-box-binding homeobox 1 expression and increased E-cadherin levels in lung cancer H1975 cells, consequently suppressing cell invasion and migration. ACY-241 treatment also demonstrated an inhibitory effect on cell invasion and migration by reducing HIF-1α level. This study confirms that HDAC6 knockdown and ACY-241 treatment effectively decrease HIF-1α expression under normoxia, thereby suppressing the epithelial-mesenchymal transition. These findings highlight the potential of selective HDAC6 inhibition as an innovative therapeutic strategy for lung cancer.
Collapse
Affiliation(s)
- Seong-Jun Park
- College of Pharmacy, Keimyung University, Daegu 42601, Korea
| | - Naeun Lee
- College of Pharmacy, Keimyung University, Daegu 42601, Korea
| | - Chul-Ho Jeong
- College of Pharmacy, Keimyung University, Daegu 42601, Korea
| |
Collapse
|
27
|
Wu K, Huang S, Zheng F, Liu Y. The CEBPA-FGF21 regulatory network may participate in the T2DM-induced skeletal muscle atrophy by regulating the autophagy-lysosomal pathway. Acta Diabetol 2023; 60:1491-1503. [PMID: 37392202 DOI: 10.1007/s00592-023-02131-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/05/2023] [Indexed: 07/03/2023]
Abstract
AIMS Recent years have witnessed an increasing research interest in the roles of transcription factor (TF)-gene regulatory network in type 2 diabetes mellitus (T2DM). Thus, we sought to characterize the mechanistic insights based on the TF-gene regulatory network in skeletal muscle atrophy in T2DM. METHODS Differentially expressed TFs (DETFs) and mRNAs (DEmRNAs) were obtained in T2DM-related gene expression profiles (GSE12643, GSE55650, GSE166502, and GSE29221), followed by WGCNA, and GO and KEGG enrichment analyses. Next, the iRegulon plug-in unit of Cytoscape software was used to construct a TF-mRNA regulatory network. Besides, RT-qPCR and ChIP-seq were utilized to measure the expression of CEBPA and FGF21 in the skeletal muscle tissues or cells of T2DM rat models. At last, the effect of overexpression of FGF21 on the autophagy-lysosomal pathway was examined in skeletal muscle cells of T2DM rats. RESULTS Totally, 12 DETFs and 102 DEmRNAs were found in the skeletal muscle tissues of T2DM samples. The DEmRNAs were mainly enriched in the autophagy-lysosomal pathway. CEBPA affected the skeletal muscle atrophy in T2DM by regulating 5 target genes via the autophagy-lysosomal pathway. CEBPA could target FGF21. In addition, the expression of CEBPA was elevated, while the expression of FGF21 was diminished in the skeletal muscle tissues or cells of T2DM rats. The CEBPA-FGF21 regulatory network promoted skeletal muscle atrophy in T2DM by activating the autophagy-lysosomal pathway. CONCLUSION The CEBPA-FGF21 regulatory network may participate in the T2DM-induced skeletal muscle atrophy by regulating the autophagy-lysosomal pathway. Thus, our study provides interesting targets for prevention of skeletal muscle atrophy in T2DM.
Collapse
Affiliation(s)
- Kai Wu
- Department of Physical Medicine and Rehabilitation, Xiangya Hospital of Central South University, No. 87, Xiang-Ya Road, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Sha Huang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Fan Zheng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
- Health Management Department, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Yuan Liu
- Department of Physical Medicine and Rehabilitation, Xiangya Hospital of Central South University, No. 87, Xiang-Ya Road, Changsha, 410008, Hunan Province, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China.
| |
Collapse
|
28
|
Zhang YS, Tu B, Song K, Lin LC, Liu ZY, Lu D, Chen Q, Tao H. Epigenetic hallmarks in pulmonary fibrosis: New advances and perspectives. Cell Signal 2023; 110:110842. [PMID: 37544633 DOI: 10.1016/j.cellsig.2023.110842] [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: 06/07/2023] [Revised: 07/25/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Epigenetics indicates that certain phenotypes of an organism can undergo heritable changes in the absence of changes in the genetic DNA sequence. Many studies have shown that epigenetic patterns play an important role in the lung and lung diseases. Pulmonary fibrosis (PF) is also a type of lung disease. PF is an end-stage change of a large group of lung diseases, characterized by fibroblast proliferation and massive accumulation of extracellular matrix, accompanied by inflammatory injury and histological destruction, that is, structural abnormalities caused by abnormal repair of normal alveolar tissue. It causes loss of lung function in patients with multiple complex diseases, leading to respiratory failure and subsequent death. However, current treatment options for IPF are very limited and no drugs have been shown to significantly prolong the survival of patients. Therefore, based on a systematic understanding of the disease mechanisms of PF, this review integrates the role of epigenetics in the development and course of PF, describes preventive and potential therapeutic targets for PF, and provides a theoretical basis for further exploration of the mechanisms of PF.
Collapse
Affiliation(s)
- Yun-Sen Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Bin Tu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Kai Song
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Dong Lu
- Department of Interventional Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China.
| | - Qi Chen
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| |
Collapse
|
29
|
Li J, Wei Q, Song K, Wang Y, Yang Y, Li M, Yu J, Su G, Peng L, Fu B, Yi P. Tangeretin attenuates bleomycin-induced pulmonary fibrosis by inhibiting epithelial-mesenchymal transition via the PI3K/Akt pathway. Front Pharmacol 2023; 14:1247800. [PMID: 37781713 PMCID: PMC10540689 DOI: 10.3389/fphar.2023.1247800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/06/2023] [Indexed: 10/03/2023] Open
Abstract
Background: Pulmonary fibrosis (PF) is a terminal pathological change in a variety of lung diseases characterized by excessive deposition of extracellular matrix, for which effective treatment is lacking. Tangeretin (Tan), a flavonoid derived from citrus, has been shown to have a wide range of pharmacological effects. This study aimed to investigate the role and potential mechanisms of Tan on pulmonary fibrosis. Methods: A model of pulmonary fibrosis was established by administering bleomycin through tracheal drip, followed by administering Tan or pirfenidone through gavage. HE and Masson staining were employed to assess the extent of pulmonary fibrosis. Subsequently, Western blot, enzyme-linked immunosorbent assay (ELISA), RNA sequencing, and immunohistochemistry techniques were employed to uncover the protective mechanism of Tan in PF mice. Furthermore, A549 cells were stimulated with TGF-β1 to induce epithelial-mesenchymal transition (EMT) and demonstrate the effectiveness of Tan in mitigating PF. Results: Tan significantly ameliorated bleomycin-induced pulmonary fibrosis, improved fibrotic pathological changes, and collagen deposition in the lungs, and reduced lung inflammation and oxidative stress. The KEGG pathway enrichment analysis revealed a higher number of enriched genes in the PI3K/Akt pathway. Additionally, Tan can inhibit the EMT process related to pulmonary fibrosis. Conclusion: Taken together, the above research results indicate that Tan suppresses inflammation, oxidative stress, and EMT in BLM-induced pulmonary fibrosis via the PI3K/Akt pathway and is a potential agent for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Jiang Li
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qian Wei
- Department of Internal Medicine-Cardiovascular, The Third Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Ke Song
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Youxin Wang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuxin Yang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Miao Li
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jiaying Yu
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Guangxu Su
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Luyuan Peng
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Bendong Fu
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Pengfei Yi
- College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
30
|
Yu WC, Yeh TY, Ye CH, Chong PCT, Ho YH, So DK, Yap KY, Peng GR, Shao CH, Jagtap AD, Chern JW, Lin CS, Lin SP, Lin SL, Yu SH, Yu CW. Discovery of HDAC6, HDAC8, and 6/8 Inhibitors and Development of Cell-Based Drug Screening Models for the Treatment of TGF-β-Induced Idiopathic Pulmonary Fibrosis. J Med Chem 2023; 66:10528-10557. [PMID: 37463500 DOI: 10.1021/acs.jmedchem.3c00644] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Idiopathic pulmonary fibrosis is incurable, and its progression is difficult to control and thus can lead to pulmonary deterioration. Pan-histone deacetylase inhibitors such as SAHA have shown potential for modulating pulmonary fibrosis yet with off-target effects. Therefore, selective HDAC inhibitors would be beneficial for reducing side effects. Toward this goal, we designed and synthesized 24 novel HDAC6, HDAC8, or dual HDAC6/8 inhibitors and established a two-stage screening platform to rapidly screen for HDAC inhibitors that effectively mitigate TGF-β-induced pulmonary fibrosis. The first stage consisted of a mouse NIH-3T3 fibroblast prescreen and yielded five hits. In the second stage, human pulmonary fibroblasts (HPFs) were used, and four out of the five hits were tested for caco-2 permeability and liver microsome stability to give two potential leads: J27644 (15) and 20. This novel two-stage screen platform will accelerate the discovery and reduce the cost of developing HDAC inhibitors to mitigate TGF-β-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Wei-Chieh Yu
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan
| | - Tsung-Yu Yeh
- National Taiwan University, School of Pharmacy, College of Medicine, Taipei 100, Taiwan
| | - Chih-Hung Ye
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan
| | | | - Yi-Hsun Ho
- National Taiwan University, School of Pharmacy, College of Medicine, Taipei 100, Taiwan
| | - Dorothy Kazuno So
- Institute of Biotechnology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei 106, Taiwan
| | - Kah Yi Yap
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan
| | - Guan-Ru Peng
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan
| | - Chi-Hsuan Shao
- National Taiwan University, School of Pharmacy, College of Medicine, Taipei 100, Taiwan
| | - Ajit Dhananjay Jagtap
- National Taiwan University, School of Pharmacy, College of Medicine, Taipei 100, Taiwan
| | - Ji-Wang Chern
- National Taiwan University, School of Pharmacy, College of Medicine, Taipei 100, Taiwan
| | - Chen-Si Lin
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan
| | - Shau-Ping Lin
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan
- Center of Systems Biology, National Taiwan University, Taipei 106, Taiwan
- The Research Center of Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei 106, Taiwan
| | - Shuei-Liong Lin
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
- Department of Integrated Diagnostics & Therapeutics, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Shu-Han Yu
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan
| | - Chao-Wu Yu
- National Taiwan University, School of Pharmacy, College of Medicine, Taipei 100, Taiwan
| |
Collapse
|
31
|
Zhang QQ, Zhang WJ, Chang S. HDAC6 inhibition: a significant potential regulator and therapeutic option to translate into clinical practice in renal transplantation. Front Immunol 2023; 14:1168848. [PMID: 37545520 PMCID: PMC10401441 DOI: 10.3389/fimmu.2023.1168848] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 05/30/2023] [Indexed: 08/08/2023] Open
Abstract
Histone deacetylase 6 (HDAC6), an almost exclusively cytoplasmic enzyme, plays an essential role in many biological processes and exerts its deacetylation-dependent/independent effects on a variety of target molecules, which has contributed to the flourishing growth of relatively isoform-specific enzyme inhibitors. Renal transplantation (RT) is one of the alternatively preferred treatments and the most cost-effective treatment approaches for the great majority of patients with end-stage renal disease (ESRD). HDAC6 expression and activity have recently been shown to be increased in kidney disease in a number of studies. To date, a substantial amount of validated studies has identified HDAC6 as a pivotal modulator of innate and adaptive immunity, and HDAC6 inhibitors (HDAC6i) are being developed and investigated for use in arrays of immune-related diseases, making HDAC6i a promising therapeutic candidate for the management of a variety of renal diseases. Based on accumulating evidence, HDAC6i markedly open up new avenues for therapeutic intervention to protect against oxidative stress-induced damage, tip the balance in favor of the generation of tolerance-related immune cells, and attenuate fibrosis by inhibiting multiple activations of cell profibrotic signaling pathways. Taken together, we have a point of view that targeting HDAC6 may be a novel approach for the therapeutic strategy of RT-related complications, including consequences of ischemia-reperfusion injury, induction of immune tolerance in transplantation, equilibrium of rejection, and improvement of chronic renal graft interstitial fibrosis after transplantation in patients. Herein, we will elaborate on the unique function of HDAC6, which focuses on therapeutical mechanism of action related to immunological events with a general account of the tantalizing potential to the clinic.
Collapse
Affiliation(s)
- Qian-qian Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Wei-jie Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Sheng Chang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
32
|
da Silva CO, de Souza Nogueira J, do Nascimento AP, Victoni T, Bártholo TP, da Costa CH, Costa AMA, Valença SDS, Schmidt M, Porto LC. COPD Patients Exhibit Distinct Gene Expression, Accelerated Cellular Aging, and Bias to M2 Macrophages. Int J Mol Sci 2023; 24:9913. [PMID: 37373058 DOI: 10.3390/ijms24129913] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
COPD, one of world's leading contributors to morbidity and mortality, is characterized by airflow limitation and heterogeneous clinical features. Three main phenotypes are proposed: overlapping asthma/COPD (ACO), exacerbator, and emphysema. Disease severity can be classified as mild, moderate, severe, and very severe. The molecular basis of inflammatory amplification, cellular aging, and immune response are critical to COPD pathogenesis. Our aim was to investigate EP300 (histone acetylase, HAT), HDAC 2 (histone deacetylase), HDAC3, and HDAC4 gene expression, telomere length, and differentiation ability to M1/M2 macrophages. For this investigation, 105 COPD patients, 42 smokers, and 73 non-smoker controls were evaluated. We identified a reduced HDAC2 expression in patients with mild, moderate, and severe severity; a reduced HDAC3 expression in patients with moderate and severe severity; an increased HDAC4 expression in patients with mild severity; and a reduced EP300 expression in patients with severe severity. Additionally, HDAC2 expression was reduced in patients with emphysema and exacerbator, along with a reduced HDAC3 expression in patients with emphysema. Surprisingly, smokers and all COPD patients showed telomere shortening. COPD patients showed a higher tendency toward M2 markers. Our data implicate genetic changes in COPD phenotypes and severity, in addition to M2 prevalence, that might influence future treatments and personalized therapies.
Collapse
Affiliation(s)
- Camila Oliveira da Silva
- Laboratory of Histocompatibility and Cryopreservation, University of the State of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil
| | - Jeane de Souza Nogueira
- Laboratory of Histocompatibility and Cryopreservation, University of the State of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil
| | | | - Tatiana Victoni
- VetAgro Sup, University of Lyon, APCSe, 69280 Marcy l'Étoile, France
| | - Thiago Prudente Bártholo
- Department of Thorax, University of the State of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil
| | | | - Andrea Monte Alto Costa
- Tissue Repair Laboratory, University of the State of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil
| | - Samuel Dos Santos Valença
- Laboratory of Redox Biology, ICB, Federal University of Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Luís Cristóvão Porto
- Laboratory of Histocompatibility and Cryopreservation, University of the State of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil
| |
Collapse
|
33
|
Sun HN, Ren CX, Lee DH, Wang WH, Guo XY, Hao YY, Wang XM, Zhang HN, Xiao WQ, Li N, Cong J, Han YH, Kwon T. PRDX1 negatively regulates bleomycin-induced pulmonary fibrosis via inhibiting the epithelial-mesenchymal transition and lung fibroblast proliferation in vitro and in vivo. Cell Mol Biol Lett 2023; 28:48. [PMID: 37268886 DOI: 10.1186/s11658-023-00460-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/12/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Pulmonary fibrosis is a major category of end-stage changes in lung diseases, characterized by lung epithelial cell damage, proliferation of fibroblasts, and accumulation of extracellular matrix. Peroxiredoxin 1 (PRDX1), a member of the peroxiredoxin protein family, participates in the regulation of the levels of reactive oxygen species in cells and various other physiological activities, as well as the occurrence and development of diseases by functioning as a chaperonin. METHODS Experimental methods including MTT assay, morphological observation of fibrosis, wound healing assay, fluorescence microscopy, flow cytometry, ELISA, western blot, transcriptome sequencing, and histopathological analysis were used in this study. RESULTS PRDX1 knockdown increased ROS levels in lung epithelial cells and promoted epithelial-mesenchymal transition (EMT) through the PI3K/Akt and JNK/Smad signalling pathways. PRDX1 knockout significantly increased TGF-β secretion, ROS production, and cell migration in primary lung fibroblasts. PRDX1 deficiency also increased cell proliferation, cell cycle circulation, and fibrosis progression through the PI3K/Akt and JNK/Smad signalling pathways. BLM treatment induced more severe pulmonary fibrosis in PRDX1-knockout mice, mainly through the PI3K/Akt and JNK/Smad signalling pathways. CONCLUSIONS Our findings strongly suggest that PRDX1 is a key molecule in BLM-induced lung fibrosis progression and acts through modulating EMT and lung fibroblast proliferation; therefore, it may be a therapeutic target for the treatment of BLM-induced lung fibrosis.
Collapse
Affiliation(s)
- Hu-Nan Sun
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China.
| | - Chen-Xi Ren
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Dong Hun Lee
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Wei-Hao Wang
- National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Xiao-Yu Guo
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Ying-Ying Hao
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Xiao-Ming Wang
- Yabian Academy of Agricultural Science, Longjing, Jilin, 1334000, China
| | - Hui-Na Zhang
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Wan-Qiu Xiao
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Nan Li
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Jie Cong
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China
| | - Ying-Hao Han
- Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing, 163319, Heilongjiang, China.
| | - Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 351-33 Neongme-Gil, Ibam-Myeon, Jeongeup-Si, Jeonbuk, 56216, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
34
|
Marzoog BA. Local Lung Fibroblast Autophagy in the Context of Lung Fibrosis Pathogenesis. CURRENT RESPIRATORY MEDICINE REVIEWS 2023; 19:6-11. [DOI: 10.2174/1573398x19666221130141600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 09/28/2022] [Accepted: 10/13/2022] [Indexed: 12/05/2022]
Abstract
Abstract:
The current molecular advances in lung fibrosis pathogenesis distend beyond the cellular to involve subcellular and molecular levels. Lung fibrogenesis and autophagy impairment are tight-ly associated. Autophagy is involved in cell cycle control and regulation of the intracellular micro-environment. Degradation of impaired intracellular organelles and biproducts is crucial to maintain-ing a healthy cell and preventing its metaplasia / transdifferentiation to a pathological cell. Autoph-agy modifies the metabolism of alveolar epithelial cells, endothelial cells, and lung fibroblasts. Au-tophagy upregulation induces local lung fibroblast hyperactivity and fibrosis. Several molecular triggers were found to induce lung fibroblast autophagy including TGFβ by inhibition of the PI3K/AKT/mTOR. However, physiologically, a balance is retained between autophagy inducers and inhibitors. Each type of autophagy plays its role in the initiation and progression of lung fibro-sis. The pathogenesis of pulmonary fibrosis is multifactorial and involves dysfunction / dysregula-tion of alveolar epithelial cells, fibroblasts, monocyte-derived macrophages, and endothelial cells. The deposition of extracellular matrix proteins, the remodeling of the lung architecture and the mo-lecular changes include impaired glycolysis, mitochondrial oxidation, gene expression modification, altered phospholipid and sphingolipid metabolism, and dysregulated protein folding lead to repro-gramming of lung fibroblast into myofibroblast and their activation. The paper thoroughly addresses the molecular triggers and inhibitors of lung fibroblast autophagy in lung fibrosis.
Collapse
Affiliation(s)
- Basheer Abdullah Marzoog
- Department of Medicine, National Research Mordovia State University, Bolshevitskaya Street-68, Saransk, Rep, Mordovia-
430005, Russia
| |
Collapse
|
35
|
Pharmacological inhibition of HDAC6 improves muscle phenotypes in dystrophin-deficient mice by downregulating TGF-β via Smad3 acetylation. Nat Commun 2022; 13:7108. [PMID: 36402791 PMCID: PMC9675748 DOI: 10.1038/s41467-022-34831-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 11/01/2022] [Indexed: 11/21/2022] Open
Abstract
The absence of dystrophin in Duchenne muscular dystrophy disrupts the dystrophin-associated glycoprotein complex resulting in skeletal muscle fiber fragility and atrophy, associated with fibrosis as well as microtubule and neuromuscular junction disorganization. The specific, non-conventional cytoplasmic histone deacetylase 6 (HDAC6) was recently shown to regulate acetylcholine receptor distribution and muscle atrophy. Here, we report that administration of the HDAC6 selective inhibitor tubastatin A to the Duchenne muscular dystrophy, mdx mouse model increases muscle strength, improves microtubule, neuromuscular junction, and dystrophin-associated glycoprotein complex organization, and reduces muscle atrophy and fibrosis. Interestingly, we found that the beneficial effects of HDAC6 inhibition involve the downregulation of transforming growth factor beta signaling. By increasing Smad3 acetylation in the cytoplasm, HDAC6 inhibition reduces Smad2/3 phosphorylation, nuclear translocation, and transcriptional activity. These findings provide in vivo evidence that Smad3 is a new target of HDAC6 and implicate HDAC6 as a potential therapeutic target in Duchenne muscular dystrophy.
Collapse
|
36
|
Nie H, Liu H, Shi Y, Lai W, Liu X, Xi Z, Lin B. Effects of Different Concentrations of Oil Mist Particulate Matter on Pulmonary Fibrosis In Vivo and In Vitro. TOXICS 2022; 10:647. [PMID: 36355939 PMCID: PMC9695344 DOI: 10.3390/toxics10110647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
Oil-mist particulate matter (OMPM) refers to oily particles with a small aerodynamic equivalent diameter in ambient air. Since the pathogenesis of pulmonary fibrosis (PF) has not been fully elucidated, this study aims to explore the potential molecular mechanisms of the adverse effects of exposure to OMPM at different concentrations in vivo and in vitro on PF. In this study, rats and cell lines were treated with different concentrations of OMPM in vivo and in vitro. Sirius Red staining analysis shows that OMPM exposure could cause pulmonary lesions and fibrosis symptoms. The expression of TGF-β1, α-SMA, and collagen I was increased in the lung tissue of rats. The activities of MMP2 and TIMP1 were unbalanced, and increased N-Cadherin and decreased E-Cadherin upon OMPM exposure in a dose-dependent manner. In addition, OMPM exposure could activate the TGF-β1/Smad3 and TGF-β1/MAPK p38 signaling pathways, and the differentiation of human lung fibroblast HFL-1 cells. Therefore, OMPM exposure could induce PF by targeting the lung epithelium and fibroblasts, and activating the TGF-β1/Smad3 and TGF-β1/MAPK p38 signaling pathways.
Collapse
|
37
|
Sehgal M, Jakhete SM, Manekar AG, Sasikumar S. Specific epigenetic regulators serve as potential therapeutic targets in idiopathic pulmonary fibrosis. Heliyon 2022; 8:e09773. [PMID: 36061031 PMCID: PMC9434059 DOI: 10.1016/j.heliyon.2022.e09773] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/27/2022] [Accepted: 06/17/2022] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF), a disorder observed mostly in older human beings, is characterised by chronic and progressive lung scarring leading to an irreversible decline in lung function. This health condition has a dismal prognosis and the currently available drugs only delay but fail to reverse the progression of lung damage. Consequently, it becomes imperative to discover improved therapeutic compounds and their cellular targets to cure IPF. In this regard, a number of recent studies have targeted the epigenetic regulation by histone deacetylases (HDACs) to develop and categorise antifibrotic drugs for lungs. Therefore, this review focuses on how aberrant expression or activity of Classes I, II and III HDACs alter TGF-β signalling to promote events such as epithelial-mesenchymal transition, differentiation of activated fibroblasts into myofibroblasts, and excess deposition of the extracellular matrix to propel lung fibrosis. Further, this study describes how certain chemical compounds or dietary changes modulate dysregulated HDACs to attenuate five faulty TGF-β-dependent profibrotic processes, both in animal models and cell lines replicating IPF, thereby identifying promising means to treat this lung disorder.
Collapse
Affiliation(s)
- Manas Sehgal
- Genetics and Molecular Biology Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, PIN - 411033, India
| | - Sharayu Manish Jakhete
- Genetics and Molecular Biology Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, PIN - 411033, India
| | - Amruta Ganesh Manekar
- Genetics and Molecular Biology Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, PIN - 411033, India
| | - Satish Sasikumar
- Genetics and Molecular Biology Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune, Maharashtra, PIN - 411033, India
| |
Collapse
|
38
|
Ma H, Wu X, Li Y, Xia Y. Research Progress in the Molecular Mechanisms, Therapeutic Targets, and Drug Development of Idiopathic Pulmonary Fibrosis. Front Pharmacol 2022; 13:963054. [PMID: 35935869 PMCID: PMC9349351 DOI: 10.3389/fphar.2022.963054] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/24/2022] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal interstitial lung disease. Recent studies have identified the key role of crosstalk between dysregulated epithelial cells, mesenchymal, immune, and endothelial cells in IPF. In addition, genetic mutations and environmental factors (e.g., smoking) have also been associated with the development of IPF. With the recent development of sequencing technology, epigenetics, as an intermediate link between gene expression and environmental impacts, has also been reported to be implicated in pulmonary fibrosis. Although the etiology of IPF is unknown, many novel therapeutic targets and agents have emerged from clinical trials for IPF treatment in the past years, and the successful launch of pirfenidone and nintedanib has demonstrated the promising future of anti-IPF therapy. Therefore, we aimed to gain an in-depth understanding of the underlying molecular mechanisms and pathogenic factors of IPF, which would be helpful for the diagnosis of IPF, the development of anti-fibrotic drugs, and improving the prognosis of patients with IPF. In this study, we summarized the pathogenic mechanism, therapeutic targets and clinical trials from the perspective of multiple cell types, gene mutations, epigenetic and environmental factors.
Collapse
Affiliation(s)
- Hongbo Ma
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xuyi Wu
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
| | - Yi Li
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
| | - Yong Xia
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
- *Correspondence: Yong Xia,
| |
Collapse
|
39
|
Xu Y, Wang X, Han D, Wang J, Luo Z, Jin T, Shi C, Zhou X, Lin L, Shan J. Revealing the mechanism of Jiegeng decoction attenuates bleomycin-induced pulmonary fibrosis via PI3K/Akt signaling pathway based on lipidomics and transcriptomics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 102:154207. [PMID: 35660351 DOI: 10.1016/j.phymed.2022.154207] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a serious lung disease with unknown etiology and irreversible course. Jiegeng decoction (JGD), a traditional prescription, is widely used to treat lung diseases due to its anti-inflammatory and expectorant effects. PURPOSE To explore the effect of JGD on mice with PF and its underlying mechanism. For this purpose, we established a mouse model with PF by bleomycin (BLM) and then administered JGD and pirfenidone at different concentrations. RESULTS In vivo, JGD was found to reduce lung inflammation, improve lung function and decrease collagen deposition to alleviate bleomycin-induced PF in mice. The mouse lung tissue was analyzed using lipidomics and transcriptomics. We found phosphatidylinositol was decreased after JGD treatment in lipidomics results, while transcriptomics results showed the critical roles of PI3K/Akt signaling pathway in JGD treatment group. Then, Western Blot and Immunohistochemistry were used to validate that JGD may regulate the expression of Bax, Caspase3, Caspase8, Caspase9 and Bcl-2 apoptosis-related proteins via PI3K/Akt signaling pathway. TUNEL staining revealed that apoptosis mainly occurs on AEC IIs. CONCLUSION Our results showed that JGD inhibits apoptosis through the PI3K/Akt signaling pathway, thereby protecting against BLM-induced PF. Hence, JGD is expected to be a potential drug candidate for the treatment of PF.
Collapse
Affiliation(s)
- Yong Xu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuan Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Di Han
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Junyi Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zichen Luo
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tianzi Jin
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chen Shi
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xianmei Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Lili Lin
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
40
|
Korfei M, Mahavadi P, Guenther A. Targeting Histone Deacetylases in Idiopathic Pulmonary Fibrosis: A Future Therapeutic Option. Cells 2022; 11:1626. [PMID: 35626663 PMCID: PMC9139813 DOI: 10.3390/cells11101626] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 02/07/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease with limited therapeutic options, and there is a huge unmet need for new therapies. A growing body of evidence suggests that the histone deacetylase (HDAC) family of transcriptional corepressors has emerged as crucial mediators of IPF pathogenesis. HDACs deacetylate histones and result in chromatin condensation and epigenetic repression of gene transcription. HDACs also catalyse the deacetylation of many non-histone proteins, including transcription factors, thus also leading to changes in the transcriptome and cellular signalling. Increased HDAC expression is associated with cell proliferation, cell growth and anti-apoptosis and is, thus, a salient feature of many cancers. In IPF, induction and abnormal upregulation of Class I and Class II HDAC enzymes in myofibroblast foci, as well as aberrant bronchiolar epithelium, is an eminent observation, whereas type-II alveolar epithelial cells (AECII) of IPF lungs indicate a significant depletion of many HDACs. We thus suggest that the significant imbalance of HDAC activity in IPF lungs, with a "cancer-like" increase in fibroblastic and bronchial cells versus a lack in AECII, promotes and perpetuates fibrosis. This review focuses on the mechanisms by which Class I and Class II HDACs mediate fibrogenesis and on the mechanisms by which various HDAC inhibitors reverse the deregulated epigenetic responses in IPF, supporting HDAC inhibition as promising IPF therapy.
Collapse
Affiliation(s)
- Martina Korfei
- Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, D-35392 Giessen, Germany; (P.M.); (A.G.)
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), D-35392 Giessen, Germany
| | - Poornima Mahavadi
- Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, D-35392 Giessen, Germany; (P.M.); (A.G.)
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), D-35392 Giessen, Germany
| | - Andreas Guenther
- Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, D-35392 Giessen, Germany; (P.M.); (A.G.)
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), D-35392 Giessen, Germany
- Lung Clinic, Evangelisches Krankenhaus Mittelhessen, D-35398 Giessen, Germany
- European IPF Registry and Biobank, D-35392 Giessen, Germany
| |
Collapse
|
41
|
Liu L, Sun Q, Davis F, Mao J, Zhao H, Ma D. Epithelial-mesenchymal transition in organ fibrosis development: current understanding and treatment strategies. BURNS & TRAUMA 2022; 10:tkac011. [PMID: 35402628 PMCID: PMC8990740 DOI: 10.1093/burnst/tkac011] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/16/2021] [Indexed: 01/10/2023]
Abstract
Organ fibrosis is a process in which cellular homeostasis is disrupted and extracellular matrix is excessively deposited. Fibrosis can lead to vital organ failure and there are no effective treatments yet. Although epithelial–mesenchymal transition (EMT) may be one of the key cellular mechanisms, the underlying mechanisms of fibrosis remain largely unknown. EMT is a cell phenotypic process in which epithelial cells lose their cell-to-cell adhesion and polarization, after which they acquire mesenchymal features such as infiltration and migration ability. Upon injurious stimulation in different organs, EMT can be triggered by multiple signaling pathways and is also regulated by epigenetic mechanisms. This narrative review summarizes the current understanding of the underlying mechanisms of EMT in fibrogenesis and discusses potential strategies for attenuating EMT to prevent and/or inhibit fibrosis. Despite better understanding the role of EMT in fibrosis development, targeting EMT and beyond in developing therapeutics to tackle fibrosis is challenging but likely feasible.
Collapse
Affiliation(s)
- Lexin Liu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK.,Department of Nephrology and Urology, Pediatric Urolith Center, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province, 310003, China
| | - Qizhe Sun
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Frank Davis
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Jianhua Mao
- Department of Nephrology, The Children Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang Province, 310003, China
| | - Hailin Zhao
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| |
Collapse
|
42
|
Khezri MR, Moloodsouri D, Hodaei D, Ghasemnejad-Berenji M. Therapeutic potential of loureirin A against SARS-CoV-2 infection. Phytother Res 2022; 36:3011-3012. [PMID: 35355342 PMCID: PMC9110998 DOI: 10.1002/ptr.7453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/10/2022] [Accepted: 03/14/2022] [Indexed: 12/17/2022]
Affiliation(s)
- Mohammad Rafi Khezri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Donya Moloodsouri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Darya Hodaei
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Ghasemnejad-Berenji
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran.,Research Center for Experimental and Applied Pharmaceutical Sciences, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
43
|
Margaria JP, Moretta L, Alves-Filho JC, Hirsch E. PI3K Signaling in Mechanisms and Treatments of Pulmonary Fibrosis Following Sepsis and Acute Lung Injury. Biomedicines 2022; 10:756. [PMID: 35453505 PMCID: PMC9028704 DOI: 10.3390/biomedicines10040756] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/11/2022] [Accepted: 03/22/2022] [Indexed: 12/12/2022] Open
Abstract
Pulmonary fibrosis is a pathological fibrotic process affecting the lungs of five million people worldwide. The incidence rate will increase even more in the next years due to the long-COVID-19 syndrome, but a resolving treatment is not available yet and usually prognosis is poor. The emerging role of the phosphatidylinositol 3-kinase (PI3K)/AKT signaling in fibrotic processes has inspired the testing of drugs targeting the PI3K/Akt pathway that are currently under clinical evaluation. This review highlights the progress in understanding the role of PI3K/Akt in the development of lung fibrosis and its causative pathological context, including sepsis as well as acute lung injury (ALI) and its consequent acute respiratory distress syndrome (ARDS). We further summarize current knowledge about PI3K inhibitors for pulmonary fibrosis treatment, including drugs under development as well as in clinical trials. We finally discuss how the design of inhaled compounds targeting the PI3K pathways might potentiate efficacy and improve tolerability.
Collapse
Affiliation(s)
- Jean Piero Margaria
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126 Torino, Italy; (J.P.M.); (L.M.)
| | - Lucia Moretta
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126 Torino, Italy; (J.P.M.); (L.M.)
| | - Jose Carlos Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirao Preto 14049-900, Brazil;
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126 Torino, Italy; (J.P.M.); (L.M.)
| |
Collapse
|
44
|
Liu Y, Wang R, Han H, Li L. Tubastatin A suppresses the proliferation of fibroblasts in epidural fibrosis through phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signalling pathway. J Pharm Pharmacol 2022; 74:rgab106. [PMID: 35230444 DOI: 10.1093/jpp/rgab106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 02/21/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVES This study was designed to explore the effect of tubastatin A (Tub A) on epidural fibrosis and the underlying mechanism. METHODS Histone deacetylase 6 (HDAC6)-overexpressed fibroblasts were constructed, and the effect of Tub A on the proliferation of activated fibroblasts was detected by Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine (EdU) and cell cycle assay. Besides, 20 Sprague-Dawley rats were subjected to animal laminectomy model construction and then randomly treated with 4% dimethyl sulfoxide (DMSO) (diluted in 0.9% saline) or Tub A (10 mg/kg/day), separately. The expression of HDAC6 and phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway-related proteins was measured in epidural fibrosis tissues. KEY FINDINGS HDAC6 was overexpressed in activated fibroblasts and epidural scar tissues of rat models. Cell proliferation was remarkably elevated in HDAC6-overexpressed fibroblasts, which was reflected by cell viability, EdU and flow cytometry-based cell cycle assay, and paralleled with the increased expression of phosphorylated PI3K, AKT and mTOR, which was remarkably reversed following Tub A treatment. 740Y-P activator addition significantly reversed the declined fibroblast proliferation induced by Tub A. The expressions of PI3K/AKT/mTOR pathway-related proteins were also reduced in epidural tissues in rat models with Tub A treatment. CONCLUSION Tub A could prevent epidural fibrosis formation by inhibiting fibroblast proliferation through mediating PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Yang Liu
- Department of Spine Surgery, Weifang People's Hospital, Weifang, Shandong, China
| | - Ruihong Wang
- Department of Spine Surgery, Weifang People's Hospital, Weifang, Shandong, China
| | - Huimin Han
- Department of Spine Surgery, Weifang People's Hospital, Weifang, Shandong, China
| | - Li Li
- Department of Spine Surgery, Weifang People's Hospital, Weifang, Shandong, China
| |
Collapse
|
45
|
Barone S, Cassese E, Alfano AI, Brindisi M, Summa V. Chasing a Breath of Fresh Air in Cystic Fibrosis (CF): Therapeutic Potential of Selective HDAC6 Inhibitors to Tackle Multiple Pathways in CF Pathophysiology. J Med Chem 2022; 65:3080-3097. [PMID: 35148101 PMCID: PMC8883472 DOI: 10.1021/acs.jmedchem.1c02067] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
![]()
Compelling new support
has been provided for histone deacetylase
isoform 6 (HDAC6) as a common thread in the generation of the dysregulated
proinflammatory and fibrotic phenotype in cystic fibrosis (CF). HDAC6
also plays a crucial role in bacterial clearance or killing as a direct
consequence of its effects on CF immune responses. Inhibiting HDAC6
functions thus eventually represents an innovative and effective strategy
to tackle multiple aspects of CF-associated lung disease. In this
Perspective, we not only showcase the latest evidence linking HDAC(6)
activity and expression with CF phenotype but also track the new dawn
of HDAC(6) modulators in CF and explore potentialities and future
perspectives in the field.
Collapse
Affiliation(s)
- Simona Barone
- Department of Pharmacy, Department of Excellence 2018-2022, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| | - Emilia Cassese
- Department of Pharmacy, Department of Excellence 2018-2022, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| | - Antonella Ilenia Alfano
- Department of Pharmacy, Department of Excellence 2018-2022, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| | - Margherita Brindisi
- Department of Pharmacy, Department of Excellence 2018-2022, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| | - Vincenzo Summa
- Department of Pharmacy, Department of Excellence 2018-2022, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, I-80131 Naples, Italy
| |
Collapse
|
46
|
Shen H, Zhang N, Liu Y, Yang X, He Y, Li Q, Shen X, Zhu Y, Yang Y. The Interaction Between Pulmonary Fibrosis and COVID-19 and the Application of Related Anti-Fibrotic Drugs. Front Pharmacol 2022; 12:805535. [PMID: 35069217 PMCID: PMC8766975 DOI: 10.3389/fphar.2021.805535] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is a highly contagious respiratory disease, which mainly affects the lungs. Critically ill patients are easily complicated by cytokine storms, acute respiratory distress syndrome (ARDS), and respiratory failure, which seriously threaten their lives. Pulmonary fibrosis (PF) is a common interstitial lung disease, and its pathogenesis may involve the participation of a variety of immune cells and inflammatory factors. Current studies have shown that patients with COVID-19 may be complicated by pulmonary fibrosis, and patients with pulmonary fibrosis may also be at higher risk of contracting COVID-19 than healthy people. Pulmonary fibrosis is an important risk factor leading to the aggravation of COVID-19 disease. COVID-19 complicated by cytokine storm and ARDS mechanism pathways are similar to the pathogenesis of pulmonary fibrosis. The potential interaction between pulmonary fibrosis and COVID-19 can cause acute exacerbation of the patient's condition, but the potential mechanism between the two has not been fully elucidated. Most of the drug treatment programs for COVID-19-related pulmonary fibrosis are currently formulated about the relevant guidelines for idiopathic pulmonary fibrosis (IPF), and there is no clear drug treatment program recommendation. This article aims to summarize the relevant mechanism pathways of COVID-19 and pulmonary fibrosis, explore the interrelationships and possible mechanisms, and discuss the value and risks of existing and potential COVID-19-related pulmonary fibrosis treatment drugs, to provide reference for anti-fibrosis treatment for patients.
Collapse
Affiliation(s)
- Hao Shen
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Nu Zhang
- Department of Pharmacy, People’s Hospital of Fushun County, Fushun, China
| | - Yuqing Liu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xuerong Yang
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuanyuan He
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qi Li
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoyan Shen
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yulian Zhu
- Department of Pharmacy, Ziyang People’s Hospital, Ziyang, China
| | - Yong Yang
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
47
|
Wang J, Hu K, Cai X, Yang B, He Q, Wang J, Weng Q. Targeting PI3K/AKT signaling for treatment of idiopathic pulmonary fibrosis. Acta Pharm Sin B 2022; 12:18-32. [PMID: 35127370 PMCID: PMC8799876 DOI: 10.1016/j.apsb.2021.07.023] [Citation(s) in RCA: 224] [Impact Index Per Article: 74.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/13/2021] [Accepted: 07/09/2021] [Indexed: 01/03/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive fibrotic interstitial pneumonia with unknown causes. The incidence rate increases year by year and the prognosis is poor without cure. Recently, phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB/AKT) signaling pathway can be considered as a master regulator for IPF. The contribution of the PI3K/AKT in fibrotic processes is increasingly prominent, with PI3K/AKT inhibitors currently under clinical evaluation in IPF. Therefore, PI3K/AKT represents a critical signaling node during fibrogenesis with potential implications for the development of novel anti-fibrotic strategies. This review epitomizes the progress that is being made in understanding the complex interpretation of the cause of IPF, and demonstrates that PI3K/AKT can directly participate to the greatest extent in the formation of IPF or cooperate with other pathways to promote the development of fibrosis. We further summarize promising PI3K/AKT inhibitors with IPF treatment benefits, including inhibitors in clinical trials and pre-clinical studies and natural products, and discuss how these inhibitors mitigate fibrotic progression to explore possible potential agents, which will help to develop effective treatment strategies for IPF in the near future.
Collapse
Affiliation(s)
- Jincheng Wang
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kaili Hu
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xuanyan Cai
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Wang
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
48
|
Walker CJ, Crocini C, Ramirez D, Killaars AR, Grim JC, Aguado BA, Clark K, Allen MA, Dowell RD, Leinwand LA, Anseth KS. Nuclear mechanosensing drives chromatin remodelling in persistently activated fibroblasts. Nat Biomed Eng 2021; 5:1485-1499. [PMID: 33875841 PMCID: PMC9102466 DOI: 10.1038/s41551-021-00709-w] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 03/07/2021] [Indexed: 02/02/2023]
Abstract
Fibrotic disease is caused by the continuous deposition of extracellular matrix by persistently activated fibroblasts (also known as myofibroblasts), even after the resolution of the injury. Using fibroblasts from porcine aortic valves cultured on hydrogels that can be softened via exposure to ultraviolet light, here we show that increased extracellular stiffness activates the fibroblasts, and that cumulative tension on the nuclear membrane and increases in the activity of histone deacetylases transform transiently activated fibroblasts into myofibroblasts displaying condensed chromatin with genome-wide alterations. The condensed structure of the myofibroblasts is associated with cytoskeletal stability, as indicated by the inhibition of chromatin condensation and myofibroblast persistence after detachment of the nucleus from the cytoskeleton via the displacement of endogenous nesprins from the nuclear envelope. We also show that the chromatin structure of myofibroblasts from patients with aortic valve stenosis is more condensed than that of myofibroblasts from healthy donors. Our findings suggest that nuclear mechanosensing drives distinct chromatin signatures in persistently activated fibroblasts.
Collapse
Affiliation(s)
- Cierra J Walker
- Materials Science and Engineering Program, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Claudia Crocini
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Daniel Ramirez
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Anouk R Killaars
- Materials Science and Engineering Program, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Joseph C Grim
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Brian A Aguado
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Kyle Clark
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Mary A Allen
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Robin D Dowell
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Leslie A Leinwand
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA.
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA.
| | - Kristi S Anseth
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA.
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA.
| |
Collapse
|
49
|
Morin inhibits the transformation of fibroblasts towards myofibroblasts through regulating "PPAR-γ-glutaminolysis-DEPTOR" pathway in pulmonary fibrosis. J Nutr Biochem 2021; 101:108923. [PMID: 34843935 DOI: 10.1016/j.jnutbio.2021.108923] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/06/2021] [Accepted: 10/22/2021] [Indexed: 12/14/2022]
Abstract
Morin, a natural flavonoid exists in many foods and dietary plants, owns good bioactivities. Herein, we investigated its effect on pulmonary fibrosis (PF), and further explored the mechanisms. Results showed that morin remarkably improved the pathologic alterations, and inhibited the transformation of fibroblasts towards myofibroblasts in lungs of mice with bleomycin-induced PF as well as TGF-β1 or hypoxia-stimulated NIH-3T3 cells. Mechanistic studies revealed that morin activated peroxisome proliferator activated receptor-gamma (PPAR-γ), and GW9662 or siPPAR-γ significantly weakened the inhibition of morin on the transformation of NIH-3T3 cells. Furthermore, morin restricted glutaminolysis by down-regulating the level of glutaminase 1 (GLS1), which was confirmed by glutamine deprivation, and GLS1 overexpression. Replenishment of metabolite α-ketoglutarate (α-KG) and 2-hydroxyglutarate (2-HG) inhibited morin-prevented transformation of fibroblasts, but neither TGF-β1 nor hypoxia could induce the transformation of IDH2-knockdown fibroblasts, suggesting 2-HG was directly involved in the action of morin. Then, ubiquitination of DEPTOR was demonstrated to be prevented by morin, which was attributed to KDM4A, an enzyme inactivated by 2-HG, and leucine as well as KDM4A inhibitor obstructed the effect of morin. Finally, the mechanisms of morin were further confirmed in vivo. Collectively, morin inhibited PF through intervening in "PPAR-γ-glutaminolysis-DEPTOR" signals, and subsequent restriction on the transformation of fibroblasts towards myofibroblasts.
Collapse
|
50
|
Karki P, Birukova AA. Microtubules as Major Regulators of Endothelial Function: Implication for Lung Injury. Front Physiol 2021; 12:758313. [PMID: 34777018 PMCID: PMC8582326 DOI: 10.3389/fphys.2021.758313] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/06/2021] [Indexed: 01/04/2023] Open
Abstract
Endothelial dysfunction has been attributed as one of the major complications in COVID-19 patients, a global pandemic that has already caused over 4 million deaths worldwide. The dysfunction of endothelial barrier is characterized by an increase in endothelial permeability and inflammatory responses, and has even broader implications in the pathogenesis of acute respiratory syndromes such as ARDS, sepsis and chronic illnesses represented by pulmonary arterial hypertension and interstitial lung disease. The structural integrity of endothelial barrier is maintained by cytoskeleton elements, cell-substrate focal adhesion and adhesive cell junctions. Agonist-mediated changes in endothelial permeability are directly associated with reorganization of actomyosin cytoskeleton leading to cell contraction and opening of intercellular gaps or enhancement of cortical actin cytoskeleton associated with strengthening of endothelial barrier. The role of actin cytoskeleton remodeling in endothelial barrier regulation has taken the central stage, but the impact of microtubules in this process remains less explored and under-appreciated. This review will summarize the current knowledge on the crosstalk between microtubules dynamics and actin cytoskeleton remodeling, describe the signaling mechanisms mediating this crosstalk, discuss epigenetic regulation of microtubules stability and its nexus with endothelial barrier maintenance, and overview a role of microtubules in targeted delivery of signaling molecules regulating endothelial permeability and inflammation.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Anna A Birukova
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|