1
|
He Y, Shen X, Zhai K, Nian S. Advances in understanding the role of interleukins in pulmonary fibrosis (Review). Exp Ther Med 2025; 29:25. [PMID: 39650776 PMCID: PMC11619568 DOI: 10.3892/etm.2024.12775] [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: 05/14/2024] [Accepted: 10/03/2024] [Indexed: 12/11/2024] Open
Abstract
Pulmonary fibrosis (PF) is a progressive, irreversible disease characterized by heterogeneous interstitial lung tissue damage. It originates from persistent or repeated lung epithelial injury and leads to the activation and differentiation of fibroblasts into myofibroblasts. Interleukins (ILs) are a group of lymphokines crucial for immunomodulation that are implicated in the pathogenesis of PF. However, different types of ILs exert disparate effects on PF. In the present review, based on the effect on PF, ILs are classified into three categories: i) Promotors of PF; ii) inhibitors of PF; and iii) those that exert dual effects on PF. Several types of ILs can promote PF by provoking inflammation, initiating proliferation and transdifferentiation of epithelial cells, exacerbating lung injury, while other ILs can inhibit PF through suppressing expression of inflammatory factors, modulating the Th1/Th2 balance and autophagy. The present review summarizes the association of ILs and PF, focusing on the roles and mechanisms of ILs underlying PF.
Collapse
Affiliation(s)
- Yuqing He
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Xuebin Shen
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Kefeng Zhai
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, P.R. China
| | - Sihui Nian
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| |
Collapse
|
2
|
Manavi MA, Fathian Nasab MH, Mohammad Jafari R, Dehpour AR. Mechanisms underlying dose-limiting toxicities of conventional chemotherapeutic agents. J Chemother 2024; 36:623-653. [PMID: 38179685 DOI: 10.1080/1120009x.2023.2300217] [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: 09/20/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Dose-limiting toxicities (DLTs) are severe adverse effects that define the maximum tolerated dose of a cancer drug. In addition to the specific mechanisms of each drug, common contributing factors include inflammation, apoptosis, ion imbalances, and tissue-specific enzyme deficiencies. Among various DLTs are bleomycin-induced pulmonary fibrosis, doxorubicin-induced cardiomyopathy, cisplatin-induced nephrotoxicity, methotrexate-induced hepatotoxicity, vincristine-induced neurotoxicity, paclitaxel-induced peripheral neuropathy, and irinotecan, which elicits severe diarrhea. Currently, specific treatments beyond dose reduction are lacking for most toxicities. Further research on cellular and molecular pathways is imperative to improve their management. This review synthesizes preclinical and clinical data on the pharmacological mechanisms underlying DLTs and explores possible treatment approaches. A comprehensive perspective reveals knowledge gaps and emphasizes the need for future studies to develop more targeted strategies for mitigating these dose-dependent adverse effects. This could allow the safer administration of fully efficacious doses to maximize patient survival.
Collapse
Affiliation(s)
- Mohammad Amin Manavi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Han SY, Im DS. Selonsertib, an ASK1 Inhibitor, Ameliorates Ovalbumin-Induced Allergic Asthma during Challenge and Sensitization Periods. Biomol Ther (Seoul) 2024; 32:451-459. [PMID: 38844790 PMCID: PMC11214966 DOI: 10.4062/biomolther.2023.203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 06/26/2024] Open
Abstract
Apoptosis signal-regulating kinase 1 (ASK1) is an upstream signaling molecule in oxidative stress-induced responses. Because oxidative stress is involved in asthma pathogenesis, ASK1 gene deficiency was investigated in animal models of allergic asthma. However, there is no study to investigate whether ASK1 inhibitors could be applied for asthma to date. Selonsertib, a potent and selective ASK1 inhibitor, was applied to BALB/c mice of an ovalbumin (OVA)-induced allergic asthma model. Selonsertib suppressed antigen-induced degranulation of RBL-2H3 mast cells in a concentration-dependent manner. The administration of selonsertib both before OVA sensitization and OVA challenge significantly reduced airway hyperresponsiveness, and suppressed eosinophil numbers and inflammatory cytokine levels in the bronchoalveolar lavage fluid. Histopathologic examination elucidated less inflammatory responses and reduced mucin-producing cells around the peribronchial regions of the lungs. Selonsertib also suppressed the IgE levels in serum and the protein levels of IL-13 in the bronchoalveolar lavage fluid. These results suggest that selonsertib may ameliorate allergic asthma by suppressing immune responses and be applicable to allergic asthma.
Collapse
Affiliation(s)
- So-Young Han
- Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02446, Republic of Korea
| | - Dong-Soon Im
- Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02446, Republic of Korea
| |
Collapse
|
4
|
Mohanan A, Washimkar KR, Mugale MN. Unraveling the interplay between vital organelle stress and oxidative stress in idiopathic pulmonary fibrosis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119676. [PMID: 38242330 DOI: 10.1016/j.bbamcr.2024.119676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 12/22/2023] [Accepted: 01/10/2024] [Indexed: 01/21/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung disease characterized by excessive accumulation of extracellular matrix, leading to irreversible fibrosis. Emerging evidence suggests that endoplasmic reticulum (ER) stress, mitochondrial stress, and oxidative stress pathways play crucial roles in the pathogenesis of IPF. ER stress occurs when the protein folding capacity of the ER is overwhelmed, triggering the unfolded protein response (UPR) and contributing to protein misfolding and cellular stress in IPF. Concurrently, mitochondrial dysfunction involving dysregulation of key regulators, including PTEN-induced putative kinase 1 (PINK1), Parkin, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), and sirtuin 3 (SIRT3), disrupts mitochondrial homeostasis and impairs cellular energy metabolism. This leads to increased reactive oxygen species (ROS) production, release of pro-fibrotic mediators, and activation of fibrotic pathways, exacerbating IPF progression. The UPR-induced ER stress further disrupts mitochondrial metabolism, resulting in altered mitochondrial mechanisms that increase the generation of ROS, resulting in further ER stress, creating a feedback loop that contributes to the progression of IPF. Oxidative stress also plays a pivotal role in IPF, as ROS-mediated activation of TGF-β, NF-κB, and MAPK pathways promotes inflammation and fibrotic responses. This review mainly focuses on the links between ER stress, mitochondrial dysfunctions, and oxidative stress with different signaling pathways involved in IPF. Understanding these mechanisms and targeting key molecules within these pathways may offer promising avenues for intervention.
Collapse
Affiliation(s)
- Anushree Mohanan
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India
| | - Kaveri R Washimkar
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Madhav Nilakanth Mugale
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
5
|
Ji A, Trumbauer AC, Noffsinger VP, Meredith LW, Dong B, Wang Q, Guo L, Li X, De Beer FC, Webb NR, Tannock LR, Starr ME, Waters CM, Shridas P. Deficiency of Acute-Phase Serum Amyloid A Exacerbates Sepsis-Induced Mortality and Lung Injury in Mice. Int J Mol Sci 2023; 24:17501. [PMID: 38139330 PMCID: PMC10744229 DOI: 10.3390/ijms242417501] [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: 11/10/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Serum amyloid A (SAA) is a family of proteins, the plasma levels of which may increase >1000-fold in acute inflammatory states. We investigated the role of SAA in sepsis using mice deficient in all three acute-phase SAA isoforms (SAA-TKO). SAA deficiency significantly increased mortality rates in the three experimental sepsis mouse models: cecal ligation and puncture (CLP), cecal slurry (CS) injection, and lipopolysaccharide (LPS) treatments. SAA-TKO mice had exacerbated lung pathology compared to wild-type (WT) mice after CLP. A bulk RNA sequencing performed on lung tissues excised 24 h after CLP indicated significant enrichment in the expression of genes associated with chemokine production, chemokine and cytokine-mediated signaling, neutrophil chemotaxis, and neutrophil migration in SAA-TKO compared to WT mice. Consistently, myeloperoxidase activity and neutrophil counts were significantly increased in the lungs of septic SAA-TKO mice compared to WT mice. The in vitro treatment of HL-60, neutrophil-like cells, with SAA or SAA bound to a high-density lipoprotein (SAA-HDL), significantly decreased cellular transmigration through laminin-coated membranes compared to untreated cells. Thus, SAA potentially prevents neutrophil transmigration into injured lungs, thus reducing exacerbated tissue injury and mortality. In conclusion, we demonstrate for the first time that endogenous SAA plays a protective role in sepsis, including ameliorating lung injury.
Collapse
Affiliation(s)
- Ailing Ji
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Andrea C. Trumbauer
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Victoria P. Noffsinger
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Luke W. Meredith
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Brittany Dong
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; (B.D.); (C.M.W.)
| | - Qian Wang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Ling Guo
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
| | - Xiangan Li
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; (B.D.); (C.M.W.)
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA;
| | - Frederick C. De Beer
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA;
| | - Nancy R. Webb
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA;
| | - Lisa R. Tannock
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA;
| | - Marlene E. Starr
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA;
- Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher M. Waters
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; (B.D.); (C.M.W.)
| | - Preetha Shridas
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (A.J.); (A.C.T.); (V.P.N.); (L.W.M.); (Q.W.); (L.G.); (X.L.); (N.R.W.); (L.R.T.)
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA;
| |
Collapse
|
6
|
Zeng Q, Zhou TT, Huang WJ, Huang XT, Huang L, Zhang XH, Sang XX, Luo YY, Tian YM, Wu B, Liu L, Luo ZQ, He B, Liu W, Tang SY. Asarinin attenuates bleomycin-induced pulmonary fibrosis by activating PPARγ. Sci Rep 2023; 13:14706. [PMID: 37679587 PMCID: PMC10485066 DOI: 10.1038/s41598-023-41933-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease that lacks effective treatment modalities. Once patients are diagnosed with IPF, their median survival is approximately 3-5 years. PPARγ is an important target for the prevention and treatment of pulmonary fibrosis. Asarinin is a lignan compound that can be extracted from food plant Asarum heterotropoides. In this study, we investigated the therapeutic effects of asarinin in a pulmonary fibrosis model constructed using bleomycin in mice and explored the underlying mechanisms. Intraperitoneal administration of asarinin to mice with pulmonary fibrosis showed that asarinin effectively attenuated pulmonary fibrosis, and this effect was significantly inhibited by the PPARγ inhibitor GW9662. Asarinin inhibited TGF-β1-induced fibroblast-to-myofibroblast transition in vitro, while GW9662 and PPARγ gene silencing significantly inhibited this effect. In addition, asarinin inhibited not only the canonical Smad pathway of TGF-β but also the non-canonical AKT and MAPK pathways by activating PPARγ. Our study demonstrates that asarinin can be used as a therapeutic agent for pulmonary fibrosis, and that PPARγ is its key target.
Collapse
Affiliation(s)
- Qian Zeng
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Ting-Ting Zhou
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Wen-Jie Huang
- School of Nursing, Hunan University of Medicine, Huaihua, Hunan, China
| | - Xiao-Ting Huang
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Lei Huang
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Xiao-Hua Zhang
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Xiao-Xue Sang
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yu-Yang Luo
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yu-Mei Tian
- School of Nursing, Hunan University of Medicine, Huaihua, Hunan, China
| | - Bin Wu
- School of Nursing, Hunan University of Medicine, Huaihua, Hunan, China
| | - Lin Liu
- School of Nursing, Hunan University of Medicine, Huaihua, Hunan, China
| | - Zi-Qiang Luo
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Bin He
- School of Nursing, Hunan University of Medicine, Huaihua, Hunan, China.
| | - Wei Liu
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
7
|
Bahri S, Abidi A, Nahdi A, Abdennabi R, Mlika M, Ben Ali R, Jameleddine S. Olea europaea L. Leaf Extract Alleviates Fibrosis Progression and Oxidative Stress Induced by Bleomycin on a Murine Model of Lung Fibrosis. Dose Response 2023; 21:15593258231200972. [PMID: 37667683 PMCID: PMC10475267 DOI: 10.1177/15593258231200972] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023] Open
Abstract
In this study, we aim to investigate the effect of industrial Olea europaea L. leaf extract (OLE) against bleomycin (BLM)-induced pulmonary fibrosis (PF) in rats. Male Wistar rats were treated with a single intratracheal injection of BLM (4 mg/kg) and a daily intraperitoneal injection of OLE (10, 20, and 40 mg/kg) for 4 weeks. Results of HPLC and LC-MS analysis revealed a large amount of oleuropein (15.43%/DW) in OLE. BLM induced apparent damage of lung architecture with condensed collagen bundles, increased lipid peroxidation which has been deduced from malondialdehyde (MDA) levels: (.9 ± .13 vs .25 ± .12 nmol/mg protein) and hydroxyproline content (.601 ± .22 vs .154 ± .139 mg/g of lung tissue) and decreased catalase (CAT) (5.93.10-5 ± 4.23.10-5 vs 6.41.10-4 ± 2.33.10-4 μmol/min/mg protein) and superoxide dismutase (SOD) (28.73 ± 3.34 vs 50.13 ± 2.1 USOD/min/mg protein) levels compared to the control. OLE treatment (40 mg/kg) stabilized MDA content (.32 ± .15 and .27 ± .13 vs .9 ± .13 nmol/mg protein), normalized SOD (61.27 ± 13.37 vs 28.73 ± 3.34 USOD/min/mg protein), and CAT (5.2.10-4 ±1.8.10-4 vs 5.93.10-5 ± 4.23.10-5 μmol/min/mg protein) activities and counteracted collagen accumulation and hydroxyproline content (.222 ± .07 vs .601 ± .22 mg/g of lung tissue) in the lung parenchyma. Finally, OLE might have a potent protective effect against PF by regulating oxidative parameters and attenuating collagen deposition, due to the existence of large amount of bioactive phenolic molecules.
Collapse
Affiliation(s)
- Sana Bahri
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Physiopathology, Food and Biomolecules (LR-17-ES-03), Technology Center of Sidi Thabet, University of Manouba, Tunis, Tunisia
- Laboratory of Quality Control, HERBES DE TUNISIE, Company AYACHI-Group, Mansoura, Siliana-Tunisia
| | - Anouar Abidi
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
- Unit of Functional Physiology and Valorization of Bio-Resources of the Higher Institute of Biotechnology of Béja, University of Jendouba, Jendouba, Tunisia
| | - Afef Nahdi
- Research Unit n° 17/ES/13, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Raed Abdennabi
- Laboratory of Plant Biotechnology, Faculty of Science, University of Sfax, Sfax, Tunisia
| | - Mona Mlika
- Laboratory of Anatomy and Pathology, Abderhaman Mami Hospital, Ariana, Tunisia
| | - Ridha Ben Ali
- Laboratory of Experimental Medicine, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Saloua Jameleddine
- Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Physiopathology, Food and Biomolecules (LR-17-ES-03), Technology Center of Sidi Thabet, University of Manouba, Tunis, Tunisia
| |
Collapse
|
8
|
Zhang X, Su J, Lin J, Liu L, Wu J, Yuan W, Zhang Y, Chen Q, Su ZJ, Xu G, Sun M, Zhang Y, Chen X, Zhang W. Fu-Zheng-Tong-Luo formula promotes autophagy and alleviates idiopathic pulmonary fibrosis by controlling the Janus kinase 2/signal transducer and activator of transcription 3 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116633. [PMID: 37207878 DOI: 10.1016/j.jep.2023.116633] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/21/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fu-Zheng-Tong-Luo (FZTL) formula is a Chinese herbal prescription which is used to treat idiopathic pulmonary fibrosis (IPF). We previously reported that the FZTL formula could improve IPF injury in rats; however, the mechanism remains unelucidated. AIM OF THE STUDY To elucidate the effects and mechanisms of the FZTL formula on IPF. MATERIALS AND METHODS The bleomycin-induced pulmonary fibrosis rat model and transforming growth factor-β-induced lung fibroblast model were used. Histological changes and fibrosis formation were detected in the rat model after treatment with the FZTL formula. Furthermore, the effects of the FZTL formula on autophagy and lung fibroblast activation were determined. Moreover, the mechanism of FZTL was explored using transcriptomics analysis. RESULTS We observed that FZTL alleviated IPF injury in rats and inhibited inflammatory responses and fibrosis formation in rats. Moreover, it promoted autophagy and inhibited lung fibroblast activation in vitro. Transcriptomics analysis revealed that FZTL regulates the Janus kinase 2 (JAK)/signal transducer and activator of the transcription 3 (STAT) signaling pathway. The JAK2/STAT3 signaling activator interleukin 6 inhibited the anti-fibroblast activation effect of the FZTL formula. Combined treatment with the JAK2 inhibitor (AZD1480) and autophagy inhibitor (3-methyladenine) did not enhance the antifibrotic effect of FZTL. CONCLUSIONS The FZTL formula can inhibit IPF injury and lung fibroblast activation. Its effects are mediated via the JAK2/STAT3 signaling pathway. The FZTL formula may be a potential complementary therapy for pulmonary fibrosis.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jie Su
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Jiacheng Lin
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Lujiong Liu
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jiamin Wu
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Wenli Yuan
- Department of Nephrology Diseases, YueYang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yibao Zhang
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Qi Chen
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zi Jian Su
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Guihua Xu
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Meng Sun
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yile Zhang
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xuan Chen
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Wei Zhang
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
9
|
Qin J, Cao M, Hu X, Tan W, Ma B, Cao Y, Chen Z, Li Q, Hu G. Dual inhibitors of ASK1 and PDK1 kinases: Design, synthesis, molecular docking and mechanism studies of N-benzyl pyridine-2-one containing derivatives as anti-fibrotic agents. Eur J Med Chem 2023; 247:115057. [PMID: 36603508 DOI: 10.1016/j.ejmech.2022.115057] [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: 11/10/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
Utilizing fragment-based hybrid designing strategies, 24 N-benzyl pyridine-2-one containing derivatives were synthesized by successfully incorporating 6-(4H-1,2,4-triazol-3-yl) pyridin-2-amine of scaffold of ASK1 inhibitor (GS-444217). These newly synthesized compounds were screened in cell-free ASK1 and PDK1 kinase and cellular vitality assays. Among all compounds tested, both 21c and 21d displayed single digit potency of 9.13, 1.73 nM in inhibiting ASK1, and exhibited excellent enzyme inhibitory activity against PDK1 (the inhibition rates at 10 μM were 13.63% and 23.80%, respectively). Specifically, both compounds inhibited the TGF-β1 induced fibrotic response and blocked the up-regulated protein expression levels of ASK1-p38/JNK signaling pathways and possessed the potency in reducing PDK1/Akt phosphorylation. The results herein showed the potential lead characteristics of 21c or 21d as dual inhibitors ASK1/PDK1 kinases.
Collapse
Affiliation(s)
- Jia Qin
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Meng Cao
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Xinlan Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Wenhua Tan
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Binghao Ma
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Yuanyuan Cao
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Zhuo Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China.
| | - Gaoyun Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China.
| |
Collapse
|
10
|
Oxidative Stress and Antioxidative Therapy in Pulmonary Arterial Hypertension. Molecules 2022; 27:molecules27123724. [PMID: 35744848 PMCID: PMC9229274 DOI: 10.3390/molecules27123724] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is clinically characterized by a progressive increase in pulmonary artery pressure, followed by right ventricular hypertrophy and subsequently right heart failure. The underlying mechanism of PAH includes endothelial dysfunction and intimal smooth muscle proliferation. Numerous studies have shown that oxidative stress is critical in the pathophysiology of PAH and involves changes in reactive oxygen species (ROS), reactive nitrogen (RNS), and nitric oxide (NO) signaling pathways. Disrupted ROS and NO signaling pathways cause the proliferation of pulmonary arterial endothelial cells (PAECs) and pulmonary vascular smooth muscle cells (PASMCs), resulting in DNA damage, metabolic abnormalities, and vascular remodeling. Antioxidant treatment has become a main area of research for the treatment of PAH. This review mainly introduces oxidative stress in the pathogenesis of PAH and antioxidative therapies and explains why targeting oxidative stress is a valid strategy for PAH treatment.
Collapse
|
11
|
Baig MH, Yousuf M, Khan MI, Khan I, Ahmad I, Alshahrani MY, Hassan MI, Dong JJ. Investigating the Mechanism of Inhibition of Cyclin-Dependent Kinase 6 Inhibitory Potential by Selonsertib: Newer Insights Into Drug Repurposing. Front Oncol 2022; 12:865454. [PMID: 35720007 PMCID: PMC9204300 DOI: 10.3389/fonc.2022.865454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/06/2022] [Indexed: 12/23/2022] Open
Abstract
Cyclin-dependent kinases (CDKs) play significant roles in numerous physiological, and are considered an attractive drug target for cancer, neurodegenerative, and inflammatory diseases. In the present study, we have aimed to investigate the binding affinity and inhibitory potential of selonsertib toward CDK6. Using the drug repurposing approach, we performed molecular docking of selonsertib with CDK6 and observed a significant binding affinity. To ascertain, we further performed essential dynamics analysis and free energy calculation, which suggested the formation of a stable selonsertib-CDK6 complex. The in-silico findings were further experimentally validated. The recombinant CDK6 was expressed, purified, and treated with selonsertib. The binding affinity of selonsertib to CDK6 was estimated by fluorescence binding studies and enzyme inhibition assay. The results indicated an appreciable binding of selonsertib against CDK6, which subsequently inhibits its activity with a commendable IC50 value (9.8 μM). We concluded that targeting CDK6 by selonsertib can be an efficient therapeutic approach to cancer and other CDK6-related diseases. These observations provide a promising opportunity to utilize selonsertib to address CDK6-related human pathologies.
Collapse
Affiliation(s)
- Mohammad Hassan Baig
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Mohd. Yousuf
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Mohd. Imran Khan
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Imran Khan
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, BezmialemVakif University, Istanbul, Turkey
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Y. Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Jae-June Dong
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
12
|
Affiliation(s)
- Michael Schuliga
- College of Health, Medicine and Wellbeing
- School of Biomedical Sciences and Pharmacy The University of Newcastle Callaghan, New South Wales, Australia
| | - Satish K Madala
- Department of Pediatrics University of Cincinnati College of Medicine Cincinnati, Ohio
- Division of Pulmonary Medicine Cincinnati Children's Hospital Medical Center Cincinnati, Ohio
| |
Collapse
|