1
|
Ren S, Liu F, Chi M, Liu J, Huang Y, Huang W, Gu W, Yuan Y, Hou S, Chen X, Ma L. Discovery of a selective and potent inhibitor of c-Jun N-terminal kinase 1 with anti-pulmonary fibrosis effect. Bioorg Med Chem Lett 2025; 116:130044. [PMID: 39608686 DOI: 10.1016/j.bmcl.2024.130044] [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: 09/22/2024] [Revised: 11/18/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
We synthesized and evaluated a series of derivatives based on the pyrimidine-2,4-diamine scaffold as potential JNK1 inhibitors, incorporating bridging rings and spirocyclic modifications to enhance their inhibitory activity. These compounds were biologically assessed through JNK enzyme inhibition assays and Western Blot analysis. Compounds 13, 14 and 19 demonstrated significant inhibitory activity at both the enzyme and cellular level compared to the lead compound 1 and clinical candidate CC-90001. Notably, 14 exhibited strong inhibitory potency against JNK1 with sub-nanomolar efficacy and suppresses TGF-β-induced epithelial-mesenchymal transition, indicating its potential as a promising candidate for further development as an anti-pulmonary fibrosis agent targeting JNK1.
Collapse
Affiliation(s)
- Shuhua Ren
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Fengling Liu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Man Chi
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jinfeng Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yi Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Weiwei Huang
- Hangzhou Matrix Biopharmaceutical Co., Ltd, Hangzhou, Zhejiang 311121, China
| | - Wenjing Gu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yaxia Yuan
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, TX 78229, USA
| | - Shurong Hou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Xiabin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Lei Ma
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| |
Collapse
|
2
|
Guo Y, Liu F, Chi M, Qian H, Zhang Y, Yuan Y, Hou S, Chen X, Ma L. Design and synthesis of JNK1-targeted PROTACs and research on the activity. Bioorg Chem 2025; 154:108044. [PMID: 39700830 DOI: 10.1016/j.bioorg.2024.108044] [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: 10/20/2024] [Revised: 11/24/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024]
Abstract
Kinase dysregulation is greatly associated with cell growth, proliferation, differentiation and apoptosis, which indicates their great potential as therapeutic targets for treatment of numerous progressive disorders, including inflammatory, metabolic and autoimmune disorders, organ fibrosis and cancer. The c‑Jun N‑Terminal Kinase (JNK), as a member of MAPK family, is proved to be a potential target for the treatment of pulmonary fibrosis, which is the most common progressive and fatal fibrotic lung disease. As a new strategy, small-molecule-mediated targeted protein degradation pathway has the advantages of catalytic properties, overcoming drug resistance and expanding target space, which can circumvent the limitations associated with kinase inhibitors. Proteolysis targeting chimeras (PROTAC) contains a linker to concatenate a ligand of E3 ubiquitin ligase and a ligand for a protein of interest (POI). We developed a total of 20 JNK1-targeted PROTACs that induce proteasomal degradation of JNK1 components. The most active PROTAC molecule PA2 was then investigated by JNK1 enzyme assay and protein degradation assay, which suggested that PA2 had an anti-JNK1 ability and provided insights for the future use of JNK1-targeted PROTAC as treatment drugs for pulmonary fibrosis.
Collapse
Affiliation(s)
- Yue Guo
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Fengling Liu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Man Chi
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Hewen Qian
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Ye Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yaxia Yuan
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, TX 78229, USA
| | - Shurong Hou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Xiabin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Lei Ma
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| |
Collapse
|
3
|
Li T, Xiong Y, Li J, Tang X, Zhong Y, Tang Z, Zhang Q, Luo Y. Mapping and Analysis of Protein and Gene Profile Identification of the Important Role of Transforming Growth Factor Beta in Synovial Invasion in Patients With Pigmented Villonodular Synovitis. Arthritis Rheumatol 2024; 76:1679-1695. [PMID: 38973550 DOI: 10.1002/art.42946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 05/06/2024] [Accepted: 07/01/2024] [Indexed: 07/09/2024]
Abstract
OBJECTIVE Pigmented villonodular synovitis (PVNS) is a rare benign proliferative disease affecting the soft-tissue lining the synovial joints and tendons. Its etiology is poorly understood, largely limiting the availability of current therapeutic options. Here, we mapped the synovial gene and protein profiles of patients with PVNS, revealed a link between synovial inflammation and invasion, and elucidated the potential molecular mechanism involved. METHODS The expression of synovial genes from 6 control individuals, 7 patients with osteoarthritis (OA), and 19 patients with PVNS was analyzed via RNA sequencing. Protein profiles from 5 control individuals, 10 patients with OA, and 32 patients with PVNS were analyzed using label-free proteomics. Microarray and reverse transcription-polymerase chain reaction analyses and immunohistochemical staining were used to evaluate inflammatory cytokine and target gene expression levels in synovial tissue, epithelial cells, and synovial fibroblasts (FLSs) derived from tissue of patients with PVNS. Various signaling pathway inhibitors, small interfering RNAs, and Western blots were used for molecular mechanism studies. Transwell migration and invasion assays were subsequently performed. RESULTS In total, 522 differentially expressed proteins were identified in the tissues of patients with PVNS. By integrating RNA sequencing and microarray analyses, significant changes in the expression of epithelial-mesenchymal transition (EMT)-related genes, including transforming growth factor TGF-b induced, neural cadherin, epithelial cadherin, SNAIL, and TWIST, were confirmed in the tissue of patients with PVNS compared to the control tissue. In vitro, TGFβ induced EMT and increased epithelial cell migration and invasion. Moreover, TGFβ not only promoted interactions between epithelial cells and FLSs but also directly increased the migration and invasion abilities of FLSs by activating the classical Smad2/3 and nonclassical JNK/AKT signaling pathways. CONCLUSION This study provides overall protein and gene profiles of PVNS and identifies the crucial role of TGFβ in synovial invasion pathology. Exploring the related molecular mechanism may also reveal a new strategy or target for PVNS therapy.
Collapse
Affiliation(s)
- Tao Li
- West China Hospital, Sichuan University, Chengdu, China
| | - Yan Xiong
- West China Hospital, Sichuan University, Chengdu, China
| | - Jian Li
- West China Hospital, Sichuan University, Chengdu, China
| | - Xin Tang
- West China Hospital, Sichuan University, Chengdu, China
| | - Yutong Zhong
- West China Hospital, Sichuan University, Chengdu, China
| | - Zhigang Tang
- West China Hospital, Sichuan University, Chengdu, China
| | - Qiuping Zhang
- West China Hospital, Sichuan University, Chengdu, China
| | - Yubin Luo
- West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Huang Y, Liu F, Ren S, Ding Y, Chi M, Huang W, Gu W, Qian H, Yuan Y, Hou S, Chen X, Ma L. Structure Optimization of c-Jun N-terminal Kinase 1 Inhibitors for Treating Idiopathic Pulmonary Fibrosis. J Med Chem 2024; 67:17713-17737. [PMID: 39303278 DOI: 10.1021/acs.jmedchem.4c01764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and lethal lung disease with an elusive etiology. Aberrant activation of c-Jun N-terminal kinase 1 (JNK1) has been implicated in its pathogenesis. Through a combination of structure-based drug design and structure-activity relationship (SAR) optimization, a series of pyrimidine-2,4-diamine scaffold derivatives have been developed as potent JNK1 inhibitors. Compound E1 was identified with low nanomolar JNK1 inhibitory potency (IC50 = 2.7 nM). The introduction of a dimethylamine side chain has significantly enhanced the ability of E1 to inhibit c-Jun phosphorylation, surpassing the clinical candidate CC-90001. Molecular dynamics simulations revealed a binding free energy of -50.46 kcal/mol for E1. Moreover, E1 displayed satisfactory pharmacokinetic properties, with a bioavailability of 69% in rats. Furthermore, compound E1 exerted significant antifibrotic effects in a bleomycin-induced IPF mouse model and prevented a TGF-β-induced epithelial-to-mesenchymal transition in vitro. These findings position E1 as a promising lead for further drug development targeting IPF.
Collapse
Affiliation(s)
- Yi Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Fengling Liu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, and Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Shuhua Ren
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yuanqing Ding
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, and Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Man Chi
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, and Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Weiwei Huang
- Hangzhou Matrix Biopharmaceutical Co., Ltd, Hangzhou, Zhejiang 311121, China
| | - Wenjing Gu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Hewen Qian
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yaxia Yuan
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, United States
| | - Shurong Hou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, and Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xiabin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, and Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Lei Ma
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
5
|
Mohamed SK, Karthikeyan S, A Omran O, Ahsin A, Salah H, Mague JT, Al-Salahi R, El Bakri Y. Insights into the crystal structure investigation and virtual screening approach of quinoxaline derivatives as potent against c-Jun N-terminal kinases 1. J Biomol Struct Dyn 2024:1-20. [PMID: 38321917 DOI: 10.1080/07391102.2024.2305317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024]
Abstract
Quinoxaline derivatives are an important class of heterocyclic compounds in which N replaces one or more carbon atoms of the naphthalene ring and exhibit a wide spectrum of biological activities and therapeutic applications. As a result, we were encouraged to explore a new synthetic approach to quinoxaline derivatives. In this work, we synthesized two new derivatives namely, ethyl 4-(2-ethoxy-2-oxoethyl)-3-oxo-3,4-dihydroquinoxaline-2-carboxylate (2) and 3-oxo-3,4-dihydroquinoxaline-2-carbohydrazide (3) respectively. Their structures were confirmed by single-crystal X-ray analysis. Hirshfeld surface (HS) analysis is performed to understand the nature and magnitude of intermolecular interactions in the crystal packing. Density functional theory using the wb97xd/def2-TZVP method was chosen to explore their reactivity, electronic stability and optical properties. Charge transfer (CT) and orbital energies were analyzed via natural population analysis (NPA), and frontier molecular orbital (FMO) theory. The calculated excellent static hyperpolarizability (βo) indicates nonlinear optical (NLO) properties for 2 and 3. Both compounds show potent activity against c-Jun N-terminal kinases 1 (JNK 1) based on structural activity relationship studies, further subjected to molecular docking, molecular dynamics and ADMET analysis to understand their potential as drug candidates.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shaaban K Mohamed
- Chemistry and Environmental Division, Manchester Metropolitan University, Manchester, England
| | - Subramani Karthikeyan
- Center for Healthcare Advancement, Innovation and Research, Vellore Institute of Technology University, Chennai, India
| | - Omran A Omran
- Department of Chemistry, Faculty of Science, Sohag University, Sohag, Egypt
| | - Atazaz Ahsin
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hanan Salah
- Department of Chemistry, Faculty of Science, Sohag University, Sohag, Egypt
| | - Joel T Mague
- Department of Chemistry, Tulane University, New Orleans, LA, USA
| | - Rashad Al-Salahi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Youness El Bakri
- Department of Theoretical and Applied Chemistry, South Ural State University, Chelyabinsk, Russian Federation
| |
Collapse
|
6
|
Cao M, Zhan M, Jing H, Wang Z, Wang Y, Li X, Miao M. Network pharmacology and experimental evidence: MAPK signaling pathway is involved in the anti-asthma roles of Perilla frutescens leaf. Heliyon 2024; 10:e22971. [PMID: 38163225 PMCID: PMC10755271 DOI: 10.1016/j.heliyon.2023.e22971] [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: 03/04/2023] [Revised: 11/22/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024] Open
Abstract
Perilla frutescens (PF) leaf is a traditional Chinese medicine and food with beneficial effects on allergic asthma. We sought to elucidate the active compounds, the targets, and underlying mechanisms of PF leaf in the treatment of allergic asthma by using experimental pharmacology and network pharmacology. An OVA-allergic asthma murine model was constructed to evaluate the effect of PF leaf on allergic asthma. And the network pharmacology and western blotting were performed to evaluate its underlying mechanisms in allergic asthma. PF leaf treatment significantly improved the lung function of OVA model mice and mitigated lung injury by significantly reducing of OVA-specific immunoglobulin E in serum, and interleukin 4, interleukin 5 and tumor necrosis factor alpha in the bronchoalveolar lavage fluid. 50 core targets were screened based on 8 compounds (determined by high performance liquid chromatography) through compound-target- disease network. Furthermore, MAPK signaling pathway was identified as the pathway mediated by PF leaf with the most potential against allergic asthma. And the WB results showed that PF leaf could down-regulate the expression of p-ERK, p-JNK and p-p38, which was highly consistent with the predicted targets and pathway network. In conclusion, this study provides the evidence to support the molecular mechanisms of PF leaf on the treatment of allergic asthma using network pharmacology and in vivo experiments.
Collapse
Affiliation(s)
- Mingzhuo Cao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450058, China
| | - Mengling Zhan
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450058, China
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450058, China
| | - Heyun Jing
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450058, China
| | - Zeqian Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450058, China
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450058, China
| | - Yuan Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450058, China
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450058, China
| | - Xiumin Li
- Department of Microbiology and Immunology, and Otolaryngology, New York Medical College, Valhalla, NY, 10595, USA
| | - Mingsan Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450058, China
| |
Collapse
|
7
|
Luo W, Gu Y, Fu S, Wang J, Zhang J, Wang Y. Emerging opportunities to treat idiopathic pulmonary fibrosis: Design, discovery, and optimizations of small-molecule drugs targeting fibrogenic pathways. Eur J Med Chem 2023; 260:115762. [PMID: 37683364 DOI: 10.1016/j.ejmech.2023.115762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common fibrotic form of idiopathic diffuse lung disease. Due to limited treatment options, IPF patients suffer from poor survival. About ten years ago, Pirfenidone (Shionogi, 2008; InterMune, 2011) and Nintedanib (Boehringer Ingelheim, 2014) were approved, greatly changing the direction of IPF drug design. However, limited efficacy and side effects indicate that neither can reverse the process of IPF. With insights into the occurrence of IPF, novel targets and agents have been proposed, which have fundamentally changed the treatment of IPF. With the next-generation agents, targeting pro-fibrotic pathways in the epithelial-injury model offers a promising approach. Besides, several next-generation IPF drugs have entered phase II/III clinical trials with encouraging results. Due to the rising IPF treatment requirements, there is an urgent need to completely summarize the mechanisms, targets, problems, and drug design strategies over the past ten years. In this review, we summarize known mechanisms, target types, drug design, and novel technologies of IPF drug discovery, aiming to provide insights into the future development and clinical application of next-generation IPF drugs.
Collapse
Affiliation(s)
- Wenxin Luo
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yilin Gu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Siyu Fu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Jifa Zhang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
8
|
Aboushousha R, van der Velden J, Hamilton N, Peng Z, MacPherson M, Erickson C, White S, Wouters EFM, Reynaert NL, Seward DJ, Li J, Janssen-Heininger YMW. Glutaredoxin attenuates glutathione levels via deglutathionylation of Otub1 and subsequent destabilization of system x C. SCIENCE ADVANCES 2023;9:eadi5192. [PMID: 37703360 PMCID: PMC10499329 DOI: 10.1126/sciadv.adi5192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/10/2023] [Indexed: 09/15/2023]
Abstract
Glutathione (GSH) is a critical component of the cellular redox system that combats oxidative stress. The glutamate-cystine antiporter, system xC-, is a key player in GSH synthesis that allows for the uptake of cystine, the rate-limiting building block of GSH. It is unclear whether GSH or GSH-dependent protein oxidation [protein S-glutathionylation (PSSG)] regulates the activity of system xC-. We demonstrate that an environment of enhanced PSSG promotes GSH increases via a system xC--dependent mechanism. Absence of the deglutathionylase, glutaredoxin (GLRX), augmented SLC7A11 protein and led to significant increases of GSH content. S-glutathionylation of C23 or C204 of the deubiquitinase OTUB1 promoted interaction with the E2-conjugating enzyme UBCH5A, leading to diminished ubiquitination and proteasomal degradation of SLC7A11 and augmentation of GSH, effects that were reversed by GLRX. These findings demonstrate an intricate link between GLRX and GSH via S-glutathionylation of OTUB1 and system xC- and illuminate a previously unknown feed-forward regulatory mechanism whereby enhanced GSH protein oxidation augments cellular GSH.
Collapse
Affiliation(s)
- Reem Aboushousha
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Jos van der Velden
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Nicholas Hamilton
- Department of Chemistry, University of Vermont, Burlington, VT 05405, USA
| | - Zhihua Peng
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Maximilian MacPherson
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Cuixia Erickson
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Sheryl White
- Department of Neurological Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Emiel F. M. Wouters
- Department of Respiratory Medicine, NUTRIM School of nutrition and translational research in metabolism, Maastricht University Medical Center, Maastricht, Netherlands
- Ludwig Boltzmann Institute for Lung Research, Vienna, Austria
| | - Niki L. Reynaert
- Department of Respiratory Medicine, NUTRIM School of nutrition and translational research in metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - David J. Seward
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Jianing Li
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | | |
Collapse
|
9
|
Li XY, Chen HR, Kuang DD, Pan LH, Li QM, Luo JP, Zha XQ. Laminaria japonica polysaccharide attenuates podocyte epithelial-mesenchymal transformation via TGF-β1-mediated Smad3 and p38MAPK pathways. Int J Biol Macromol 2023; 241:124637. [PMID: 37121417 DOI: 10.1016/j.ijbiomac.2023.124637] [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: 12/03/2022] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/02/2023]
Abstract
In the present work, we explored the interventional effect and potential mechanism of a purified Laminaria japonica polysaccharide (LJP61A) on podocyte epithelial-mesenchymal transition (EMT) in TGF-β1-induced podocytes and adriamycin-treated mice. Results showed that compared to the model groups, LJP61A significantly up-regulated the levels of epithelial markers (Nephrin, WT-1, podocin) and down-regulated the levels of mesenchymal markers (α-SMA, FN1) in vitro and in vivo, thus preventing EMT-like morphological changes of podocytes, proteinuria and kidney injury. Smad3 and p38MAPK are two central pathways mediating podocyte EMT activated by TGF-β1. We found that LJP61A suppressed TGF-β1-induced activation of Smad3, Smad4 and p38MAPK in vitro and in vivo. Moreover, the inhibitory actions of LJP61A on podocyte EMT were synergistically strengthened by Smad3 inhibitor SIS3 and p38MAPK inhibitor SB203580. Taken together, these findings revealed that LJP61A could prevent podocyte EMT, which might be related to the inhibition of TGF-β1-mediated Smad3 and p38MAPK pathways.
Collapse
Affiliation(s)
- Xue-Ying Li
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Hao-Ran Chen
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Dan-Dan Kuang
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Li-Hua Pan
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Qiang-Ming Li
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Jian-Ping Luo
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Xue-Qiang Zha
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; Key Laboratory of Metabolism and Regulation for Major Disease of Anhui Higher Education Institutes, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China.
| |
Collapse
|
10
|
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
|
11
|
Dragon J, Hoaglund M, Badireddy AR, Nielsen G, Schlezinger J, Shukla A. Perfluoroalkyl Substances (PFAS) Affect Inflammation in Lung Cells and Tissues. Int J Mol Sci 2023; 24:8539. [PMID: 37239886 PMCID: PMC10218140 DOI: 10.3390/ijms24108539] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Adverse lung outcomes from exposure to per-and polyfluoroalkyl substances (PFAS) are known; however, the mechanism of action is poorly understood. To explore this, human bronchial epithelial cells were grown and exposed to varied concentrations of short-chain (perfluorobutanoic acid, perflurobutane sulfonic acid and GenX) or long-chain (PFOA and perfluorooctane sulfonic acid (PFOS)) PFAS, alone or in a mixture to identify cytotoxic concentrations. Non-cytotoxic concentrations of PFAS from this experiment were selected to assess NLRP3 inflammasome activation and priming. We found that PFOA and PFOS alone or in a mixture primed and activated the inflammasome compared with vehicle control. Atomic force microscopy showed that PFOA but not PFOS significantly altered the membrane properties of cells. RNA sequencing was performed on the lungs of mice that had consumed PFOA in drinking water for 14 weeks. Wild type (WT), PPARα knock-out (KO) and humanized PPARα (KI) were exposed to PFOA. We found that multiple inflammation- and immune-related genes were affected. Taken together, our study demonstrated that PFAS exposure could alter lung biology in a significant manner and may contribute to asthma/airway hyper-responsiveness.
Collapse
Affiliation(s)
- Julie Dragon
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA; (J.D.); (M.H.); (A.R.B.); (G.N.); (J.S.)
| | - Michael Hoaglund
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA; (J.D.); (M.H.); (A.R.B.); (G.N.); (J.S.)
| | - Appala Raju Badireddy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA; (J.D.); (M.H.); (A.R.B.); (G.N.); (J.S.)
| | - Greylin Nielsen
- Department of Environmental Health, School of Public Health, Boston University, Boston, MA 02118, USA
| | - Jennifer Schlezinger
- Department of Environmental Health, School of Public Health, Boston University, Boston, MA 02118, USA
| | - Arti Shukla
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA; (J.D.); (M.H.); (A.R.B.); (G.N.); (J.S.)
| |
Collapse
|
12
|
Qian H, Ding Y, Deng X, Huang W, Li Z, Liu F, Zhang J, Wang L, Liu J, Yuan Y, Hou S, Chen X, Ma L. Synthesis-accessibility-oriented design of c-Jun N-terminal kinase 1 inhibitor. Eur J Med Chem 2023; 256:115442. [PMID: 37156184 DOI: 10.1016/j.ejmech.2023.115442] [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: 03/10/2023] [Revised: 04/21/2023] [Accepted: 04/30/2023] [Indexed: 05/10/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a severe and progressive lung disease with poor prognosis and limited treatment options. The c-Jun N-Terminal Kinase 1 (JNK1), a key component of the MAPK pathway, has been implicated in the pathogenesis of IPF and represents a potential therapeutic target. However, the development of JNK1 inhibitors has been slowed, partly due to synthetic complexity in medicinal chemistry modification. Here, we report a synthesis-accessibility-oriented strategy for designing JNK1 inhibitors based on computational prediction of synthetic feasibility and fragment-based molecule generation. This strategy led to the discovery of several potent JNK1 inhibitors, such as compound C6 (IC50 = 33.5 nM), which exhibited comparable activity to the clinical candidate CC-90001 (IC50 = 24.4 nM). The anti-fibrotic effect of C6 was further confirmed in animal model of pulmonary fibrosis. Moreover, compound C6 could be synthesized in only two steps, compared to nine steps for CC-90001. Our findings suggest that compound C6 is a promising lead for further optimization and development as a novel anti-fibrotic agent targeting JNK1. In addition, the discovery of C6 also demonstrates the feasibility of synthesis-accessibility-oriented strategy in lead discovery.
Collapse
Affiliation(s)
- Hewen Qian
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Yuanqing Ding
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Xingyu Deng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Weiwei Huang
- Hangzhou Matrix Biopharmaceutical Co., Ltd, Hangzhou, Zhejiang, 311121, China
| | - Zhenzhen Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Fengling Liu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Jie Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Lihui Wang
- Institute of Ageing Research, Hangzhou Normal University, School of Medicine, Hangzhou, Zhejiang Province, 311121, China
| | - Junping Liu
- Institute of Ageing Research, Hangzhou Normal University, School of Medicine, Hangzhou, Zhejiang Province, 311121, China
| | - Yaxia Yuan
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, Texas, 78229, USA
| | - Shurong Hou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| | - Xiabin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| | - Lei Ma
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China.
| |
Collapse
|
13
|
Ye Y, Gaudy A, Thomas M, Reyes J, Burkhardt B, Horan G, Liu L, Chen J, Ghosh A, Carayannopoulos LN, Tatosian DA, Palmisano M. Safety, Pharmacokinetics, and Pharmacodynamics of CC-90001 (BMS-986360), a c-Jun N-terminal Kinase Inhibitor, in Phase 1 Studies in Healthy Participants. Clin Pharmacol Drug Dev 2022; 11:1394-1404. [PMID: 36256505 PMCID: PMC10092235 DOI: 10.1002/cpdd.1178] [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: 02/24/2022] [Accepted: 08/29/2022] [Indexed: 01/28/2023]
Abstract
CC-90001 selectively inhibits c-Jun N-terminal kinase (JNK), a stress-activated protein implicated in fibrosis. In 3 phase 1 trials evaluating CC-90001 pharmacokinetics, pharmacodynamics, and safety, healthy adults (N = 184) received oral CC-90001 in a single dose (10-720 mg) or multiple doses (30-480 mg once daily for 7-18 days) or placebo. CC-90001 was rapidly absorbed (median time to maximum concentration, 1-4 hours) and eliminated with a mean terminal elimination half-life of 12-28 hours. Steady state was reached on day 5, with a mean accumulation ratio of 1.5- to 2-fold following daily dosing. Exposure was similar in fed versus fasted participants and in Japanese versus non-Japanese participants. CC-90001 demonstrated dose- and exposure-dependent inhibition of JNK as determined by histopathological analysis of c-Jun phosphorylation in ultraviolet-irradiated skin. The most common treatment-emergent adverse events were nausea and headache; all were mild or moderate in intensity. Based on exposure-response analysis using high-quality electrocardiogram data, no clinically relevant QT prolongation liability for CC-90001 was observed. Overall, single- and multiple-dose CC-90001 were generally safe and well tolerated at the tested doses and demonstrated JNK pathway engagement. These results support further clinical evaluation of CC-90001.
Collapse
Affiliation(s)
- Ying Ye
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | | | | | | | - Gerald Horan
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Liangang Liu
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Jian Chen
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | | | | | | |
Collapse
|
14
|
Xie Q, Liu R, Zou Z, Feng Y, Huang Y, Xu G, Sun W, Liang Y, Zhong W. MYPT1 inhibits the metastasis of renal clear cell carcinoma via the MAPK8/N-cadherin pathway. FEBS Open Bio 2022; 12:2083-2095. [PMID: 36106411 PMCID: PMC9623519 DOI: 10.1002/2211-5463.13487] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 08/23/2022] [Accepted: 09/14/2022] [Indexed: 01/25/2023] Open
Abstract
Myosin phosphatase target subunit 1 (MYPT1) is a subunit of myosin phosphatase that is capable of regulating smooth muscle contraction. MYPT1 has been reported to be involved in a wide variety of tumours, but its expression and biological functions in renal clear cell carcinoma (ccRCC) remain obscure. Herein, we analysed the relationship between patient clinicopathological characteristics and MYPT1 expression levels in ccRCC patients using a tissue microarray (TMA) and data retrieved from the TCGA-KIRC dataset. MYPT1 was overexpressed or depleted using siRNA in ccRCC cells to assess the effects on migration and invasion in vitro and in vivo. Additionally, RNA-sequencing and bioinformatics analysis were performed to investigate the precise mechanism. MYPT1 expression in ccRCC tissues was observed to be lower than that in nonmalignant tissues (P < 0.05). In addition, MYPT1 downregulation was closely linked to advanced pathological stage (P < 0.05), and poor OS (overall survival; P < 0.05). Functionally, increased expression of MYPT1 suppressed ccRCC migration and invasion in vitro, and inhibited tumour metastasis in vivo. In addition, MYPT1 overexpression exerted its suppressive effects via the MAPK8/N-cadherin pathway in ccRCC.
Collapse
Affiliation(s)
- Qingling Xie
- Guangdong Provincial Institute of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ren Liu
- Guangdong Provincial Institute of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Zhihao Zou
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yuanfa Feng
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yiqiao Huang
- Department of UrologyThe Fifth Affiliated Hospital of Guangzhou Medical UniversityChina
| | - Guibin Xu
- Department of UrologyThe Fifth Affiliated Hospital of Guangzhou Medical UniversityChina
| | - Wei Sun
- Department of Urology, Huadu District People's HospitalSouthern Medical UniversityGuangzhouChina
| | - Yuxiang Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Weide Zhong
- Guangdong Provincial Institute of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina,Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| |
Collapse
|
15
|
Fathimath Muneesa M, Barki RR, Shaikh SB, Bhandary YP. Curcumin intervention during progressive fibrosis controls inflammatory cytokines and the fibrinolytic system in pulmonary fibrosis. Toxicol Appl Pharmacol 2022; 449:116116. [PMID: 35716765 DOI: 10.1016/j.taap.2022.116116] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 11/30/2022]
Abstract
Persistent injuries and chronic inflammation paired with dysregulated healing process in the lungs leads to scarring and stiffening of the tissue leading to a condition called pulmonary fibrosis. There is no efficacious therapy against the condition because of the poorly understood pathophysiology of the disease. Curcumin is well known anti-inflammatory natural compound and is shown to have beneficial effects in many diseases. It is also reported to show antifibrotic activities in pulmonary fibrosis. There are evidences that fibrinolytic system plays a crucial role in the development of pulmonary fibrosis. We aimed to see whether curcumin could regulate inflammation and fibrinolysis in murine model of pulmonary fibrosis. We prepared BLM induced pulmonary fibrosis model by administering BLM at a dose of 2 mg/ kg bodyweight. Curcumin (75 mg/kg body wt) was instilled intraperitoneally on different time points. The effect of curcumin on inflammatory cytokines and fibrinolytic system was studied using molecular biology techniques like RT-PCR, western blot and immunohistochemistry/immunofluorescence. We observed that BLM brought changes in the expressions of components in the fibrinolytic system, i.e. BLM favoured fibrin deposition by increasing the expression of PAI-1 (plasminogen activator inhibitor) and decreasing the expression of uPA (Urokinase plasminogen activator) and uPAR (Urokinase plasminogen activator receptor). We also demonstrate that curcumin could restore the normal expression of fibrinolytic components, uPA, uPAR and PAI-1. Curcumin could also minimize the expression of key enzymes in tissue remodeling in pulmonary fibrosis, MMP-2 and MMP-9, which were elevated in the BLM treated group. Our data suggest that curcumin exerts an anti-inflammatory and antifibrotic effect in lungs. We highlight curcumin as a feasible adjuvant therapy option against pulmonary fibrosis.
Collapse
Affiliation(s)
- M Fathimath Muneesa
- Yenepoya Research Centre, Yenepoya University, Deralakatte, Mangalore 575018, Karnataka, India
| | - Rashmi R Barki
- Yenepoya Research Centre, Yenepoya University, Deralakatte, Mangalore 575018, Karnataka, India
| | - Sadiya Bi Shaikh
- Yenepoya Research Centre, Yenepoya University, Deralakatte, Mangalore 575018, Karnataka, India; Rahman Lab, Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, United States of America
| | - Yashodhar P Bhandary
- Yenepoya Research Centre, Yenepoya University, Deralakatte, Mangalore 575018, Karnataka, India.
| |
Collapse
|
16
|
van de Wetering C, Manuel AM, Sharafi M, Aboushousha R, Qian X, Erickson C, MacPherson M, Chan G, Adcock IM, ZounematKermani N, Schleich F, Louis R, Bohrnsen E, D'Alessandro A, Wouters EF, Reynaert NL, Li J, Wolf CR, Henderson CJ, Lundblad LKA, Poynter ME, Dixon AE, Irvin CG, van der Vliet A, van der Velden JL, Janssen-Heininger YM. Glutathione-S-transferase P promotes glycolysis in asthma in association with oxidation of pyruvate kinase M2. Redox Biol 2021; 47:102160. [PMID: 34624602 PMCID: PMC8502950 DOI: 10.1016/j.redox.2021.102160] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/02/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Interleukin-1-dependent increases in glycolysis promote allergic airways disease in mice and disruption of pyruvate kinase M2 (PKM2) activity is critical herein. Glutathione-S-transferase P (GSTP) has been implicated in asthma pathogenesis and regulates the oxidation state of proteins via S-glutathionylation. We addressed whether GSTP-dependent S-glutathionylation promotes allergic airways disease by promoting glycolytic reprogramming and whether it involves the disruption of PKM2. METHODS We used house dust mite (HDM) or interleukin-1β in C57BL6/NJ WT or mice that lack GSTP. Airway basal cells were stimulated with interleukin-1β and the selective GSTP inhibitor, TLK199. GSTP and PKM2 were evaluated in sputum samples of asthmatics and healthy controls and incorporated analysis of the U-BIOPRED severe asthma cohort database. RESULTS Ablation of Gstp decreased total S-glutathionylation and attenuated HDM-induced allergic airways disease and interleukin-1β-mediated inflammation. Gstp deletion or inhibition by TLK199 decreased the interleukin-1β-stimulated secretion of pro-inflammatory mediators and lactate by epithelial cells. 13C-glucose metabolomics showed decreased glycolysis flux at the pyruvate kinase step in response to TLK199. GSTP and PKM2 levels were increased in BAL of HDM-exposed mice as well as in sputum of asthmatics compared to controls. Sputum proteomics and transcriptomics revealed strong correlations between GSTP, PKM2, and the glycolysis pathway in asthma. CONCLUSIONS GSTP contributes to the pathogenesis of allergic airways disease in association with enhanced glycolysis and oxidative disruption of PKM2. Our findings also suggest a PKM2-GSTP-glycolysis signature in asthma that is associated with severe disease.
Collapse
Affiliation(s)
- Cheryl van de Wetering
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA; Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Allison M Manuel
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Mona Sharafi
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - Reem Aboushousha
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Xi Qian
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Cuixia Erickson
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Maximilian MacPherson
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Garrett Chan
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Ian M Adcock
- National Heart & Lung Institute & Data Science Institute, Imperial College London, UK
| | | | - Florence Schleich
- Department of Respiratory Medicine, CHU Sart-TilmanB35, Liege, Belgium
| | - Renaud Louis
- Department of Respiratory Medicine, CHU Sart-TilmanB35, Liege, Belgium
| | - Eric Bohrnsen
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| | - Emiel F Wouters
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
| | - Niki L Reynaert
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Jianing Li
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - C Roland Wolf
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Center, Ninewells Hospital Dundee DD1 9SY, Scotland, United Kingdom
| | - Colin J Henderson
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Center, Ninewells Hospital Dundee DD1 9SY, Scotland, United Kingdom
| | - Lennart K A Lundblad
- Meakins-Christie Laboratories, McGill University & THORASYS Thoracic Medical Systems Inc., Montréal, QC, Canada
| | - Matthew E Poynter
- Department of Medicine, College of Medicine, University of Vermont, Burlington, VT, USA
| | - Anne E Dixon
- Department of Medicine, College of Medicine, University of Vermont, Burlington, VT, USA
| | - Charles G Irvin
- Department of Medicine, College of Medicine, University of Vermont, Burlington, VT, USA
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Jos L van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | | |
Collapse
|
17
|
Aboushousha R, Elko E, Chia SB, Manuel AM, van de Wetering C, van der Velden J, MacPherson M, Erickson C, Reisz JA, D'Alessandro A, Wouters EFM, Reynaert NL, Lam YW, Anathy V, van der Vliet A, Seward DJ, Janssen-Heininger YMW. Glutathionylation chemistry promotes interleukin-1 beta-mediated glycolytic reprogramming and pro-inflammatory signaling in lung epithelial cells. FASEB J 2021; 35:e21525. [PMID: 33817836 DOI: 10.1096/fj.202002687rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 12/28/2022]
Abstract
Glycolysis is a well-known process by which metabolically active cells, such as tumor or immune cells meet their high metabolic demands. Previously, our laboratory has demonstrated that in airway epithelial cells, the pleiotropic cytokine, interleukin-1 beta (IL1B) induces glycolysis and that this contributes to allergic airway inflammation and remodeling. Activation of glycolysis is known to increase NADPH reducing equivalents generated from the pentose phosphate pathway, linking metabolic reprogramming with redox homeostasis. In addition, numerous glycolytic enzymes are known to be redox regulated. However, whether and how redox chemistry regulates metabolic reprogramming more generally remains unclear. In this study, we employed a multi-omics approach in primary mouse airway basal cells to evaluate the role of protein redox biochemistry, specifically protein glutathionylation, in mediating metabolic reprogramming. Our findings demonstrate that IL1B induces glutathionylation of multiple proteins involved in metabolic regulation, notably in the glycolysis pathway. Cells lacking Glutaredoxin-1 (Glrx), the enzyme responsible for reversing glutathionylation, show modulation of multiple metabolic pathways including an enhanced IL1B-induced glycolytic response. This was accompanied by increased secretion of thymic stromal lymphopoietin (TSLP), a cytokine important in asthma pathogenesis. Targeted inhibition of glycolysis prevented TSLP release, confirming the functional relevance of enhanced glycolysis in cells stimulated with IL1B. Collectively, data herein point to an intriguing link between glutathionylation chemistry and glycolytic reprogramming in epithelial cells and suggest that glutathionylation chemistry may represent a therapeutic target in pulmonary pathologies with perturbations in the glycolysis pathway.
Collapse
Affiliation(s)
- Reem Aboushousha
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Evan Elko
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Shi B Chia
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Allison M Manuel
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Cheryl van de Wetering
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Jos van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Maximilian MacPherson
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Cuixia Erickson
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO, USA
| | - Emiel F M Wouters
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands.,Ludwig Boltzmann Institute for Lung Research, Vienna, Austria
| | - Niki L Reynaert
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Ying-Wai Lam
- Department of Biology, University of Vermont College of Medicine, Burlington, VT, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - David J Seward
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | | |
Collapse
|
18
|
Ather JL, Van Der Vliet KE, Mank MM, Reed LF, Dixon AE, Poynter ME. Obese adipose tissue modulates proinflammatory responses of mouse airway epithelial cells. Am J Physiol Regul Integr Comp Physiol 2021; 321:R79-R90. [PMID: 34105399 DOI: 10.1152/ajpregu.00316.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although recognized as an important endocrine organ, little is known about the mechanisms through which adipose tissue can regulate inflammatory responses in distant tissues, such as lung that are affected by obesity. To explore potential mechanisms, male C57BL/6J mice were provided either high-fat diet, low-fat diet, or were provided a high-fat diet then switched to the low-fat diet to promote weight loss. Visceral adipocytes were then cultured in vitro to generate conditioned media (CM) that was used to treat both primary (mouse tracheal epithelial cells; MTECs) and immortalized (mouse-transformed club cells; MTCCs) airway epithelial cells. Adiponectin levels were greatly depressed in the CM from both obese and diet-switched adipocytes relative to mice continually fed the low-fat diet. MTECs from mice with obesity secreted higher baseline levels of inflammatory cytokines than MTECs from lean or diet-switched mice. MTECs treated with obese adipocyte CM increased their secretion of these cytokines compared with MTECs treated with lean CM. Diet-switched CM modestly decreased the production of cytokines compared with obese CM, and these effects were recapitulated when the CM was used to treat MTCCs. Adipose stromal vascular cells from mice with obesity expressed genes consistent with an M1 macrophage phenotype and decreased eosinophil abundance compared with lean stromal vascular fraction, a profile that persisted in the lean diet-switched mice despite substantial weight loss. Soluble factors secreted from obese adipocytes exert a proinflammatory effect on airway epithelial cells, and these alterations are attenuated by diet-induced weight loss, which could have implications for the airway dysfunction related to obese asthma and its mitigation by weight loss.
Collapse
Affiliation(s)
- Jennifer L Ather
- Division of Pulmonary Disease and Critical Care, Vermont Lung Center, Department of Medicine, The University of Vermont, Burlington, Vermont
| | - Katherine E Van Der Vliet
- Division of Pulmonary Disease and Critical Care, Vermont Lung Center, Department of Medicine, The University of Vermont, Burlington, Vermont
| | - Madeleine M Mank
- Division of Pulmonary Disease and Critical Care, Vermont Lung Center, Department of Medicine, The University of Vermont, Burlington, Vermont
| | - Leah F Reed
- Division of Pulmonary Disease and Critical Care, Vermont Lung Center, Department of Medicine, The University of Vermont, Burlington, Vermont
| | - Anne E Dixon
- Division of Pulmonary Disease and Critical Care, Vermont Lung Center, Department of Medicine, The University of Vermont, Burlington, Vermont
| | - Matthew E Poynter
- Division of Pulmonary Disease and Critical Care, Vermont Lung Center, Department of Medicine, The University of Vermont, Burlington, Vermont
| |
Collapse
|
19
|
Kasuya Y, Kim JD, Hatano M, Tatsumi K, Matsuda S. Pathophysiological Roles of Stress-Activated Protein Kinases in Pulmonary Fibrosis. Int J Mol Sci 2021; 22:ijms22116041. [PMID: 34204949 PMCID: PMC8199902 DOI: 10.3390/ijms22116041] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/22/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is one of the most symptomatic progressive fibrotic lung diseases, in which patients have an extremely poor prognosis. Therefore, understanding the precise molecular mechanisms underlying pulmonary fibrosis is necessary for the development of new therapeutic options. Stress-activated protein kinases (SAPKs), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38) are ubiquitously expressed in various types of cells and activated in response to cellular environmental stresses, including inflammatory and apoptotic stimuli. Type II alveolar epithelial cells, fibroblasts, and macrophages are known to participate in the progression of pulmonary fibrosis. SAPKs can control fibrogenesis by regulating the cellular processes and molecular functions in various types of lung cells (including cells of the epithelium, interstitial connective tissue, blood vessels, and hematopoietic and lymphoid tissue), all aspects of which remain to be elucidated. We recently reported that the stepwise elevation of intrinsic p38 signaling in the lungs is correlated with a worsening severity of bleomycin-induced fibrosis, indicating an importance of this pathway in the progression of pulmonary fibrosis. In addition, a transcriptome analysis of RNA-sequencing data from this unique model demonstrated that several lines of mechanisms are involved in the pathogenesis of pulmonary fibrosis, which provides a basis for further studies. Here, we review the accumulating evidence for the spatial and temporal roles of SAPKs in pulmonary fibrosis.
Collapse
Affiliation(s)
- Yoshitoshi Kasuya
- Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (M.H.); (S.M.)
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Correspondence: ; Tel.: +81-432-262-193; Fax: +81-432-262-196
| | - Jun-Dal Kim
- Department of Research and Development, Institute of Natural Medicine (INM), University of Toyama, Toyama 930-0194, Japan;
| | - Masahiko Hatano
- Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (M.H.); (S.M.)
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan;
| | - Shuichi Matsuda
- Department of Biomedical Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (M.H.); (S.M.)
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan;
| |
Collapse
|
20
|
Busch SM, Lorenzana Z, Ryan AL. Implications for Extracellular Matrix Interactions With Human Lung Basal Stem Cells in Lung Development, Disease, and Airway Modeling. Front Pharmacol 2021; 12:645858. [PMID: 34054525 PMCID: PMC8149957 DOI: 10.3389/fphar.2021.645858] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/29/2021] [Indexed: 12/18/2022] Open
Abstract
The extracellular matrix (ECM) is not simply a quiescent scaffold. This three-dimensional network of extracellular macromolecules provides structural, mechanical, and biochemical support for the cells of the lung. Throughout life, the ECM forms a critical component of the pulmonary stem cell niche. Basal cells (BCs), the primary stem cells of the airways capable of differentiating to all luminal cell types, reside in close proximity to the basolateral ECM. Studying BC-ECM interactions is important for the development of therapies for chronic lung diseases in which ECM alterations are accompanied by an apparent loss of the lung's regenerative capacity. The complexity and importance of the native ECM in the regulation of BCs is highlighted as we have yet to create an in vitro culture model that is capable of supporting the long-term expansion of multipotent BCs. The interactions between the pulmonary ECM and BCs are, therefore, a vital component for understanding the mechanisms regulating BC stemness during health and disease. If we are able to replicate these interactions in airway models, we could significantly improve our ability to maintain basal cell stemness ex vivo for use in in vitro models and with prospects for cellular therapies. Furthermore, successful, and sustained airway regeneration in an aged or diseased lung by small molecules, novel compounds or via cellular therapy will rely upon both manipulation of the airway stem cells and their immediate niche within the lung. This review will focus on the current understanding of how the pulmonary ECM regulates the basal stem cell function, how this relationship changes in chronic disease, and how replicating native conditions poses challenges for ex vivo cell culture.
Collapse
Affiliation(s)
- Shana M. Busch
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Zareeb Lorenzana
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
21
|
Schiffers C, Lundblad LKA, Hristova M, Habibovic A, Dustin CM, Daphtary N, Aliyeva M, Seward DJ, Janssen-Heininger YMW, Wouters EFM, Reynaert NL, van der Vliet A. Downregulation of DUOX1 function contributes to aging-related impairment of innate airway injury responses and accelerated senile emphysema. Am J Physiol Lung Cell Mol Physiol 2021; 321:L144-L158. [PMID: 33951398 DOI: 10.1152/ajplung.00021.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Aging is associated with a gradual loss of lung function due to increased cellular senescence, decreased regenerative capacity, and impaired innate host defense. One important aspect of innate airway epithelial host defense to nonmicrobial triggers is the secretion of alarmins such as IL-33 and activation of type 2 inflammation, which were previously found to depend on activation of the NADPH oxidase (NOX) homolog DUOX1, and redox-dependent signaling pathways that promote alarmin secretion. Here, we demonstrate that normal aging of C57BL/6J mice resulted in markedly decreased lung innate epithelial type 2 responses to exogenous triggers such as the airborne allergen Dermatophagoides pteronyssinus, which was associated with marked downregulation of DUOX1, as well as DUOX1-mediated redox-dependent signaling. DUOX1 deficiency was also found to accelerate age-related airspace enlargement and decline in lung function but did not consistently affect other features of lung aging such as senescence-associated inflammation. Intriguingly, observations of age-related DUOX1 downregulation and enhanced airspace enlargement due to DUOX1 deficiency in C57BL/6J mice, which lack a functional mitochondrial nicotinamide nucleotide transhydrogenase (NNT), were much less dramatic in C57BL/6NJ mice with normal NNT function, although the latter mice also displayed impaired innate epithelial injury responses with advancing age. Overall, our findings indicate a marked aging-dependent decline in (DUOX1-dependent) innate airway injury responses to external nonmicrobial triggers, but the impact of aging on DUOX1 downregulation and its significance for age-related senile emphysema development was variable between different C57BL6 substrains, possibly related to metabolic alterations due to differences in NNT function.
Collapse
Affiliation(s)
- Caspar Schiffers
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont.,Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Lennart K A Lundblad
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Milena Hristova
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Aida Habibovic
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Christopher M Dustin
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Nirav Daphtary
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Minara Aliyeva
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - David J Seward
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Yvonne M W Janssen-Heininger
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Emiel F M Wouters
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands.,Ludwig Boltzman Institute for Lung Health, Vienna, Austria
| | - Niki L Reynaert
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
22
|
Morgan EL, Scarth JA, Patterson MR, Wasson CW, Hemingway GC, Barba-Moreno D, Macdonald A. E6-mediated activation of JNK drives EGFR signalling to promote proliferation and viral oncoprotein expression in cervical cancer. Cell Death Differ 2021; 28:1669-1687. [PMID: 33303976 PMCID: PMC8166842 DOI: 10.1038/s41418-020-00693-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Human papillomaviruses (HPV) are a major cause of malignancy worldwide, contributing to ~5% of all human cancers including almost all cases of cervical cancer and a growing number of ano-genital and oral cancers. HPV-induced malignancy is primarily driven by the viral oncogenes, E6 and E7, which manipulate host cellular pathways to increase cell proliferation and enhance cell survival, ultimately predisposing infected cells to malignant transformation. Consequently, a more detailed understanding of viral-host interactions in HPV-associated disease offers the potential to identify novel therapeutic targets. Here, we identify that the c-Jun N-terminal kinase (JNK) signalling pathway is activated in cervical disease and in cervical cancer. The HPV E6 oncogene induces JNK1/2 phosphorylation in a manner that requires the E6 PDZ binding motif. We show that blockade of JNK1/2 signalling using small molecule inhibitors, or knockdown of the canonical JNK substrate c-Jun, reduces cell proliferation and induces apoptosis in cervical cancer cells. We further demonstrate that this phenotype is at least partially driven by JNK-dependent activation of EGFR signalling via increased expression of EGFR and the EGFR ligands EGF and HB-EGF. JNK/c-Jun signalling promoted the invasive potential of cervical cancer cells and was required for the expression of the epithelial to mesenchymal transition (EMT)-associated transcription factor Slug and the mesenchymal marker Vimentin. Furthermore, JNK/c-Jun signalling is required for the constitutive expression of HPV E6 and E7, which are essential for cervical cancer cell growth and survival. Together, these data demonstrate a positive feedback loop between the EGFR signalling pathway and HPV E6/E7 expression, identifying a regulatory mechanism in which HPV drives EGFR signalling to promote proliferation, survival and EMT. Thus, our study has identified a novel therapeutic target that may be beneficial for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Ethan L. Morgan
- grid.9909.90000 0004 1936 8403School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire LS2 9JT UK ,grid.9909.90000 0004 1936 8403Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire LS2 9JT UK ,grid.94365.3d0000 0001 2297 5165Present Address: Tumor Biology Section, Head and Neck Surgery Branch, National Institute of Deafness and Other Communication Disorders, National Institute of Health, Bethesda, MD USA
| | - James A. Scarth
- grid.9909.90000 0004 1936 8403School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire LS2 9JT UK ,grid.9909.90000 0004 1936 8403Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire LS2 9JT UK
| | - Molly R. Patterson
- grid.9909.90000 0004 1936 8403School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire LS2 9JT UK ,grid.9909.90000 0004 1936 8403Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire LS2 9JT UK
| | - Christopher W. Wasson
- grid.9909.90000 0004 1936 8403School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire LS2 9JT UK ,grid.9909.90000 0004 1936 8403Present Address: Leeds Institute of Rheumatic and Musculoskeletal Medicine, School of Medicine, University of Leeds, St-James University Teaching Hospital, Leeds, West Yorkshire UK
| | - Georgia C. Hemingway
- grid.9909.90000 0004 1936 8403School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire LS2 9JT UK
| | - Diego Barba-Moreno
- grid.9909.90000 0004 1936 8403School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire LS2 9JT UK ,grid.9909.90000 0004 1936 8403Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire LS2 9JT UK
| | - Andrew Macdonald
- grid.9909.90000 0004 1936 8403School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire LS2 9JT UK ,grid.9909.90000 0004 1936 8403Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire LS2 9JT UK
| |
Collapse
|
23
|
Cyano Enone-Bearing Triterpenoid Soloxolone Methyl Inhibits Epithelial-Mesenchymal Transition of Human Lung Adenocarcinoma Cells In Vitro and Metastasis of Murine Melanoma In Vivo. Molecules 2020; 25:molecules25245925. [PMID: 33327637 PMCID: PMC7765109 DOI: 10.3390/molecules25245925] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/04/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Introduction of α-cyano α,β-unsaturated carbonyl moiety into natural cyclic compounds markedly improves their bioactivities, including inhibitory potential against tumor growth and metastasis. Previously, we showed that cyano enone-bearing derivatives of 18βH-glycyrrhetinic (GA) and deoxycholic acids displayed marked cytotoxicity in different tumor cell lines. Moreover, GA derivative soloxolone methyl (SM) was found to induce ER stress and apoptosis in tumor cells in vitro and inhibit growth of carcinoma Krebs-2 in vivo. In this work, we studied the effects of these compounds used in non-toxic dosage on the processes associated with metastatic potential of tumor cells. Performed screening revealed SM as a hit compound, which inhibits motility of murine melanoma B16 and human lung adenocarcinoma A549 cells and significantly suppresses colony formation of A549 cells. Further study showed that SM effectively blocked transforming growth factor β (TGF-β)-induced epithelial-mesenchymal transition (EMT) of A549 cells: namely, inhibited TGF-β-stimulated motility and invasion of tumor cells as well as loss of their epithelial characteristics, such as, an acquisition of spindle-like phenotype, up- and down-regulation of mesenchymal (vimentin, fibronectin) and epithelial (E-cadherin, zona occludens-1 (ZO-1)) markers, respectively. Network pharmacology analysis with subsequent verification by molecular modeling revealed that matrix metalloproteinases MMP-2/-9 and c-Jun N-terminal protein kinase 1 (JNK1) can be considered as hypothetical primary targets of SM, mediating its marked anti-EMT activity. The inhibitory effect of SM on EMT revealed in vitro was further confirmed in a metastatic model of murine B16 melanoma: SM was found to effectively block metastatic dissemination of melanoma B16 cells in vivo, increase expression of E-cadherin and suppress expression of MMP-9 in lung metastatic foci. Altogether, our data provided valuable information for a better understanding of the antitumor activity of cyano enone-bearing semisynthetic compounds and revealed SM as a promising anti-metastatic drug candidate.
Collapse
|
24
|
Chia SB, Nolin JD, Aboushousha R, Erikson C, Irvin CG, Poynter ME, van der Velden J, Taatjes DJ, van der Vliet A, Anathy V, Janssen-Heininger YMW. Glutaredoxin deficiency promotes activation of the transforming growth factor beta pathway in airway epithelial cells, in association with fibrotic airway remodeling. Redox Biol 2020; 37:101720. [PMID: 32971362 PMCID: PMC7509797 DOI: 10.1016/j.redox.2020.101720] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
S-glutathionylation of reactive protein cysteines is a post-translational event that plays a critical role in transducing signals from oxidants into biological responses. S-glutathionylation can be reversed by the deglutathionylating enzyme glutaredoxin (GLRX). We have previously demonstrated that ablation of Glrx sensitizes mice to the development of parenchymal lung fibrosis(1). It remains unclear whether GLRX also controls airway fibrosis, a clinical feature relevant to asthma and chronic obstructive pulmonary disease, and whether GLRX controls the biology of airway epithelial cells, which have been implicated in the pathophysiology of these diseases. In the present study we utilized a house dust mite (HDM) model of allergic airway disease in wild type (WT) and Glrx-/- mice on a C57BL/6 background prone to develop airway fibrosis, and tracheal basal stem cells derived from WT mice, global Glrx-/- mice, or bi-transgenic mice allowing conditional ablation of the Glrx gene. Herein we show that absence of Glrx led to enhanced HDM-induced collagen deposition, elevated levels of transforming growth factor beta 1 (TGFB1) in the bronchoalveolar lavage, and resulted in increases in airway hyperresponsiveness. Airway epithelial cells isolated from Glrx-/- mice or following conditional ablation of Glrx showed spontaneous increases in secretion of TGFB1. Glrx-/- basal cells also showed spontaneous TGFB pathway activation, in association with increased expression of mesenchymal genes, including collagen 1a1 and fibronectin. Overall, these findings suggest that GLRX regulates airway fibrosis via a mechanism(s) that involve the plasticity of basal cells, the stem cells of the airways.
Collapse
Affiliation(s)
- Shi B Chia
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, 05405, USA
| | - James D Nolin
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, 05405, USA; Center for Genes, Environment and Health, National Jewish Health, Denver, CO, 80206, USA
| | - Reem Aboushousha
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, 05405, USA
| | - Cuixia Erikson
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, 05405, USA
| | - Charles G Irvin
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, 05405, USA
| | - Matthew E Poynter
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, 05405, USA
| | - Jos van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, 05405, USA
| | - Douglas J Taatjes
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, 05405, USA
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, 05405, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, 05405, USA
| | - Yvonne M W Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, 05405, USA.
| |
Collapse
|
25
|
EGF-mediated reduced miR-92a-1-5p controls HTR-8/SVneo cell invasion through activation of MAPK8 and FAS which in turn increase MMP-2/-9 expression. Sci Rep 2020; 10:12274. [PMID: 32703964 PMCID: PMC7378053 DOI: 10.1038/s41598-020-68966-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/03/2020] [Indexed: 02/08/2023] Open
Abstract
The members of human miR-17-92 cluster are implicated in several cancers and are known to increase cancer cells invasiveness. The present study reports reduced expression of miR-92a-1-5p in EGF treated HTR-8/SVneo trophoblastic cells by NGS and qRT-PCR. Overexpression of miR-92a-1-5p led to significantly reduced EGF-mediated HTR-8/SVneo cells invasion. MAPK8 and FAS were predicted to be miR-92a-1-5p targets, and confirmed to be reduced by qRT-PCR and Western blotting in trophoblast cells overexpressing miR-92a-1-5p. The binding of miR-92a-1-5p to MAPK8 and FAS 3'-UTR was confirmed by Luciferase reporter assay and Rescue assay. EGF increases MMP-2 & MMP-9 expression and reduces TIMP1 expression in HTR-8/SVneo cells. Inhibition of MAPK8 (by SP600125) reduced EGF-mediated MMP-9/TIMP1 ratio and invasion. Similarly, silencing of FAS by siRNA reduced EGF-mediated MMP-2/TIMP1 ratio and invasion. Treatment of HTR-8/SVneo cells with STAT1/3 inhibitors or siRNAs led to loss of EGF-mediated reduction in miR-92a-1-5p levels. Inserting the predicted binding sites of STAT3 present in promoter region of miR-92a-1-5p upstream of Luciferase promoter reduced its expression in presence of STAT3 expression vector. Thus, EGF leads to reduced miR-92a-1-5p expression which may be regulated by STAT1/STAT3 and controls HTR-8/SVneo cells invasion by targeting MAPK8 and FAS, which in turn increases MMP-2/MMP-9 expression.
Collapse
|
26
|
Spagnolo P, Bonella F, Ryerson CJ, Tzouvelekis A, Maher TM. Shedding light on developmental drugs for idiopathic pulmonary fibrosis. Expert Opin Investig Drugs 2020; 29:797-808. [PMID: 32538186 DOI: 10.1080/13543784.2020.1782885] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is an age-related disease of unknown cause. The disease is characterized by relentless scarring of the lung parenchyma resulting in respiratory failure and death. Two antifibrotic drugs (pirfenidone and nintedanib) are approved for the treatment of IPF worldwide, but they do not offer a cure and are associated with tolerability issues. Owing to its high unmet medical need, IPF is an area of dynamic research activity. AREAS COVERED There is a growing portfolio of novel therapies that target different pathways involved in the complex pathogenesis of IPF. In this review, we discuss the mechanisms of action and available data for compounds in the most advanced stages of clinical development. We searched PubMed for articles on this topic published from 1 January 2000, to 6 June 2020. EXPERT OPINION The approval of pirfenidone and nintedanib has fueled IPF drug discovery and development. New drugs are likely to reach the clinic in the near future. However, numerous challenges remain; the lack of animal models that reproduce the complexity of human disease and the poor translation of preclinical and early-phase positive effects to late stage clinical trials must be tackled.
Collapse
Affiliation(s)
- Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac Thoracic, Vascular Sciences and Public Health, University of Padova , Padova, Italy
| | - Francesco Bonella
- Center for Interstitial and Rare Lung Diseases, Ruhrlandklinik University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Christopher J Ryerson
- Department of Medicine, University of British Columbia and Centre for Heart Lung Innovation, St Paul's Hospital , Vancouver, Canada
| | - Argyris Tzouvelekis
- Department of Pneumology, Medical School, National and Kapodistrian University of Athens , Athens, Greece
| | - Toby M Maher
- NIHR Respiratory Clinical Research Facility, Royal Brompton Hospital , London, UK.,National Heart and Lung Institute, Imperial College, Sir Alexander Fleming Building , London, UK
| |
Collapse
|
27
|
Duong MTH, Lee JH, Ahn HC. C-Jun N-terminal kinase inhibitors: Structural insight into kinase-inhibitor complexes. Comput Struct Biotechnol J 2020; 18:1440-1457. [PMID: 32637042 PMCID: PMC7327381 DOI: 10.1016/j.csbj.2020.06.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/07/2020] [Accepted: 06/07/2020] [Indexed: 12/12/2022] Open
Abstract
The activation of c-Jun N-terminal kinases (JNKs) plays an important role in physiological processes including neuronal function, immune activity, and development, and thus, JNKs have been a therapeutic target for various diseases such as neurodegenerative diseases, inflammation, and cancer. Efforts to develop JNK-specific inhibitors have been ongoing for several decades. In this process, the structures of JNK in complex with various inhibitors have contributed greatly to the design of novel compounds and to the elucidation of structure-activity relationships. Almost 100 JNK structures with various compounds have been determined. Here we summarize the information gained from these structures and classify the inhibitors into several groups based on the binding mode. These groups include inhibitors in the open conformation and closed conformation of the gatekeeper residue, non-ATP site binders, peptides, covalent inhibitors, and type II kinase inhibitors. Through this work, deep insight into the interaction of inhibitors with JNKs can be gained and this will be helpful for developing novel, potent, and selective inhibitors.
Collapse
Affiliation(s)
- Men Thi Hoai Duong
- Department of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi 10326, South Korea
| | - Joon-Hwa Lee
- Department of Chemistry and RINS, Gyeongsang National University, Jinju, Gyeongnam 52828, South Korea
| | - Hee-Chul Ahn
- Department of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi 10326, South Korea
| |
Collapse
|
28
|
Shimada C, Xu R, Al-Alem L, Stasenko M, Spriggs DR, Rueda BR. Galectins and Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12061421. [PMID: 32486344 PMCID: PMC7352943 DOI: 10.3390/cancers12061421] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is known for its aggressive pathological features, including the capacity to undergo epithelial to mesenchymal transition, promoting angiogenesis, metastatic potential, chemoresistance, inhibiting apoptosis, immunosuppression and promoting stem-like features. Galectins, a family of glycan-binding proteins defined by a conserved carbohydrate recognition domain, can modulate many of these processes, enabling them to contribute to the pathology of ovarian cancer. Our goal herein was to review specific galectin members identified in the context of ovarian cancer, with emphasis on their association with clinical and pathological features, implied functions, diagnostic or prognostic potential and strategies being developed to disrupt their negative actions.
Collapse
Affiliation(s)
- Chisa Shimada
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA; (C.S.); (R.X.); (L.A.-A.); (D.R.S.)
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Rui Xu
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA; (C.S.); (R.X.); (L.A.-A.); (D.R.S.)
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Linah Al-Alem
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA; (C.S.); (R.X.); (L.A.-A.); (D.R.S.)
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Marina Stasenko
- Gynecology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York City, NY 10065, USA;
| | - David R. Spriggs
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA; (C.S.); (R.X.); (L.A.-A.); (D.R.S.)
- Department of Hematology/Medical Oncology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Bo R. Rueda
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA; (C.S.); (R.X.); (L.A.-A.); (D.R.S.)
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA
- Correspondence:
| |
Collapse
|
29
|
Degradation of different molecular weight fucoidans and their inhibition of TGF-β1 induced epithelial-mesenchymal transition in mouse renal tubular epithelial cells. Int J Biol Macromol 2020; 151:545-553. [PMID: 32057857 DOI: 10.1016/j.ijbiomac.2020.02.068] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/31/2020] [Accepted: 02/08/2020] [Indexed: 12/12/2022]
Abstract
In order to investigate the anti-fibrotic effect of different molecular weight (Mw) fucoidans on TGF-β1-induced mouse renal tubular epithelial cell (MTEC) mode. Oxidative degradation method was used to obtain fucoidans with different molecular weights and the reaction time, reaction temperature and the concentration of oxidants were investigated. Cell viability was detected by CCK-8, and EMT markers expression was detected by Western-bolt and Cell immunofluorescence assay. As a result, after chemical analysis of three independent batches of prepared samples, one batch of fucoidan sample (LHX 1-9) which chemical contents are similar but Mw ranging from 3.3 KDa to 49.3 KDa were selected to do further research. We found LHX1 (Mw = 3.3 KDa) and LHX 3-9 (Mw = 6.6 KDa, 8.3 KDa, 11.3 KDa, 14.9 KDa, 25.2 KDa, 35.4 KDa, 49.3 KDa) could resist the TGF-β1-induced depithelial-mesenchymal transition (EMT) by decreased expression of Fn and CTGF and maintained epithelial cell morphology in MTEC. However, the relationship between the Mw of fucoidans and their anti-EMT effect is not simply linear. Among the samples, LHX 1, 5 and 8 showed significant anti-EMT effects than others by de-regulated Fn and CTGF expression on MTEC cells.
Collapse
|
30
|
van der Velden JL, Alcorn JF, Chapman DG, Lundblad LKA, Irvin CG, Davis RJ, Butnor K, Janssen-Heininger YMW. Airway epithelial specific deletion of Jun-N-terminal kinase 1 attenuates pulmonary fibrosis in two independent mouse models. PLoS One 2020; 15:e0226904. [PMID: 31935227 PMCID: PMC6959564 DOI: 10.1371/journal.pone.0226904] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/07/2019] [Indexed: 01/06/2023] Open
Abstract
The stress-induced kinase, c-Jun-N-terminal kinase 1 (JNK1) has previously been implicated in the pathogenesis of lung fibrosis. However, the exact cell type(s) wherein JNK1 exerts its pro-fibrotic role(s) remained enigmatic. Herein we demonstrate prominent activation of JNK in bronchial epithelia using the mouse models of bleomycin- or AdTGFβ1-induced fibrosis. Furthermore, in lung tissues of patients with idiopathic pulmonary fibrosis (IPF), active JNK was observed in various regions including type I and type II pneumocytes and fibroblasts. No JNK activity was observed in adjacent normal tissue or in normal control tissue. To address the role of epithelial JNK1, we ablated Jnk1 form bronchiolar and alveolar type II epithelial cells using CCSP-directed Cre recombinase-mediated ablation of LoxP-flanked Jnk1 alleles. Our results demonstrate that ablation of Jnk1 from airway epithelia resulted in a strong protection from bleomycin- or adenovirus expressing active transforming growth factor beta-1 (AdTGFβ1)-induced fibrosis. Ablation of the Jnk1 allele at a time when collagen increases were already present showed a reversal of existing increases in collagen content. Epithelial Jnk1 ablation resulted in attenuation of mesenchymal genes and proteins in lung tissue and preserved expression of epithelial genes. Collectively, these data suggest that epithelial JNK1 contributes to the pathogenesis of pulmonary fibrosis. Given the presence of active JNK in lungs from patients with IPF, targeting JNK1 in airway epithelia may represent a potential treatment strategy to combat this devastating disease.
Collapse
Affiliation(s)
- Jos L. van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - John F. Alcorn
- Children’s Hospital of Pittsburgh University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - David G. Chapman
- Departments of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Lennart K. A. Lundblad
- Departments of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Charles G. Irvin
- Departments of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Roger J. Davis
- Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Kelly Butnor
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Yvonne M. W. Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, United States of America
- * E-mail:
| |
Collapse
|
31
|
van de Wetering C, Aboushousha R, Manuel AM, Chia SB, Erickson C, MacPherson MB, van der Velden JL, Anathy V, Dixon AE, Irvin CG, Poynter ME, van der Vliet A, Wouters EFM, Reynaert NL, Janssen-Heininger YMW. Pyruvate Kinase M2 Promotes Expression of Proinflammatory Mediators in House Dust Mite-Induced Allergic Airways Disease. THE JOURNAL OF IMMUNOLOGY 2020; 204:763-774. [PMID: 31924651 DOI: 10.4049/jimmunol.1901086] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022]
Abstract
Asthma is a chronic disorder characterized by inflammation, mucus metaplasia, airway remodeling, and hyperresponsiveness. We recently showed that IL-1-induced glycolytic reprogramming contributes to allergic airway disease using a murine house dust mite model. Moreover, levels of pyruvate kinase M2 (PKM2) were increased in this model as well as in nasal epithelial cells from asthmatics as compared with healthy controls. Although the tetramer form of PKM2 converts phosphoenolpyruvate to pyruvate, the dimeric form of PKM2 has alternative, nonglycolysis functions as a transcriptional coactivator to enhance the transcription of several proinflammatory cytokines. In the current study, we examined the impact of PKM2 on the pathogenesis of house dust mite-induced allergic airways disease in C57BL/6NJ mice. We report, in this study, that activation of PKM2, using the small molecule activator, TEPP46, augmented PKM activity in lung tissues and attenuated airway eosinophils, mucus metaplasia, and subepithelial collagen. TEPP46 attenuated IL-1β-mediated airway inflammation and expression of proinflammatory mediators. Exposure to TEPP46 strongly decreased the IL-1β-mediated increases in thymic stromal lymphopoietin (TSLP) and GM-CSF in primary tracheal epithelial cells isolated from C57BL/6NJ mice. We also demonstrate that IL-1β-mediated increases in nuclear phospho-STAT3 were decreased by TEPP46. Finally, STAT3 inhibition attenuated the IL-1β-induced release of TSLP and GM-CSF, suggesting that the ability of PKM2 to phosphorylate STAT3 contributes to its proinflammatory function. Collectively, these results demonstrate that the glycolysis-inactive form of PKM2 plays a crucial role in the pathogenesis of allergic airways disease by increasing IL-1β-induced proinflammatory signaling, in part, through phosphorylation of STAT3.
Collapse
Affiliation(s)
- Cheryl van de Wetering
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405.,Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6211 LK Maastricht, the Netherlands; and
| | - Reem Aboushousha
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Allison M Manuel
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Shi B Chia
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Cuixia Erickson
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Maximilian B MacPherson
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Jos L van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Anne E Dixon
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Charles G Irvin
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Matthew E Poynter
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Emiel F M Wouters
- Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6211 LK Maastricht, the Netherlands; and
| | - Niki L Reynaert
- Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6211 LK Maastricht, the Netherlands; and
| | - Yvonne M W Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405;
| |
Collapse
|
32
|
Guigni BA, van der Velden J, Kinsey CM, Carson JA, Toth MJ. Effects of conditioned media from murine lung cancer cells and human tumor cells on cultured myotubes. Am J Physiol Endocrinol Metab 2020; 318:E22-E32. [PMID: 31689144 PMCID: PMC6985792 DOI: 10.1152/ajpendo.00310.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Factors secreted from tumors/tumor cells are hypothesized to cause skeletal muscle wasting in cancer patients. We examined whether cancer cells secrete factors to promote atrophy by evaluating the effects of conditioned media (CM) from murine lung cancer cells and primary cultures of human lung tumor cells on cultured myotubes. We evaluated murine Lewis lung carcinoma (LLC) and KRASG12D cells, and primary cell lines derived from tumor biopsies from patients with lung cancer (hTCM; n = 6). In all experiments, serum content was matched across treatment groups. We hypothesized that CM from murine and human tumor cells would reduce myotube myosin content, decrease mitochondrial content, and increase mitochondrial reactive oxygen species (ROS) production. Treatment of myotubes differentiated for 7 days with CM from LLC and KRASG12D cells did not alter any of these variables. Effects of murine tumor cell CM were observed when myotubes differentiated for 4 days were treated with tumor cell CM and compared with undiluted differentiation media. However, these effects were not apparent if tumor cell CM treatments were compared with control cell CM or dilution controls. Finally, CM from human lung tumor primary cell lines did not modify myosin content or mitochondrial content or ROS production compared with either undiluted differentiated media, control cell CM, or dilution controls. Our results do not support the hypothesis that factors released from cultured lung cancer/tumor cells promote myotube wasting or mitochondrial abnormalities, but we cannot dismiss the possibility that these cells could secrete such factors in vivo within the native tumor microenvironment.
Collapse
MESH Headings
- Adenocarcinoma/metabolism
- Aged
- Aged, 80 and over
- Animals
- Cachexia/etiology
- Cachexia/metabolism
- Carcinoma, Lewis Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Squamous Cell/metabolism
- Cell Line, Tumor
- Culture Media, Conditioned/pharmacology
- Female
- Humans
- Lung Neoplasms/metabolism
- Male
- Mice
- Middle Aged
- Mitochondria, Muscle/drug effects
- Mitochondria, Muscle/metabolism
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Myoblasts, Skeletal
- Myosins/metabolism
- Neoplasms/complications
- Neoplasms/metabolism
- Primary Cell Culture
- Reactive Oxygen Species/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Blas A Guigni
- Department of Medicine, College of Medicine, University of Vermont, Burlington, Vermont
- Department of Molecular Physiology and Biophysics, College of Medicine, University of Vermont, Burlington, Vermont
| | - Jos van der Velden
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - C Matthew Kinsey
- Department of Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - James A Carson
- Integrative Muscle Biology Laboratory, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Michael J Toth
- Department of Medicine, College of Medicine, University of Vermont, Burlington, Vermont
- Department of Molecular Physiology and Biophysics, College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
33
|
Tripathi V, Shin JH, Stuelten CH, Zhang YE. TGF-β-induced alternative splicing of TAK1 promotes EMT and drug resistance. Oncogene 2019; 38:3185-3200. [PMID: 30626936 PMCID: PMC6486402 DOI: 10.1038/s41388-018-0655-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/20/2018] [Accepted: 12/07/2018] [Indexed: 02/07/2023]
Abstract
Transforming growth factor-β (TGF-β) is major inducer of epithelial to mesenchymal transition (EMT), which associates with cancer cell metastasis and resistance to chemotherapy and targeted drugs, through both transcriptional and non-transcriptional mechanisms. We previously reported that in cancer cells, heightened mitogenic signaling allows TGF-β-activated Smad3 to interact with poly(RC) binding protein 1 (PCBP1) and together they regulate many alternative splicing events that favors expression of protein isoforms essential for EMT, cytoskeletal rearrangement, and adherens junction signaling. Here, we show that the exclusion of TGF-β-activated kinase 1 (TAK1) variable exon 12 requires another RNA-binding protein, Fox-1 homolog 2 (Rbfox2), which binds intronic sequences in front of exon 12 independently of the Smad3-PCBP1 complex. Functionally, exon 12-excluded TAK1∆E12 and full length TAK1FL are distinct. The short isoform TAK1∆E12 is constitutively active and supports TGF-β-induced EMT and nuclear factor kappa B (NF-κB) signaling, whereas the full-length isoform TAK1FL promotes TGF-β-induced apoptosis. These observations offer a harmonious explanation for how a single TAK1 kinase can mediate the opposing responses of cell survival and apoptosis in response to TGF-β. They also reveal a propensity of the alternatively spliced TAK1 isoform TAK1∆E12 to cause drug resistance due to its activity in supporting EMT and NF-κB survival signaling.
Collapse
Affiliation(s)
- Veenu Tripathi
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jee-Hye Shin
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Christina H Stuelten
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ying E Zhang
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
34
|
Li Z, Liu X, Tian F, Li J, Wang Q, Gu C. MKP2 inhibits TGF-β1-induced epithelial-to-mesenchymal transition in renal tubular epithelial cells through a JNK-dependent pathway. Clin Sci (Lond) 2018; 132:2339-2355. [PMID: 30322849 DOI: 10.1042/cs20180602] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/13/2018] [Accepted: 10/15/2018] [Indexed: 02/08/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is a phenotypic conversion that plays a crucial role in renal fibrosis leading to chronic renal failure. Mitogen-activated protein kinase phosphatase 2 (MKP2) is a member of the dual-specificity MKPs that regulate the MAP kinase pathway involved in transforming growth factor-β1 (TGF-β1)-induced EMT. However, the function of MKP2 in the regulation of EMT and the underlying mechanisms are still largely unknown. In the present study, we detected the expression of MKP2 in an animal model of renal fibrosis and evaluated the potential role of MKP2 in tubular EMT induced by TGF-β1. We found that the expression of MKP2 was up-regulated in the tubular epithelial of unilateral ureter obstruction rats. Meanwhile, we also demonstrated that TGF-β1 up-regulated MKP2 expression in NRK-52E cells during their EMT phenotype acquisition. Importantly, overexpression of MKP2 inhibited c-Jun amino terminal kinase (JNK) signaling and partially reversed EMT induced by TGF-β1. Moreover, reducing MKP2 expression enhanced JNK phosphorylation, promoted the E-cadherin suppression and induced α-SMA expression and fibronectin secretion in response to TGF-β1, which could be rescued by a JNK inhibitor. These results provide the first evidence that MKP2 is a negative feedback molecule induced by TGF-β1, and MKP2 overexpression inhibits TGF-β1-induced EMT through the JNK signaling pathway. MKP2 could be a promising target to be used in gene therapy for renal fibrosis.
Collapse
Affiliation(s)
- Zhenzhen Li
- Department of Nephrology, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xianghua Liu
- Pathological Experiment Center, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Fengyan Tian
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ji Li
- Pediatric Urology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingwei Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chaohui Gu
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
35
|
Jemaà M, Abassi Y, Kifagi C, Fezai M, Daams R, Lang F, Massoumi R. Reversine inhibits Colon Carcinoma Cell Migration by Targeting JNK1. Sci Rep 2018; 8:11821. [PMID: 30087398 PMCID: PMC6081478 DOI: 10.1038/s41598-018-30251-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 07/26/2018] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer is one of the most commonly diagnosed cancers and the third most common cause of cancer-related death. Metastasis is the leading reason for the resultant mortality of these patients. Accordingly, development and characterization of novel anti-cancer drugs limiting colorectal tumor cell dissemination and metastasis are needed. In this study, we found that the small molecule Reversine reduces the migration potential of human colon carcinoma cells in vitro. A coupled kinase assay with bio-informatics approach identified the c-Jun N-terminal kinase (JNK) cascade as the main pathway inhibited by Reversine. Knockdown experiments and pharmacological inhibition identified JNK1 but not JNK2, as a downstream effector target in cancer cell migration. Xenograft experiments confirm the effect of JNK inhibition in the metastatic potential of colon cancer cells. These results highlight the impact of individual JNK isoforms in cancer cell metastasis and propose Reversine as a novel anti-cancer molecule for treatment of colon cancer patients.
Collapse
Affiliation(s)
- Mohamed Jemaà
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, 22381, Sweden. .,Department of Physiology I, Tübingen University, Gmelinstr. 5, D-72076, Tübingen, Germany.
| | - Yasmin Abassi
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, 22381, Sweden
| | - Chamseddine Kifagi
- Division of Immunology and Vaccinology, Technical University of Denmark, Copenhagen, Denmark
| | - Myriam Fezai
- Department of Physiology I, Tübingen University, Gmelinstr. 5, D-72076, Tübingen, Germany
| | - Renée Daams
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, 22381, Sweden
| | - Florian Lang
- Department of Physiology I, Tübingen University, Gmelinstr. 5, D-72076, Tübingen, Germany. .,Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany.
| | - Ramin Massoumi
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, Lund, 22381, Sweden.
| |
Collapse
|
36
|
Qian X, Aboushousha R, van de Wetering C, Chia SB, Amiel E, Schneider RW, van der Velden JLJ, Lahue KG, Hoagland DA, Casey DT, Daphtary N, Ather JL, Randall MJ, Aliyeva M, Black KE, Chapman DG, Lundblad LKA, McMillan DH, Dixon AE, Anathy V, Irvin CG, Poynter ME, Wouters EFM, Vacek PM, Henket M, Schleich F, Louis R, van der Vliet A, Janssen-Heininger YMW. IL-1/inhibitory κB kinase ε-induced glycolysis augment epithelial effector function and promote allergic airways disease. J Allergy Clin Immunol 2018; 142:435-450.e10. [PMID: 29108965 PMCID: PMC6278819 DOI: 10.1016/j.jaci.2017.08.043] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 06/30/2017] [Accepted: 08/23/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND Emerging studies suggest that enhanced glycolysis accompanies inflammatory responses. Virtually nothing is known about the relevance of glycolysis in patients with allergic asthma. OBJECTIVES We sought to determine whether glycolysis is altered in patients with allergic asthma and to address its importance in the pathogenesis of allergic asthma. METHODS We examined alterations in glycolysis in sputum samples from asthmatic patients and primary human nasal cells and used murine models of allergic asthma, as well as primary mouse tracheal epithelial cells, to evaluate the relevance of glycolysis. RESULTS In a murine model of allergic asthma, glycolysis was induced in the lungs in an IL-1-dependent manner. Furthermore, administration of IL-1β into the airways stimulated lactate production and expression of glycolytic enzymes, with notable expression of lactate dehydrogenase A occurring in the airway epithelium. Indeed, exposure of mouse tracheal epithelial cells to IL-1β or IL-1α resulted in increased glycolytic flux, glucose use, expression of glycolysis genes, and lactate production. Enhanced glycolysis was required for IL-1β- or IL-1α-mediated proinflammatory responses and the stimulatory effects of IL-1β on house dust mite (HDM)-induced release of thymic stromal lymphopoietin and GM-CSF from tracheal epithelial cells. Inhibitor of κB kinase ε was downstream of HDM or IL-1β and required for HDM-induced glycolysis and pathogenesis of allergic airways disease. Small interfering RNA ablation of lactate dehydrogenase A attenuated HDM-induced increases in lactate levels and attenuated HDM-induced disease. Primary nasal epithelial cells from asthmatic patients intrinsically produced more lactate compared with cells from healthy subjects. Lactate content was significantly higher in sputum supernatants from asthmatic patients, notably those with greater than 61% neutrophils. A positive correlation was observed between sputum lactate and IL-1β levels, and lactate content correlated negatively with lung function. CONCLUSIONS Collectively, these findings demonstrate that IL-1β/inhibitory κB kinase ε signaling plays an important role in HDM-induced glycolysis and pathogenesis of allergic airways disease.
Collapse
Affiliation(s)
- Xi Qian
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Reem Aboushousha
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Cheryl van de Wetering
- Department of Pulmonology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Shi B Chia
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Eyal Amiel
- Department of Medical Laboratory and Radiation, University of Vermont College of Nursing and Health Sciences, Burlington, Vt
| | - Robert W Schneider
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Jos L J van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Karolyn G Lahue
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Daisy A Hoagland
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Dylan T Casey
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Nirav Daphtary
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Jennifer L Ather
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Matthew J Randall
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Minara Aliyeva
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Kendall E Black
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - David G Chapman
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vt; Woolcock Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Lennart K A Lundblad
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - David H McMillan
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Anne E Dixon
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Charles G Irvin
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Matthew E Poynter
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vt
| | - Emiel F M Wouters
- Department of Pulmonology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Pamela M Vacek
- Medical Biostatistics Unit, University of Vermont College of Medicine, Burlington, Vt
| | - Monique Henket
- Department of Respiratory Medicine, CHU Sart-TilmanB35, Liege, Belgium
| | - Florence Schleich
- Department of Respiratory Medicine, CHU Sart-TilmanB35, Liege, Belgium
| | - Renaud Louis
- Department of Respiratory Medicine, CHU Sart-TilmanB35, Liege, Belgium
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vt
| | | |
Collapse
|
37
|
Das M, Zawada WM, West J, Stenmark KR. JNK2 regulates vascular remodeling in pulmonary hypertension. Pulm Circ 2018; 8:2045894018778156. [PMID: 29718758 PMCID: PMC6055330 DOI: 10.1177/2045894018778156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 04/26/2018] [Indexed: 01/04/2023] Open
Abstract
Pulmonary arterial (PA) wall modifications are key pathological features of pulmonary hypertension (PH). Although such abnormalities correlate with heightened phosphorylation of c-Jun N-terminal kinases 1/2 (JNK1/2) in a rat model of PH, the contribution of specific JNK isoforms to the pathophysiology of PH is unknown. Hence, we hypothesized that activation of either one, or both JNK isoforms regulates PA remodeling in PH. We detected increased JNK1/2 phosphorylation in the thickened vessels of PH patients' lungs compared to that in lungs of healthy individuals. JNK1/2 phosphorylation paralleled a marked reduction in MAP kinase phosphatase 1 (JNK dephosphorylator) expression in patients' lungs. Association of JNK1/2 activation with vascular modification was confirmed in the calf model of severe hypoxia-induced PH. To ascertain the role of each JNK isoform in pathophysiology of PH, wild-type (WT), JNK1 null (JNK1-/-), and JNK2 null (JNK2-/-) mice were exposed to chronic hypoxia (10% O2 for six weeks) to develop PH. In hypoxic WT lungs, an increase in JNK1/2 phosphorylation was associated with PH-like pathology. Hallmarks of PH pathophysiology, i.e. excessive accumulation of extracellular matrix and vessel muscularization with medial wall thickening, was also detected in hypoxic JNK1-/- lungs, but not in hypoxia-exposed JNK2-/- lungs. However, hypoxia-induced increases in right ventricular systolic pressure (RVSP) and in right ventricular hypertrophy (RVH) were similar in all three genotypes. Our findings suggest that JNK2 participates in PA remodeling (but likely not in vasoconstriction) in murine hypoxic PH and that modulating JNK2 actions might quell vascular abnormalities and limit the course of PH.
Collapse
Affiliation(s)
- Mita Das
- Department of Internal Medicine, College of Medicine Phoenix, University of Arizona, Phoenix, AZ, USA
| | - W. Michael Zawada
- Department of Basic Medical Sciences, A. T. Still University, School of Osteopathic Medicine Arizona, Mesa, AZ, USA
| | - James West
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kurt R. Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
38
|
van der Velden JL, Wagner DE, Lahue KG, Abdalla ST, Lam YW, Weiss DJ, Janssen-Heininger YMW. TGF-β1-induced deposition of provisional extracellular matrix by tracheal basal cells promotes epithelial-to-mesenchymal transition in a c-Jun NH 2-terminal kinase-1-dependent manner. Am J Physiol Lung Cell Mol Physiol 2018; 314:L984-L997. [PMID: 29469614 PMCID: PMC6032072 DOI: 10.1152/ajplung.00053.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 02/14/2018] [Accepted: 02/15/2018] [Indexed: 12/16/2022] Open
Abstract
Epithelial cells have been suggested as potential drivers of lung fibrosis, although the epithelial-dependent pathways that promote fibrogenesis remain unknown. Extracellular matrix is increasingly recognized as an environment that can drive cellular responses in various pulmonary diseases. In this study, we demonstrate that transforming growth factor-β1 (TGF-β1)-stimulated mouse tracheal basal (MTB) cells produce provisional matrix proteins in vitro, which initiate mesenchymal changes in subsequently freshly plated MTB cells via Rho kinase- and c-Jun NH2-terminal kinase (JNK1)-dependent processes. Repopulation of decellularized lung scaffolds, derived from mice with bleomycin-induced fibrosis or from patients with idiopathic pulmonary fibrosis, with wild-type MTB cells resulted in a loss of epithelial gene expression and augmentation of mesenchymal gene expression compared with cells seeded into decellularized normal lungs. In contrast, Jnk1-/- basal cells seeded into fibrotic lung scaffolds retained a robust epithelial expression profile, failed to induce mesenchymal genes, and differentiated into club cell secretory protein-expressing cells. This new paradigm wherein TGF-β1-induced extracellular matrix derived from MTB cells activates a JNK1-dependent mesenchymal program, which impedes subsequent normal epithelial cell homeostasis, provides a plausible scenario of chronic aberrant epithelial repair, thought to be critical in lung fibrogenesis. This study identifies JNK1 as a possible target for inhibition in settings wherein reepithelialization is desired.
Collapse
Affiliation(s)
- Jos L van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont , Burlington, Vermont
| | - Darcy E Wagner
- Department of Medicine, University of Vermont , Burlington, Vermont
- Department of Experimental Medical Sciences, Lung Bioengineering, and Regeneration, Lund University , Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University , Lund, Sweden
| | - Karolyn G Lahue
- Department of Pathology and Laboratory Medicine, University of Vermont , Burlington, Vermont
| | - Sarah T Abdalla
- Department of Pathology and Laboratory Medicine, University of Vermont , Burlington, Vermont
| | - Ying-Wai Lam
- Department of Biology, University of Vermont , Burlington, Vermont
- Vermont Genetics Networks Proteomics Facility, University of Vermont , Burlington, Vermont
| | - Daniel J Weiss
- Department of Medicine, University of Vermont , Burlington, Vermont
| | | |
Collapse
|
39
|
Ying Q, Wu G. Molecular mechanisms involved in podocyte EMT and concomitant diabetic kidney diseases: an update. Ren Fail 2017; 39:474-483. [PMID: 28413908 PMCID: PMC6014344 DOI: 10.1080/0886022x.2017.1313164] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) is a tightly regulated process by which epithelial cells lose their hallmark epithelial characteristics and gain the features of mesenchymal cells. For podocytes, expression of nephrin, podocin, P-cadherin, and ZO-1 is downregulated, the slit diaphragm (SD) will be altered, and the actin cytoskeleton will be rearranged. Diabetes, especially hyperglycemia, has been demonstrated to incite podocyte EMT through several molecular mechanisms such as TGF-β/Smad classic pathway, Wnt/β-catenin signaling pathway, Integrins/integrin-linked kinase (ILK) signaling pathway, MAPKs signaling pathway, Jagged/Notch signaling pathway, and NF-κB signaling pathway. As one of the most fundamental prerequisites to develop ground-breaking therapeutic options to prevent the development and progression of diabetic kidney disease (DKD), a comprehensive understanding of the molecular mechanisms involved in the pathogenesis of podocyte EMT is compulsory. Therefore, the purpose of this paper is to update the research progress of these underlying signaling pathways and expound the podocyte EMT-related DKDs.
Collapse
Affiliation(s)
- Qidi Ying
- a Department of Pharmacology, Pharmacy , China Pharmaceutical University , Nanjing , Jiangsu , China
| | - Guanzhong Wu
- a Department of Pharmacology, Pharmacy , China Pharmaceutical University , Nanjing , Jiangsu , China
| |
Collapse
|
40
|
Ebelt ND, Kaoud TS, Edupuganti R, Van Ravenstein S, Dalby KN, Van Den Berg CL. A c-Jun N-terminal kinase inhibitor, JNK-IN-8, sensitizes triple negative breast cancer cells to lapatinib. Oncotarget 2017; 8:104894-104912. [PMID: 29285221 PMCID: PMC5739608 DOI: 10.18632/oncotarget.20581] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 08/04/2017] [Indexed: 12/11/2022] Open
Abstract
Triple negative breast cancers (TNBC) have poor prognosis compared to other breast cancer subtypes and represent 15-20% of breast cancers diagnosed. Unique targets and new molecularly-targeted therapies are urgently needed for this subtype. Despite high expression of Epidermal Growth Factor Receptor, inhibitors such as lapatinib have not shown therapeutic efficacy in TNBC patients. Herein, we report that treatment with the covalent JNK inhibitor, JNK-IN-8, synergizes with lapatinib to cause cell death, while these compounds as single agents have little effect. The combination significantly increases survival of mice bearing xenografts of MDA-MB-231 human TNBC cells. Our studies demonstrate that lapatinib treatment increases c-Jun and JNK phosphorylation indicating a mechanism of resistance. Combined, these compounds significantly reduce transcriptional activity of Nuclear Factor kappa B, Activating Protein 1, and Nuclear factor erythroid 2-Related Factor 2. As master regulators of antioxidant response, their decreased activity induces a 10-fold increase in reactive oxygen species that is cytotoxic, and is rescued by addition of exogenous antioxidants. Over expression of p65 or Nrf2 also significantly rescues viability during JNK-IN-8 and lapatinib treatment. Further studies combining JNK-IN-8 and lapatinib may reveal a benefit for patients with TNBC, fulfilling a critical medical need.
Collapse
Affiliation(s)
- Nancy D Ebelt
- Institute of Cellular & Molecular Biology, University of Texas at Austin, Dell Pediatric Research Institute, Austin, TX 78723, USA.,Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Tamer S Kaoud
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA.,Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, El-Minia 61519, Egypt
| | - Ramakrishna Edupuganti
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Sabrina Van Ravenstein
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Kevin N Dalby
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Carla L Van Den Berg
- Institute of Cellular & Molecular Biology, University of Texas at Austin, Dell Pediatric Research Institute, Austin, TX 78723, USA.,Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Dell Pediatric Research Institute, Austin, TX 78723, USA
| |
Collapse
|
41
|
Eurlings IMJ, Reynaert NL, van de Wetering C, Aesif SW, Mercken EM, de Cabo R, van der Velden JL, Janssen-Heininger YM, Wouters EFM, Dentener MA. Involvement of c-Jun N-Terminal Kinase in TNF-α-Driven Remodeling. Am J Respir Cell Mol Biol 2017; 56:393-401. [PMID: 27875656 DOI: 10.1165/rcmb.2015-0195oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Lung tissue remodeling in chronic obstructive pulmonary disease (COPD) is characterized by airway wall thickening and/or emphysema. Although the bronchial and alveolar compartments are functionally independent entities, we recently showed comparable alterations in matrix composition comprised of decreased elastin content and increased collagen and hyaluronan contents of alveolar and small airway walls. Out of several animal models tested, surfactant protein C (SPC)-TNF-α mice showed remodeling in alveolar and airway walls similar to what we observed in patients with COPD. Epithelial cells are able to undergo a phenotypic shift, gaining mesenchymal properties, a process in which c-Jun N-terminal kinase (JNK) signaling is involved. Therefore, we hypothesized that TNF-α induces JNK-dependent epithelial plasticity, which contributes to lung matrix remodeling. To this end, the ability of TNF-α to induce a phenotypic shift was assessed in A549, BEAS2B, and primary bronchial epithelial cells, and phenotypic markers were studied in SPC-TNF-α mice. Phenotypic markers of mesenchymal cells were elevated both in vitro and in vivo, as shown by the expression of vimentin, plasminogen activator inhibitor-1, collagen, and matrix metalloproteinases. Concurrently, the expression of the epithelial markers, E-cadherin and keratin 7 and 18, was attenuated. A pharmacological inhibitor of JNK attenuated this phenotypic shift in vitro, demonstrating involvement of JNK signaling in this process. Interestingly, activation of JNK signaling was also clearly present in lungs of SPC-TNF-α mice and patients with COPD. Together, these data show a role for TNF-α in the induction of a phenotypic shift in vitro, resulting in increased collagen production and the expression of elastin-degrading matrix metalloproteinases, and provide evidence for involvement of the TNF-α-JNK axis in extracellular matrix remodeling.
Collapse
Affiliation(s)
- Irene M J Eurlings
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Niki L Reynaert
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Cheryl van de Wetering
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Scott W Aesif
- 2 Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Evi M Mercken
- 3 Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland; and
| | - Rafael de Cabo
- 3 Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland; and
| | - Jos L van der Velden
- 4 Department of Pathology, University of Vermont College of Medicine, Burlington, Vermont
| | | | - Emiel F M Wouters
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Mieke A Dentener
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
42
|
Park JH, Park B, Park KK. Suppression of Hepatic Epithelial-to-Mesenchymal Transition by Melittin via Blocking of TGFβ/Smad and MAPK-JNK Signaling Pathways. Toxins (Basel) 2017; 9:138. [PMID: 28406452 PMCID: PMC5408212 DOI: 10.3390/toxins9040138] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 01/27/2023] Open
Abstract
Transforming growth factor (TGF)-β1 plays a crucial role in the epithelial-to-mesenchymal transition (EMT) in hepatocytes and hepatic stellate cells (HSC), which contributes to the pathogenesis of liver fibrosis. Melittin (MEL) is a major component of bee venom and is effective in rheumatoid arthritis, pain relief, cancer cell proliferation, fibrosis and immune modulating activity. In this study, we found that MEL inhibits hepatic EMT in vitro and in vivo, regulating the TGFβ/Smad and TGFβ/nonSmad signaling pathways. MEL significantly inhibited TGF-β1-induced expression of EMT markers (E-cadherin reduction and vimentin induction) in vitro. These results were confirmed in CCl₄-induced liver in vivo. Treatment with MEL almost completely blocked the phosphorylation of Smad2/3, translocation of Smad4 and phosphorylation of JNK in vitro and in vivo. Taken together, these results suggest that MEL suppresses EMT by inhibiting the TGFβ/Smad and TGFβ/nonSmad-c-Jun N-terminal kinase (JNK)/Mitogen-activated protein kinase (MAPK) signaling pathways. These results indicated that MEL possesses potent anti-fibrotic and anti-EMT properties, which may be responsible for its effects on liver diseases.
Collapse
Affiliation(s)
- Ji-Hyun Park
- College of Pharmacy, Keimyung University, Dalgubeoldaero, Dalseo-Gu, Daegu 42601, Korea.
| | - Byoungduck Park
- College of Pharmacy, Keimyung University, Dalgubeoldaero, Dalseo-Gu, Daegu 42601, Korea.
| | - Kwan-Kyu Park
- Department of Pathology, Daegu Catholic University Medical Center, Duryugongwon-ro, Nam-gu, Daegu 42472, Korea.
| |
Collapse
|
43
|
Danyal K, de Jong W, O'Brien E, Bauer RA, Heppner DE, Little AC, Hristova M, Habibovic A, van der Vliet A. Acrolein and thiol-reactive electrophiles suppress allergen-induced innate airway epithelial responses by inhibition of DUOX1 and EGFR. Am J Physiol Lung Cell Mol Physiol 2016; 311:L913-L923. [PMID: 27612966 DOI: 10.1152/ajplung.00276.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/05/2016] [Indexed: 01/27/2023] Open
Abstract
Acrolein is a major thiol-reactive component of cigarette smoke (CS) that is thought to contribute to increased asthma incidence associated with smoking. Here, we explored the effects of acute acrolein exposure on innate airway responses to two common airborne allergens, house dust mite and Alternaria alternata, and observed that acrolein exposure of C57BL/6 mice (5 ppm, 4 h) dramatically inhibited innate airway responses to subsequent allergen challenge, demonstrated by attenuated release of the epithelial-derived cytokines IL-33, IL-25, and IL-1α. Acrolein and other anti-inflammatory thiol-reactive electrophiles, cinnamaldehyde, curcumin, and sulforaphane, similarly inhibited allergen-induced production of these cytokines from human or murine airway epithelial cells in vitro. Based on our previous observations indicating the importance of Ca2+-dependent signaling, activation of the NADPH oxidase DUOX1, and Src/EGFR-dependent signaling in allergen-induced epithelial secretion of these cytokines, we explored the impact of acrolein on these pathways. Acrolein and other thiol-reactive electrophiles were found to dramatically prevent allergen-induced activation of DUOX1 as well as EGFR, and acrolein was capable of inhibiting EGFR tyrosine kinase activity via modification of C797. Biotin-labeling strategies indicated increased cysteine modification and carbonylation of Src, EGFR, as well as DUOX1, in response to acrolein exposure in vitro and in vivo, suggesting that direct alkylation of these proteins on accessible cysteine residues may be responsible for their inhibition. Collectively, our findings indicate a novel anti-inflammatory mechanism of CS-derived acrolein and other thiol-reactive electrophiles, by directly inhibiting DUOX1- and EGFR-mediated airway epithelial responses to airborne allergens.
Collapse
Affiliation(s)
- Karamatullah Danyal
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Willem de Jong
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Edmund O'Brien
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Robert A Bauer
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - David E Heppner
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Andrew C Little
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Milena Hristova
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Aida Habibovic
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
44
|
Yu CY, Kuo HC. The Trans-Spliced Long Noncoding RNA ts RMST Impedes Human Embryonic Stem Cell Differentiation Through WNT5A-Mediated Inhibition of the Epithelial-to-Mesenchymal Transition. Stem Cells 2016; 34:2052-2062. [DOI: 10.1002/stem.2386] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
The trans-spliced noncoding RNA RMST (tsRMST) is an emerging regulatory lncRNA in the human pluripotency circuit. Previously, we found that tsRMST represses lineage-specific transcription factors through the PRC2 complex and NANOG in human pluripotent stem cells (hESCs). Here, we demonstrate that tsRMST also modulates noncanonical Wnt signaling to suppress the epithelial-to-mesenchymal transition (EMT) and in vitro differentiation of embryonic stem cells (ESCs). Our results demonstrate that disruption of tsRMST expression in hESCs results in the upregulation of WNT5A, EMT, and lineage-specific genes/markers. Furthermore, we found that the PKC inhibitors Go6983 and Go6976 inhibited the effects of WNT5A, indicating that WNT5A promotes the EMT and in vitro differentiation although conventional and novel PKC activation in hESCs. Finally, we showed that either antiserum neutralization of WNT5A or Go6983 treatment in tsRMST knockdown cells decreased the expression of mesenchymal and lineage-specific markers. Together, these findings indicate that tsRMST regulates Wnt and EMT signaling pathways in hESCs by repressing WNT5A, which is a potential EMT inducer for promoting in vitro differentiation of hESCs through PKC activation. Our findings provide further insights into the role of trans-spliced RNA and WNT5A in hESC differentiation, in which EMT plays an important role.
Collapse
Affiliation(s)
- Chun-Ying Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Hung-Chih Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
45
|
Moustakas A, Heldin CH. Mechanisms of TGFβ-Induced Epithelial-Mesenchymal Transition. J Clin Med 2016; 5:jcm5070063. [PMID: 27367735 PMCID: PMC4961994 DOI: 10.3390/jcm5070063] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 02/07/2023] Open
Abstract
Transitory phenotypic changes such as the epithelial–mesenchymal transition (EMT) help embryonic cells to generate migratory descendants that populate new sites and establish the distinct tissues in the developing embryo. The mesenchymal descendants of diverse epithelia also participate in the wound healing response of adult tissues, and facilitate the progression of cancer. EMT can be induced by several extracellular cues in the microenvironment of a given epithelial tissue. One such cue, transforming growth factor β (TGFβ), prominently induces EMT via a group of specific transcription factors. The potency of TGFβ is partly based on its ability to perform two parallel molecular functions, i.e. to induce the expression of growth factors, cytokines and chemokines, which sequentially and in a complementary manner help to establish and maintain the EMT, and to mediate signaling crosstalk with other developmental signaling pathways, thus promoting changes in cell differentiation. The molecules that are activated by TGFβ signaling or act as cooperating partners of this pathway are impossible to exhaust within a single coherent and contemporary report. Here, we present selected examples to illustrate the key principles of the circuits that control EMT under the influence of TGFβ.
Collapse
Affiliation(s)
- Aristidis Moustakas
- Ludwig Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE 751 24 Uppsala, Sweden.
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, SE 751 23 Uppsala, Sweden.
| | - Carl-Henrik Heldin
- Ludwig Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE 751 24 Uppsala, Sweden.
| |
Collapse
|
46
|
Jones JT, Qian X, van der Velden JLJ, Chia SB, McMillan DH, Flemer S, Hoffman SM, Lahue KG, Schneider RW, Nolin JD, Anathy V, van der Vliet A, Townsend DM, Tew KD, Janssen-Heininger YMW. Glutathione S-transferase pi modulates NF-κB activation and pro-inflammatory responses in lung epithelial cells. Redox Biol 2016; 8:375-82. [PMID: 27058114 PMCID: PMC4827796 DOI: 10.1016/j.redox.2016.03.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 03/22/2016] [Indexed: 01/30/2023] Open
Abstract
Nuclear Factor kappa B (NF-κB) is a transcription factor family critical in the activation of pro- inflammatory responses. The NF-κB pathway is regulated by oxidant-induced post-translational modifications. Protein S-glutathionylation, or the conjugation of the antioxidant molecule, glutathione to reactive cysteines inhibits the activity of inhibitory kappa B kinase beta (IKKβ), among other NF-κB proteins. Glutathione S-transferase Pi (GSTP) is an enzyme that has been shown to catalyze protein S-glutathionylation (PSSG) under conditions of oxidative stress. The objective of the present study was to determine whether GSTP regulates NF-κB signaling, S-glutathionylation of IKK, and subsequent pro-inflammatory signaling. We demonstrated that, in unstimulated cells, GSTP associated with the inhibitor of NF-κB, IκBα. However, exposure to LPS resulted in a rapid loss of association between IκBα and GSTP, and instead led to a protracted association between IKKβ and GSTP. LPS exposure also led to increases in the S-glutathionylation of IKKβ. SiRNA-mediated knockdown of GSTP decreased IKKβ-SSG, and enhanced NF-κB nuclear translocation, transcriptional activity, and pro-inflammatory cytokine production in response to lipopolysaccharide (LPS). TLK117, an isotype-selective inhibitor of GSTP, also enhanced LPS-induced NF-κB transcriptional activity and pro-inflammatory cytokine production, suggesting that the catalytic activity of GSTP is important in repressing NF-κB activation. Expression of both wild-type and catalytically-inactive Y7F mutant GSTP significantly attenuated LPS- or IKKβ-induced production of GM-CSF. These studies indicate a complex role for GSTP in modulating NF-κB, which may involve S-glutathionylation of IKK proteins, and interaction with NF-κB family members. Our findings suggest that targeting GSTP is a potential avenue for regulating the activity of this prominent pro-inflammatory and immunomodulatory transcription factor.
Collapse
Affiliation(s)
- Jane T Jones
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Xi Qian
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Jos L J van der Velden
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Shi Biao Chia
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - David H McMillan
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Stevenson Flemer
- Department of Chemistry, The University of Vermont, Burlington, VT, United States
| | - Sidra M Hoffman
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Karolyn G Lahue
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Robert W Schneider
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - James D Nolin
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, The University of Vermont, Burlington, VT, United States
| | - Danyelle M Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | | |
Collapse
|
47
|
Sandhu V, Bowitz Lothe IM, Labori KJ, Skrede ML, Hamfjord J, Dalsgaard AM, Buanes T, Dube G, Kale MM, Sawant S, Kulkarni-Kale U, Børresen-Dale AL, Lingjærde OC, Kure EH. Differential expression of miRNAs in pancreatobiliary type of periampullary adenocarcinoma and its associated stroma. Mol Oncol 2016; 10:303-316. [PMID: 26590090 PMCID: PMC5528959 DOI: 10.1016/j.molonc.2015.10.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 09/22/2015] [Accepted: 10/08/2015] [Indexed: 02/08/2023] Open
Abstract
Periampullary adenocarcinomas can be of two histological subtypes, intestinal or pancreatobiliary. The latter is more frequent and aggressive, and characterized by a prominent desmoplastic stroma, which is tightly related to the biology of the cancer, including its poor response to chemotherapy. Whereas miRNAs are known to regulate various cellular processes and interactions between cells, their exact role in periampullary carcinoma remains to be characterized, especially with respect to the prominent stromal component of pancreatobiliary type cancers. The present study aimed at elucidating this role by miRNA expression profiling of the carcinomatous and stromal component in twenty periampullary adenocarcinomas of pancreatobiliary type. miRNA expression profiles were compared between carcinoma cells, stromal cells and normal tissue samples. A total of 43 miRNAs were found to be differentially expressed between carcinoma and stroma of which 11 belong to three miRNA families (miR-17, miR-15 and miR-515). The levels of expression of miRNAs miR-17, miR-20a, miR-20b, miR-223, miR-10b, miR-2964a and miR-342 were observed to be higher and miR-519e to be lower in the stromal component compared to the carcinomatous and normal components. They follow a trend where expression in stroma is highest followed by carcinoma and then normal tissue. Pathway analysis revealed that pathways regulating tumor-stroma interactions such as ECM interaction remodeling, epithelial-mesenchymal transition, focal adhesion pathway, TGF-beta, MAPK signaling, axon guidance and endocytosis were differently regulated. The miRNA-mRNA mediated interactions between carcinoma and stromal cells add new knowledge regarding tumor-stroma interactions.
Collapse
Affiliation(s)
- V Sandhu
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department for Environmental Health and Science, Telemark University College, Bø in Telemark, Norway
| | - I M Bowitz Lothe
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - K J Labori
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Oslo, Norway
| | - M L Skrede
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - J Hamfjord
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - A M Dalsgaard
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - T Buanes
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - G Dube
- Bioinformatics Centre, Savitribai Phule Pune University (Formerly University of Pune), Pune, India
| | - M M Kale
- Department of Statistics, Savitribai Phule Pune University (Formerly University of Pune), Pune, India
| | - S Sawant
- Bioinformatics Centre, Savitribai Phule Pune University (Formerly University of Pune), Pune, India
| | - U Kulkarni-Kale
- Bioinformatics Centre, Savitribai Phule Pune University (Formerly University of Pune), Pune, India
| | - A-L Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - O C Lingjærde
- K.G. Jebsen Centre for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Computer Science, University of Oslo, Oslo, Norway
| | - E H Kure
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department for Environmental Health and Science, Telemark University College, Bø in Telemark, Norway.
| |
Collapse
|
48
|
Hristova M, Habibovic A, Veith C, Janssen-Heininger YMW, Dixon AE, Geiszt M, van der Vliet A. Airway epithelial dual oxidase 1 mediates allergen-induced IL-33 secretion and activation of type 2 immune responses. J Allergy Clin Immunol 2015; 137:1545-1556.e11. [PMID: 26597162 DOI: 10.1016/j.jaci.2015.10.003] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 09/23/2015] [Accepted: 10/02/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND The IL-1 family member IL-33 plays a critical role in type 2 innate immune responses to allergens and is an important mediator of allergic asthma. The mechanisms by which allergens provoke epithelial IL-33 secretion are still poorly understood. OBJECTIVE Based on previous findings indicating involvement of the NADPH oxidase dual oxidase 1 (DUOX1) in epithelial wound responses, we explored the potential involvement of DUOX1 in allergen-induced IL-33 secretion and potential alterations in airways of asthmatic patients. METHODS Cultured human or murine airway epithelial cells or mice were subjected to acute challenge with Alternaria alternata or house dust mite, and secretion of IL-33 and activation of subsequent type 2 responses were determined. The role of DUOX1 was explored by using small interfering RNA approaches and DUOX1-deficient mice. Cultured nasal epithelial cells from healthy subjects or asthmatic patients were evaluated for DUOX1 expression and allergen-induced responses. RESULTS In vitro or in vivo allergen challenge resulted in rapid airway epithelial IL-33 secretion, which depended critically on DUOX1-mediated activation of epithelial epidermal growth factor receptor and the protease calpain-2 through a redox-dependent mechanism involving cysteine oxidation within epidermal growth factor receptor and the tyrosine kinase Src. Primary nasal epithelial cells from patients with allergic asthma were found to express increased DUOX1 and IL-33 levels and demonstrated enhanced IL-33 secretion in response to allergen challenge compared with values seen in nasal epithelial cells from nonasthmatic subjects. CONCLUSION Our findings implicate epithelial DUOX1 as a pivotal mediator of IL-33-dependent activation of innate airway type 2 immune responses to common airborne allergens and indicate that enhanced DUOX1 expression and IL-33 secretion might present important contributing features of allergic asthma.
Collapse
Affiliation(s)
- Milena Hristova
- Department of Pathology and Laboratory Medicine, Vermont Lung Center, University of Vermont, Burlington, Vt
| | - Aida Habibovic
- Department of Pathology and Laboratory Medicine, Vermont Lung Center, University of Vermont, Burlington, Vt
| | - Carmen Veith
- Department of Pathology and Laboratory Medicine, Vermont Lung Center, University of Vermont, Burlington, Vt
| | | | - Anne E Dixon
- Department of Medicine, Vermont Lung Center, University of Vermont, Burlington, Vt
| | - Miklos Geiszt
- Department of Physiology and Lendület Peroxidase Enzyme Research Group, Semmelweis University, Budapest, Hungary
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Vermont Lung Center, University of Vermont, Burlington, Vt.
| |
Collapse
|
49
|
Sahu SK, Garding A, Tiwari N, Thakurela S, Toedling J, Gebhard S, Ortega F, Schmarowski N, Berninger B, Nitsch R, Schmidt M, Tiwari VK. JNK-dependent gene regulatory circuitry governs mesenchymal fate. EMBO J 2015; 34:2162-81. [PMID: 26157010 PMCID: PMC4557668 DOI: 10.15252/embj.201490693] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 06/05/2015] [Indexed: 12/14/2022] Open
Abstract
The epithelial to mesenchymal transition (EMT) is a biological process in which cells lose cell–cell contacts and become motile. EMT is used during development, for example, in triggering neural crest migration, and in cancer metastasis. Despite progress, the dynamics of JNK signaling, its role in genomewide transcriptional reprogramming, and involved downstream effectors during EMT remain largely unknown. Here, we show that JNK is not required for initiation, but progression of phenotypic changes associated with EMT. Such dependency resulted from JNK-driven transcriptional reprogramming of critical EMT genes and involved changes in their chromatin state. Furthermore, we identified eight novel JNK-induced transcription factors that were required for proper EMT. Three of these factors were also highly expressed in invasive cancer cells where they function in gene regulation to maintain mesenchymal identity. These factors were also induced during neuronal development and function in neuronal migration in vivo. These comprehensive findings uncovered a kinetically distinct role for the JNK pathway in defining the transcriptome that underlies mesenchymal identity and revealed novel transcription factors that mediate these responses during development and disease.
Collapse
Affiliation(s)
| | | | - Neha Tiwari
- Institute of Physiological Chemistry University Medical Center Johannes Gutenberg University, Mainz, Germany
| | | | | | - Susanne Gebhard
- Department of Obstetrics and Gynecology, Johannes Gutenberg University, Mainz, Germany
| | - Felipe Ortega
- Institute of Physiological Chemistry University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Nikolai Schmarowski
- Institute for Microscopic Anatomy and Neurobiology University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Benedikt Berninger
- Institute of Physiological Chemistry University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Robert Nitsch
- Institute for Microscopic Anatomy and Neurobiology University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Marcus Schmidt
- Department of Obstetrics and Gynecology, Johannes Gutenberg University, Mainz, Germany
| | | |
Collapse
|
50
|
Kim EJ, Kim HJ, Park MK, Kang GJ, Byun HJ, Lee H, Lee CH. Cardamonin Suppresses TGF-β1-Induced Epithelial Mesenchymal Transition via Restoring Protein Phosphatase 2A Expression. Biomol Ther (Seoul) 2015; 23:141-8. [PMID: 25767682 PMCID: PMC4354315 DOI: 10.4062/biomolther.2014.117] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/12/2014] [Accepted: 12/16/2014] [Indexed: 12/20/2022] Open
Abstract
Epithelial mesenchymal transition (EMT) is the first step in metastasis and implicated in the phenotype of cancer stem cells. Therefore, understanding and controlling EMT, are essential to the prevention and cure of metastasis. In the present study, we examined, by Western blot, reverse transcription polymerase chain reaction (RT-PCR), and confocal microscopy, the effects of cardamonin (CDN) on transforming growth factor-β1 (TGF-β1)-induced EMT of A549 lung adenocarcinoma cell lines. TGF-β1 induced expression of N-cadherin and decreased expression of E-cadherin. CDN suppressed N-cadherin expression and restored E-cadherin expression. Further, TGF-β1 induced migration and invasion of A549 cancer cells, which was suppressed by CDN. TGF-β1 induced c-Jun N-terminal kinase (JNK) activation during EMT, but CDN blocked it. Protein serine/threonine phosphatase 2A (PP2A) expression in A549 cancer cells was reduced by TGF-β1 but CDN restored it. The overall data suggested that CDN suppresses TGF-β1-induced EMT via PP2A restoration, making it a potential new drug candidate that controls metastasis.
Collapse
Affiliation(s)
- Eun Ji Kim
- BK21PLUS R-FIND Team, College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Hyun Ji Kim
- BK21PLUS R-FIND Team, College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Mi Kyung Park
- BK21PLUS R-FIND Team, College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Gyeung Jin Kang
- BK21PLUS R-FIND Team, College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Hyun Jung Byun
- BK21PLUS R-FIND Team, College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Ho Lee
- National Cancer Center, Goyang 410-769, Republic of Korea
| | - Chang Hoon Lee
- BK21PLUS R-FIND Team, College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| |
Collapse
|