1
|
Niu M, Wang YZ, Deng XM, Wu X, Hua ZY, Lv TT. Tryptanthrin alleviate lung fibrosis via suppression of MAPK/NF-κB and TGF-β1/SMAD signaling pathways in vitro and in vivo. Toxicol Appl Pharmacol 2025; 498:117285. [PMID: 40089192 DOI: 10.1016/j.taap.2025.117285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/17/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF), a progressive interstitial lung disease of unknown etiology, remains a therapeutic challenge with limited treatment options. This study investigates the therapeutic potential and molecular mechanisms of Tryptanthrin, a bioactive indole quinazoline alkaloid derived from Isatis tinctoria L., in pulmonary fibrosis. In a bleomycin-induced murine IPF model, Tryptanthrin administration (5 and 10 mg/kg/day for 28 days) significantly improved pulmonary function parameters and attenuated histological evidence of fibrosis. Mechanistic analysis revealed dual pathway modulation: Tryptanthrin suppressed MAPK/NF-κB signaling through inhibition of phosphorylation events, subsequently reducing pulmonary levels of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6). Concurrently, it attenuated TGF-β1/Smad pathway activation by decreasing TGF-β1 expression and Smad2/3 phosphorylation, thereby downregulating fibrotic markers including COL1A1, α-smooth muscle actin (α-SMA), and fibronectin in lung tissues. Complementary in vitro studies using Lipopolysaccharide (LPS) or TGF-β1-stimulated NIH3T3 fibroblasts confirmed these anti-inflammatory and anti-fibrotic effects through analogous pathway inhibition. Our findings demonstrate that Tryptanthrin exerts therapeutic effects against pulmonary fibrosis via coordinated modulation of both inflammatory (MAPK/NF-κB) and fibrotic (TGF-β1/Smad) signaling cascades, suggesting its potential as a novel multi-target therapeutic agent for IPF management.
Collapse
Affiliation(s)
- Min Niu
- College of Pharmacy & Traditional Chinese Medicine, Jiangsu College of Nursing, Jiangsu, China.
| | | | - Xiang-Min Deng
- College of Pharmacy & Traditional Chinese Medicine, Jiangsu College of Nursing, Jiangsu, China
| | - Xin Wu
- College of Pharmacy & Traditional Chinese Medicine, Jiangsu College of Nursing, Jiangsu, China
| | - Zheng-Ying Hua
- College of Pharmacy & Traditional Chinese Medicine, Jiangsu College of Nursing, Jiangsu, China
| | - Ting-Ting Lv
- College of Pharmacy & Traditional Chinese Medicine, Jiangsu College of Nursing, Jiangsu, China
| |
Collapse
|
2
|
Wang Y, Liu C, Wang N, Weng D, Zhao Y, Yang H, Wang H, Xu S, Gao J, Lang C, Fan Z, Yu L, He Z. hAMSCs regulate EMT in the progression of experimental pulmonary fibrosis through delivering miR-181a-5p targeting TGFBR1. Stem Cell Res Ther 2025; 16:2. [PMID: 39757225 DOI: 10.1186/s13287-024-04095-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a common and multidimensional devastating interstitial lung disease. The development of novel and more effective interventions for PF is an urgent clinical need. A previous study has found that miR-181a-5p plays an important role in the development of PF, and human amniotic mesenchymal stem cells (hAMSCs) exert potent therapeutic potential on PF. However, whether hAMSCs act on PF by delivering miR-181a-5p and its detailed mechanism still remain unknown. Thus, this study was designed to investigate the underlying possible mechanism of hAMSCs on PF in bleomycin (BLM)-induced mouse PF model, and a co-culture system of hAMSCs and A549 cells epithelial mesenchymal transition (EMT) model, focusing on its effects on collagen deposition, EMT, and epithelial cell cycle regulation. METHODS hAMSCs with different miR-181a-5p expression levels were constructed. BLM (4 mg/kg) was used to create a PF model, while TGF-β1 was used to induce A549 cells to construct an EMT model. Furthermore, the effects of different miR-181a-5p expression in hAMSCs on collagen deposition and EMT during lung fibrosis were assessed in vivo and in vitro. RESULTS We found that hAMSCs exerted anti-fibrotic effect in BLM-induced mouse PF model. Moreover, hAMSCs also exerted protective effect on TGFβ1-induced A549 cell EMT model. Furthermore, hAMSCs ameliorated PF by promoting epithelial cell proliferation, reducing epithelial cell apoptosis, and attenuating EMT of epithelial cells through paracrine effects. hAMSCs regulated EMT in PF through delivering miR-181a-5p targeting TGFBR1. CONCLUSIONS Our findings reveal for the first time that hAMSCs inhibit PF by promoting epithelial cell proliferation, reducing epithelial cell apoptosis, and attenuating EMT. Mechanistically, the therapeutic effect of hMASCs on PF is achieved through delivering miR-181a-5p targeting TGFBR1.
Collapse
Affiliation(s)
- Yanyang Wang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Chan Liu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Nuoxin Wang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Dong Weng
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Yan Zhao
- Department of Prevention Healthcare, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, 400038, China
| | - Hongyu Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Haoyuan Wang
- Department of Cardiothoracic Surgery, Liuzhou People's Hospital, Liuzhou, 545001, Guangxi, China
| | - Shangfu Xu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Jianmei Gao
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi, 563000, Guizhou, China
| | - Changhui Lang
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Zhenhai Fan
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Limei Yu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Zhixu He
- Center of Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, 550025, Guizhou, China.
- Department of Pediatric Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
3
|
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
|
4
|
Aripova N, Duryee MJ, Zhou W, England BR, Hunter CD, Klingemann LE, Aripova N, Nelson AJ, Katafiasz D, Bailey KL, Poole JA, Thiele GM, Mikuls TR. Citrullinated and malondialdehyde-acetaldehyde-modified fibrinogen activates macrophages and promotes profibrotic responses in human lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 2025; 328:L134-L147. [PMID: 39560968 PMCID: PMC11905797 DOI: 10.1152/ajplung.00153.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
The objective of this study was to assess fibrinogen (FIB) comodified with citrulline (CIT) and/or malondialdehyde-acetaldehyde (MAA) initiates macrophage-fibroblast interactions, leading to extracellular matrix (ECM) deposition that characterizes rheumatoid arthritis-associated interstitial lung disease (RA-ILD). Macrophages (Mϕ) were stimulated with native-FIB, FIB-CIT, FIB-MAA, or FIB-MAA-CIT. Supernatants (SNs) [Mϕ-SN (U-937-derived) or MϕP-SN (PBMC-derived)] or direct antigens were coincubated with human lung fibroblasts (HLFs). Gene expression was examined using RT-PCR. ECM deposition was quantified using immunohistochemistry and Western blot; cell signaling mechanisms were delineated. Platelet-derived growth factor (PDGF)-BB and TGF-β were measured in macrophage supernatants, and inhibition studies were performed using Su16f and SB431542, respectively. HLF gene expression of CD36, COL6A3, MMP-9, MMP-10, and MMP-12 was increased following stimulations with Mϕ-SN generated from modified FIB but not from direct antigens. HLF stimulated with MϕP-SNFIB-MAA-CIT derived from patients with RA-ILD resulted in 4- to 30-fold increases in COL6A3 and MMP12 expression; upregulation was greater in HLFs stimulated with MϕP-SN derived from RA-ILD versus controls. HLF exposure to Mϕ-SNFIB-MAA-CIT increased types I/VI collagen deposition versus all other Mϕ-SN groups and was greater than FIB-MAA-CIT stimulation. PDGF-BB and TGF-β signaling had the highest concentrations identified in Mϕ-SNFIB-MAA-CIT and MϕP-SNFIB-MAA-CIT, particularly from RA-ILD-derived cells. PDGF-BB and TGF-β inhibitors, alone and in combination, significantly reduced HLF-mediated ECM deposition from Mϕ-SN stimulations. These results show that comodified fibrinogen activates macrophages to produce PDGF-BB and TGF-β that promotes an aggressive HLF phenotype characterized by increased ECM deposition. These results suggest that targeting CIT and/or MAA modifications or downstream cellular signals could represent novel approaches to RA-ILD treatment.NEW & NOTEWORTHY This report demonstrates that fibrinogen simultaneously harboring two common posttranslational modifications activates macrophages to secrete platelet-derived growth factor (PDGF)-BB and transforming growth factor (TGF)-β. Resulting cross talk between activated macrophages and human lung fibroblasts leads to marked increases in extracellular matrix deposition. These protein modifications are abundant and colocalize in lung tissues from patients with rheumatoid arthritis-associated interstitial lung disease (RA-ILD), and the results suggest that agents targeting citrullination and/or malondialdehyde-acetaldehyde (MAA) adduct formation could represent novel therapeutic strategies.
Collapse
Affiliation(s)
- Nozima Aripova
- Division of Rheumatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Michael J Duryee
- Division of Rheumatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Veteran Affairs Nebraska-Western Iowa Health Care System, Research Services 151, Omaha, Nebraska, United States
| | - Wenxian Zhou
- Division of Rheumatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Bryant R England
- Division of Rheumatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Veteran Affairs Nebraska-Western Iowa Health Care System, Research Services 151, Omaha, Nebraska, United States
| | - Carlos D Hunter
- Division of Rheumatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Veteran Affairs Nebraska-Western Iowa Health Care System, Research Services 151, Omaha, Nebraska, United States
| | - Lauren E Klingemann
- Division of Rheumatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Nigina Aripova
- Department of Biology, Washington University in Saint Louis, Saint Louis, Missouri, United States
| | - Amy J Nelson
- Division of Allergy & Immunology, Department of Internal Medicine, Omaha, Nebraska, United States
| | - Dawn Katafiasz
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Omaha, Nebraska, United States
| | - Kristina L Bailey
- Veteran Affairs Nebraska-Western Iowa Health Care System, Research Services 151, Omaha, Nebraska, United States
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Omaha, Nebraska, United States
| | - Jill A Poole
- Division of Allergy & Immunology, Department of Internal Medicine, Omaha, Nebraska, United States
| | - Geoffrey M Thiele
- Division of Rheumatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Veteran Affairs Nebraska-Western Iowa Health Care System, Research Services 151, Omaha, Nebraska, United States
| | - Ted R Mikuls
- Division of Rheumatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Veteran Affairs Nebraska-Western Iowa Health Care System, Research Services 151, Omaha, Nebraska, United States
| |
Collapse
|
5
|
Velázquez-Enríquez JM, Santos-Álvarez JC, Ramírez-Hernández AA, Reyes-Jiménez E, Pérez-Campos Mayoral L, Romero-Tlalolini MDLÁ, Jiménez-Martínez C, Arellanes-Robledo J, Villa-Treviño S, Vásquez-Garzón VR, Baltiérrez-Hoyos R. Chlorogenic acid attenuates idiopathic pulmonary fibrosis: An integrated analysis of network pharmacology, molecular docking, and experimental validation. Biochem Biophys Res Commun 2024; 734:150672. [PMID: 39260206 DOI: 10.1016/j.bbrc.2024.150672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/26/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
AIMS Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung condition, the cause of which remains unknown and for which no effective therapeutic treatment is currently available. Chlorogenic acid (CGA), a natural polyphenolic compound found in different plants and foods, has emerged as a promising agent due to its anti-inflammatory, antioxidant, and antifibrotic properties. However, the molecular mechanisms underlying the therapeutic effect of CGA in IPF remain unclear. The purpose of this study was to analyze the pharmacological impact and underlying mechanisms of CGA in IPF. MAIN METHODS Using network pharmacology analysis, genes associated with IPF and potential molecular targets of CGA were identified through specialized databases, and a protein-protein interaction (PPI) network was constructed. Molecular docking was performed to accurately select potential therapeutic targets. To investigate the effects of CGA on lung histology and key gene expression, a murine model of bleomycin-induced lung fibrosis was used. KEY FINDINGS Network pharmacology analysis identified 384 were overlapped between CGA and IPF. Key targets including AKT1, TP53, JUN, CASP3, BCL2, MMP9, NFKB1, EGFR, HIF1A, and IL1B were identified. Pathway analysis suggested the involvement of cancer, atherosclerosis, and inflammatory processes. Molecular docking confirmed the stable binding between CGA and targets. CGA regulated the expression mRNA of EGFR, MMP9, AKT1, BCL2 and IL1B and attenuated pulmonary fibrosis in the mouse model. SIGNIFICANCE CGA is a promising multi-target therapeutic agent for IPF, which is supported by its efficacy in reducing fibrosis through the modulation of key pathways. This evidence provides a basis to further investigate CGA as an IPF potential treatment.
Collapse
Affiliation(s)
- Juan Manuel Velázquez-Enríquez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico.
| | - Jovito Cesar Santos-Álvarez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico
| | - Alma Aurora Ramírez-Hernández
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico
| | - Edilburga Reyes-Jiménez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico
| | - Laura Pérez-Campos Mayoral
- Facultad Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico
| | - María de Los Ángeles Romero-Tlalolini
- CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico
| | - Cristian Jiménez-Martínez
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Zacatenco, Av. Wilfrido Massieu Esq. Cda. Miguel Stampa S/N, Alcaldía Gustavo A. Madero, Mexico City, 07738, Mexico
| | - Jaime Arellanes-Robledo
- Laboratorio de Enfermedades Hepáticas, Instituto Nacional de Medicina Genómica - INMEGEN, México City, 14610, Mexico; Dirección Adjunta de Investigación Humanística y Científica, Consejo Nacional de Humanidades, Ciencias y Tecnologías - CONAHCYT, México City, 03940, Mexico
| | - Saúl Villa-Treviño
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, C.P. 07360, Mexico
| | - Verónica Rocío Vásquez-Garzón
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico; CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico
| | - Rafael Baltiérrez-Hoyos
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico; CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico.
| |
Collapse
|
6
|
Yao Z, Lu Y, Wang P, Chen Z, Zhou L, Sang X, Yang Q, Wang K, Hao M, Cao G. The role of JNK signaling pathway in organ fibrosis. J Adv Res 2024:S2090-1232(24)00431-4. [PMID: 39366483 DOI: 10.1016/j.jare.2024.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Fibrosis is a tissue damage repair response caused by multiple pathogenic factors which could occur in almost every apparatus and leading to the tissue structure damage, physiological abnormality, and even organ failure until death. Up to now, there is still no specific drugs or strategies can effectively block or changeover tissue fibrosis. JNKs, a subset of mitogen-activated protein kinases (MAPK), have been reported that participates in various biological processes, such as genetic expression, DNA damage, and cell activation/proliferation/death pathways. Increasing studies indicated that abnormal regulation of JNK signal pathway has strongly associated with tissue fibrosis. AIM OF REVIEW This review designed to sum up the molecular mechanism progresses in the role of JNK signal pathway in organ fibrosis, hoping to provide a novel therapy strategy to tackle tissue fibrosis. KEY SCIENTIFIC CONCEPTS OF REVIEW Recent evidence shows that JNK signaling pathway could modulates inflammation, immunoreaction, oxidative stress and Multiple cell biological functions in organ fibrosis. Therefore, targeting the JNK pathway may be a useful strategy in cure fibrosis.
Collapse
Affiliation(s)
- Zhouhui Yao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yandan Lu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Pingping Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ziyan Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Licheng Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xianan Sang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qiao Yang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Kuilong Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Min Hao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Songyang Research Institute of Zhejiang Chinese Medical University, Songyang, 323400, China.
| | - Gang Cao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
7
|
Nemeth J, Skronska-Wasek W, Keppler S, Schundner A, Groß A, Schoenberger T, Quast K, El Kasmi KC, Ruppert C, Günther A, Frick M. Adiponectin suppresses stiffness-dependent, profibrotic activation of lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 2024; 327:L487-L502. [PMID: 39104319 PMCID: PMC11482465 DOI: 10.1152/ajplung.00037.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/05/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, irreversible respiratory disease with limited therapeutic options. A hallmark of IPF is excessive fibroblast activation and extracellular matrix (ECM) deposition. The resulting increase in tissue stiffness amplifies fibroblast activation and drives disease progression. Dampening stiffness-dependent activation of fibroblasts could slow disease progression. We performed an unbiased, next-generation sequencing (NGS) screen to identify signaling pathways involved in stiffness-dependent lung fibroblast activation. Adipocytokine signaling was downregulated in primary lung fibroblasts (PFs) cultured on stiff matrices. Re-activating adipocytokine signaling with adiponectin suppressed stiffness-dependent activation of human PFs. Adiponectin signaling depended on CDH13 expression and p38 mitogen-activated protein kinase gamma (p38MAPKγ) activation. CDH13 expression and p38MAPKγ activation were strongly reduced in lungs from IPF donors. Our data suggest that adiponectin-signaling via CDH13 and p38MAPKγ activation suppresses profibrotic activation of fibroblasts in the lung. Targeting of the adiponectin signaling cascade may provide therapeutic benefits in IPF.NEW & NOTEWORTHY A hallmark of idiopathic pulmonary fibrosis (IPF) is excessive fibroblast activation and extracellular matrix (ECM) deposition. The resulting increase in tissue stiffness amplifies fibroblast activation and drives disease progression. Dampening stiffness-dependent activation of fibroblasts could slow disease progression. We found that activation of the adipocytokine signaling pathway halts and reverses stiffness-induced, profibrotic fibroblast activation. Specific targeting of this signaling cascade may therefore provide therapeutic benefits in IPF.
Collapse
Affiliation(s)
- Julia Nemeth
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | | - Sophie Keppler
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | | - Alexander Groß
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | | | - Karsten Quast
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Clemens Ruppert
- Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
| | - Andreas Günther
- Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
- Center for Interstitial and Rare Lung Diseases, Justus-Liebig University Giessen, Giessen, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
8
|
Fuster-Martínez I, Calatayud S. The current landscape of antifibrotic therapy across different organs: A systematic approach. Pharmacol Res 2024; 205:107245. [PMID: 38821150 DOI: 10.1016/j.phrs.2024.107245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Fibrosis is a common pathological process that can affect virtually all the organs, but there are hardly any effective therapeutic options. This has led to an intense search for antifibrotic therapies over the last decades, with a great number of clinical assays currently underway. We have systematically reviewed all current and recently finished clinical trials involved in the development of new antifibrotic drugs, and the preclinical studies analyzing the relevance of each of these pharmacological strategies in fibrotic processes affecting tissues beyond those being clinically studied. We analyze and discuss this information with the aim of determining the most promising options and the feasibility of extending their therapeutic value as antifibrotic agents to other fibrotic conditions.
Collapse
Affiliation(s)
- Isabel Fuster-Martínez
- Departamento de Farmacología, Universitat de València, Valencia 46010, Spain; FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia 46020, Spain.
| | - Sara Calatayud
- Departamento de Farmacología, Universitat de València, Valencia 46010, Spain; CIBERehd (Centro de Investigación Biomédica en Red - Enfermedades Hepáticas y Digestivas), Spain.
| |
Collapse
|
9
|
Li L, Zhang G, Yang Z, Kang X. Stress-Activated Protein Kinases in Intervertebral Disc Degeneration: Unraveling the Impact of JNK and p38 MAPK. Biomolecules 2024; 14:393. [PMID: 38672411 PMCID: PMC11047866 DOI: 10.3390/biom14040393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Intervertebral disc degeneration (IDD) is a major cause of lower back pain. The pathophysiological development of IDD is closely related to the stimulation of various stressors, including proinflammatory cytokines, abnormal mechanical stress, oxidative stress, metabolic abnormalities, and DNA damage, among others. These factors prevent normal intervertebral disc (IVD) development, reduce the number of IVD cells, and induce senescence and apoptosis. Stress-activated protein kinases (SAPKs), particularly, c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38 MAPK), control cell signaling in response to cellular stress. Previous studies have shown that these proteins are highly expressed in degenerated IVD tissues and are involved in complex biological signal-regulated processes. Therefore, we summarize the research reports on IDD related to JNK and p38 MAPK. Their structure, function, and signal regulation mechanisms are comprehensively and systematically described and potential therapeutic targets are proposed. This work could provide a reference for future research and help improve molecular therapeutic strategies for IDD.
Collapse
Affiliation(s)
- Lei Li
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (L.L.); (G.Z.); (Z.Y.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou 730030, China
| | - Guangzhi Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (L.L.); (G.Z.); (Z.Y.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou 730030, China
| | - Zhili Yang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (L.L.); (G.Z.); (Z.Y.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou 730030, China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (L.L.); (G.Z.); (Z.Y.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou 730030, China
| |
Collapse
|
10
|
Fortier SM, Walker NM, Penke LR, Baas JD, Shen Q, Speth JM, Huang SK, Zemans RL, Bennett AM, Peters-Golden M. MAPK phosphatase 1 inhibition of p38α within lung myofibroblasts is essential for spontaneous fibrosis resolution. J Clin Invest 2024; 134:e172826. [PMID: 38512415 PMCID: PMC11093610 DOI: 10.1172/jci172826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 03/15/2024] [Indexed: 03/23/2024] Open
Abstract
Fibrosis following tissue injury is distinguished from normal repair by the accumulation of pathogenic and apoptosis-resistant myofibroblasts (MFs), which arise primarily by differentiation from resident fibroblasts. Endogenous molecular brakes that promote MF dedifferentiation and clearance during spontaneous resolution of experimental lung fibrosis may provide insights that could inform and improve the treatment of progressive pulmonary fibrosis in patients. MAPK phosphatase 1 (MKP1) influences the cellular phenotype and fate through precise and timely regulation of MAPK activity within various cell types and tissues, yet its role in lung fibroblasts and pulmonary fibrosis has not been explored. Using gain- and loss-of-function studies, we found that MKP1 promoted lung MF dedifferentiation and restored the sensitivity of these cells to apoptosis - effects determined to be mainly dependent on MKP1's dephosphorylation of p38α MAPK (p38α). Fibroblast-specific deletion of MKP1 following peak bleomycin-induced lung fibrosis largely abrogated its subsequent spontaneous resolution. Such resolution was restored by treating these transgenic mice with the p38α inhibitor VX-702. We conclude that MKP1 is a critical antifibrotic brake whose inhibition of pathogenic p38α in lung fibroblasts is necessary for fibrosis resolution following lung injury.
Collapse
Affiliation(s)
- Sean M. Fortier
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Natalie M. Walker
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Loka R. Penke
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jared D. Baas
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Qinxue Shen
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jennifer M. Speth
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Steven K. Huang
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Rachel L. Zemans
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Anton M. Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
11
|
Pillai M, Lafortune P, Dabo A, Yu H, Park SS, Taluru H, Ahmed H, Bobrow D, Sattar Z, Jundi B, Reece J, Ortega RR, Soto B, Yewedalsew S, Foronjy R, Wyman A, Geraghty P, Ohlmeyer M. Small-Molecule Activation of Protein Phosphatase 2A Counters Bleomycin-Induced Fibrosis in Mice. ACS Pharmacol Transl Sci 2023; 6:1659-1672. [PMID: 37974628 PMCID: PMC10644462 DOI: 10.1021/acsptsci.3c00117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Indexed: 11/19/2023]
Abstract
The activity of protein phosphatase 2A (PP2A), a serine-threonine phosphatase, is reduced in the lung fibroblasts of idiopathic pulmonary fibrosis (IPF) patients. The objective of this study was to determine whether the reactivation of PP2A could reduce fibrosis and preserve the pulmonary function in a bleomycin (BLM) mouse model. Here, we present a new class of direct small-molecule PP2A activators, diarylmethyl-pyran-sulfonamide, exemplified by ATUX-1215. ATUX-1215 has improved metabolic stability and bioavailability compared to our previously described PP2A activators. Primary human lung fibroblasts were exposed to ATUX-1215 and an older generation PP2A activator in combination with TGFβ. ATUX-1215 treatment enhanced the PP2A activity, reduced the phosphorylation of ERK and JNK, and reduced the TGFβ-induced expression of ACTA2, FN1, COL1A1, and COL3A1. C57BL/6J mice were administered 5 mg/kg ATUX-1215 daily following intratracheal instillation of BLM. Three weeks later, forced oscillation and expiratory measurements were performed using the Scireq Flexivent System. ATUX-1215 prevented BLM-induced lung physiology changes, including the preservation of normal PV loop, compliance, tissue elastance, and forced vital capacity. PP2A activity was enhanced with ATUX-1215 and reduced collagen deposition within the lungs. ATUX-1215 also prevented the BLM induction of Acta2, Ccn2, and Fn1 gene expression. Treatment with ATUX-1215 reduced the phosphorylation of ERK, p38, JNK, and Akt and the secretion of IL-12p70, GM-CSF, and IL1α in BLM-treated animals. Delayed treatment with ATUX-1215 was also observed to slow the progression of lung fibrosis. In conclusion, our study indicates that the decrease in PP2A activity, which occurs in fibroblasts from the lungs of IPF subjects, could be restored with ATUX-1215 administration as an antifibrotic agent.
Collapse
Affiliation(s)
- Meshach Pillai
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Pascale Lafortune
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Abdoulaye Dabo
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Howard Yu
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Sangmi S. Park
- Department
of Cell Biology, The State University of
New York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Harsha Taluru
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Huma Ahmed
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Dylan Bobrow
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Zeeshan Sattar
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Bakr Jundi
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Joshua Reece
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Romy Rodriguez Ortega
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Brian Soto
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Selome Yewedalsew
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Robert Foronjy
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Anne Wyman
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | - Patrick Geraghty
- Department
of Medicine, The State University of New
York Downstate Health Sciences University, Brooklyn, New York 11203, United States
- Department
of Cell Biology, The State University of
New York Downstate Health Sciences University, Brooklyn, New York 11203, United States
| | | |
Collapse
|
12
|
Yu D, Xiang Y, Gou T, Tong R, Xu C, Chen L, Zhong L, Shi J. New therapeutic approaches against pulmonary fibrosis. Bioorg Chem 2023; 138:106592. [PMID: 37178650 DOI: 10.1016/j.bioorg.2023.106592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023]
Abstract
Pulmonary fibrosis is the end-stage change of a large class of lung diseases characterized by the proliferation of fibroblasts and the accumulation of a large amount of extracellular matrix, accompanied by inflammatory damage and tissue structure destruction, which also shows the normal alveolar tissue is damaged and then abnormally repaired resulting in structural abnormalities (scarring). Pulmonary fibrosis has a serious impact on the respiratory function of the human body, and the clinical manifestation is progressive dyspnea. The incidence of pulmonary fibrosis-related diseases is increasing year by year, and no curative drugs have appeared so far. Nevertheless, research on pulmonary fibrosis have also increased in recent years, but there are no breakthrough results. Pathological changes of pulmonary fibrosis appear in the lungs of patients with coronavirus disease 2019 (COVID-19) that have not yet ended, and whether to improve the condition of patients with COVID-19 by means of the anti-fibrosis therapy, which are the questions we need to address now. This review systematically sheds light on the current state of research on fibrosis from multiple perspectives, hoping to provide some references for design and optimization of subsequent drugs and the selection of anti-fibrosis treatment plans and strategies.
Collapse
Affiliation(s)
- Dongke Yu
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yu Xiang
- College of Medicine, University of Electronic Science and Technology, Chengdu 610072, China
| | - Tingting Gou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Chuan Xu
- Department of Oncology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Lu Chen
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| | - Ling Zhong
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu 610072, China.
| | - Jianyou Shi
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| |
Collapse
|
13
|
Mohammed OA, Abdel-Reheim MA, Saleh LA, Alamri MMS, Alfaifi J, Adam MIE, Farrag AA, AlQahtani AAJ, BinAfif WF, Hashish AA, Abdel-Ghany S, Elmorsy EA, El-wakeel HS, Doghish AS, Hamad RS, Saber S. Alvespimycin Exhibits Potential Anti-TGF-β Signaling in the Setting of a Proteasome Activator in Rats with Bleomycin-Induced Pulmonary Fibrosis: A Promising Novel Approach. Pharmaceuticals (Basel) 2023; 16:1123. [PMID: 37631038 PMCID: PMC10458542 DOI: 10.3390/ph16081123] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an irreversible and life-threatening lung disease of unknown etiology presenting only a few treatment options. TGF-β signaling orchestrates a cascade of events driving pulmonary fibrosis (PF). Notably, recent research has affirmed the augmentation of TGF-β receptor (TβR) signaling via HSP90 activation. HSP90, a molecular chaperone, adeptly stabilizes and folds TβRs, thus intricately regulating TGF-β1 signaling. Our investigation illuminated the impact of alvespimycin, an HSP90 inhibitor, on TGF-β-mediated transcriptional responses by inducing destabilization of TβRs. This outcome stems from the explicit interaction of TβR subtypes I and II with HSP90, where they are clients of this cellular chaperone. It is worth noting that regulation of proteasome-dependent degradation of TβRs is a critical standpoint in the termination of TGF-β signal transduction. Oleuropein, the principal bioactive compound found in Olea europaea, is acknowledged for its role as a proteasome activator. In this study, our aim was to explore the efficacy of a combined therapy involving oleuropein and alvespimycin for the treatment of PF. We employed a PF rat model that was induced by intratracheal bleomycin infusion. The application of this dual therapy yielded a noteworthy impediment to the undesired activation of TGF-β/mothers against decapentaplegic homologs 2 and 3 (SMAD2/3) signaling. Consequently, this novel combination showcased improvements in both lung tissue structure and function while also effectively restraining key fibrosis markers such as PDGF-BB, TIMP-1, ACTA2, col1a1, and hydroxyproline. On a mechanistic level, our findings unveiled that the antifibrotic impact of this combination therapy likely stemmed from the enhanced degradation of both TβRI and TβRII. In conclusion, the utilization of proteasomal activators in conjunction with HSP90 inhibitors ushers in a promising frontier for the management of PF.
Collapse
Affiliation(s)
- Osama A. Mohammed
- Department of Clinical Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt
| | - Lobna A. Saleh
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt;
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | | | - Jaber Alfaifi
- Department of Child Health, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Masoud I. E. Adam
- Department of Medical Education and Internal Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Alshaimaa A. Farrag
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt;
- Unit of Anatomy, Department of Basic Medical Sciences, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - AbdulElah Al Jarallah AlQahtani
- Department of Internal Medicine, Division of Dermatology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Waad Fuad BinAfif
- Department of Internal Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Abdullah A. Hashish
- Department of Basic Medical Sciences, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Sameh Abdel-Ghany
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (S.A.-G.); (E.A.E.)
| | - Elsayed A. Elmorsy
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (S.A.-G.); (E.A.E.)
- Pharmacology and Therapeutics Department, Qassim College of Medicine, Qassim University, Buraydah 51452, Saudi Arabia
| | - Hend S. El-wakeel
- Physiology Department, Benha Faculty of Medicine, Benha University, Benha 13518, Egypt;
- Physiology Department, Albaha Faculty of Medicine, Albaha University, Al Baha 65799, Saudi Arabia
| | - Ahmed S. Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo, Cairo 11829, Egypt;
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11231, Egypt
| | - Rabab S. Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia;
- Central Laboratory, Theodor Bilharz Research Institute, Giza 12411, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| |
Collapse
|
14
|
Chu KA, Yeh CC, Hsu CH, Hsu CW, Kuo FH, Tsai PJ, Fu YS. Reversal of Pulmonary Fibrosis: Human Umbilical Mesenchymal Stem Cells from Wharton's Jelly versus Human-Adipose-Derived Mesenchymal Stem Cells. Int J Mol Sci 2023; 24:ijms24086948. [PMID: 37108112 PMCID: PMC10139084 DOI: 10.3390/ijms24086948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Pulmonary fibrosis (PF) is a progressive, non-reversible illness with various etiologies. Currently, effective treatments for fibrotic lungs are still lacking. Here, we compared the effectiveness of transplantation of human mesenchymal stem cells from umbilical cord Wharton's jelly (HUMSCs) versus those from adipose tissue (ADMSCs) in reversing pulmonary fibrosis in rats. Bleomycin 5 mg was intratracheally injected to establish a severe, stable, single left lung animal model with PF. On Day 21 post-BLM administration, one single transplantation of 2.5 × 107 HUMSCs or ADMSCs was performed. Lung function examination of Injury and Injury+ADMSCs rats displayed significantly decreased blood oxygen saturation and increased respiratory rates, while Injury+HUMSCs rats showed statistical amelioration in blood oxygen saturation and significant alleviation in respiratory rates. Reduced cell number in the bronchoalveolar lavage and lower myofibroblast activation appeared in the rats transplanted with either ADMSCs or HUMSCS than that in the Injury group. However, ADMSC transplantation stimulated more adipogenesis. Furthermore, matrix-metallopeptidase-9 over-expression for collagen degradation, and the elevation of Toll-like receptor-4 expression for alveolar regeneration were observed only in the Injury+HUMSCs. In comparison with the transplantation of ADMSCs, transplantation of HUMSCs exhibited a much more effective therapeutic effect on PF, with significantly better results in alveolar volume and lung function.
Collapse
Affiliation(s)
- Kuo-An Chu
- Division of Chest Medicine, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
- Department of Nursing, Shu-Zen Junior College of Medicine and Management, Kaohsiung 821004, Taiwan
- School of Nursing, Fooyin University, Kaohsiung 831301, Taiwan
| | - Chang-Ching Yeh
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Department of Obstetrics and Gynecology, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Nurse-Midwifery and Women Health, National Taipei University of Nursing and Health Sciences, Taipei 112303, Taiwan
| | - Chun-Hsiang Hsu
- Division of Chest Medicine, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
| | - Chien-Wei Hsu
- Division of Chest Medicine, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Medical Intensive Unit, Department of Critical Care Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
| | - Fu-Hsien Kuo
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Pei-Jiun Tsai
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Trauma Center, Department of Surgery, Veterans General Hospital, Taipei 112201, Taiwan
- Department of Critical Care Medicine, Veterans General Hospital, Taipei 112201, Taiwan
| | - Yu-Show Fu
- Department of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
15
|
Ligresti G, Raslan AA, Hong J, Caporarello N, Confalonieri M, Huang SK. Mesenchymal cells in the Lung: Evolving concepts and their role in fibrosis. Gene 2023; 859:147142. [PMID: 36603696 PMCID: PMC10068350 DOI: 10.1016/j.gene.2022.147142] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 01/03/2023]
Abstract
Mesenchymal cells in the lung are crucial during development, but also contribute to the pathogenesis of fibrotic disorders, including idiopathic pulmonary fibrosis (IPF), the most common and deadly form of fibrotic interstitial lung diseases. Originally thought to behave as supporting cells for the lung epithelium and endothelium with a singular function of producing basement membrane, mesenchymal cells encompass a variety of cell types, including resident fibroblasts, lipofibroblasts, myofibroblasts, smooth muscle cells, and pericytes, which all occupy different anatomic locations and exhibit diverse homeostatic functions in the lung. During injury, each of these subtypes demonstrate remarkable plasticity and undergo varying capacity to proliferate and differentiate into activated myofibroblasts. Therefore, these cells secrete high levels of extracellular matrix (ECM) proteins and inflammatory cytokines, which contribute to tissue repair, or in pathologic situations, scarring and fibrosis. Whereas epithelial damage is considered the initial trigger that leads to lung injury, lung mesenchymal cells are recognized as the ultimate effector of fibrosis and attempts to better understand the different functions and actions of each mesenchymal cell subtype will lead to a better understanding of why fibrosis develops and how to better target it for future therapy. This review summarizes current findings related to various lung mesenchymal cells as well as signaling pathways, and their contribution to the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Giovanni Ligresti
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US.
| | - Ahmed A Raslan
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Jeongmin Hong
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, US
| | - Marco Confalonieri
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, US
| |
Collapse
|
16
|
Chen S, Gao D, Sun R, Bao J, Lu C, Zhang Z, Xiao T, Gu X, Zhou H. Anlotinib prove to be a potential therapy for the treatment of pulmonary fibrosis complicated with lung adenocarcinoma. Pulm Pharmacol Ther 2023; 80:102202. [PMID: 36906117 DOI: 10.1016/j.pupt.2023.102202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 02/13/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023]
Abstract
Pulmonary fibrosis is a chronic interstitial fibrosis lung disease with high mortality, which is often complicated with lung cancer. The incidence of IPF complicated with lung cancer is getting higher and higher. At present, there is no consensus on the management and treatment of pulmonary fibrosis patients with lung cancer. There is an urgent need to develop preclinical drug evaluation methods for IPF with lung cancer and potential therapeutic drugs for IPF with lung cancer. The pathogenic mechanism of IPF is similar to that of lung cancer, and the multi-effect drugs with anticancer and anti-fibrosis will have potential value in the treatment of IPF complicated with lung cancer. In this study, we established an animal model of IPF complicated with lung cancer in situ to evaluate the therapeutic effect of the antiangiogenic drug anlotinib. The pharmacodynamic results in vivo showed that anlotinib could significantly improve the lung function of IPF-LC mice, reduce the content of collagen in lung tissue, increase the survival rate of mice, and inhibit the growth of lung tumor in mice. The results of Western blot and immunohistochemical analysis of lung tissue showed that anlotinib significantly inhibited the expression of fibrosis marker protein α-SMA, Collagen I and Fibronectin and tumor proliferation marker protein PCNA in mouse lung tissue, and down-regulated the content of serum tumor marker CEA. Through transcriptome analysis, we found that anlotinib regulates MAPK signal pathway, PARP signal pathway and coagulation cascade signal pathway in lung cancer and pulmonary fibrosis, which all play an important role in lung cancer and pulmonary fibrosis. In addition, there is crosstalk between the signal pathway participated by the target of anlotinib and MAPK, JAK/STAT and mTOR signal pathway. In summary, anlotinib will be a candidate for IPF-LC treatment.
Collapse
Affiliation(s)
- Shanshan Chen
- The First Affiliated Hospital of Zhengzhou University, 1 Longhu Middle Ring Road, Zhengzhou, Jinshui District, Henan Province, People's Republic of China.
| | - Dandi Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China.
| | - Ronghao Sun
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China.
| | - Jiali Bao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China.
| | - Chunya Lu
- The First Affiliated Hospital of Zhengzhou University, 1 Longhu Middle Ring Road, Zhengzhou, Jinshui District, Henan Province, People's Republic of China
| | - Zihui Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Ting Xiao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China.
| | - Xiaoting Gu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China.
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China.
| |
Collapse
|
17
|
Lu Y, Tang K, Wang S, Tian Z, Fan Y, Li B, Wang M, Zhao J, Xie J. Dach1 deficiency drives alveolar epithelium apoptosis in pulmonary fibrosis via modulating C-Jun/Bim activity. Transl Res 2023; 257:54-65. [PMID: 36754276 DOI: 10.1016/j.trsl.2023.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/10/2023] [Accepted: 01/30/2023] [Indexed: 02/08/2023]
Abstract
Dysregulation of type II alveolar epithelial cells (AECII) plays a vital role in the initiation and development of pulmonary fibrosis (PF). Dachshund homolog 1 (Dach1), frequently expressed in epithelial cells with stem cell potential, controls cell proliferation, apoptosis, and cell cycle in tissue development and disease process. In this study, we demonstrated that the lungs collected from PF patients and mice of Bleomycin (BLM)-treated were characterized by low expression of Dachshund homolog 1 (Dach1), especially in AECII. Dach1 deficiency in the alveolar epithelium exacerbated PF in BLM-treated mice, as evidenced by reduced pulmonary function and increased expression of fibrosis markers. Rather, treatment with lung-specific overexpression of Dach1 alleviated histopathological damage, lung compliance, and fibrosis in BLM-treated mice. Moreover, overexpression of Dach1 could inhibit epithelial apoptosis in vitro. Conversely, primary AECII with Dach1 depletion were more susceptible to apoptosis in vivo. Mechanically, Dach1 combined with C-Jun protooncogene selectively bound to the promoter of B-cell lymphoma 2 interacting mediators of cell death (Bim), by which it repressed Bim expression and alleviated epithelial apoptosis. Taken together, our data support that Dach1 in AECII contributes to the progression of PF and may be a viable target for the prevention and treatment of PF.
Collapse
Affiliation(s)
- Yanjiao Lu
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kum Tang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shanshan Wang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhen Tian
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Fan
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Boyu Li
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meijia Wang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jianping Zhao
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
18
|
Sadiq IZ. Free Radicals and Oxidative Stress: Signaling Mechanisms, Redox Basis for Human Diseases, and Cell Cycle Regulation. Curr Mol Med 2023; 23:13-35. [PMID: 34951363 DOI: 10.2174/1566524022666211222161637] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 12/16/2022]
Abstract
Free radicals contain one or more unpaired electrons in their valence shell, thus making them unstable, short-lived, and highly reactive species. Excessive generation of these free radicals ultimately leads to oxidative stress causing oxidation and damage to significant macromolecules in the living system and essentially disrupting signal transduction pathways and antioxidants equilibrium. At lower concentrations, ROS serves as "second messengers," influencing many physiological processes in the cell. However, higher concentrations beyond cell capacity cause oxidative stress, contributing to human pathologies such as diabetes, cancer, Parkinson's disease, cardiovascular diseases, cataract, asthma, hypertension, atherosclerosis, arthritis, and Alzheimer's disease. Signaling pathways such as NF-κB, MAPKs, PI3K/Akt/ mTOR, and Keap1-Nrf2- ARE modulate the detrimental effects of oxidative stress by increasing the expression of cellular antioxidant defenses, phase II detoxification enzymes, and decreased production of ROS. Free radicals such as H2O2 are indeed needed for the advancement of the cell cycle as these molecules influence DNA, proteins, and enzymes in the cell cycle pathway. In the course of cell cycle progression, the cellular redox environment becomes more oxidized, moving from the G1 phase, becoming higher in G2/M and moderate in the S phase. Signals in the form of an increase in cellular pro-oxidant levels are required, and these signals are often terminated by a rise in the amount of antioxidants and MnSOD with a decrease in the level of cyclin D1 proteins. Therefore, understanding the mechanism of cell cycle redox regulation will help in the therapy of many diseases.
Collapse
Affiliation(s)
- Idris Zubairu Sadiq
- Department of Biochemistry, Faculty of life Sciences, Ahmadu Bello University, Zaria-Nigeria
- Department of Biochemistry, Faculty of Sciences, Maryam Abacha American University of Niger, ADS Avenue, Roi Muhammad VI Du Maroc Maradi, Republique Du Niger
| |
Collapse
|
19
|
Sánchez-Roncancio C, García B, Gallardo-Hidalgo J, Yáñez JM. GWAS on Imputed Whole-Genome Sequence Variants Reveal Genes Associated with Resistance to Piscirickettsia salmonis in Rainbow Trout ( Oncorhynchus mykiss). Genes (Basel) 2022; 14:114. [PMID: 36672855 PMCID: PMC9859203 DOI: 10.3390/genes14010114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Genome-wide association studies (GWAS) allow the identification of associations between genetic variants and important phenotypes in domestic animals, including disease-resistance traits. Whole Genome Sequencing (WGS) data can help increase the resolution and statistical power of association mapping. Here, we conduced GWAS to asses he facultative intracellular bacterium Piscirickettsia salmonis, which affects farmed rainbow trout, Oncorhynchus mykiss, in Chile using imputed genotypes at the sequence level and searched for candidate genes located in genomic regions associated with the trait. A total of 2130 rainbow trout were intraperitoneally challenged with P. salmonis under controlled conditions and genotyped using a 57K single nucleotide polymorphism (SNP) panel. Genotype imputation was performed in all the genotyped animals using WGS data from 102 individuals. A total of 488,979 imputed WGS variants were available in the 2130 individuals after quality control. GWAS revealed genome-wide significant quantitative trait loci (QTL) in Omy02, Omy03, Omy25, Omy26 and Omy27 for time to death and in Omy26 for binary survival. Twenty-four (24) candidate genes associated with P. salmonis resistance were identified, which were mainly related to phagocytosis, innate immune response, inflammation, oxidative response, lipid metabolism and apoptotic process. Our results provide further knowledge on the genetic variants and genes associated with resistance to intracellular bacterial infection in rainbow trout.
Collapse
Affiliation(s)
- Charles Sánchez-Roncancio
- Doctorado en Acuicultura, Programa Cooperativo: Universidad de Chile. Universidad Católica del Norte. Pontificia Universidad Católica de Valparaíso, Chile
- Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago 8820808, Chile
| | - Baltasar García
- Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago 8820808, Chile
- Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, La Pintana, Santiago 8820808, Chile
| | - Jousepth Gallardo-Hidalgo
- Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago 8820808, Chile
- Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, La Pintana, Santiago 8820808, Chile
| | - José M. Yáñez
- Center for Research and Innovation in Aquaculture (CRIA), Universidad de Chile, Santiago 8820808, Chile
- Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, La Pintana, Santiago 8820808, Chile
- Núcleo Milenio de Salmonidos Invasores Australes (INVASAL), Concepcion 4030000, Chile
| |
Collapse
|
20
|
Xu G, Feng S, Sun R, Ding Q, Shi Y. Systematic Analysis Strategy Based on Network Pharmacology to Investigate the Potential Mechanism of Fritillaria thunbergii Miq. against Idiopathic Pulmonary Fibrosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2996878. [PMID: 36479180 PMCID: PMC9722288 DOI: 10.1155/2022/2996878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 01/05/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a long-term, distressing, and age-related interstitial lung disease characterized by a complicated etiology and irreversible progression. Fritillaria thunbergii Miq. (Zhe Beimu, ZBM) is frequently used for its heat-clearing and phlegm-resolving properties in herbal compounds for the treatment of IPF. However, the specific mechanisms underlying the effects of ZBM against IPF have not yet been reported. In this study, we applied a systematic analysis strategy based on network pharmacology to explore the probable core targets and major pathways of ZBM against IPF. In addition, molecular docking simulation and quantitative real-time polymerase chain reaction (qRT-PCR) were performed to preliminarily investigate the possible mechanisms underlying the therapeutic effects of ZBM on IPF. We collected a total of 86 components of ZBM and used network pharmacology analysis to screen nine presumptive targets of ZBM against IPF. The molecular-docking results indicated that the components of ZBM exhibited good binding activity with presumptive targets. The qRT-PCR results also suggested that ZBM may partly alleviate IPF by regulating the expression of presumptive targets. This study laid the foundation for further clinical applications of ZBM and the development of IPF-related therapeutic products.
Collapse
Affiliation(s)
- Gonghao Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Siwen Feng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Rui Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qi Ding
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen 518118, China
| | - Yuanyuan Shi
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen 518118, China
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
21
|
Liu F, Song C, Cai W, Chen J, Cheng K, Guo D, Duan DD, Liu Z. Shared mechanisms and crosstalk of COVID-19 and osteoporosis via vitamin D. Sci Rep 2022; 12:18147. [PMID: 36307516 PMCID: PMC9614744 DOI: 10.1038/s41598-022-23143-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/25/2022] [Indexed: 12/31/2022] Open
Abstract
Recently accumulated evidence implicates a close association of vitamin D (VitD) insufficiency to the incidence and clinical manifestations of the COVID-19 caused by severe acute respiratory syndrome coronavirus-2 (SARS-COV-2). Populations with insufficient VitD including patients with osteoporosis are more susceptible to SARS-COV-2 infection and patients with COVID-19 worsened or developed osteoporosis. It is currently unknown, however, whether osteoporosis and COVID-19 are linked by VitD insufficiency. In this study, 42 common targets for VitD on both COVID-19 and osteoporosis were identified among a total of 243 VitD targets. Further bioinformatic analysis revealed 8 core targets (EGFR, AR, ESR1, MAPK8, MDM2, EZH2, ERBB2 and MAPT) in the VitD-COVID-19-osteoporosis network. These targets are involved in the ErbB and MAPK signaling pathways critical for lung fibrosis, bone structural integrity, and cytokines through a crosstalk between COVID-19 and osteoporosis via the VitD-mediated conventional immune and osteoimmune mechanisms. Molecular docking confirmed that VitD binds tightly to the predicted targets. These findings support that VitD may target common signaling pathways in the integrated network of lung fibrosis and bone structural integrity as well as the immune systems. Therefore, VitD may serve as a preventive and therapeutic agent for both COVID-19 and osteoporosis.
Collapse
Affiliation(s)
- Fei Liu
- grid.410578.f0000 0001 1114 4286Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000 Sichuan China
| | - Chao Song
- grid.410578.f0000 0001 1114 4286Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000 Sichuan China
| | - Weiye Cai
- grid.410578.f0000 0001 1114 4286Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000 Sichuan China
| | - Jingwen Chen
- grid.410578.f0000 0001 1114 4286Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000 Sichuan China
| | - Kang Cheng
- grid.410578.f0000 0001 1114 4286Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000 Sichuan China
| | - Daru Guo
- grid.410578.f0000 0001 1114 4286Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000 Sichuan China
| | - Dayue Darrel Duan
- grid.410578.f0000 0001 1114 4286Center for Phenomics of Traditional Chinese Medicine, and the Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000 Sichuan China
| | - Zongchao Liu
- grid.410578.f0000 0001 1114 4286Department of Orthopedics, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, 646000 Sichuan China
| |
Collapse
|
22
|
Ma H, Liu S, Li S, Xia Y. Targeting Growth Factor and Cytokine Pathways to Treat Idiopathic Pulmonary Fibrosis. Front Pharmacol 2022; 13:918771. [PMID: 35721111 PMCID: PMC9204157 DOI: 10.3389/fphar.2022.918771] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/06/2022] [Indexed: 02/05/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease of unknown origin that usually results in death from secondary respiratory failure within 2–5 years of diagnosis. Recent studies have identified key roles of cytokine and growth factor pathways in the pathogenesis of IPF. Although there have been numerous clinical trials of drugs investigating their efficacy in the treatment of IPF, only Pirfenidone and Nintedanib have been approved by the FDA. However, they have some major limitations, such as insufficient efficacy, undesired side effects and poor pharmacokinetic properties. To give more insights into the discovery of potential targets for the treatment of IPF, this review provides an overview of cytokines, growth factors and their signaling pathways in IPF, which have important implications for fully exploiting the therapeutic potential of targeting cytokine and growth factor pathways. Advances in the field of cytokine and growth factor pathways will help slow disease progression, prolong life, and improve the quality of life for IPF patients in the future.
Collapse
Affiliation(s)
- Hongbo Ma
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Shengming Liu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Shanrui Li
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yong Xia
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
| |
Collapse
|
23
|
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: 0] [Impact Index Per Article: 0] [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
|
24
|
Mohamad EA, Mohamed ZN, Hussein MA, Elneklawi MS. GANE can Improve Lung Fibrosis by Reducing Inflammation via Promoting p38MAPK/TGF-β1/NF-κB Signaling Pathway Downregulation. ACS OMEGA 2022; 7:3109-3120. [PMID: 35097306 PMCID: PMC8792938 DOI: 10.1021/acsomega.1c06591] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/24/2021] [Indexed: 05/30/2023]
Abstract
There is a trend to use nanoparticles as distinct treatments for cancer treatment because they have overcome many of the limitations of traditional drug delivery systems. Gallic acid (GA) is an effective polyphenol in the treatment of tissue injuries. In this study, GA was loaded onto niosomes to produce gallic acid nanoemulsion (GANE) using a green synthesis technique. GANE's efficiency, morphology, UV absorption, release, and Fourier-transform infrared spectroscopy (FTIR) analysis were evaluated. An in vitro study was conducted on the A549 lung carcinoma cell line to determine the GANE cytotoxicity. Also, our study was extended to evaluate the protective effect of GANE against lipopolysaccharide (LPS)-induced pulmonary fibrosis in rats. GANE showed higher encapsulation efficiency and strong absorption at 280 nm. Transmission electron microscopy presented a spherical shape of the prepared nanoparticles, and FTIR demonstrated different spectra for the free gallic acid sample compared to GANE. GANE showed cytotoxicity for the A549 carcinoma lung cell line with a low IC50 value. It was found that oral administration of GANE at 32.8 and 82 mg/kg.b.w. and dexamethasone (0.5 mg/kg) provided significant protection against LPS-induced pulmonary fibrosis. GANE enhanced production of superoxide dismutase, GPx, and GSH. It simultaneously reduced the MDA level. The GANE and dexamethasone, induced the production of IL-4, but suppressed TNF-α and IL-6. On the other hand, the lung p38MAPK, TGF-β1, and NF-κB gene expression was downregulated in rats administrated with GANE when compared with the LPS-treated rats. Histological studies confirmed the effective effect of GANE as it had a lung-protective effect against LPS-induced lung fibrosis. It was noticed that GANE can inhibit oxidative stress, lipid peroxidation, and cytokines and downregulate p38MAPK, TGF-β1, and NF-κB gene expression to suppress the proliferation and migration of lung fibrotic cells.
Collapse
Affiliation(s)
- Ebtesam A. Mohamad
- Biophysics
Department, Faculty of Science, Cairo University, Cairo University Street, Giza 12613, Egypt
| | - Zahraa N. Mohamed
- Medical
Laboratory Department, Faculty of Applied Medical Sciences, October 6 University, 6th of October City 28125, Giza, Egypt
| | - Mohammed A. Hussein
- Biochemistry
Department, Faculty of Applied Medical Sciences, October 6 University, 6th of
October City 28125, Giza, Egypt
| | - Mona S. Elneklawi
- Biomedical
Equipment Department, Faculty of Applied Medical Sciences, October 6 University, 6th of October City 28125, Giza, Egypt
| |
Collapse
|
25
|
Pro-Inflammatory Serum Amyloid a Stimulates Renal Dysfunction and Enhances Atherosclerosis in Apo E-Deficient Mice. Int J Mol Sci 2021; 22:ijms222212582. [PMID: 34830462 PMCID: PMC8623330 DOI: 10.3390/ijms222212582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 11/16/2022] Open
Abstract
Acute serum amyloid A (SAA) is an apolipoprotein that mediates pro-inflammatory and pro-atherogenic pathways. SAA-mediated signalling is diverse and includes canonical and acute immunoregulatory pathways in a range of cell types and organs. This study aimed to further elucidate the roles for SAA in the pathogenesis of vascular and renal dysfunction. Two groups of male ApoE-deficient mice were administered SAA (100 µL, 120 µg/mL) or vehicle control (100 µL PBS) and monitored for 4 or 16 weeks after SAA treatment; tissue was harvested for biochemical and histological analyses at each time point. Under these conditions, SAA administration induced crosstalk between NF-κB and Nrf2 transcriptional factors, leading to downstream induction of pro-inflammatory mediators and antioxidant response elements 4 weeks after SAA administration, respectively. SAA treatment stimulated an upregulation of renal IFN-γ with a concomitant increase in renal levels of p38 MAPK and matrix metalloproteinase (MMP) activities, which is linked to tissue fibrosis. In the kidney of SAA-treated mice, the immunolocalisation of inducible nitric oxide synthase (iNOS) was markedly increased, and this was localised to the parietal epithelial cells lining Bowman’s space within glomeruli, which led to progressive renal fibrosis. Assessment of aortic root lesion at the study endpoint revealed accelerated atherosclerosis formation; animals treated with SAA also showed evidence of a thinned fibrous cap as judged by diffuse collagen staining. Together, this suggests that SAA elicits early renal dysfunction through promoting the IFN-γ-iNOS-p38 MAPK axis that manifests as the fibrosis of renal tissue and enhanced cardiovascular disease.
Collapse
|
26
|
Verma K, Pant M, Paliwal S, Dwivedi J, Sharma S. An Insight on Multicentric Signaling of Angiotensin II in Cardiovascular system: A Recent Update. Front Pharmacol 2021; 12:734917. [PMID: 34489714 PMCID: PMC8417791 DOI: 10.3389/fphar.2021.734917] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022] Open
Abstract
The multifaceted nature of the renin-angiotensin system (RAS) makes it versatile due to its involvement in pathogenesis of the cardiovascular disease. Angiotensin II (Ang II), a multifaceted member of RAS family is known to have various potential effects. The knowledge of this peptide has immensely ameliorated after meticulous research for decades. Several studies have evidenced angiotensin I receptor (AT1 R) to mediate the majority Ang II-regulated functions in the system. Functional crosstalk between AT1 R mediated signal transduction cascades and other signaling pathways has been recognized. The review will provide an up-to-date information and recent discoveries involved in Ang II receptor signal transduction and their functional significance in the cardiovascular system for potential translation in therapeutics. Moreover, the review also focuses on the role of stem cell-based therapies in the cardiovascular system.
Collapse
Affiliation(s)
- Kanika Verma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | - Malvika Pant
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | - Sarvesh Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Banasthali, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| |
Collapse
|