1
|
Wang JH, Li M, Xie PF, Si JY, Feng ZJ, Tang CF, Li JM. Procyanidin C1 ameliorates aging-related skin fibrosis through targeting EGFR to inhibit TGFβ/SMAD pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156787. [PMID: 40315640 DOI: 10.1016/j.phymed.2025.156787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/03/2025] [Accepted: 04/17/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND Aging-related skin fibrosis (SF) is a complex condition with limited treatment options. Procyanidin C1 (PCC1), a natural polyphenolic compound with demonstrated senolytic activity, has emerged as a potential therapeutic agent for fibrotic disorders through its selective elimination of senescent cells. However, its therapeutic efficacy and mechanisms in aging-related SF remain unclear. PURPOSE This study aimed to investigate the mechanisms of PCC1 in aging-related SF. RESULTS In D-galactose-induced L929 cells, PCC1 treatment significantly attenuated the expression of both senescence-associated markers (IL-1β, P16, P21 and LMNB1) and fibrosis-related markers (α-SMA, LOXL2 and COL1). Network pharmacology and experimental validation (molecular docking, DARTS, CETSA, MST) identified EGFR as a primary target, with PCC1 directly binding to and inhibiting EGFR phosphorylation. Furthermore, PCC1 treatment effectively down-regulated TGFβ1 expression and suppressed SMAD2/3 phosphorylation in D-galactose-induced L929 cells. Notably, PCC1 blocked NSC228155-induced EGFR phosphorylation and inhibited ERK/MAPK, AKT/mTOR and TGFβ/SMAD pathway activation. In bleomycin-induced SF mice, PCC1 significantly attenuated epidermal hyperplasia, improved collagen structure, restored the collagen I/III ratio, and reduced EGFR phosphorylation along with TGFβ1 expression and SMAD2/3 phosphorylation. CONCLUSION This study elucidates that PCC1 exerts its anti-fibrotic effects through dual mechanisms: resistance to cellular senescence and modulation of fibroblast heterogeneity. By directly binding to EGFR and inhibiting its phosphorylation, PCC1 subsequently suppresses multiple downstream signaling cascades, ultimately ameliorating TGFβ/SMAD-mediated SF. These findings establish PCC1 as a promising therapeutic candidate for aging-related skin fibrosis, offering a novel approach through targeted EGFR inhibition and comprehensive pathway modulation.
Collapse
Affiliation(s)
- Jun-Han Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Min Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Peng-Fei Xie
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Jia-Yao Si
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Zhen-Jie Feng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Chuan-Feng Tang
- State Key Laboratory of Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jian-Mei Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
2
|
Dagnachew YM, Lim HY, Wupeng L, Lim SY, Lim SJN, Thiam CH, Tan SW, Eng JLJ, Mei D, Hazwany Mohammad Azhar S, Ong WS, Tan QHC, Wong WSF, Angeli V. Collagen deposition in lung parenchyma driven by depletion of interstitial Lyve-1 + macrophages prevents cigarette smoke-induced emphysema and loss of airway function. Front Immunol 2025; 15:1493395. [PMID: 39830508 PMCID: PMC11738928 DOI: 10.3389/fimmu.2024.1493395] [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: 09/09/2024] [Accepted: 12/10/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction Collagen is essential for maintaining lung structure and function and its remodeling has been associated with respiratory diseases including chronic obstructive pulmonary disease (COPD). However, the cellular mechanisms driving collagen remodeling and the functional implications of this process in the pathophysiology of pulmonary diseases remain poorly understood. Methods To address this question, we employed Lyve1wt/cre ; Csf1rflox/flox mice with specific depletion of Lyve-1+ macrophages and assessed the content, types and organization of collagen in lung compartments at steady state and after chronic exposure to cigarette smoke (CS). Results Using this mouse model, we found that the absence of this subpopulation of tissue resident macrophage led to the deposition of type I collagen fibers around the alveoli and bronchi at steady state. Further analysis by polarized light microscopy and Sircol collagen assay revealed that the collagen fibers accumulating in the lungs depleted of Lyve-1+ macrophages were thicker and crosslinked. A decrease in MMP-9 gene expression and proteolytic activity together with an increase in Col1a1, Timp-3 and Lox expression accompanied the collagen alterations. Next, we investigated the effect of the collagen remodeling on the pathophysiology of COPD and airway function in mice lacking Lyve-1+ macrophages exposed chronically to cigarette smoke (CS), a well-established animal model of COPD. We found that deposition of collagen prior CS exposure protected these mice against destruction of alveoli (emphysema), and bronchi thickening and prevented loss of airway function. Discussion Thus, we uncover that interstitial Lyve-1+ macrophages regulate the composition, amount, and architecture of collagen network in the lungs at steady state and that such collagen remodeling functionally impacts the development of COPD. This study further supports the potential of targeting collagen as promising approaches to treat respiratory diseases.
Collapse
Affiliation(s)
- Yinebeb Mezgebu Dagnachew
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology & Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Biomedical Sciences, School of Medicine, Bahir Dar University, Bahir Dar, Ethiopia
| | - Hwee Ying Lim
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology & Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Liao Wupeng
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore-Hebrew University of Jerusalem Alliance for Research and Enterprise, National University of Singapore, Singapore, Singapore
| | - Sheau Yng Lim
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology & Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Sheng Jie Natalie Lim
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology & Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Chung Hwee Thiam
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology & Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Shu Wen Tan
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology & Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Joan Lau Joo Eng
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology & Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Dan Mei
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Wei Siong Ong
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Qi Hui Caris Tan
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Wai-Shiu Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore-Hebrew University of Jerusalem Alliance for Research and Enterprise, National University of Singapore, Singapore, Singapore
- Drug Discovery and Optimization Platform (DDOP), Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Veronique Angeli
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology & Immunology, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
3
|
Ghaderi DD, Aronson MR, Mehta A, Friedman RM, McDaid KS, Giordano T, Jacobs IN, Gottardi R. Azithromycin Prevents Subglottic Stenosis in Mice. Laryngoscope 2025; 135:409-415. [PMID: 39276033 PMCID: PMC11635149 DOI: 10.1002/lary.31754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/06/2024] [Accepted: 07/24/2024] [Indexed: 09/16/2024]
Abstract
OBJECTIVE Pediatric subglottic stenosis (SGS) is characterized by subglottic narrowing which occurs when pathological fibroblasts deposit extracellular matrix that reduces airway patency. Recent clinical observations have suggested that azithromycin may have favorable impacts on SGS reduction while treating airway infections; furthermore, our recent work in mice demonstrated that the airway microbiome influences SGS. In this work, we characterize the protective effect of azithromycin as an immunomodulatory and antibacterial therapeutic against subglottic stenosis. METHODS Immunomodulatory and antifibrotic effects of azithromycin were assessed on TGF-β1-stimulated airway fibroblasts at 10 μg/mL for 5 days. Changes in gene expression were quantified by RT-qPCR and myofibroblast differentiation by α-SMA immunostaining. Murine airways were pretreated (2-weeks) with intranasal azithromycin before SGS injury by a twisted wire brush. Disease severity and immune response were characterized by histology and immunostaining for immune cells. RESULTS In vitro, azithromycin treatment of TGF-β1-stimulated fibroblasts exhibited strong reductions in extracellular matrix (COL1A1, LOX) and myofibroblast-related gene expression (ACTA2). Notably, there was a significant reduction in pro-fibrotic expression, which was observed with 10 μg/mL azithromycin. Immunostaining of fibroblasts for α-SMA revealed strong reductions in the number of positive-staining cells and the intensity of each positive cell. In vivo, azithromycin exhibited a significant decrease in lamina propria thickness indicative of reduced stenosis with associated changes in T-cell infiltration. CONCLUSIONS Overall, we show azithromycin prevents pro-fibrotic gene expression and myofibroblast differentiation and can help protect mice from developing SGS. This introduces azithromycin as a potential treatment for SGS. LEVEL OF EVIDENCE NA Laryngoscope, 135:409-415, 2025.
Collapse
Affiliation(s)
- Daniel D. Ghaderi
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Matthew R. Aronson
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Otolaryngology, Department of SurgeryChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Amrita Mehta
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ryan M. Friedman
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Otolaryngology, Department of SurgeryChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Kendra S. McDaid
- Department of Veterinary ResourcesChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Terri Giordano
- Division of Otolaryngology, Department of SurgeryChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Ian N. Jacobs
- Division of Otolaryngology, Department of SurgeryChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Department of Otorhinolaryngology‐Head and Neck SurgeryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Riccardo Gottardi
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Otolaryngology, Department of SurgeryChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Department of Otorhinolaryngology‐Head and Neck SurgeryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Division of Pulmonary Medicine, Department of PediatricsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Orthopaedic SurgeryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Ri.MED FoundationPalermoItaly
| |
Collapse
|
4
|
Park SH, Choi SH, Park HY, Ko J, Yoon JS. Role of Lysyl Oxidase-Like Protein 3 in the Pathogenesis of Graves' Orbitopathy in Orbital Fibroblasts. Invest Ophthalmol Vis Sci 2024; 65:33. [PMID: 39546293 DOI: 10.1167/iovs.65.13.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024] Open
Abstract
Purpose The lysyl oxidase (LOX) family has been implicated in the pathogenesis of diseases caused by inflammation and fibrosis. Therefore, we aimed to examine the role of lysyl oxidase-like protein 3 (LOXL3) in Graves' orbitopathy (GO) pathogenesis and its potential as a treatment target. Methods Quantitative real-time polymerase chain reaction compared the transcript levels of the five LOX family subtypes in orbital tissue explants obtained from patients with GO (n = 18) and healthy controls (n = 15). The effects of LOXL3 inhibition on interleukin (IL)-1β-induced proinflammatory cytokines, transforming growth factor (TGF)-β-induced profibrotic proteins, intracellular signaling molecules, and adipogenic markers were evaluated using Western blotting. Adipogenic differentiation was identified using Oil Red O staining. Results LOX and LOXL3 transcript levels were high in GO tissues. Stimulation with IL-1β, TGF-β, and insulin-like growth factor-1 significantly increased LOXL3 messenger RNA expression in GO fibroblasts. Furthermore, silencing LOXL3 attenuated the IL-1β-induced production of proinflammatory cytokines (IL-6, IL-8, and intercellular adhesion molecule-1) and TGF-β-induced production of profibrotic proteins (fibronectin, collagen 1α, and alpha-smooth muscle actin). It also reduced the IL-1β or TGF-β-induced expression of phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells, protein kinase B, extracellular signal-regulated kinase, and c-Jun N-terminal kinase. Additionally, LOXL3 silencing suppressed adipocyte differentiation and the expression of adipogenic transcription factors (leptin, AP-2, peroxisome proliferator-activated receptor gamma, and CCAAT/enhancer-binding protein). Conclusions LOXL3 is crucial in GO pathogenesis. LOXL3 inhibition reduced inflammatory cytokine production, fibrotic protein expression, and fibroblast differentiation into adipocytes. This study highlights LOXL3 as a potential therapeutic target for GO.
Collapse
Affiliation(s)
| | - Soo Hyun Choi
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Young Park
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - JaeSang Ko
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Sook Yoon
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
5
|
Tirunavalli SK, Andugulapati SB. Geneticin ameliorates pulmonary fibrosis by attenuating the TGF-β/Smad via modulating AMPK/SIRT1 signaling. Life Sci 2024; 346:122626. [PMID: 38614295 DOI: 10.1016/j.lfs.2024.122626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/18/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
AIM Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive condition with unknown aetiology that causes the lung parenchyma to scar incessantly, lowering the quality of life and hastening death. In this investigation, we studied the anti-fibrotic activity of Geneticin (a derivative of gentamycin) using in vitro and in vivo models. MAIN METHODS The TGF-β-mediated differentiation model was adopted to investigate (fibrotic marker's levels/expression) the anti-fibrotic activity of geneticin (GNC) in in-vitro scenarios (LL29 and DHLF cells). In vivo, the bleomycin (BLM)-induced pulmonary fibrosis model was employed by administering BLM intratracheally. Post 14 days of BLM administration, animals were treated with geneticin (6.25, 12.5, and 25 mg·kg-1) for another 14 days, and their therapeutic effect was investigated using a spectrum of techniques. KEY FINDINGS RTqPCR and western-blot results revealed that geneticin treatment significantly attenuated the TGF-β/BLM mediated fibrotic cascade of markers in both in-vitro and in-vivo models respectively. Further, the BLM-induced pulmonary fibrosis model revealed, that geneticin dose-dependently reduced the BLM-induced inflammatory cell infiltrations, and thickness of the alveoli walls, improved the structural distortion of the lung, and aided in improving the survival rate of the rats. Picrosirus and Masson's trichrome staining indicated that geneticin therapy reduced collagen deposition and, as a result, lung functional characteristics were improved as assessed by flexivent. Mechanistic studies have shown that geneticin reduced fibrosis by attenuating the TGF-β/Smad through modulating the AMPK/SIRT1 signaling. SIGNIFICANCE These findings suggest that geneticin may be a promising therapeutic agent for the treatment of pulmonary fibrosis in clinical settings.
Collapse
Affiliation(s)
- Satya Krishna Tirunavalli
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Sai Balaji Andugulapati
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India.
| |
Collapse
|
6
|
Wu Y, Shi W, Li H, Liu C, Shimizu K, Li R, Zhang C. Specneuzhenide improves bleomycin-induced pulmonary fibrosis in mice via AMPK-dependent reduction of PD-L1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155318. [PMID: 38493719 DOI: 10.1016/j.phymed.2023.155318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/29/2023] [Accepted: 12/25/2023] [Indexed: 03/19/2024]
Abstract
BACKGROUND Pulmonary fibrosis (PF) is an escalating global health issue, characterized by rising rates of morbidity and mortality annually. Consequently, further investigation of potential damage mechanisms and potential preventive strategies for PF are warranted. Specnuezhenide (SPN), a prominent secoiridoid compound derived from Ligustrum lucidum Ait, exhibits anti-inflammatory and anti-oxidative capacities, indicating the potential therapeutic actions on PF. However, the underlying mechanisms of SPN on PF remain unclear. PURPOSE This work was aimed at investigating the protective actions of SPN on PF and the potential mechanism. METHODS In vivo, mice were administrated with bleomycin (BLM) to establish PF model. PF mice were treated with SPN (45/90 mg/kg) by gavage. In vitro, we employed TGF-β1 (10 ng/mL)-induced MLE-12 and PLFs cells, which then were treated with SPN (5, 10, 20 µM). DARTS assay, biofilm interference experiment and molecular docking were performed to investigate the molecular target of SPN. RESULTS In vivo, we found SPN treatment improved survival rate, alleviated pathological changes through reducing BLM-induced extracellular matrix (ECM) deposition, as well as BLM-induced epithelial-mesenchymal transition (EMT). In vitro, SPN inhibited EMT and lung fibroblast transdifferentiation. Mechanistically, SPN activated the AMPK protein to decrease the abnormally high level of PD-L1. Furthermore, the compound C, known as an AMPK inhibitor, exhibited a significant hindrance to the inhibition of SPN on TGF-β1-caused fibroblast transdifferentiation and proliferation. This outcome could be attributed to the fact that compound C could eliminate the inhibitory effects of SPN on PD-L1 expression. Interestingly, DARTS assay, biofilm interference experiment and molecular docking results all indicated that SPN could bind to AMPK, which suggested that SPN might be a potential agonist targeting AMPK protein. CONCLUSION Altogether, the results in our work illustrated that SPN promoted AMPK-dependent reduction of PD-L1 protein, contributing to the inhibition of fibrosis progression. Thus, SPN may represent a potential AMPK agonist for PF treatment.
Collapse
Affiliation(s)
- Yanliang Wu
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Chinese Medicine Resources, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Wen Shi
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Chinese Medicine Resources, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Haini Li
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Chinese Medicine Resources, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Chang Liu
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Chinese Medicine Resources, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Kuniyoshi Shimizu
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Forest and Forest Products Sciences, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka 812-8581, Japan
| | - Renshi Li
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Chinese Medicine Resources, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Chaofeng Zhang
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Chinese Medicine Resources, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
7
|
Xu J, Zhang J, Chen W, Ni X. The tumor-associated fibrotic reactions in microenvironment aggravate glioma chemoresistance. Front Oncol 2024; 14:1388700. [PMID: 38863628 PMCID: PMC11165034 DOI: 10.3389/fonc.2024.1388700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/10/2024] [Indexed: 06/13/2024] Open
Abstract
Malignant gliomas are one of the most common and lethal brain tumors with poor prognosis. Most patients with glioblastoma (GBM) die within 2 years of diagnosis, even after receiving standard treatments including surgery combined with concomitant radiotherapy and chemotherapy. Temozolomide (TMZ) is the first-line chemotherapeutic agent for gliomas, but the frequent acquisition of chemoresistance generally leads to its treatment failure. Thus, it's urgent to investigate the strategies for overcoming glioma chemoresistance. Currently, many studies have elucidated that cancer chemoresistance is not only associated with the high expression of drug-resistance genes in glioma cells but also can be induced by the alterations of the tumor microenvironment (TME). Numerous studies have explored the use of antifibrosis drugs to sensitize chemotherapy in solid tumors, and surprisingly, these preclinical and clinical attempts have exhibited promising efficacy in treating certain types of cancer. However, it remains unclear how tumor-associated fibrotic alterations in the glioma microenvironment (GME) mediate chemoresistance. Furthermore, the possible mechanisms behind this phenomenon are yet to be determined. In this review, we have summarized the molecular mechanisms by which tumor-associated fibrotic reactions drive glioma transformation from a chemosensitive to a chemoresistant state. Additionally, we have outlined antitumor drugs with antifibrosis functions, suggesting that antifibrosis strategies may be effective in overcoming glioma chemoresistance through TME normalization.
Collapse
Affiliation(s)
- Jiaqi Xu
- The Second Clinical Medical School, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ji Zhang
- Department of Neurosurgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wubing Chen
- Department of Radiology, Wuxi Fifth People’s Hospital, Jiangnan University, Wuxi, China
| | - Xiangrong Ni
- The Second Clinical Medical School, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Plastic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Kaplan JL, Rivas VN, Connolly DJ. Advancing Treatments for Feline Hypertrophic Cardiomyopathy: The Role of Animal Models and Targeted Therapeutics. Vet Clin North Am Small Anim Pract 2023; 53:1293-1308. [PMID: 37414693 DOI: 10.1016/j.cvsm.2023.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Feline HCM is the most common cardiovascular disease in cats, leading to devastating outcomes, including congestive heart failure (CHF), arterial thromboembolism (ATE), and sudden death. Evidence demonstrating long-term survival benefit with currently available therapies is lacking. Therefore, it is imperative to explore intricate genetic and molecular pathways that drive HCM pathophysiology to inspire the development of novel therapeutics. Several clinical trials exploring new drug therapies are currently underway, including those investigating small molecule inhibitors and rapamycin. This article outlines the key work performed using cellular and animal models that has led to and continues to guide the development of new innovative therapeutic strategies.
Collapse
Affiliation(s)
- Joanna L Kaplan
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA, USA.
| | - Victor N Rivas
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - David J Connolly
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield, Hertfordshire, UK
| |
Collapse
|
9
|
Zeng Q, Zhou TT, Huang WJ, Huang XT, Huang L, Zhang XH, Sang XX, Luo YY, Tian YM, Wu B, Liu L, Luo ZQ, He B, Liu W, Tang SY. Asarinin attenuates bleomycin-induced pulmonary fibrosis by activating PPARγ. Sci Rep 2023; 13:14706. [PMID: 37679587 PMCID: PMC10485066 DOI: 10.1038/s41598-023-41933-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease that lacks effective treatment modalities. Once patients are diagnosed with IPF, their median survival is approximately 3-5 years. PPARγ is an important target for the prevention and treatment of pulmonary fibrosis. Asarinin is a lignan compound that can be extracted from food plant Asarum heterotropoides. In this study, we investigated the therapeutic effects of asarinin in a pulmonary fibrosis model constructed using bleomycin in mice and explored the underlying mechanisms. Intraperitoneal administration of asarinin to mice with pulmonary fibrosis showed that asarinin effectively attenuated pulmonary fibrosis, and this effect was significantly inhibited by the PPARγ inhibitor GW9662. Asarinin inhibited TGF-β1-induced fibroblast-to-myofibroblast transition in vitro, while GW9662 and PPARγ gene silencing significantly inhibited this effect. In addition, asarinin inhibited not only the canonical Smad pathway of TGF-β but also the non-canonical AKT and MAPK pathways by activating PPARγ. Our study demonstrates that asarinin can be used as a therapeutic agent for pulmonary fibrosis, and that PPARγ is its key target.
Collapse
Affiliation(s)
- Qian Zeng
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Ting-Ting Zhou
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Wen-Jie Huang
- School of Nursing, Hunan University of Medicine, Huaihua, Hunan, China
| | - Xiao-Ting Huang
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Lei Huang
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Xiao-Hua Zhang
- Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Xiao-Xue Sang
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yu-Yang Luo
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yu-Mei Tian
- School of Nursing, Hunan University of Medicine, Huaihua, Hunan, China
| | - Bin Wu
- School of Nursing, Hunan University of Medicine, Huaihua, Hunan, China
| | - Lin Liu
- School of Nursing, Hunan University of Medicine, Huaihua, Hunan, China
| | - Zi-Qiang Luo
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Bin He
- School of Nursing, Hunan University of Medicine, Huaihua, Hunan, China.
| | - Wei Liu
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, 172 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
10
|
Xu M, Zhao C, Song H, Wang C, Li H, Qiu X, Jing H, Zhuang W. Inhibitory effects of Schisandrin C on collagen behavior in pulmonary fibrosis. Sci Rep 2023; 13:13475. [PMID: 37596361 PMCID: PMC10439186 DOI: 10.1038/s41598-023-40631-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023] Open
Abstract
Pulmonary fibrosis (PF) is a serious progressive fibrotic disease that is characterized by excessive accumulation of extracellular matrix (ECM), thus resulting in stiff lung tissues. Lysyl oxidase (LOX) is an enzyme involved in fibrosis by catalyzing collagen cross-linking. Studies found that the ingredients in schisandra ameliorated bleomycin (BLM)-induced PF, but it is unknown whether the anti-PF of schisandra is related to LOX. In this study, we established models of PF including a mouse model stimulated by BLM and a HFL1 cell model induced by transforming growth factor (TGF)-β1 to evaluate the inhibition effects of Schisandrin C (Sch C) on PF. We observed that Sch C treatment decreased pulmonary indexes compared to control group. Treatment of Sch C showed a significant reduction in the accumulation of ECM as evidenced by decreased expressions of α-SMA, FN, MMP2, MMP9, TIMP1 and collagen proteins such as Col 1A1, and Col 3A1. In addition, the expression of LOX in the lung tissue of mice after Sch C treatment was effectively decreased compared with the MOD group. The inhibition effects in vitro were consistent with those in vivo. Mechanistic studies revealed that Sch C significantly inhibited TGF-β1/Smad2/3 and TNF-α/JNK signaling pathways. In conclusion, our data demonstrated that Sch C significantly ameliorated PF in vivo and vitro, which may play an important role by reducing ECM deposition and inhibiting the production of LOX.
Collapse
Affiliation(s)
- Mingchen Xu
- Department of Molecular Biology Test Technique, College of Medical Technology, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin, 132013, China
| | - Chenghe Zhao
- Department of Molecular Biology Test Technique, College of Medical Technology, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin, 132013, China
| | - Haiming Song
- Department of Molecular Biology Test Technique, College of Medical Technology, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin, 132013, China
| | - Chunmei Wang
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, China
| | - He Li
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, China
| | - Xudong Qiu
- Department of Hand Surgery, Affiliated Hospital, Beihua University, Jilin, China
| | - He Jing
- Department of Hand Surgery, Affiliated Hospital, Beihua University, Jilin, China
| | - Wenyue Zhuang
- Department of Molecular Biology Test Technique, College of Medical Technology, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin, 132013, China.
| |
Collapse
|
11
|
Ming S, Qu S, Wu Y, Wei J, Zhang G, Jiang G, Huang X. COVID-19 Metabolomic-Guided Amino Acid Therapy Protects from Inflammation and Disease Sequelae. Adv Biol (Weinh) 2023; 7:e2200265. [PMID: 36775870 DOI: 10.1002/adbi.202200265] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/06/2022] [Indexed: 02/14/2023]
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) has caused a worldwide pandemic since 2019. A metabolic disorder is a contributing factor to deaths from COVID-19. However, the underlying mechanism of metabolic dysfunction in COVID-19 patients and the potential interventions are not elucidated. Here targeted plasma metabolomic is performed, and the metabolite profiles among healthy controls, and asymptomatic, moderate, and severe COVID-19 patients are compared. Among the altered metabolites, arachidonic acid and linolenic acid pathway metabolites are profoundly up-regulated in COVID-19 patients. Arginine biosynthesis, alanine, aspartate, and glutamate metabolism pathways are significantly disturbed in asymptomatic patients. In the comparison of metabolite variances among the groups, higher levels of l-citrulline and l-glutamine are found in asymptomatic carriers and moderate or severe patients at the remission stage. Furthermore, l-citrulline and l-glutamine combination therapy is demonstrated to effectively protect mice from coronavirus infection and endotoxin-induced sepsis, and is observed to efficiently prevent the occurrence of pulmonary fibrosis and central nervous system damage. Collectively, the data reveal the metabolite profile of asymptomatic COVID-19 patients and propose a potential strategy for COVID-19 treatment.
Collapse
Affiliation(s)
- Siqi Ming
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong Province, 518100, China
| | - Siying Qu
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
| | - Jiayou Wei
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
| | - Guoliang Zhang
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong Province, 518100, China
| | - Guanmin Jiang
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong Province, 518100, China
| |
Collapse
|
12
|
Zhang S, Tong X, Liu S, Huang J, Zhang L, Zhang T, Wang D, Fan H. AAV9-Tspyl2 gene therapy retards bleomycin-induced pulmonary fibrosis by modulating downstream TGF-β signaling in mice. Cell Death Dis 2023; 14:389. [PMID: 37391440 PMCID: PMC10313802 DOI: 10.1038/s41419-023-05889-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 05/19/2023] [Accepted: 06/14/2023] [Indexed: 07/02/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating fibrotic lung disease characterized by scarring and destruction of the lung architecture, with limited treatment options. Targeted gene therapy to restore cell division autoantigen-1 (CDA1) expression may be a potential treatment approach to delay the progression of pulmonary fibrosis (PF). Here, we focused on CDA1, which was significantly decreased in human IPF, in a mouse model of bleomycin (BLM)-induced PF, and in transforming growth factor (TGF-β)-challenged lung fibroblasts. In vitro, CDA1 overexpression by lentivirus infection in human embryonic lung fibroblasts (HFL1 cells) inhibited the production of pro-fibrotic and pro-inflammatory cytokines, lung fibroblast-to-myofibroblast transition, and extracellular matrix protein expression induced by exogenous TGF-β1 treatment, whereas CDA1 knockdown with small interfering RNA promoted this effect. CDA1 overexpression also inhibited cell proliferation and migration. In a mouse model of BLM-induced PF, we provided novel evidence that the intratracheal delivery of adeno-associated virus serotype 9 carrying the mouse Tspyl2 gene reduced lung tissue inflammation and fibrosis. Mechanistically, CDA1, as a transcription regulator, could repress the TGF-β signal transduction in vivo and in vitro. In conclusion, our results show that Tspyl2 gene therapy plays an antifibrotic role by inhibiting the lung fibroblast-to-myofibroblast transition and downstream TGF-β/Smad3 signaling transduction in BLM-induced PF in mice, suggesting that CDA1 is an appropriate and promising therapeutic target for PF.
Collapse
Affiliation(s)
- Shijie Zhang
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, China
| | - Xiang Tong
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, China
| | - Sitong Liu
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, China
| | - Jizhen Huang
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, China
| | - Tianli Zhang
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, China
| | - Dongguang Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, China
| | - Hong Fan
- Department of Respiratory and Critical Care Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
13
|
Tian L, Sun T, Fan M, Lu H, Sun C. Novel silk protein/hyaluronic acid hydrogel loaded with azithromycin as an immunomodulatory barrier to prevent postoperative adhesions. Int J Biol Macromol 2023; 235:123811. [PMID: 36841387 DOI: 10.1016/j.ijbiomac.2023.123811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/12/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023]
Abstract
Peritoneal adhesions, a common postoperative complication of laparotomy, are still treated with physical barriers, but their efficacy and ease of use are controversial. In this paper, we developed a wound microenvironment-responsive hydrogel composed of Antheraea pernyi silk protein (ASF) from wild cocoons and tyramine-modified hyaluronic acid (HA-Ph) loaded with azithromycin (AZI), glucose oxidase (GOX), and horseradish peroxidase (HRP). In addition, GOX-catalyzed oxygen production enhanced the antibacterial ability of the hydrogel. Moreover, the drug-loaded hydrogel increased macrophage CD206 expression while decreasing IL-6 and TNF-α expression. More importantly, the retarding effect of this novel hydrogel system on AZI almost eliminated the appearance of postoperative adhesions in rats. It was also found that the novel hydrogel enhanced the modulation of the TLR-4/Myd88/NF-κB pathway and TGF-β/Smad2/3 pathway by azithromycin in the locally damaged peritoneum of rats, which accelerated the remodeling of damaged tissues and dramatically reduced the deposition of collagen. Therefore, spraying the novel drug-loaded hydrogel on postoperative abdominal wounds can effectively inhibit the formation of postoperative adhesions.
Collapse
Affiliation(s)
- Linan Tian
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Tongtong Sun
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Mengyao Fan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Hongyan Lu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Changshan Sun
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|