1
|
Yin X, Wang Y, Li M, Zhang E, Huang L, Yang C. Yi-Qi-Qing-Shi-Hua-Yu method improves uterine inflammation in rats with sequelae of pelvic inflammatory disease through the TLR4/NF-κB signaling pathway and regulates intestinal flora. Tissue Cell 2025; 95:102918. [PMID: 40253799 DOI: 10.1016/j.tice.2025.102918] [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: 11/07/2024] [Revised: 03/04/2025] [Accepted: 04/11/2025] [Indexed: 04/22/2025]
Abstract
Sequelae of pelvic inflammatory disease (SPID) is a common gynecological disease, which is often accompanied by pathological changes and inflammation, and may even lead to infertility. The Yi-Qi-Qing-Shi-Hua-Yu method (YQQSHY), as a traditional Chinese medicine treatment method, is considered to have potential therapeutic effects on SPID. This study will explore the efficacy and potential mechanism of YQQSHY on SPID. This study established a SPID rat model by mixed bacteria and evaluated the regulatory effect of YQQSHY on uterine tissue pathology, immune status, and intestinal flora in SPID rats through histopathology, molecular experiments, immunology, and intestinal flora sequencing analysis. H&E staining showed that YQQSHY significantly reduced the pathological changes and fibrosis in the uterine tissue of SPID rats. ELISA results showed that YQQSHY could significantly reduce the levels of pro-inflammatory cytokines in the serum and vaginal secretions of SPID rats and increase the expression of anti-inflammatory factors. Flow cytometry analysis showed that the YQQSHY treatment group significantly adjusted the proportion of T cells in the peripheral blood of SPID rats. Western blot showed that YQQSHY can regulate TLR4, MyD88, p-NF-KB p65, and induce the transcription of p65 into the nucleus. Immunofluorescence examination of T cell subsets in uterine tissue. In addition, intestinal flora sequencing results showed that YQQSHY significantly modulated the composition and diversity of the intestinal flora of SPID rats. In conclusion, YQQSHY inhibits inflammatory response, regulates T cell ratio, and improves intestinal flora structure through the TLR4/NF-κB signaling pathway, which is the main mechanism for improving uterine inflammation and fibrosis in SPID rats. These findings provide experimental basis and theoretical support for further exploring the application of YQQSHY in clinical treatment.
Collapse
Affiliation(s)
- Xiaolan Yin
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yan Wang
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Maoya Li
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Enfeng Zhang
- Chengdu University of Traditional Chinese Medicine, China
| | - Li Huang
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Chengcheng Yang
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| |
Collapse
|
2
|
Yang C, Qin Z, Ma H, Liu H, Hou M, Wei J, Guo H, An X, Yang F, Yang A, Dang Y, Zhang F. Epithelial cells and fibroblasts are both activated via TGF-β1 and GSK-3β pathways differentially in the comorbidity of pulmonary fibrosis with lung adenocarcinoma. Life Sci 2025; 374:123696. [PMID: 40349653 DOI: 10.1016/j.lfs.2025.123696] [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: 11/20/2024] [Revised: 04/06/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
AIMS Pulmonary fibrosis (PF) is always exacerbated by the comorbidity of lung adenocarcinoma (LUAD), and patients frequently died from the complications of PF instead of lung cancer. Although many studies have unveiled the mechanisms underlying PF exacerbation due to lung cancer resection and radiotherapy, the influence of lung cancer itself on PF remains enigmatic. MATERIALS AND METHODS We cocultivated mouse pulmonary cells with mouse LUAD cells to explore the influence of LUAD on the pathogenesis and progression of PF. Additionally, a comorbidity model of PF with LUAD was established in mice via intratracheal injection of bleomycin (BLM) followed by in situ transplantation of LUAD cells. Furthermore, immunofluorescence, immunohistochemistry, and molecular analyses were employed to elucidate the mechanisms underlying the exacerbation of PF by the comorbidity of LUAD. KEY FINDINGS We found that PF was significantly exacerbated by LUAD. In the microenvironment of LUAD, the epithelial-mesenchymal transition (EMT) was predominantly activated in lung epithelial cells, while the transformation of lung fibroblasts into myofibroblasts was markedly induced. The TGF-β and GSK-3β pathways were differentially activated in lung epithelial cells and fibroblasts. Furthermore, clinical samples confirmed the involvement of these pathways in the process of PF exacerbation induced by LUAD in patients' lung lesions of PF with LUAD. SIGNIFICANCE This study initially reveals that LUAD exacerbates PF by modulating epithelial cells and fibroblasts through TGF-β and GSK-3β pathways differentially. Practically, targeting the pathways of TGF-β and GSK-3β may promise a potential strategy for the prophylaxis of PF exacerbation in patients with LUAD.
Collapse
Affiliation(s)
- Chenguang Yang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu, China; School of Medicine, Tarim University, Alar 843300, Xinjiang, China
| | - Zijian Qin
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu, China; Lanzhou University First Affiliated Hospital, Lanzhou 730000, Gansu, China
| | - Hu Ma
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu, China
| | - Huanqin Liu
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu, China
| | - Mengdan Hou
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu, China
| | - Jing Wei
- Gansu Provincial People's Hospital, Lanzhou 730000, Gansu, China
| | - Hongyan Guo
- Gansu Second Provincial People's Hospital, Lanzhou 730000, Gansu, China
| | - Xiang An
- Lanzhou University First Affiliated Hospital, Lanzhou 730000, Gansu, China
| | - Feng Yang
- Gansu Provincial People's Hospital, Lanzhou 730000, Gansu, China
| | - Aijun Yang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu, China
| | - Yamei Dang
- Gansu Provincial People's Hospital, Lanzhou 730000, Gansu, China
| | - Fangfang Zhang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, Gansu, China.
| |
Collapse
|
3
|
Tian J, Yang X, Fan S, Peng X, Su H, Bi H, Qiu M. Highly oxygenated lanostane triterpenoids from Ganoderma applanatum and their anti-liver fibrosis effects. Bioorg Chem 2025; 161:108497. [PMID: 40288012 DOI: 10.1016/j.bioorg.2025.108497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 04/08/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Inspired by the intriguing structures and significant activities of Ganoderma triterpenoids (GTs), the EtOAc extract of Ganoderma applanatum was phytochemically investigated, leading to the isolation of 11 GTs, including 10 new one (1-10). Their structures including absolute configurations, were elucidated through IR, UV, HRESIMS, 1D NMR and 2D NMR data analyses. Notably, applanoids J (1) and K (2) represent the first example of GTs with an unprecedented B-seco-lanostane architecture featuring 1,4-cyclohexanedione motif. Meanwhile, compounds 1-7 and 9-11 were evaluated for their hepatoprotective activity using TGF-β1-induced liver fibrosis model, and some of them such as compounds 2, 4, 6, 7 and 10 significantly suppressed the abnormal upregulation of fibrosis-related genes FN, ACTA2 (encode α-SMA) and COL1A1. Mechanistic studies suggested that the anti-liver fibrosis effect of compound 7 may be mediated through inhibition of the TGF-β/Smad signaling pathway. This study not only illustrates the structural diversity of GTs but also highlights their potential as promising anti-liver fibrosis agents.
Collapse
Affiliation(s)
- Jianing Tian
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xiao Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen 518055, China
| | - Shicheng Fan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen 518055, China
| | - Xingrong Peng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Haiguo Su
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Huichang Bi
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen 518055, China.
| | - Minghua Qiu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| |
Collapse
|
4
|
Liang L, Dong Z, Shen Z, Zang Y, Yang W, Wu L, Bao L. Inhibitory effects of umbelliferone on carbon tetrachloride-induced hepatic fibrosis in rats through the TGF‑β1‑Smad signaling pathway. Mol Med Rep 2025; 32:171. [PMID: 40242963 PMCID: PMC12020354 DOI: 10.3892/mmr.2025.13536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Hepatic fibrosis (HF) is a critical marker of advanced‑stage chronic liver disease and involves pivotal contributions from hepatic stellate cells (HSCs). Currently, there are no effective treatments for HF. Umbelliferone (7‑hydroxycoumarin; UMB) is a natural compound with significant anti‑inflammatory, antioxidant and anti‑tumor activities. However, its potential efficacy in treating HF has not been studied. The present study explored the protective effects of UMB against HF, targeting the TGF‑β1‑Smad signaling pathway to explore the underlying mechanisms of UMB. Carbon tetrachloride (CCl4) was injected intraperitoneally to induce HF in rats and primary HSCs were treated in vitro with UMB to investigate the improvement effect of UMB on HF. The levels of fibrosis markers, inflammation, oxidative stress and TGF‑β1‑Smad signaling pathway in the rat liver tissue and HSCs were detected using hematoxylin and eosin staining, enzyme‑linked immunosorbent assay, reverse transcription‑quantitative PCR, Cell Counting Kit‑8 and western blotting. The improvement in liver histopathology, liver function indexes and fibrosis markers demonstrated that UMB markedly inhibited the CCl4‑induced HF and inflammation in the rats. Additionally, UMB prominently reduced the pro‑inflammatory factors and oxidative stress levels. In vitro, UMB markedly inhibited primary HSC activation and decreased alpha‑smooth muscle actin and collagen I expression. The mechanism experiment proved that UMB inhibited the TGF‑β1‑Smad signaling pathway and ameliorated HF. The present study was the first to demonstrate, to the best of the authors' knowledge, that UMB might be a promising natural active compound for treating HF. Its therapeutic effect is associated with its modulation of the TGF‑β1‑Smad signaling pathway.
Collapse
Affiliation(s)
- Lijuan Liang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010110, P.R. China
| | - Zhiheng Dong
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010030, P.R. China
| | - Ziqing Shen
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010110, P.R. China
| | - Yifan Zang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010110, P.R. China
| | - Wenlong Yang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010110, P.R. China
| | - Lan Wu
- Mongolia Medical School, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010110, P.R. China
| | - Lidao Bao
- Department of Pharmacy, Hohhot First Hospital, Hohhot, Inner Mongolia Autonomous Region 010030, P.R. China
| |
Collapse
|
5
|
Jin J, Xue Y, Tian L. Network toxicological analysis of sodium dehydroacetate in food safety. Food Chem Toxicol 2025; 201:115469. [PMID: 40274010 DOI: 10.1016/j.fct.2025.115469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/19/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
Sodium dehydroacetate (Na-DHA), a synthetic preservative under tightened regulations, was evaluated for multi-organ toxicity using network toxicology. ADMETlab3.0 predicted genotoxicity, hepatotoxicity, and carcinogenicity risks. Target mining identified 13 cancer-related, 11 liver injury-related, and 8 genotoxicity-related core genes, with shared hubs (ALOX5, PTGS2, SMAD3, TNF) across pathologies. Functional analyses revealed inflammation, oxidative stress, and immune dysregulation as central mechanisms. KEGG pathway analysis linked cancer/liver injury to AGE-RAGE signaling (TNF, NOX4) and genotoxicity to efferocytosis impairment (PTGS2, ALOX5), suggesting DNA repair disruption. The integrated network demonstrated Na-DHA's pleiotropic effects through convergent pathways, transcending organ-specific toxicity. This systemic profile challenges conventional single-endpoint assessments, advocating comprehensive multi-organ risk evaluation.
Collapse
Affiliation(s)
- Jing Jin
- Center for Rehabilitation Medicine, Department of Rehabilitation, Neurotumor Rehabilitation Ward, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yan Xue
- The Nanomedicine Research Laboratory, Hand Surgery Research Center, Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Liang Tian
- Center for Rehabilitation Medicine, Department of Rehabilitation, Neurotumor Rehabilitation Ward, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Yang F, Zhang X, Dai W, Xu K, Mei Y, Liu T, Wang K, Liang Q, Guo P, Liang C, Meng J. Multivariate genome-wide analysis of sarcopenia reveals genetic comorbidity with urological diseases. Exp Gerontol 2025; 206:112783. [PMID: 40378933 DOI: 10.1016/j.exger.2025.112783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/23/2025] [Accepted: 05/12/2025] [Indexed: 05/19/2025]
Abstract
INTRODUCTION Sarcopenia is a prevalent age-related disorder characterized by progressive loss of muscle mass, strength, and physical performance. While genome-wide association studies (GWAS) have explored isolated traits, the multifactorial genetic architecture underlying sarcopenia remains poorly defined. In this study, we constructed a comprehensive genetic factor to explain the genetic architecture of sarcopenia, and explore causal associations, genetic comorbidities, and mediating pathways linking sarcopenia to 30 urological diseases. METHODS Based on the European Working Group on Sarcopenia in Older People (EWGSOP) criteria, six sarcopenia-related phenotypes were selected. A multivariate GWAS framework using genomic structural equation modeling (genomic SEM) was constructed, with an effective sample size of 651,820 individuals. Bidirectional Mendelian randomization (MR) was employed to assess causal relationships between sarcopenia and 30 urological diseases. Genetic correlation, tissue-specific heritability enrichment, shared risk genes, and gene set enrichment analyses were conducted to dissect genetic comorbidities between sarcopenia and urological diseases. Multi-omic mediation analysis was conducted to identify potential pathways mediated by blood proteins, metabolites, and immune traits. RESULTS Multivariate GWAS identified 215 loci and 30,869 single-nucleotide polymorphisms (SNPs) associated with polygenic architecture of sarcopenia. Bidirectional Mendelian randomization revealed causal links between sarcopenia and urological diseases, notably hyperplasia of prostate (BPH; OR = 1.17, P = 0.043) and acute tubulointerstitial nephritis (ATIN; OR = 1.14, P = 0.028). Genetic comorbidity analyses identified local genetic correlations between sarcopenia and BPH, and highlighted tissue-specific heritability enrichment in Cells Cultured fibroblasts tissue for both traits, while no genetic correlation was found with ATIN. We identified 75 shared risk genes for sarcopenia and BPH, which were enriched in cellular component biogenesis, RNA binding, and metabolic pathways. Multi-omic mediation analyses prioritized 17 metabolites and proteins linking sarcopenia to BPH and ATIN, though no significant immune mediators were identified. CONCLUSION These findings unveil a shared genetic architecture between sarcopenia and urological diseases, especially BPH, with heritability enrichment in fibroblast tissue and metabolic dysfunction emerging as the significant overlapping pathway.
Collapse
Affiliation(s)
- Feixiang Yang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei 230001, China
| | - Xiangyu Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei 230001, China; First School of Clinical Medicine, Anhui Medical University, Hefei 230032, China
| | - Wei Dai
- Department of Urology, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu 241001, China
| | - Ke Xu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei 230001, China; Second School of Clinical Medicine, Anhui Medical University, Hefei 230032, China
| | - Yunyun Mei
- Fudan University Shanghai Cancer Center (Xiamen), Xiamen 361000, China
| | - Tianrui Liu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei 230001, China; Second School of Clinical Medicine, Anhui Medical University, Hefei 230032, China
| | - Kun Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei 230001, China; Second School of Clinical Medicine, Anhui Medical University, Hefei 230032, China
| | - Qianjun Liang
- Department of Urology, Lu'an Hospital of Anhui Medical University, Lu'an People's Hospital, Lu'an 237005, China
| | - Peng Guo
- Department of Urology, The Affiliated Jiangyin People's Hospital of Nantong University, Jiangyin 214400, China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei 230001, China.
| | - Jialin Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei 230001, China; School of Life Sciences, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
7
|
Wu S, Chen Q, Yang X, Zhang L, Huang X, Huang J, Wu J, Sun C, Zhang W, Wang J. The KSR1/MEK/ERK signaling pathway promotes the progression of intrauterine adhesions. Cell Signal 2025; 131:111730. [PMID: 40089092 DOI: 10.1016/j.cellsig.2025.111730] [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: 11/24/2024] [Revised: 02/24/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Kinase suppressor of Ras 1 (KSR1) serves as a scaffold protein within the RAS-RAF pathway and plays a role in tumorigenesis, immune regulation, cell proliferation, and apoptosis. However, the specific role of KSR1 in the formation and progression of fibrotic diseases, such as intrauterine adhesions (IUA), remains unclear. This study aims to investigate KSR1 expression in IUA and the mechanisms underlying its role in promoting IUA progression. KSR1 was found to be significantly overexpressed in the endometrium of both IUA model rats and patients with IUA. KSR1 is positively involved in the regulation of proliferation, migration, and fibrosis (FN1, Collagen I, α-SMA) in immortalized human endometrial stromal cells (THESCs). Furthermore, KSR1 knockdown was observed to inhibit the fibrosis, proliferation, and migration of transforming growth factor-β1 (TGF-β1)-induced THESCs. Further studies demonstrated that the key proteins of the MEK/ERK signaling pathway, p-MEK1 and p-ERK1/2, were significantly overexpressed in the uterus of IUA rats. In vitro rescue experiments confirmed that the MEK/ERK pathway inhibitor U0126 (An ERK inhibitor) effectively suppressed the enhanced fibrosis, proliferation, and migration induced by KSR1 overexpression. In conclusion, this study demonstrates that KSR1 promotes IUA by enhancing proliferation, migration, and fibrosis of endometrial stromal cells via the MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Shasha Wu
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Qiuhong Chen
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Xiao Yang
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Lulu Zhang
- Medical Sciences Research Center, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Xiyue Huang
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Jinglin Huang
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Jiangling Wu
- Medical Sciences Research Center, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Congcong Sun
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China.
| | - Wenwen Zhang
- Department of Pathology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China.
| | - Jia Wang
- Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China.
| |
Collapse
|
8
|
Gao Z. New insights into Smad3 in cardiac fibrosis. Gene 2025; 952:149418. [PMID: 40089084 DOI: 10.1016/j.gene.2025.149418] [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: 01/26/2025] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/17/2025]
Abstract
Damage to myocardial tissues, leading to myocardial fibrosis, is a significant pathological hallmark across various heart diseases. SMAD3, a central transcriptional regulator within the transforming growth factor-beta (TGF-β) signaling pathway, plays a pivotal role in the pathological progression of myocardial fibrosis and cardiac remodeling. It intricately regulates physiological and pathological processes encompassing cell proliferation, differentiation, tissue repair, and fibrosis. Notably, SMAD3 exerts crucial influences in myocardial fibrosis subsequent to myocardial infarction, pressure overload-induced myocardial fibrosis, diabetic cardiomyopathy (DCM), aging-associated cardiac fibrosis and myocarditis-related myocardial fibrosis. The targeted modulation of genes or the utilization of compounds, including traditional Chinese medicine (paeoniflorin, baicalin, and genistein et al.) and other pharmaceutical agents that modulate SMAD3, may offer avenues for restraining the pathological cascade of myocardial fibrosis. Consequently, targeted regulation of SMAD3 associated with myocardial fibrosis may herald novel therapeutic paradigms for ameliorating myocardial diseases.
Collapse
Affiliation(s)
- Zhen Gao
- Liaocheng Vocational and Technical College, Shandong, China.
| |
Collapse
|
9
|
Dias V, Alves Â, Medeiros R, Teixeira AL, Dias F. Unveiling the TGF-β/Smad signaling in renal carcinoma progression: Prognostic and therapeutic insights. Biochim Biophys Acta Rev Cancer 2025; 1880:189372. [PMID: 40505840 DOI: 10.1016/j.bbcan.2025.189372] [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: 03/19/2025] [Revised: 06/03/2025] [Accepted: 06/09/2025] [Indexed: 06/16/2025]
Abstract
About 2 to 3 % of all new cancer cases worldwide are renal carcinomas. Nevertheless, during the past few decades, there has been a 2 % annual increase in the prevalence of this malignancy worldwide. Within three years, 20 to 40 % of individuals with early-stage malignancies may either have a local recurrence or metastasis, and 20 to 30 % will receive a diagnosis of metastatic illness. For renal cancer, traditional chemotherapy and radiation are ineffective, while metastatic-focused treatments are linked to several side effects and the quick emergence of resistance. As a silent disease with no specific and identifiable signs and symptoms and an aggressive nature, there is an urgent need to discover trustworthy prognostic biomarkers as well as novel therapeutic targets. With multiple studies suggesting that renal carcinoma is a disease with complex genomic and epigenomic backgrounds influencing tumor development and progression, it is essential to study the dysregulated cellular pathways behind this disease to address this need. In this context, the canonical TGF-β signaling pathway could reveal interesting clues. The current study attempts to outline the role of this signaling mechanism in renal cancer dynamics, with a focus on its potential prognostic, predictive, and therapeutic relevance.
Collapse
Affiliation(s)
- Vânia Dias
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal
| | - Ângela Alves
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; Research Department, Portuguese League Against Cancer Northern Branch (LPCC-NRN), 4200-172 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal; Research Department, Portuguese League Against Cancer Northern Branch (LPCC-NRN), 4200-172 Porto, Portugal; Faculty of Medicine (FMUP), University of Porto, 4200-319 Porto, Portugal; Laboratory Medicine, Clinical Pathology Department, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal; Biomedicine Research Center (CEBIMED), Research Innovation and Development Institute (FP-I3ID), Faculty of Health Sciences, Fernando Pessoa University (UFP), 4249-004 Porto, Portugal
| | - Ana Luísa Teixeira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal
| | - Francisca Dias
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal.
| |
Collapse
|
10
|
Nakagawa T, Honda T, Inagaki S, Yuasa T, Tourtas T, Schlötzer-Schrehardt U, Kruse F, Aouimeur I, Vaitinadapoule H, Travers G, He Z, Gain P, Koizumi N, Thuret G, Okumura N. Involvement of TGF-β signaling pathway-associated genes in the corneal endothelium of patients with Fuchs endothelial corneal dystrophy. Exp Eye Res 2025; 255:110334. [PMID: 40081749 DOI: 10.1016/j.exer.2025.110334] [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: 11/12/2024] [Revised: 03/01/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
This study investigated the involvement of TGF-β signaling pathway-associated genes in the pathogenesis of Fuchs endothelial corneal dystrophy (FECD). The RNA-sequencing analysis of corneal endothelial cells (CECs) from FECD patients revealed significant alterations in multiple TGF-β superfamily genes, with 9 genes upregulated (including BMP6, GDF5, and TGF-β2) and 10 genes downregulated (including BMP2, NOG, and INHBA) compared to controls. Quantitative PCR validation confirmed the elevated expression of GDF5 (3.35-fold in non-expanded and 7.66-fold in expanded TCF4), TGF-β2 (6.17-fold and 11.5-fold), and TGF-β1 (1.78-fold and 1.58-fold) in FECD patients with and without TCF4 trinucleotide repeat expansion. Ex-vivo experiments using donor corneas demonstrated that TGF-β2 stimulation significantly increased the expression of extracellular matrix (ECM) components associated with guttae formation, including fibronectin, types I and VI collagens, and other matrix proteins. Immunofluorescence confirmed increased fibronectin protein expression in the corneal endothelium following TGF-β1 or TGF-β2 treatment. This study provides the first comprehensive analysis of TGF-β superfamily involvement in FECD and suggests that GDF5, found to be upregulated in FECD, may contribute to the disease process. These findings further indicate that dysregulation of TGF-β signaling pathways drives the characteristic ECM accumulation in FECD, potentially offering new therapeutic targets for this progressive corneal disease involving fibrosis-related alterations. Future research is warranted to clarify GDF5's specific role and mechanistic impact on FECD pathogenesis.
Collapse
Affiliation(s)
- Tatsuya Nakagawa
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Tetsuro Honda
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Soichiro Inagaki
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Taichi Yuasa
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Theofilos Tourtas
- Department of Ophthalmology, University of Erlangen-Nürnberg, Erlangen, Germany
| | | | - Friedrich Kruse
- Department of Ophthalmology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ines Aouimeur
- Laboratory for Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, Health & Innovation Campus, Jean Monnet University, Saint-Etienne, France
| | - Hanielle Vaitinadapoule
- Laboratory for Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, Health & Innovation Campus, Jean Monnet University, Saint-Etienne, France
| | - Gauthier Travers
- Laboratory for Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, Health & Innovation Campus, Jean Monnet University, Saint-Etienne, France
| | - Zhiguo He
- Laboratory for Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, Health & Innovation Campus, Jean Monnet University, Saint-Etienne, France
| | - Philippe Gain
- Laboratory for Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, Health & Innovation Campus, Jean Monnet University, Saint-Etienne, France; Ophthalmology Department, University Hospital, Saint-Etienne, France
| | - Noriko Koizumi
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Gilles Thuret
- Laboratory for Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, Health & Innovation Campus, Jean Monnet University, Saint-Etienne, France; Ophthalmology Department, University Hospital, Saint-Etienne, France.
| | - Naoki Okumura
- Department of Biomedical Engineering, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan.
| |
Collapse
|
11
|
Park HY, Choi SH, Ko J, Yoon JS. Therapeutic effect of nintedanib in orbital fibroblasts in patients with Graves' orbitopathy. Immunopharmacol Immunotoxicol 2025; 47:406-418. [PMID: 40289264 DOI: 10.1080/08923973.2025.2491554] [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: 11/22/2024] [Accepted: 04/06/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Nintedanib is a potent antifibrotic angiokinase inhibitor approved for various fibrotic lung diseases. Potential therapeutic efficacy of nintedanib in various inflammatory diseases is under investigation. In this study, we investigated the therapeutic effect of nintedanib on adipogenesis and fibrosis in orbital fibroblasts in patients with Graves' orbitopathy (GO). METHODS Primary orbital fibroblasts were cultured from orbital connective tissue of patients with GO and healthy controls. The cells were pretreated with nintedanib before stimulation with either interleukin (IL)-1β, transforming growth factor (TGF)-β, insulin-like growth factor-1, or IL-11. Fibrosis-related and intracellular signaling protein expressions were assessed using western blotting. Hyaluronan and procollagen concentrations were quantified using enzyme-linked immunosorbent assay. Adipogenesis was quantified by Oil Red O staining and the levels of adipogenic transcription factors were determined by Western blot. RESULTS TGF-β-induced fibronectin and collagen 1/3 protein expression was abrogated by nintedanib treatment. Nintedanib decreased the phosphorylation of signal transducer and activator of transcription 3, SMAD 2/3, Akt, c-Jun N-terminal kinase, and extracellular regulated protein kinase. Exposure to nintedanib hindered adipocyte differentiation and expression of adipogenic transcription factors, including peroxisome proliferator-activated receptor γ, CCAAT/enhancer-binding protein α/β, adipocyte protein 2, adiponectin, and leptin. Additionally, nintedanib reduced hyaluronan and procollagen secretion. CONCLUSIONS Nintedanib suppressed profibrotic protein production, adipogenesis, and hyaluronan production in in vitro. These findings indicate the potential therapeutic efficacy of nintedanib in GO management.
Collapse
Affiliation(s)
- Hyun Young Park
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soo Hyun Choi
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - JaeSang Ko
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Sook Yoon
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
12
|
Renu K. Exosomes derived from human adipose mesenchymal stem cells act as a therapeutic target for oral submucous fibrosis. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2025; 126:102224. [PMID: 39765310 DOI: 10.1016/j.jormas.2025.102224] [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: 12/03/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
Oral submucosal fibrosis is a highly malignant oral condition that necessitates the use of sophisticated therapeutic procedures. OSF is a multifactorial precancerous condition induced by areca nut chewing, deficiencies in vitamins and trace minerals, immunological aspects, and hereditary factors. Adipose tissue-derived mesenchymal stem cells possess the capability for multidirectional activation and are extensively distributed throughout the body. They have minimal immunogenicity and are extensively utilized in cancer treatment. Exosomes are extracellular vesicles produced by the intracellular route. They are biological carriers comprising microRNA, messenger RNA, lipids and proteins crucial for intercellular communication. ADSC exosomes, serving as a vehicle for miRNA, possess accessibility and little immunogenicity. They can significantly contribute to adipose tissue regrowth, angiogenesis, immunological modulation, and tissue repair. ADSC-Exo exhibits antifibrotic properties and may serve as a potential treatment for OSF. This review presents a novel therapeutic approach and clarifies the precise mechanisms involved in the clinical management of OSF using ADSC-Exo.
Collapse
Affiliation(s)
- Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai - 600077, Tamil Nadu, India.
| |
Collapse
|
13
|
Washimkar KR, Bisen AC, Verma S, Bhatt D, Yadav M, Kumar A, Bhatta RS, Bawankule DU, Yadav PP, Mugale MN. Modulation in NF-κB-p65/NLRP3, TXNIP-mediated signaling using an ethanolic fruit extract of Withania coagulans mitigates silica-induced pulmonary fibrosis in rats. Fitoterapia 2025; 183:106578. [PMID: 40318702 DOI: 10.1016/j.fitote.2025.106578] [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: 11/13/2024] [Revised: 04/22/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Withania coagulans encompasses many active phytoconstituents, which have been used to treat many ailments. Prior research has shown that fruit extract of Withania coagulans has anti-inflammatory properties and effectively reduces oxidative stress in various diseases. Nevertheless, its effects are not obscured in the silica (SiO2) induced pulmonary fibrosis (PF). In the current study, an ethanolic fruit extract of Withania coagulans (WCE) was prepared, and its effects and underlying mechanisms on SiO2-induced PF in rats were elucidated. LC-MS/MS analysis identified various bioactive phytoconstituents, secondary plant metabolites, and flavonoids in the WCE. In vitro, results showed that the WCE exhibited no toxicity towards A549 cells, reduced the production of reactive oxygen species, and inhibited cell migration. Further, WCE abrogated alveolar wall thickening, reduced inflammatory cell infiltration, and maintained lung architecture. It also suppresses collagen accumulation and mucus production, abrogating inflammation by downregulating nuclear factor kappa B (NF-κB-p65)/ NOD-like receptor protein 3 (NLRP3) and cytokine levels. It suppresses oxidative and endoplasmic reticulum stress induced by SiO2 by downregulating thioredoxin-interacting protein (TXNIP), activating transcription factor 6 (ATF6), and C/EBP Homologous Protein (CHOP) proteins. Additionally, WCE, by suppressing EMT and transforming growth factor beta 1 (TGF-β1)/Suppressor of Mothers against Decapentaplegic (Smad) pathway, mitigated PF in rats. Taken together, WCE via anti-inflammatory and anti-oxidative properties inhibited SiO2-induced PF, and therefore, it can be envisaged as an effective antifibrotic agent to treat PF.
Collapse
Affiliation(s)
- Kaveri R Washimkar
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Amol Chhatrapati Bisen
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India; Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Sophisticated Analytical Instrument Facility and Research, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India
| | - Shobhit Verma
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Divya Bhatt
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India; In vivo Testing Facility, Bioprospection, and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, Uttar Pradesh 226015, India
| | - Manisha Yadav
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Akhilesh Kumar
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India
| | - Rabi Shankar Bhatta
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India
| | - Dnyaneshwar U Bawankule
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India; In vivo Testing Facility, Bioprospection, and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, Uttar Pradesh 226015, India
| | - Prem Prakash Yadav
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India
| | - Madhav Nilakanth Mugale
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| |
Collapse
|
14
|
Harding-Fox SL, Cellek S. The role of cyclic adenosine monophosphate (cAMP) in pathophysiology of fibrosis. Drug Discov Today 2025; 30:104368. [PMID: 40318753 DOI: 10.1016/j.drudis.2025.104368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/11/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Fibrosis, the excessive production and disorganised deposition of extracellular matrix proteins, can occur in any organ system, disrupting functionality and causing fatality. The number, efficacy and safety of antifibrotic drugs are incredibly limited. Therapeutics which elevate intracellular cyclic adenosine monophosphate (cAMP) offer a potential solution. In this review, we present the signalling mechanisms involved in fibrosis pathophysiology, how cAMP and its effectors might interact with these pathways, and the current preclinical and clinical efforts in this field. cAMP elevating agents have the potential to be future antifibrotic drug candidates, but further studies are required, particularly to develop tissue specific therapeutics.
Collapse
Affiliation(s)
- Sophie L Harding-Fox
- Fibrosis Research Group, Medical Technology Research Centre, School of Allied Health and Social Care, Faculty of Health, Medicine and Social Care, Anglia Ruskin University, Chelmsford, Essex CM1 1SQ, UK.
| | - Selim Cellek
- Fibrosis Research Group, Medical Technology Research Centre, School of Allied Health and Social Care, Faculty of Health, Medicine and Social Care, Anglia Ruskin University, Chelmsford, Essex CM1 1SQ, UK
| |
Collapse
|
15
|
Wang N, Wang Y, Zhang L, Yang W, Fu S. Molecular Mechanisms of Epithelial-Mesenchymal Transition in Retinal Pigment Epithelial Cells: Implications for Age-Related Macular Degeneration (AMD) Progression. Biomolecules 2025; 15:771. [PMID: 40563412 DOI: 10.3390/biom15060771] [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: 03/22/2025] [Revised: 05/03/2025] [Accepted: 05/16/2025] [Indexed: 06/28/2025] Open
Abstract
Age-related macular degeneration (AMD), the leading cause of irreversible blindness worldwide, represents a complex neurodegenerative disorder whose pathogenesis remains elusive. At the core of AMD pathophysiology lies the retinal pigment epithelium (RPE), whose epithelial-mesenchymal transition (EMT) has emerged as a critical pathological mechanism driving disease progression. This transformative process, characterized by RPE cell dedifferentiation and subsequent extracellular matrix remodeling, is orchestrated through a sophisticated network of molecular interactions and cellular signaling cascades. Our review provides a comprehensive analysis of the molecular landscape underlying RPE EMT in AMD, with particular emphasis on seven interconnected pathological axes: (i) oxidative stress and mitochondrial dysfunction, (ii) hypoxia-inducible factor signaling, (iii) autophagic flux dysregulation, (iv) chronic inflammatory responses, (v) complement system overactivation, (vi) epigenetic regulation through microRNA networks, and (vii) key developmental signaling pathway reactivation. Furthermore, we evaluate emerging therapeutic strategies targeting EMT modulation, providing a comprehensive perspective on potential interventions to halt AMD progression. By integrating current mechanistic insights with therapeutic prospects, this review aims to bridge the gap between fundamental research and clinical translation in AMD management.
Collapse
Affiliation(s)
- Na Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Yaqi Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Wenjing Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Songbo Fu
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Gansu Province Clinical Research Center for Endocrine Disease, Lanzhou 730000, China
| |
Collapse
|
16
|
Wu W, Tang W, Liang W, Li Q, Qi X, Gao R, Zhou P, Chen G, Xiong J, Gu X, Ding P, Yu M, Yang C, Wen S. GDF15 suppresses abdominal aortic aneurysm by upregulating AREG expression to adjust macrophage polarization. Int Immunopharmacol 2025; 159:114899. [PMID: 40414071 DOI: 10.1016/j.intimp.2025.114899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/19/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
OBJECTIVE Inflammation plays a key role in abdominal aortic aneurysm (AAA), with macrophages being crucial. Growth differentiation factor 15 (GDF15) is a new anti-inflammatory cytokine potentially useful in AAA diagnosis and treatment, but its role is unclear. METHODS AND RESULTS In mice with AAA, GDF15 expression was higher in lesioned tissues. Daily intraperitoneal injection of recombinant GDF15 (rGDF15) reduced aortic dilation, inflammation, degradation of aortic wall elastin and collagen, cellular apoptosis, and increased smooth muscle cells. GDF15 knockdown worsened AAA severity. Immunohistochemistry and immunofluorescence showed rGDF15 treatment reduced M1 macrophage polarization and enhanced M2 polarization, decreasing the M1/M2 ratio. GDF15 knockdown had the opposite effect. Additionally, Amphiregulin (AREG) expression increased with rGDF15 treatment and decreased with GDF15 knockdown. Immunofluorescence colocalization revealed lower AREG expression in M1 macrophages and higher AREG expression in M2 macrophages, suggesting that AREG may be involved in the regulation of macrophage polarization by GDF15 in AAA. Mechanistically, GDF15 upregulates AREG expression by activating the TGF-βR/SMAD2/3 signaling pathway, thereby inhibiting M1 polarization and promoting M2 polarization of macrophages. CONCLUSION This study demonstrates that exogenous injection of rGDF15 upregulates AREG expression and regulates macrophage polarization, thereby inhibiting AAA. GDF15 may not only serve as a diagnostic and prognostic marker for AAA but also as a potential molecular target for therapeutic intervention in AAA.
Collapse
Affiliation(s)
- Wanying Wu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenjing Tang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Liang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qin Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoyu Qi
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ran Gao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peng Zhou
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gezheng Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinrong Xiong
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoying Gu
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peiwu Ding
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Miao Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chao Yang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Shuang Wen
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
17
|
Yu HR, Tiao MM, Huang SC, Sheu JJC, Tain YL, Sheen JM, Lin IC, Tsai CC, Huang LT, Hsu CN, Tsai CM, Lin YH, Lee PF, Su YT. Impact of maternal microplastic exposure on offspring lung structure and function: Insights into transcriptional misregulation and the TGF-β/α-SMA pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 299:118397. [PMID: 40412254 DOI: 10.1016/j.ecoenv.2025.118397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 05/16/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
The "Developmental Origins of Health and Disease" (DOHaD) theory suggests that prenatal exposure to harmful environmental factors may impair fetal tissue development, increasing the risk of diseases later in life. This study investigated the effects of prenatal exposure to polystyrene microplastics (PS-MPs) on offspring lung development. Pregnant Sprague-Dawley rats were randomly assigned to receive PS-MPs in drinking water until delivery, with a control group receiving standard water. Offspring were assessed at 7 and 120 d after birth without further PS-MPs exposure. Histopathological examination at 7 d revealed PS-MPs deposits, alveolar collapse, and inflammation in lung tissue. Gene expression analysis showed disruptions in tight junctions, transcriptional regulation, and transforming growth factor-beta (TGF-β) pathways. By day 120, lung dysfunction and structural changes, consistent with emphysema were observed. These findings demonstrate that prenatal PS-MPs exposure adversely affects lung development potentially increasing the risk of respiratory diseases. Public health measures should address the potential hazards of microplastics to fetal health.
Collapse
Affiliation(s)
- Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan; Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Mao-Meng Tiao
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan; Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan.
| | - Shun-Chen Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Jim Jinn-Chyuan Sheu
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80761, Taiwan; School of Chinese Medicine, China Medical University, Taichung 40454, Taiwan.
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan; Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan; Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - I-Chun Lin
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan; Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Ching-Chou Tsai
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung 83301, Taiwan
| | - Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan; Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Chih-Min Tsai
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan
| | - Yu-Hsiu Lin
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Pei-Fen Lee
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Yu-Tsun Su
- Department of Pediatrics, E-Da Hospital, I-Shou University and School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan
| |
Collapse
|
18
|
Zhi Y, Shu M, Tang P, Li Y, Guo M, Deng J, Mo H, Wu M, Liu B, Mai Y, Ling J, Zhao X, Zhang X, Zuo W. Overexpression of Decorin Optimizes the Treatment Efficacy of Umbilical Cord Mesenchymal Stem Cells in Bleomycin-Induced Pulmonary Fibrosis in Rats. Stem Cells Int 2025; 2025:6324980. [PMID: 40438789 PMCID: PMC12119169 DOI: 10.1155/sci/6324980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 09/02/2024] [Accepted: 04/25/2025] [Indexed: 06/01/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a long-term, diffuse pulmonary parenchyma lesion that primarily affects middle-aged and older adults. It is characterized by pulmonary interstitial fibrosis of unknown cause. The death rate upon diagnosis is higher than that of many other cancer types. Mesenchymal stem cell (MSC) treatment of organ fibrosis is a hot topic in preclinical and clinical research because it effectively treats IPF. In recent years, decorin (DCN) has been regarded as a critical mediator for its anti-inflammatory and antifibrotic effects. The purpose of this study was to generate human umbilical cord MSCs (HUC-MSCs) that overexpress DCN and to investigate the safety, mechanism, and effectiveness of using these cells to cure pulmonary fibrosis caused by bleomycin (BLM). First, lentiviral (LV) particles carrying the therapeutic DCN gene (LV-DCN) and control LV particles were created and transfected using the plasmid vector GV208 to create a viral solution for infecting HUC-MSCs. These solutions were used to create a DCN overexpression cell line and an MSC-Con. cell line infected with the control lentivirus. Intratracheal injection of BLM was used to establish a rat model of pulmonary fibrosis. On the second day following modeling, different treatments were administered, and the body weight and survival status of the rats were noted. The relevant tests were performed on days 15 and 29 following modeling. The results demonstrated that the overexpression of DCN did not affect the properties of HUC-MSCs and that these cells were effective in treating IPF. MSC-Con. and MSC-DCN reduced systemic inflammation by reducing serum interleukin (IL) 1β. Both cell types successfully treated pulmonary fibrosis in rats, as demonstrated by hematoxylin and eosin (HE) and Masson staining. MSC-DCN showed better efficacy due to lower mortality, higher weight gain, less alveolar inflammation, and less fibrosis. The safety of venous transplantation with MSCs was established by HE staining of the heart, liver, spleen, and kidney, as well as serum lactate dehydrogenase (LDH), creatinine (CRE), alanine aminotransferase (ALT), and aspartate aminotransferase (AST) levels. Immunohistochemical (IHC) staining of CD68 and CD206 in lung tissue and in vitro experiments on THP-1-induced M2 macrophage polarization and transforming growth factor-beta 1 (TGF-β1)-induced MRC-5 fibrosis indicated that MSC-DCN may mitigate lung inflammation by altering macrophage recruitment and polarization and inhibiting TGF-β1 expression to reduce fibrous hyperplasia and collagen deposition, thereby improving the treatment of BLM-induced IPF.
Collapse
Affiliation(s)
- Yaofeng Zhi
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Minghui Shu
- Department of Geriatric Medicine, The Affiliated Jiangmen Traditional Chinese Medicine Hospital of Jinan University, Jiangmen, Guangdong, China
| | - Pinsheng Tang
- Department of Cardiovascular, The Affiliated Jiangmen Traditional Chinese Medicine Hospital of Jinan University, Jiangmen, Guangdong, China
| | - Yingjie Li
- Department of Respiratory, The Affiliated Jiangmen Traditional Chinese Medicine Hospital of Jinan University, Jiangmen, Guangdong, China
| | - Min Guo
- Traditional Chinese Medicine Department, Guangdong Jiangmen Chinese Medicine College, Jiangmen, Guangdong, China
| | - Jiongrui Deng
- Department of Pulmonary and Critical Care Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Haixin Mo
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Meimei Wu
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Baoyi Liu
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Yanyang Mai
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Jie Ling
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Xulin Zhao
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Xin Zhang
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, China
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Wanli Zuo
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
- Department of Respiratory, The Affiliated Jiangmen Traditional Chinese Medicine Hospital of Jinan University, Jiangmen, Guangdong, China
- Department of Pulmonary and Critical Care Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| |
Collapse
|
19
|
Wang Y, Chen S, Zhou Z, Jiang J, Chen S. Tetrathiomolybdate alleviates bleomycin-induced pulmonary fibrosis by reducing copper concentration and suppressing EMT. Eur J Med Res 2025; 30:394. [PMID: 40390111 PMCID: PMC12087083 DOI: 10.1186/s40001-025-02640-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/27/2025] [Indexed: 05/21/2025] Open
Abstract
Pulmonary fibrosis (PF) is a disease characterized by dysregulated extracellular matrix deposition and aberrant fibroblast activation. Emerging evidence implicates that dysregulated copper metabolism contributed to fibrotic pathogenesis, yet its role and the therapeutic potential of copper modulation remain underexplored. This study investigated the involvement of cuproptosis, a programmed cell death induced by intracellular copper overload, in PF and evaluated the therapeutic efficacy of the copper chelator tetrathiomolybdate (TTM). In a bleomycin (BLM)-induced murine PF model, intratracheal BLM administration elevated lung copper levels, upregulated oligomerized DLAT, and exacerbated fibrosis, as evidenced by collagen deposition, α-smooth muscle actin, and transforming growth factor-beta expression. TTM treatment significantly attenuated fibrotic progression, reduced oxidative stress, and suppressed Olig-DLAT accumulation. In vitro, copper ionophores induced cuproptosis in bronchial epithelial cells, characterized by reduced viability, elevated intracellular Cu⁺, and Olig-DLAT aggregation, which were reversed by TTM. Furthermore, TTM mitigated TGF-β-driven epithelial-mesenchymal transition (EMT) and fibroblast-to-myofibroblast transition (FMT), downregulating collagen-1 and restoring E-cadherin expression. These findings establish cuproptosis as a novel mechanistic contributor to PF and highlight TTM's dual role in restoring copper homeostasis and inhibiting fibrogenic pathways, offering a promising therapeutic strategy for fibrotic lung diseases.
Collapse
Affiliation(s)
- Yajun Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Shuyang Chen
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Zhou
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinjun Jiang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Shujing Chen
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
20
|
Meng W, Li N, Lv F, Chen B, Lu S, Zhang J, Zhang T, Tao Q, Zhou Y, Ma L, Guan Y. SiRNA-Targeting TGF-β1 Based on Nanoparticle-Coated Ureteral Stents to Inhibit Ureteral Stricture. ACS Biomater Sci Eng 2025. [PMID: 40359485 DOI: 10.1021/acsbiomaterials.4c01925] [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: 05/15/2025]
Abstract
Ureteral stricture is a difficult urological problem with no optimal solution and is the result of scar hyperplasia and fibrosis caused by ureteral injury. Preventing the formation of ureteral strictures around drug-loaded ureteral stents is at the heart of the current research. TGF-β1 is a key factor affecting collagen deposition and fiber formation. Therefore, in this study, we established a rabbit ureteral stricture model, implanted a ureteral stent loaded with TGF-β1-siRNA for treatment, and compared the histopathology of ureteral stricture and the protein expression of genes related to the formation of stricture between different groups to test their therapeutic effects. We used sustained- and slow-release properties of the nanoparticles that were confirmed through in vitro experiments. The results of the fluorescence immunoassay showed that siRNA loaded by ureteral stents had high transfection efficiency on human ureter epithelial cells in vivo. In addition, the rabbit ureteral stricture model experiment verified that TGF-β1-siRNA could effectively transfect into ureteral tissues and inhibit the expression of TGF-β1, thereby inhibiting ureteral stricture. At the same time, the images of rabbit gross anatomy specimens showed that the hydronephrosis could also be effectively relieved. In summary, all the results mentioned above suggest that ureteral stents combined with RNA interference technology and a nanoparticle delivery system have broad prospects for clinical application in the suppression of ureteral stricture.
Collapse
Affiliation(s)
- Wei Meng
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Ningning Li
- Xinglin College, Nantong University, Nantong 226001, China
| | - Feng Lv
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Bo Chen
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Shuaijiang Lu
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Jiayi Zhang
- Xinglin College, Nantong University, Nantong 226001, China
| | - Tong Zhang
- Xinglin College, Nantong University, Nantong 226001, China
| | - Qianyu Tao
- Xinglin College, Nantong University, Nantong 226001, China
| | - Youlang Zhou
- Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Limin Ma
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Yangbo Guan
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| |
Collapse
|
21
|
Gholizadeh N, Yousefian M, Mohammadpour H, Razavi AE, Talaei S, Sheykhbahaei N. Long non-coding RNAs PVT1, CCAT2, and TCF7L2, and miR-33 and c-Myc expression in oral squamous cell carcinoma and oral lichen planus patients. J Craniomaxillofac Surg 2025:S1010-5182(25)00136-2. [PMID: 40360347 DOI: 10.1016/j.jcms.2025.04.006] [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: 06/23/2024] [Revised: 03/09/2025] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
OBJECTIVE The objective of this study was to assess the potential of TCF7L2, CCAT2, and PVT1 LncRNAs, c-Myc, and miR-33 as biomarkers for early diagnosis and differentiation of oral squamous cell carcinoma (OSCC) and premalignant lesions. DESIGN Bioinformatics tools, including COSMIC, GeneMANIA, PathVisio, KEGG Pathway Database, IntOGen, and WikiPathways, were used to investigate the signaling pathways of cancer-associated genes. The limma package was utilized for statistical analysis to identify Differentially Expressed Genes (DEGs) between OSCC tumor and normal samples. The regulatory microRNAs were analyzed using miRDB, miRWalk, and TargetScan. The type of cancer for analysis was selected using IntOGen. The expression levels of LncRNAs, miR-33, and c-Myc were measured by polymerase chain reaction (PCR) in 28 OLP and 30 OSCC tissue samples, compared to 30 healthy and 30 OSCC-adjacent tissue specimens as control groups. Data were analyzed using the Mann-Whitney test, receiver operating characteristic (ROC) curve, and multiple linear regression. RESULTS The expression of c-Myc (3.53 ± 2.78 vs 0.93 ± 0.50), PVT1 (10.94 ± 8.49 vs 0.91 ± 0.48), CCAT2 (11.77 ± 10.00 vs 0.92 ± 0.95), and TCF7L2 (6.48 ± 4.30 vs 1.27 ± 0.96) was significantly higher in OSCC samples compared to OLP (P < 0.001). Conversely, miR-33 expression was significantly lower in OSCC samples (0.24 ± 0.25 vs 4.90 ± 3.90). There was a significant correlation between c-Myc, CCAT2, PVT1, and miR-33 expression and clinicopathological characteristics of OSCC specimens. In OSCC samples, c-Myc, PVT1, CCAT2, and TCF7L2 showed a significant positive correlation with each other, while miR-33 expression was negatively correlated with the overexpression of other genes. The area under the curve (AUC) for c-Myc, PVT-1, CCAT2, miR-33, and TCF7L2 were 0.917, 1.000, 0.979, 0.006, and 0.929, respectively. CONCLUSIONS Our findings suggest that c-Myc and LncRNAs (TCF7L2, PVT1, and CCAT2) are upregulated and miR-33 is downregulated in OSCC compared to OLP samples. These genes may serve as potential genetic biomarkers for diagnosis and prediction of clinicopathological features of OSCC.
Collapse
Affiliation(s)
- Narges Gholizadeh
- Department of Oral & Maxillofacial Medicine, School of Dentistry, Tehran University of Medical Science, Tehran, Iran; Department of Oral & Maxillofacial Surgery, School of Dentistry, Tehran University of Medical Science, Tehran, Iran
| | - Marzieh Yousefian
- Department of Oral & Maxillofacial Medicine, School of Dentistry, Tehran University of Medical Science, Tehran, Iran
| | - Hadiseh Mohammadpour
- Iran National Tumor Bank, Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirnader Emami Razavi
- Iran National Tumor Bank, Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Sanam Talaei
- Department of Oral & Maxillofacial Medicine, School of Dentistry, Tehran University of Medical Science, Tehran, Iran
| | - Nafiseh Sheykhbahaei
- Department of Oral & Maxillofacial Medicine, School of Dentistry, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
22
|
Wang C, Jiang X, Han S, Zang H, Gao X. Loganin ameliorates left ventricular fibrosis and dysfunction induced by pressure overload via the Sirt1/AKT/TGF-β1 signaling pathway. J Nat Med 2025:10.1007/s11418-025-01911-9. [PMID: 40347371 DOI: 10.1007/s11418-025-01911-9] [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: 01/26/2025] [Accepted: 04/18/2025] [Indexed: 05/12/2025]
Abstract
Loganin (LG), a natural compound derived from Cornus officinalis Sieb. et Zucc., possesses diverse pharmacological properties, such as anti-inflammatory, anti-hypertrophic, and antioxidant effects. However, the role of LG in the pathogenesis of Heart Failure (HF) remains unclear. The current work aimed to explore the underlying mechanism of LG in pressure overload-induced HF, both in vivo and in vitro, using transverse aortic constriction (TAC) surgery or isoproterenol (ISO) administration. Following eight weeks of TAC surgery, histological assessments, including hematoxylin and eosin staining, wheat germ agglutinin staining, TUNEL assay, and Masson's trichrome staining, were conducted to evaluate the extent of cardiomyocyte remodeling. Additionally, RT-PCR and WB analyses were performed to detect the levels of various targets. Furthermore, H9C2 cardiomyocytes were treated with ISO to induce hypertrophy, and the effects of LG on cell viability, α-smooth muscle actin (α-SMA) expression, and molecular targets were investigated. Our findings revealed that LG treatment at 40 mg/kg/day significantly attenuated cardiac dysfunction, decreased left ventricular collagen deposition in both interstitial and perivascular spaces. Mechanistically, LG mitigated ISO-induced toxicity in H9C2 cardiomyocytes, decreasing cellular hypertrophy and α-SMA expression. Moreover, we observed a downregulation of Sirtuin 1 (Sirt1) at the molecular level, accompanied by reduced phosphorylation of Akt and transforming growth factor-β1 (TGF-β1). Notably, the administration of the Sirt1 inhibitor, EX527, effectively abolished the protective effects of LG. Therefore, the cardio-protective effects of LG were mediated through the activation of the Sirt1/Akt/TGF-β1 signaling pathway, leading to reduced fibrosis and improved cardiac function.
Collapse
Affiliation(s)
- Changbin Wang
- Department of Cardiology, The First Rongjun Youfu Hospital of Shandong Province, No. 23 Jiefang Road, Jinan, 25000, China
| | - Xiaoli Jiang
- Department of Cardiology, The First Rongjun Youfu Hospital of Shandong Province, No. 23 Jiefang Road, Jinan, 25000, China
| | - Shuhua Han
- Station for Fengming Community Health Service, People's Hospital of Zhoucun District, No. 999 Hengxing Road, Zibo, 255300, China
| | - Huimei Zang
- Center for Gene and Immunotherapy, The Second Hospital, Cheeloo College of Medicine, Shandong University, No.44 Wenhua West Road, Jinan, 250033, China
- Multidisciplinary Innovation Center for Nephrology of the Second Hospital, Cheeloo College of Medicine, Shandong University, No.44 Wenhua West Road, Jinan, 250033, China
| | - Xiaoyuan Gao
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong First Medical University, No.105 Jiefang Road, Jinan, 250033, China.
| |
Collapse
|
23
|
Hou R, Shui W, Xing X, Chen Y, Shen W, Song Q, Wang J. Relationship Between TGF-β1 and Left Ventricular Geometry and Function in Patients With Essential Hypertension. JOURNAL OF CLINICAL ULTRASOUND : JCU 2025. [PMID: 40346927 DOI: 10.1002/jcu.24055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/22/2025] [Accepted: 03/31/2025] [Indexed: 05/12/2025]
Abstract
OBJECTIVE To explore the association of transforming growth factor β1 (TGF-β1) with left ventricular geometry (LVG) and left ventricular function (LVF) in cases having essential hypertension. METHODS This retrospective study of 213 cases of essential hypertension, according to echocardiogram measurements, were split into normal geometry (NG), concentric remodeling (CR), eccentric hypertrophy (EH), and concentric hypertrophy (CH) groups. General clinical data of each patient was analyzed and office blood pressure measurements were performed. Detection of blood biochemistry and serum TGF-β1 content was conducted. The association of TGF-β1 with LVG and LVF parameters was assessed. RESULTS In contrast to the NG and CR groups, the TGF-β1 concentration was higher in the EH and CH groups, and it was most pronounced in the CH group. The TGF-β1 was positively linked to E/e' (r = 0.506, p < 0.001), whereas left ventricular global longitudinal strain (GLS) (r = -0.447, p < 0.001) was negatively correlated. Moreover, serum TGF-β1 levels were independently linked to EH and CH. CONCLUSION TGF-β1 was associated with abnormal LVG and LVF in cases of essential hypertension, indicating that it may induce LVG and LVF abnormalities.
Collapse
Affiliation(s)
- Ran Hou
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wen Shui
- Department of Cardiopulmonary Function Examination, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Xueqing Xing
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaodong Chen
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenqian Shen
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qingfei Song
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jian Wang
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
24
|
Jiao Q, Xu X, Xu L, Wang Y, Pang S, Hao J, Liu X, Zhao Y, Qi W, Qin L, Huang T, Li J, Wang T. Knockdown of eIF3a alleviates pulmonary arterial hypertension by inhibiting endothelial-to-mesenchymal transition via TGFβ1/SMAD pathway. J Transl Med 2025; 23:524. [PMID: 40346622 PMCID: PMC12065328 DOI: 10.1186/s12967-025-06505-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/13/2025] [Indexed: 05/11/2025] Open
Abstract
OBJECTIVE Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by vascular remodeling and involves Endothelial-to-Mesenchymal transition (EndMT) in pulmonary artery endothelial cells (PAECs). EndMT is a complex cell differentiation process, mainly showing the detachment of endothelial cell migration and reducing endothelial cell characteristics to varying degrees, acquiring mesenchymal cell characteristics. In addition, numerous studies have reported that eIF3a over expression plays an important role in the occurrence and development of fibrotic diseases, cancer, and degenerative lesions, however, the mechanisms of eIF3a affecting the dysfunction of pulmonary arterial endothelial cells remains largely unknown. Therefore, we aimed to demonstrate the underlying mechanisms of eIF3a-knockdown inhibiting EndMT by regulating TGFβ1/SMAD signal pathway in PAH. METHODS In this study, we screened the potential target genes associated with idiopathic pulmonary arterial hypertension (IPAH) by WGCNA to provide a reference for the diagnosis and treatment of PAH. By constructing WGCNA, which indicated that the blue module (module-trait associations between modules and clinical feature information were calculated to selected the optimum module) is most closely associated with IPAH, we further screened out 10 up-regulated candidate biomarker genes. Male SD rats were randomly assigned to four groups: Control, Monocrotaline (MCT), AAV1-shRNA-NC group and AAV1-shRNA-eIF3a group. The eIF3a-knockdown rat model was constructed by adeno-associated virus type-1 (AAV1) infection, PAH was evaluated according to hemodynamic alteration, right heart hypertrophy and histopathological changes in the lung tissue. Hematoxylin eosin (H&E) staining was used to assess the morphological changes of pulmonary arteries in rats of each treatment group. Co-localization of eIF3a with alpha-small muscle action (α-SMA) and co-localization of eIF3a with endothelial marker (CD31) were detected by double-label immunofluorescence. Immunohistochemistry (IHC) and Western blot (WB) experiments were performed to assess the expression of eIF3a, EndMT and TGFβ1/SMAD signal related proteins. In vitro, primary rat pulmonary artery endothelial cells (PAECs) were transfected with si-eIF3a to investigate the effects of eIF3a-knockdown on hypoxia-induced EndMT in PAECs and further elucidate its underlying molecular mechanisms. RESULTS By WGCNA analysis, we screened the up-regulated hub genes of TMF1, GOLGB1, ARMC8, PRPF40 A, EIF3 A, ROCK2, EIF5B, CCP110, and KRR1 associated with PAH, and in order to verify the potential role of eIF3a in the development of pulmonary arterial hypertension, MCT-induced PAH rat model was constructed successfully. The expression of eIF3a was increased in MCT-treated lungs. Knockdown of eIF3a significantly inhibited the pulmonary arterial hypertension and vascular remodeling in MCT-induced PAH rat model, ameliorated MCT-induced increases of right ventricular systolic pressure (RVSP) and right ventricular hypertrophy (RVH) in rats. Double-labeled immunofluorescence showed eIF3a was mostly co-localized with CD31, this result indicated that the development of MCT-induced PAH was related to the regulation of PAECs function (most likely associated with the change of EndMT in endothelial cells). WB showed that the expressions of EndMT related proteins were significantly increased by regulating TGFβ1/SMAD signaling pathway in MCT-induced PAH rat lung tissues, however, knockdown of eIF3a markedly attenuated these changes. In addition, we observed the same results in rat PAECs with chronic hypoxia exposure. These results indicate that eIF3a-knockdown inhibited EndMT by regulating TGFβ1/SMAD signaling pathway in PAECs, thereby improving the development of MCT-induced PAH. CONCLUSIONS Knockdown of eIF3a inhibited EndMT in PAECs regulating TGFβ1/SMAD signaling pathway, significantly alleviated the changes of RVSP, RVH and vascular remodeling in MCT-induced PAH rats, eIF3a may be a promising and novel therapeutic target for the treatment of PAH.
Collapse
Affiliation(s)
- Qiuhong Jiao
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Xiufeng Xu
- Department of Geriatrics, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Longwu Xu
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Yuying Wang
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Shulan Pang
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Jie Hao
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Xiaohong Liu
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Yudan Zhao
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Wanpeng Qi
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Limin Qin
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Tao Huang
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Jingtian Li
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Tao Wang
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China.
| |
Collapse
|
25
|
Wang L, Qiu T, Xu F, Zhang L, Zhang C, Ye W. Fabricate heparin-mimic thin gel layers for vascular cell selective regulation using 5-hydroxydopamine cross-linked chitosan and sulfonated polymers. Int J Biol Macromol 2025; 311:144027. [PMID: 40345300 DOI: 10.1016/j.ijbiomac.2025.144027] [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: 02/17/2025] [Revised: 05/02/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
In this work, 5-hydroxydopamine was employed as a crosslinking agent to bind chitosan and sulfonated polymers to fabricate thin gel layers (TGLs) featuring heparin-mimic structures. By controlling the distribution of exposed chemical moieties (phenolic hydroxyl, amino, and sulfonic groups), the growth of the endothelial cell (EC) and smooth muscle cell (SMC) on the TGLs surfaces could be modulated. Such modulation effectively maintained the quantity and proportion of the two cell types within a reasonable range, thereby offering a potential avenue for promoting re-endothelialization. The prepared TGLs showed improved hydrophilicity as well as hemocompatibility. For cytocompatibility test, TGLs led to a notable promotion of the growth of human umbilical vein endothelial cells (HUVECs) and exerted substantial inhibitory effects on the proliferation of human umbilical artery smooth muscle cells (HUASMCs). The ratio of HUVECs to HUASMCs rose from 0.184 to 1.97. The enhanced hemocompatibility was attributed to the incorporation of exposed functional groups. Regarding the highly selective effects, these were ascribed to the synergistic influence of high sulfonation degree and the presence of amino groups and phenolic hydroxyl groups. The current work illustrated a simple method for synthesizing a multifunctional biomimetic polymer material that offers the promise of broader biomedical research applications.
Collapse
Affiliation(s)
- Lingren Wang
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Huaiyin Institute of Technology, Huaian, Jiangsu 223003, China.
| | - Tingting Qiu
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Huaiyin Institute of Technology, Huaian, Jiangsu 223003, China
| | - Fan Xu
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Huaiyin Institute of Technology, Huaian, Jiangsu 223003, China
| | - Linna Zhang
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Huaiyin Institute of Technology, Huaian, Jiangsu 223003, China
| | - Chao Zhang
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Huaiyin Institute of Technology, Huaian, Jiangsu 223003, China
| | - Wei Ye
- Department of Chemical Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu 223003, China.
| |
Collapse
|
26
|
Cao Y, Yang D, Cai S, Yang L, Yu S, Geng Q, Mo M, Li W, Wei Y, Li Y, Yin T, Diao L. Adenomyosis-associated infertility: an update of the immunological perspective. Reprod Biomed Online 2025; 50:104703. [PMID: 40175227 DOI: 10.1016/j.rbmo.2024.104703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/29/2024] [Accepted: 10/09/2024] [Indexed: 04/04/2025]
Abstract
Adenomyosis is characterized by the invasion of endometrial glands and stroma into the myometrium. Its clinical manifestations often include dysmenorrhoea, excessive menstrual bleeding and infertility. Reduced pregnancy and live birth rates and an increased miscarriage rate are observed in women with adenomyosis. This review summarizes relevant advances and presents the underlying mechanisms of adenomyosis-associated infertility from an immunological perspective. Individuals with adenomyosis exhibit imbalances in immune cell subpopulations and the endocrine hormone-immunomodulatory axis. These immunological alterations may be key contributors to, or at least accomplices in, impaired endometrial receptivity. In addition, adenomyosis often occurs in association with endometriosis, uterine leiomyoma or endometrial polyps, which are pathogenetically relevant; their similarities and differences are discussed from an immunological perspective. The clinical diagnostic criteria of adenomyosis are not perfect, and the pathogenesis remains to be fully explored. Therefore screening for effective targets for early diagnosis and treatment at the cellular and molecular levels from the immunological point of view holds great potential, which will be of great importance in preventing this disease and improving women's reproductive health.
Collapse
Affiliation(s)
- Ying Cao
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dongyong Yang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Songchen Cai
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, Guangdong, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Lingtao Yang
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, Guangdong, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Shuyi Yu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, Guangdong, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Qiang Geng
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, Guangdong, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Meilan Mo
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, Guangdong, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Wenzhu Li
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yiqiu Wei
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yuye Li
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, Guangdong, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China.
| | - Tailang Yin
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, Guangdong, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China.
| |
Collapse
|
27
|
Ding L, Bao D, Dai B, Meng Q, Lv C, Luo H, Zhu H. The role and mechanism of CRISPLD2 in skin fibrosis of systemic sclerosis. Rheumatology (Oxford) 2025; 64:3134-3141. [PMID: 39374552 DOI: 10.1093/rheumatology/keae541] [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: 04/22/2024] [Revised: 08/23/2024] [Accepted: 09/14/2024] [Indexed: 10/09/2024] Open
Abstract
OBJECTIVES SSc is an autoimmune connective tissue disease involving multiple organs. The most common clinical symptom of SSc is progressive fibrosis of the skin, and the pathologically manifestations of skin were activation and proliferation of fibroblasts and continuous proliferation of extracellular matrix. TGF-β can promote the proliferation and activation of fibroblasts, causing excessive deposition of collagen and structural proteins. Therefore, exploring the specific mechanism of TGF-β-related pathway on fibrosis is of great significance for improving skin fibrosis in SSc. METHODS Genes related to TGF-β pathway were screened through bioinformatics analysis, and SSc phenotypes were verified in vivo and in vitro. The relevant molecular mechanisms were preliminarily discussed in combination with transcriptome sequencing. RESULTS Human cysteine-rich secreted protein LCCL domain protein 2 (CRISPLD2) was found increased reactivity in TGF-β-induced fibroblasts, and the expression of ACTA2 (ɑ-SMA) decreased significantly in TGF-β-mediated fibroblasts with up-regulation of CRISPLD2. CONCLUSION CRISPLD2 was found to have increased reactivity in TGF-β-induced fibroblasts, and we further confirmed that CRISPLD2 can participate in TGF-β-induced fibroblast fibrosis from multiple perspectives and levels in negative feedback regulation, and investigated the mechanism of CRISPLD2 in fibrosis.
Collapse
Affiliation(s)
- Liqing Ding
- Department of Rheumatology and Immunology, Xiangya Hospital of Central South University, Changsha, Hunan, P.R China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, P.R China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R China
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, P.R China
| | - Ding Bao
- Department of Rheumatology and Immunology, Xiangya Hospital of Central South University, Changsha, Hunan, P.R China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, P.R China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R China
| | - Bingying Dai
- Department of Rheumatology and Immunology, Xiangya Hospital of Central South University, Changsha, Hunan, P.R China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, P.R China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R China
| | - Qiming Meng
- Department of Rheumatology and Immunology, Xiangya Hospital of Central South University, Changsha, Hunan, P.R China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, P.R China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R China
| | - Chunliu Lv
- Department of Breast Tumor Plastic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P.R China
- Department of Head and Neck Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, P.R China
| | - Hui Luo
- Department of Rheumatology and Immunology, Xiangya Hospital of Central South University, Changsha, Hunan, P.R China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, P.R China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R China
| | - Honglin Zhu
- Department of Rheumatology and Immunology, Xiangya Hospital of Central South University, Changsha, Hunan, P.R China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, P.R China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R China
| |
Collapse
|
28
|
Dos Santos ACC, Figueiredo-Vanzan D, Bentes J, Motta JM, Mata-Santos HA, Pyrrho ADS, Castelo-Branco MTL. Tetrylpyamethrazine alleviates hepatic fibrosis induced by experimental mansonic schistosomiasis. Inflammopharmacology 2025; 33:2833-2847. [PMID: 40268854 DOI: 10.1007/s10787-025-01759-1] [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: 01/14/2025] [Accepted: 04/11/2025] [Indexed: 04/25/2025]
Abstract
Hepatic fibrosis resulting from human mansonic schistosomiasis significantly impairs liver function and contributes substantially to morbidity associated with helminth infections. This pathological state develops following the deposition of helminth eggs within hepatic tissues, triggering a granulomatous inflammatory reaction. Schistosomiasis, a neglected tropical disease affecting approximately 240 million individuals globally, represents a major public health challenge. Although praziquantel (PZQ) is recommended by the World Health Organization (WHO) as the primary treatment for helminth infections, additional therapies are required to address the associated liver fibrosis. This study investigated the efficacy of tetramethylpyrazine (TMP), a natural compound known for its anti-inflammatory, antifibrotic, and hepatoprotective properties in various experimental models, in mitigating hepatic fibrosis induced by mansonic schistosomiasis. Our in vivo experiments demonstrated that TMP treatment significantly reduced hepatic granuloma size, as evidenced by histological analysis. Furthermore, our in vitro studies showed that TMP increased levels of the anti-inflammatory cytokine IL-10 while decreasing levels of the profibrotic cytokine IL-13 in a concentration-dependent manner. Immunofluorescence analysis also revealed that TMP effectively inhibited collagen deposition. Collectively, these findings suggest that TMP exhibits potential as an anti-inflammatory and antifibrotic agent for hepatic fibrosis resulting from Schistosoma mansoni infection.
Collapse
Affiliation(s)
- Ana Carolina Campos Dos Santos
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Josiane Bentes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Maria Motta
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Morgana Teixeira Lima Castelo-Branco
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
29
|
Peng L, Fan Y, Wang L, Han C, Hao Z. Luteolin 7-Glucuronide in Artemisia rupestris L. Extract Attenuates Pulmonary Fibrosis by Inhibiting Fibroblast Activation and FMT via Targeting of TGF-β1. Antioxidants (Basel) 2025; 14:533. [PMID: 40427415 PMCID: PMC12108481 DOI: 10.3390/antiox14050533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
Pulmonary fibrosis (PF) is a chronic pulmonary disease characterized by excessive extracellular matrix (ECM) deposition, with cigarette smoking being a major risk factor and no effective treatment at present. Transforming growth factor beta 1 (TGF-β1) plays a key role in PF and regulating oxidative stress. This study investigated the effects and mechanisms of Artemisia rupestris L. ethanol extract (ER) on cigarette smoke (CS)-induced PF. We used pull-down and LC-MS analyses to screen and identify compounds that bind to TGF-β1 in ER. We demonstrated that ER inhibits CS-induced PF, lung inflammation, and oxidative stress, thereby improving pulmonary structural injury. The ER inhibits fibroblast activation and fibroblast-to-myofibroblast transition (FMT), reducing collagen deposition for the treatment of PF. We identified the active ingredient in ER that binds to TGF-β1, namely, Luteolin 7-glucuronide (LG). LG inhibits the TGF-β1 signaling pathway through targeted binding to TGF-β1, downregulates the expression of downstream proteins (including collagen I, α-SMA, MMP-2, and MMP-9), and inhibits fibronectin expression. It also inhibits fibroblast activation and FMT, enhances E-cadherin expression to promote fibroblast adhesion, and suppresses collagen deposition, alleviating PF. Based on these findings, we propose that LG might be a promising therapeutic drug candidate for treating PF.
Collapse
Affiliation(s)
- Lingfeng Peng
- Chinese Veterinary Medicine Innovation Center, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (L.P.); (Y.F.); (L.W.); (C.H.)
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
- National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Yimeng Fan
- Chinese Veterinary Medicine Innovation Center, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (L.P.); (Y.F.); (L.W.); (C.H.)
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
- National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Luyao Wang
- Chinese Veterinary Medicine Innovation Center, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (L.P.); (Y.F.); (L.W.); (C.H.)
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
- National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Chao Han
- Chinese Veterinary Medicine Innovation Center, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (L.P.); (Y.F.); (L.W.); (C.H.)
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
- National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Zhihui Hao
- Chinese Veterinary Medicine Innovation Center, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (L.P.); (Y.F.); (L.W.); (C.H.)
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
- National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| |
Collapse
|
30
|
Pang X, Pan Y, Wang M, Qiu S, He Y, Ren Y, Yu T, Yu S, Cui Y. Comparison of reproductive performance and functional analysis of spermatogenesis factors between domestic yak and semi-wild blood yak. BMC Genomics 2025; 26:418. [PMID: 40301732 PMCID: PMC12038992 DOI: 10.1186/s12864-025-11594-x] [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: 12/08/2024] [Accepted: 04/10/2025] [Indexed: 05/01/2025] Open
Abstract
This study investigates differences in reproductive performance, testicular histology, and transcriptomic profiles between male Subei (SB; semi-wild) yaks and two domestic yaks, Gannan (GN) and Qinghai (QH). Key metrics including mating age, utilization time, breeding capacity, morphometric traits, and testicular indices were analyzed. SB yaks exhibited superior reproductive metrics, including earlier sexual maturity, prolonged utilization periods, and enhanced breeding capacity compared to GN and QH (P < 0.05). Morphologically, SB yaks demonstrated significantly greater body weight, and testicular dimensions. Compared with GN and QH yaks, the seminiferous tubules of SB yaks exhibited significantly larger spermatogenic cells and luminal cavities, along with a notably higher sperm density within the luminal cavity. Transcriptomic analysis identified 2,403 and 4,428 differentially expressed genes (DEGs) in GN vs. SB and QH vs. SB comparisons, respectively. Eight key genes (TPPP3, SMAD3, PAFAH1B3, BMP7, ARSA, CTNNB1, SMAD4, STAT3) and three pathways (Hippo, pluripotency regulation, TGF-β) were implicated in testicular development and spermatogenesis. These findings underscore the genetic and physiological advantages of SB yaks, offering insights for enhancing male yak reproductive performance.
Collapse
Affiliation(s)
- Xin Pang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Gansu Innovation Centre for Livestock Embryo Engineering and Technology, Lanzhou, China
| | - Yangyang Pan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Gansu Innovation Centre for Livestock Embryo Engineering and Technology, Lanzhou, China
| | - Meng Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Gansu Innovation Centre for Livestock Embryo Engineering and Technology, Lanzhou, China
| | - Shantong Qiu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- Gansu Innovation Centre for Livestock Embryo Engineering and Technology, Lanzhou, China
| | - Yulong He
- Jiuquan Animal Husbandry and Veterinary Medicine General Station, Jiuquan, China
| | - Yuchun Ren
- Central Agricultural Radio and Television School Tianzhu County Branch, Wuwei, China
| | - Tianjun Yu
- Subei Mongolian Autonomous County Animal Husbandry and Veterinary Technical Service Center, Jiuquan, China
| | - Sijiu Yu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China.
- Gansu Innovation Centre for Livestock Embryo Engineering and Technology, Lanzhou, China.
| | - Yan Cui
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China.
- Gansu Innovation Centre for Livestock Embryo Engineering and Technology, Lanzhou, China.
| |
Collapse
|
31
|
Feng Y, Mao T, Yi J, Zhang N, Gu Y, Shen H, Chen J. Runt-related transcription factors: from pathogenesis to therapeutic targets in multiple-organ fibrosis. Front Cell Dev Biol 2025; 13:1528645. [PMID: 40356603 PMCID: PMC12066561 DOI: 10.3389/fcell.2025.1528645] [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: 11/15/2024] [Accepted: 03/26/2025] [Indexed: 05/15/2025] Open
Abstract
Fibrosis is a partially manageable process that leads to scarring and tissue hardening by prompting myofibroblasts to deposit significant amounts of extracellular matrix (ECM) following injury. It results in detrimental consequences and pathological characteristics, which hinder the functioning of associated organs and increase mortality rates. Runt-related transcription factors (RUNX) are part of a highly conserved family of heterodimer transcription factors, comprising RUNX1, RUNX2, and RUNX3. They are involved in several biological processes and undergo various forms of post-translational modification. RUNX regulates multiple targets and pathways to impact fibrosis, indicating promise for clinical application. Therefore, its significance in the fibrosis process should not be disregarded. The review begins with an objective description of the structure, transcriptional mechanism, and biological function of RUNX1, RUNX2, and RUNX3. A subsequent analysis is made of their physiological relationship with heart, lung, kidney, and liver, followed by a focus on the signaling mechanism of RUNX in regulating fibrosis of these organs. Furthermore, potential agents or drugs targeting RUNX for treating organ fibrosis are summarized, along with an evaluation of the therapeutic prospects and potential value of RUNX in fibrosis. Further research into RUNX could contribute to the development of novel therapeutic approaches for fibrosis.
Collapse
Affiliation(s)
- Yuan Feng
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Tianshi Mao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jifei Yi
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Na Zhang
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Yinying Gu
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Huifen Shen
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Jie Chen
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Integrated Traditional Chinese and Western Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
32
|
Yang Y, Zhao L, Gao F, Wu G, Luo Y, An M. Modulation of renal fibrosis-related signaling pathways by traditional Chinese medicine: molecular mechanisms and experimental evidence. Int Urol Nephrol 2025:10.1007/s11255-025-04532-z. [PMID: 40293615 DOI: 10.1007/s11255-025-04532-z] [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: 02/10/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
Renal fibrosis (RF), characterized by excessive deposition of extracellular matrix leading to tissue damage and scar formation, represents a refractory disease and a pivotal pathological basis for the progression to end-stage renal disease. The pathogenesis of RF is intricate, prominently implicating multiple key signaling pathways, including adenosine monophosphate-activated protein kinase/mammalian target of rapamycin (AMPK/mTOR), phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), transforming growth factor-β1/small mother against decapentaplegic (TGF-β1/Smad), toll-like receptor 4/nuclear factor kappa B (TLR4/NF-κB), wingless integrated/β-catenin (Wnt/β-catenin), hypoxia-inducible factor-1α (HIF-1α), Hedgehog, and mitogen-activated protein kinase (MAPK). The current Western medical practices primarily rely on supportive and replacement therapies, which are often costly and suboptimal in efficacy. In contrast, traditional Chinese medicine (TCM), with its inherent advantages of multi-target, multi-pathway, and multi-effect modulation, emerges as a promising new strategy for RF treatment. However, a systematic, comprehensive, and detailed summary of these advancements remains absent. Therefore, this review consolidates the recent research progress on TCM modulation of RF-related signaling pathways, aiming to provide a theoretical foundation for further investigations into RF and the development of TCM interventions.
Collapse
Affiliation(s)
- Yufei Yang
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - Longshan Zhao
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
- Department of Pharmacy, Shenyang Pharmaceutical University, Benxi, 117004, China
| | - Fengli Gao
- Department of Pharmacy, Second Affiliated Hospital of Baotou Medical College, Baotou, 014030, China
| | - Guodong Wu
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - Yiduo Luo
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - Ming An
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China.
| |
Collapse
|
33
|
Liu B, Zhang Z, Mu Y, Ma L, Zhou H, Wang DA. Biophysical-Inspired Interpenetrated Fibrillar and Reticular Collagen Scaffold with Vascular Endothelial Cell Membrane Incorporation for Guided In Situ Spleen Tissue Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2501574. [PMID: 40277280 DOI: 10.1002/adma.202501574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/02/2025] [Indexed: 04/26/2025]
Abstract
The spleen's complex structure and limited regenerative ability hinder its regrowth at the site of injure, affecting patient quality of life and risk severe complications. The spleen's stroma primarily consists of reticular and fibrillar collagen, supporting its microvascular network. Inspired by such biophysical environment, this work develops an inducible scaffold featuring an interpenetrating network structure of fibrous and reticular collagen, which is loaded with vascular endothelial cell membranes to facilitate in situ regeneration. The regenerated parenchyma includes red pulp, white pulp, and a vascular system. The scaffold effectively reduces oxidative stress at the injury site, recruits cells to degrade the scaffold, and promotes tissue integration, thereby accelerating spleen regeneration. Additionally, the regenerated tissue compensates for the spleen's functions, enhancing its ability to clear abnormal red blood cells and platelets. Proteomics and RNA sequencing analyses reveal that the scaffold induced the upregulation of key pathways, including the Wnt signalling pathway, Statin pathway, and amino acid metabolism pathway. This activation mobilizes splenic cells metabolism, enhances immune cell activity, and facilitates the remodeling of the extracellular matrix. Moreover, the incorporated cell membrane components promote splenic blood vessels regeneration by upregulating the neural crest cell differentiation pathway within the tissue.
Collapse
Affiliation(s)
- Bangheng Liu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
- Center for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, New Territories, Hong Kong SAR, 999077, China
- Department of Biomedical Engineering, Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong SAR, 999077, China
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Liang Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
- Center for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, New Territories, Hong Kong SAR, 999077, China
| | - Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
- Center for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, New Territories, Hong Kong SAR, 999077, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
- Center for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, New Territories, Hong Kong SAR, 999077, China
- Department of Biomedical Engineering, Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong SAR, 999077, China
| |
Collapse
|
34
|
Qian R, Xu Y, Zhang L, Wang L, Chen X, Wang M, Bao Q, Yao Y, Xie L. Haizao Yuhu decoction ameliorates silica-induced lung injury by inhibiting transforming growth factor-beta1/Smad pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119669. [PMID: 40122314 DOI: 10.1016/j.jep.2025.119669] [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: 01/21/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Haizao Yuhu Decoction (HYD) is a traditional Chinese herbal formula known for regulating Qi and dispersing stasis. AIM OF THE STUDY This study investigates the effects of HYD on silica-induced lung injury and the underlying mechanisms. MATERIALS AND METHODS The main constituents of HYD were identified using ultra-performance liquid chromatography Q-Exactive mass spectrometry (UPLC-QE-MS). Network pharmacology was employed to predict the targets and pathways through which HYD ameliorates silicosis, which were validated in a silica-induced lung injury mouse model and a TGF-β1-induced alveolar epithelial cell model. Pathological evaluation was conducted using hematoxylin-eosin (H&E) and Masson staining, while inflammatory cytokines and fibrosis were assessed via enzyme-linked immunosorbent assay (ELISA) and hydroxyproline quantification. Western blotting (WB) was performed to analyse protein expression levels of targeted markers. Proliferation and migration capabilities of MLE12 cells treated with HYD and its bioactive constituents (glycitein, diosmetin, and limonin) were assessed using cell counting kit-8 (CCK-8) and wound healing assays. RESULTS HYD significantly alleviated silica-induced lung injury, reducing inflammatory response and collagen deposition. A total of 176 constituents were identified in HYD, with 111 being pharmacologically active and linked to 1397 potential therapeutic targets, 107 associated with silicosis. Enrichment analyses highlighted the TGF-β1/Smad pathway and epithelial-mesenchymal transition (EMT) in HYD's anti-silicosis effects, which was validated by the restoration of TGF-β1, p-Smad2/Smad2, p-Smad3/Smad3, E-cadherin, and Vimentin following HYD treatment. Additionally, glycitein, diosmetin, and limonin inhibited the proliferation and migration of TGF-β1-induced MLE12 cells and suppressed the activation of TGF-β1/Smad pathway and EMT. CONCLUSIONS HYD effectively alleviates silica-induced lung injury by specifically inhibiting the TGF-β1/Smad pathway and EMT process.
Collapse
Affiliation(s)
- Rui Qian
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Yunyi Xu
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Luoning Zhang
- Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Liqun Wang
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Xuxi Chen
- Molecular Toxicology Key Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Mengzhu Wang
- Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Qixue Bao
- Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Yuqin Yao
- State Key Laboratory of Biotherapy, West China School of Clinical Medicine (West China Hospital) Sichuan University, Chengdu, China; Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Linshen Xie
- Department of Occupational Health and Environmental Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
35
|
Wang W, Gao X, Niu W, Yin J, He K. Targeting Metabolism: Innovative Therapies for MASLD Unveiled. Int J Mol Sci 2025; 26:4077. [PMID: 40362316 PMCID: PMC12071536 DOI: 10.3390/ijms26094077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/01/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
The recent introduction of the term metabolic-dysfunction-associated steatotic liver disease (MASLD) has highlighted the critical role of metabolism in the disease's pathophysiology. This innovative nomenclature signifies a shift from the previous designation of non-alcoholic fatty liver disease (NAFLD), emphasizing the condition's progressive nature. Simultaneously, MASLD has become one of the most prevalent liver diseases worldwide, highlighting the urgent need for research to elucidate its etiology and develop effective treatment strategies. This review examines and delineates the revised definition of MASLD, exploring its epidemiology and the pathological changes occurring at various stages of the disease. Additionally, it identifies metabolically relevant targets within MASLD and provides a summary of the latest metabolically targeted drugs under development, including those in clinical and some preclinical stages. The review finishes with a look ahead to the future of targeted therapy for MASLD, with the goal of summarizing and providing fresh ideas and insights.
Collapse
Affiliation(s)
- Weixin Wang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.W.); (W.N.)
| | - Xin Gao
- School of Public Health, Jilin University, Changchun 130021, China;
| | - Wentong Niu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.W.); (W.N.)
| | - Jinping Yin
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130041, China;
| | - Kan He
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.W.); (W.N.)
| |
Collapse
|
36
|
de Vasconcellos JF, Westbrook P, Dingle M, Dimtchev A, Raiciulescu S, Schellhase CW, Piscoya A, Putko R, Bedrin M, Cole H, Cubbage N, Dargan LJ, Pellegrini VD, Nesti LJ. Preclinical validation of TGFβ inhibitors as a novel therapeutic strategy for post-traumatic heterotopic ossification. Sci Rep 2025; 15:14277. [PMID: 40274953 PMCID: PMC12022333 DOI: 10.1038/s41598-025-96961-0] [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: 04/16/2024] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Heterotopic ossification (HO) is characterized by the abnormal growth of ectopic bone in non-skeletal soft tissues through a fibrotic pathway and is a frequent complication in a wide variety of musculoskeletal injuries. We have previously demonstrated that TGFβ levels are elevated in the soft tissues following extremity injuries. Since TGFβ mediates the initial inflammatory and wound-healing response in the traumatized muscle bed, we hypothesized that targeted inhibition of the TGFβ pathway may be able to abrogate the unbalanced fibrotic phenotype and bone-forming response observed in post-traumatic HO. Primary mesenchymal progenitor cells (MPCs) harvested from debrided traumatized human muscle tissue were used in this study. After treatment with TGFβ inhibitors (SB431542, Galunisertib/LY2157299, Halofuginone, and SIS3) cell proliferation/survival, fibrotic formation, osteogenic induction, gene expression, and phosphorylation of SMAD2/3 were assessed. In vivo studies were performed with a Sprague-Dawley rat blast model treated with the TGFβ inhibitors. The treatment effects on the rat tissues were investigated by radiographs, histology, and gene expression analyses. Primary MPCs treated with TGFβ had a significant increase in the number of fibrotic nodules compared to the control, while TGFβ inhibitors that directly block the TGFβ extracellular receptor had the greatest effect on reducing the number of fibrotic nodules and significantly reducing the expression of fibrotic genes. In vivo studies demonstrated a trend towards a lower extent of HO formation by radiographic analysis up to 4 months after injury when animals were treated with the TGFβ inhibitors SB431542, Halofuginone and SIS3. Altogether, our results suggest that targeted inhibition of the TGFβ pathway may be a useful therapeutic strategy for post-traumatic HO patients.
Collapse
Affiliation(s)
| | - Phillip Westbrook
- Department of Orthopaedics and Physical Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Marvin Dingle
- Walter Reed National Military Medical Center, Bethesda, MD, 20889, USA
| | - Alexander Dimtchev
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Sorana Raiciulescu
- Department of Preventive Medicine and Biostatistics, Biostatistics Consulting Center, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | | | - Andres Piscoya
- Walter Reed National Military Medical Center, Bethesda, MD, 20889, USA
| | - Robert Putko
- Walter Reed National Military Medical Center, Bethesda, MD, 20889, USA
| | - Michael Bedrin
- Walter Reed National Military Medical Center, Bethesda, MD, 20889, USA
| | - Hisae Cole
- Department of Biology, James Madison University, 951 Carrier Drive, MSC 7801, Harrisonburg, VA, 22807, USA
| | - Nicole Cubbage
- Department of Biology, James Madison University, 951 Carrier Drive, MSC 7801, Harrisonburg, VA, 22807, USA
| | - Lauren Jeannette Dargan
- Department of Biology, James Madison University, 951 Carrier Drive, MSC 7801, Harrisonburg, VA, 22807, USA
| | - Vincent D Pellegrini
- Department of Orthopaedics and Physical Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, 1 Medical Center Drive, Lebanon, NH, 03756, USA.
| | - Leon J Nesti
- Head, Clinical and Experimental Orthopaedics, Alcamena Stem Cell Therapeutics, 1450 South Rolling Road, Suite 4.069, Halethorpe, MD, 21227, USA.
- Formerly at Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| |
Collapse
|
37
|
Liu C, Yu Q, Shen L, Wen X, Lian J, Wang J, Yang J, Chen L. Cepharantine prevents hypertrophic scarring by regulating the TGF-β/SMAD pathway. Arch Dermatol Res 2025; 317:734. [PMID: 40274641 DOI: 10.1007/s00403-025-04120-2] [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: 01/08/2025] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 04/26/2025]
Abstract
Hypertrophic scarring (HS) is a fibrotic skin disorder characterized by excessive deposition of extracellular matrix (ECM), leading to symptoms such as pain, itching, and skin contraction. HS can also result in restricted joint mobility and cosmetic deformities, imposing psychological and economic burdens on patients. Additionally, it increases wound care costs, and currently, no ideal treatment options exist. Therefore, HS is not only a clinical care issue but also a societal problem, with significant challenges related to its management and prevention. In this study, a custom-made cepharanthine ointment was applied to a rabbit ear scar model to investigate its effects on morphology, histology, and protein expression in HS. Additionally, the mechanism underlying the effect of cepharanthine on affected fibroblasts and the expression of ECM proteins was explored in vitro models of fibrosis. Animal experiments demonstrated that cepharanthine significantly reduced the tissue scar hypertrophy index and collagen content, improved the arrangement of fibroblasts, and inhibited ECM production. Cellular experiments indicated that cepharanthine effectively downregulated key proteins in the TGF-β/SMAD pathway, decreased ECM protein expression, and suppressed fibroblast proliferation and migration. Cepharanthine can prevent HS by reducing ECM deposition through the TGF-β/SMAD signalling pathway.
Collapse
Affiliation(s)
- Chenhuan Liu
- Zunyi Medical University, Zunyi, Guizhou, China
- Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | - Qin Yu
- Department of Orthopedics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Lingyan Shen
- Zunyi Medical University, Zunyi, Guizhou, China
- Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | - Xiaoyan Wen
- Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | - Juan Lian
- The Third Affiliated Hospital of Chengdu Medical College, Pidu District People's Hospital, Chengdu, Sichuan, China
| | - Jiani Wang
- School of Nursing, University of South China, Hengyang, Hunan, China
| | - Jin Yang
- Zunyi Medical University, Zunyi, Guizhou, China.
- Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China.
| | - Lin Chen
- Zunyi Medical University, Zunyi, Guizhou, China.
- Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China.
| |
Collapse
|
38
|
Zhou W, Wang J, Ding L, Cai R, Cheng J, Liang P, Zhu Y, Zhang Z. Autophagy inhibition enhances antifibrotic potential of placental mesenchymal stem cells of fetal origin via regulating TGF-β1 mediated protein degradation of HGF. Sci Rep 2025; 15:13805. [PMID: 40258831 PMCID: PMC12012010 DOI: 10.1038/s41598-025-97054-8] [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: 12/22/2024] [Accepted: 04/02/2025] [Indexed: 04/23/2025] Open
Abstract
Mesenchymal stem cell (MSC) therapy represents a promising strategy for pulmonary fibrosis (PF) treatment, with hepatocyte growth factor (HGF) serving as a key mediator of MSC-mediated protection. However, the therapeutic efficacy of MSCs is limited by the complex PF microenvironment, and the mechanisms underlying this limitation remain unclear. This study investigates how the PF pathological microenvironment modulates the antifibrotic potential of placental mesenchymal stem cells of fetal origin (fPMSCs) through HGF regulation and elucidates the molecular mechanisms involved. Morphological analysis, flow cytometry, and multilineage differentiation assays were employed to characterize fPMSCs. Transforming growth factor-β1 (TGF-β1) was employed to simulate the PF microenvironment and activate fPMSCs in vitro. ELISA and Western blotting were used to analyze HGF expression, autophagy markers, and Smad signaling. Autophagosome formation was visualized via confocal microscopy and transmission electron microscopy. Co-immunoprecipitation (Co-IP) assays were performed to assess the interaction between p62 and HGF. The antifibrotic function of fPMSCs was further evaluated using a transwell co-culture system with MRC-5 fibroblasts in vitro and a bleomycin-induced PF mouse model in vivo. Phenotypic characterization confirmed that fPMSCs exhibited canonical MSC morphology, expressed CD73/CD90/CD105, lacked CD14/CD34/CD45/HLA-DR, and differentiated into adipogenic, osteogenic, and chondrogenic lineages. TGF-β1 treatment robustly downregulated the antifibrotic capacity, HGF protein expression, and paracrine secretion in fPMSCs. Recombinant HGF enhanced antifibrotic effects, while an HGF-neutralizing antibody abolished them. TGF-β1 induced autophagy in fPMSCs, promoting HGF degradation via p62 interaction and impairing antifibrotic function in vitro and in vivo. Mechanistically, Smad3 phosphorylation mediated the regulation of autophagy and HGF expression in TGF-β1-treated fPMSCs. Our findings demonstrate that TGF-β1 impairs the antifibrotic function of fPMSCs via autophagy-dependent HGF degradation and Smad3 signaling. Conversely, autophagy inhibition restores HGF levels and enhances fPMSCs' therapeutic efficacy in a preclinical PF model. Targeting autophagy inhibition emerges as a promising therapeutic strategy to counteract pulmonary fibrosis.
Collapse
Affiliation(s)
- Wei Zhou
- The First School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- The People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750000, Ningxia, China
| | - Jing Wang
- The First School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Lu Ding
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, 804 W. Shengli Road, Yinchuan, 750000, Ningxia, China
| | - Ruizhi Cai
- The First School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Jie Cheng
- The Third School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Panpan Liang
- The People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750000, Ningxia, China
| | - Yongzhao Zhu
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, 804 W. Shengli Road, Yinchuan, 750000, Ningxia, China.
| | - Zaiqi Zhang
- The First School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China.
| |
Collapse
|
39
|
Zhao Q, Jin M, Zhao Q, Wang Z, Zhao C, Xue X, Qiao X, Qu P, Han D, Tao R. Natural products in traditional Chinese medicine for renal fibrosis: a comprehensive review. Front Pharmacol 2025; 16:1560567. [PMID: 40308781 PMCID: PMC12041090 DOI: 10.3389/fphar.2025.1560567] [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: 01/14/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Renal fibrosis represents the terminal pathological manifestation of most chronic kidney diseases, driving progressive loss of renal function. Natural products have emerged as promising therapeutic agents for preventing and ameliorating renal fibrosis due to their multi-target efficacy and favorable safety profiles. In this review, we conducted a comprehensive literature search on PubMed using the keywords "natural product" and "renal fibrosis" from 2004 to 2025, identifying 704 relevant articles. We systematically categorize and discuss the biological effects of key natural products and formulations with antifibrotic potential, focusing on five major classes: glycosides, flavonoids, phenolic compounds, anthraquinones, and terpenoids. Representative compounds from each category are highlighted for their mechanisms of action, including modulation of oxidative stress, inflammation, autophagy, and fibrosis signaling pathways. This review aims to provide a theoretical foundation for the development of natural product-based therapies to combat renal fibrosis, offering insights into their therapeutic potential and future research directions.
Collapse
Affiliation(s)
- Qianqian Zhao
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Meihua Jin
- Department of Immunology, Medical College, Dalian University, Dalian, Liaoning, China
| | - Qiang Zhao
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Zhimei Wang
- Department of Immunology, Medical College, Dalian University, Dalian, Liaoning, China
| | - Chun Zhao
- Department of Immunology, Medical College, Dalian University, Dalian, Liaoning, China
| | - Xiaocong Xue
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Xikai Qiao
- Department of Immunology, Medical College, Dalian University, Dalian, Liaoning, China
| | - Peng Qu
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Donghe Han
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Ran Tao
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| |
Collapse
|
40
|
He J, Yue H, Zhang S, Dong R, Zhang F, Wang X, Wang K, Zhang H, Yang D, Dong Z, Liu H. Dehydrocorydaline attenuates bleomycin-induced pulmonary fibrosis by inhibiting fibroblast activation. Respir Res 2025; 26:136. [PMID: 40221718 PMCID: PMC11992754 DOI: 10.1186/s12931-025-03218-5] [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: 11/15/2024] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) is an irreversible, progressive, chronic and fatal interstitial lung disease with limited therapeutic options. Dehydrocorydaline (DHC), derived from the traditional Chinese medicinal plant Corydalis yanhusuo, has exhibited a variety of pharmacological properties. Nevertheless, the potential function and mechanism of DHC in the management of PF have yet to be elucidated. PURPOSE To evaluate the therapeutical efficacy of DHC in different PF models and elucidate its underlying mechanism. METHODS A well-established Bleomycin-induced PF mouse model and human precision-cut lung slices (hPCLS) following fibrosis-inducing cocktail stimulation were employed. The antifibrotic effects of DHC on PF were measured by histopathological manifestation, immunofluorescent staining and expression levels of fibrosis related markers. Human primary pulmonary fibroblasts (HPFs) were used to explore the impact of DHC on fibroblast function and the underlying mechanism. RESULTS Here, we demonstrated that DHC exhibited a therapeutic efficacy in Bleomycin-induced PF mouse model with a dose dependent, as well as in hPCLS after fibrosis-inducing cocktail stimulation, as evidenced by histopathological staining, decrease of Fibronectin, Collagen 1 and α-SMA expression. Additionally, in vitro experiments indicated that DHC effectively suppressed fibroblast to myofibroblast transition, but had no significant effect on the proliferation and migration of fibroblast. Mechanistic studies revealed that the inhibitory effect of DHC on fibroblast activation was dependent on the endoplasmic reticulum stress, thereby inhibiting TGF-β/SMAD signal pathway. CONCLUSIONS Our study implied that DHC hold a promise therapeutic approach against PF by suppressing fibroblast activation. The safety and efficacy of DHC have been preliminary demonstrated in a mouse model.
Collapse
Affiliation(s)
- Jianhan He
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Huihui Yue
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Shufei Zhang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ruihan Dong
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Fengqin Zhang
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Xuewen Wang
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Ke Wang
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Huilan Zhang
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Danlei Yang
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China.
| | - Zhaoxing Dong
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, No. 41, Northwestern Street, Ningbo, 315010, China.
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China.
| |
Collapse
|
41
|
Kananivand M, Nouri F, Yousefi MH, Pajouhi A, Ghorbani H, Afkhami H, Razavi ZS. Mesenchymal stem cells and their exosomes: a novel approach to skin regeneration via signaling pathways activation. J Mol Histol 2025; 56:132. [PMID: 40208456 DOI: 10.1007/s10735-025-10394-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025]
Abstract
Accelerating wound healing is a crucial objective in surgical and regenerative medicine. The wound healing process involves three key stages: inflammation, cell proliferation, and tissue repair. Mesenchymal stem cells (MSCs) have demonstrated significant therapeutic potential in promoting tissue regeneration, particularly by enhancing epidermal cell migration and proliferation. However, the precise molecular mechanisms underlying MSC-mediated wound healing remain unclear. This review highlights the pivotal role of MSCs and their exosomes in wound repair, with a specific focus on critical signaling pathways, including PI3K/Akt, WNT/β-catenin, Notch, and MAPK. These pathways regulate essential cellular processes such as proliferation, differentiation, and angiogenesis. Moreover, in vitro and in vivo studies reveal that MSCs accelerate wound closure, enhance collagen deposition, and modulate immune responses, contributing to improved tissue regeneration. Understanding these mechanisms provides valuable insights into MSC-based therapeutic strategies for enhancing wound healing.
Collapse
Affiliation(s)
- Maryam Kananivand
- Medical Department, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Nouri
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University (SRBIAU), Tehran, Iran
| | - Mohammad Hasan Yousefi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
| | - Ali Pajouhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hakimeah Ghorbani
- Department of Sciences, Faculty of Biological Sciences, Tabriz University of Sciences, Tabriz, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran.
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran.
| | - Zahra Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
42
|
Dilger OB, Carstens MF, Bothun CE, Payne AN, Berry DJ, Sanchez-Sotelo J, Morrey ME, Thaler R, Dudakovic A, Abdel MP. Induction of cellular autophagy impairs TGF-β1-mediated extracellular matrix deposition in primary human knee fibroblasts. Bone Joint Res 2025; 14:331-340. [PMID: 40192622 PMCID: PMC11975063 DOI: 10.1302/2046-3758.144.bjr-2024-0312.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
Aims To evaluate the role of autophagy in primary knee fibroblasts undergoing myofibroblast differentiation as an in vitro model of arthrofibrosis, a complication after total knee arthroplasty characterized by aberrant intra-articular scar tissue formation and limited range of motion. Methods We conducted a therapeutic screen of autophagic-modulating therapies in primary human knee fibroblasts undergoing transforming growth factor-beta 1 (TGF-β1)-mediated myofibroblast differentiation. Autophagy was induced pharmacologically with rapamycin or by amino acid deprivation. Picrosirius red staining was performed to quantify collagen deposition. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting were conducted to evaluate fibrotic gene expression levels. Results Rapamycin, an mTOR complex 1 (mTORC1) inhibitor and autophagy inducer, reduced TGF-β1-mediated collagen deposition. Interestingly, we simultaneously report that myofibrogenic genes, including ACTA2, were highly upregulated following rapamycin-TGF-β1 treatment. When autophagy was induced through amino acid deprivation, we demonstrated suppressed extracellular matrix levels, fibrotic gene expression (e.g. ACTA2), and SMAD2 phosphorylation levels in TGF-β1-stimulated fibroblasts. Conclusion Our findings demonstrate that the induction of cellular autophagy suppresses TGF-β1-induced collagen deposition in primary human knee fibroblasts. Taken together, these data suggest that cellular autophagy may be prophylactic against the pathogenesis of arthrofibrosis.
Collapse
Affiliation(s)
- Oliver B. Dilger
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Mason F. Carstens
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Cole E. Bothun
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ashley N. Payne
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Daniel J. Berry
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Mark E. Morrey
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew P. Abdel
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
43
|
Gill SK, Gomer RH. Translational Regulators in Pulmonary Fibrosis: MicroRNAs, Long Non-Coding RNAs, and Transcript Modifications. Cells 2025; 14:536. [PMID: 40214489 PMCID: PMC11988943 DOI: 10.3390/cells14070536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Fibrosing disorders including idiopathic pulmonary fibrosis (IPF) are progressive irreversible diseases, often with poor prognoses, characterized by the accumulation of excessive scar tissue and extracellular matrix. Translational regulation has emerged as a critical aspect of gene expression control, and the dysregulation of key effectors is associated with disease pathogenesis. This review examines the current literature on translational regulators in IPF, focusing on microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and RNA transcript modifications including alternative polyadenylation and chemical modification. Some of these translational regulators potentiate fibrosis, and some of the regulators inhibit fibrosis. In IPF, some of the profibrotic regulators are upregulated, and some of the antifibrotic regulators are downregulated. Correcting these defects in IPF-associated translational regulators could be an intriguing avenue for therapeutics.
Collapse
Affiliation(s)
| | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
44
|
Chang X, Zhang Y, Deng M, Yang R, Zhang J, Hao M, Miao J. OTUD1 inhibits endometriosis fibrosis by deubiquitinating MADH7. Mol Hum Reprod 2025; 31:gaaf014. [PMID: 40279273 DOI: 10.1093/molehr/gaaf014] [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: 08/22/2024] [Revised: 03/12/2025] [Indexed: 04/27/2025] Open
Abstract
Fibrosis constitutes the principal pathophysiological mediator of pain and infertility manifestations in endometriosis, and the inhibitory factor of the TGF-β pathway, MADH7, makes a vital impact on the progression of fibrosis. Ovarian tumor domain-containing protein 1 (OTUD1) deubiquitinase binds to the MADH7 protein, although its specific role in endometriosis needs to be investigated. This study is the first to explore the role of OTUD1 in endometriosis and to investigate its impact on the growth of endometriosis lesions in vitro and in vivo, using C57BL/6N female mice and human primary stromal endometriosis cells (HEMCs). Moreover, the obtained results demonstrated that OTUD1 inhibited the expression of fibrosis-related proteins in HEMCs in vitro, and the mechanistic execution of this phenotype was achieved via coordinated deubiquitination coupled with MADH7-mediated transcriptional reprogramming. These events stopped the growth of lesions in vivo and reduced abdominal inflammation. The study demonstrated the critical role of the deubiquitinating enzyme OTUD1 in endometriosis, indicating its potential therapeutic effect on endometriosis.
Collapse
Affiliation(s)
- Xiangyu Chang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yanqin Zhang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Mengqi Deng
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Ruiye Yang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Jiamin Zhang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Menglin Hao
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Jinwei Miao
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| |
Collapse
|
45
|
Hao MY, Li HJ, Han HS, Chu T, Wang YW, Si WR, Jiang QY, Wu DD. Recent advances in the role of gasotransmitters in necroptosis. Apoptosis 2025; 30:616-635. [PMID: 39833633 DOI: 10.1007/s10495-024-02057-x] [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] [Accepted: 12/13/2024] [Indexed: 01/22/2025]
Abstract
Necroptosis is a finely regulated programmed cell death process involving complex molecular mechanisms and signal transduction networks. Among them, receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like protein are the key molecules regulating this process. In recent years, gasotransmitters such as nitric oxide, carbon monoxide and hydrogen sulfide have been suggested to play a regulatory role in necroptosis. This paper reviews the evidence that these gasotransmitters are involved in the regulation of necroptosis by influencing the production of reactive oxygen species, regulating the modification of S subunits of RIPK1 and RIPK3, regulating inflammatory mediators, and signal transduction. In addition, this review explores the potential therapeutic applications of these gasotransmitters in pathological conditions such as cardiovascular disease and ischemia-reperfusion injury. Although some studies have revealed the important role of gasotransmitters in necroptosis, the specific mechanism of action is still not fully understood. Future research is needed to further elucidate the molecular mechanisms of gasotransmitters in precisely regulating necroptosis, which will help develop new therapeutic strategies to prevent and treat related diseases.
Collapse
Affiliation(s)
- Meng-Yuan Hao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Hong-Jie Li
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Hang-Shen Han
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Ti Chu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yan-Wen Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Wei-Rong Si
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.
| | - Qi-Ying Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China.
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China.
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
46
|
Zhang H, Hong Z, Jiang Z, Hu W, Hu J, Zhu R. miR-29b-3p Affects the Hypertrophy of Ligamentum Flavum in Lumbar Spinal Stenosis and its Mechanism. Biochem Genet 2025; 63:1824-1838. [PMID: 38625592 DOI: 10.1007/s10528-024-10811-8] [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: 12/18/2023] [Accepted: 04/10/2024] [Indexed: 04/17/2024]
Abstract
To explore the effect of miR-29b-3p on fibrosis and hypertrophy of ligamentum flavum (LF) in lumbar spinal stenosis (LSS) and its underlying mechanism. Patients with LSS and lumbar disc herniation (LDH) (control) undergoing posterior lumbar laminectomy were included in this study. Human LF samples were obtained for LF cell isolation, RNA, and protein extraction. Histomorphological analysis of LF was performed using hematoxylin-eosin (HE) staining. After isolation, culture, and transfection of primary LF cells, different transfection groups were constructed: NC-mimic, miR-29b-3p-mimic, NC-inhibitor, and miR-29b-3p-inhibitor. Quantitative real time polymerase chain reaction (qRT-PCR) was performed to detect the expression of miR-29b-3p in LF and LF cells. Western blot analysis detected the protein expressions of P16 and CyclinD1. ELISA detected the protein expressions of TGF-β1, Smad2, Smad3, TLR4, Type I collagen, and Type III collagen. Finally, LF cell viability was detected using the Cell Counting Kit-8 (CCK8) assay. The thickness of LF was significantly thicker in the LSS group compared to the LDH group (p < 0.05), accompanied by a higher calcification degree, more fibroblasts, and a larger area of collagen fiber proliferation. miR-29b-3p expression was significantly lower in LSS-derived LF tissues and cells than in LDH-derived tissues and cells (both p < 0.05). Compared to the NC-mimic group, the miR-29b-3p-mimic group exhibited significantly higher miR-29b-3p expression, decreased protein expressions of Type I collagen, Type III collagen, TGF-β1, Smad2, Smad3, TLR4, P16, and CyclinD1, and inhibited LF cell proliferation (all p < 0.05). As expected, the miR-29b-3p-inhibitor group displayed contrasting expression patterns (all p < 0.05). Compared to the phosphate buffer saline (PBS) group, the Trimethylamine-N-Oxide (TMAO) group showed significantly increased expressions of TGF-β1, Smad2, Smad3, TLR4, Type I collagen, Type III collagen, P16, and CyclinD1, as well as enhanced LF cell proliferation (all p < 0.05). However, there was no significant difference between the TMAO group and the Ang II group (all p > 0.05). Upregulation of miR-29b-3p expression may play a role in improving LF fibrosis and hypertrophy in LSS by inhibiting P16 expression and suppressing the activation of the TGF-β/Smad signaling pathway. This finding offers new insights into future gene modification therapy for this patient population.
Collapse
Affiliation(s)
- Hongjie Zhang
- Department of Orthopedics, Dehong People's Hospital, Kunming Medical University Affiliated Dehong Hospital, Dehong, No.13 Yonghan Road, Mangshi District, 678400, China
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, No.10, Poyang Lake Road, Jinghai District, Tianjin, 301617, China
| | - Zhixiong Hong
- Department of Orthopedics, Dehong People's Hospital, Kunming Medical University Affiliated Dehong Hospital, Dehong, No.13 Yonghan Road, Mangshi District, 678400, China
| | - Zehua Jiang
- Department of Spine Surgery, Tianjin Union Medical Center, No.190 Jieyuan Road, Hongqiao District, Tianjin, 300122, China
| | - Wei Hu
- Department of Spine Surgery, Tianjin Union Medical Center, No.190 Jieyuan Road, Hongqiao District, Tianjin, 300122, China
| | - Jiashao Hu
- Department of Orthopedics, Dehong People's Hospital, Kunming Medical University Affiliated Dehong Hospital, Dehong, No.13 Yonghan Road, Mangshi District, 678400, China
| | - Rusen Zhu
- Department of Spine Surgery, Tianjin Union Medical Center, No.190 Jieyuan Road, Hongqiao District, Tianjin, 300122, China.
| |
Collapse
|
47
|
Zhang J, Zhang X, Wu R, Dong CS. Unveiling purine metabolism dysregulation orchestrated immunosuppression in advanced pancreatic cancer and concentrating on the central role of NT5E. Front Immunol 2025; 16:1569088. [PMID: 40236698 PMCID: PMC11996659 DOI: 10.3389/fimmu.2025.1569088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/07/2025] [Indexed: 04/17/2025] Open
Abstract
Background The dismal efficacy of immunotherapy for Pancreatic cancer (PC) can be predominantly ascribed to its distinctive cold-tumor properties. The by-products of purine metabolic reprogramming are extensively engaged in tumor immune modulation, influencing the functions and recruitment of immune cells and molding an immune microenvironment that is propitious for tumor growth. Methods We harnessed single-cell transcriptomics and spatial transcriptomics to concurrently analyze the purine metabolism (PM) features of the PC microenvironment. We quantitatively appraised the PM traits of diverse cell subsets via scoring algorithms such as AUCell and Ucell. Moreover, cell development and cell-cell interaction analysis elucidated the alterations in TME induced by PM dysregulation. Additionally, we defined the PM disorder characteristics of PC patients and utilized this to assess the immune phenotypes and prognoses of the patient population. Also, we identified the crucial intermediate genes that impact PM reprogramming and the establishment of an immunosuppressive environment within the TME of PC, and validated them through spatial sectioning and cell co-culture experiments. Results Multi - dimensional transcriptome data elucidated the unique heterogeneity of PM in the PC microenvironment, which manifested that tumor cells and fibroblasts demonstrating higher PM scores in the TME. Cellchat analysis revealed that malignant cells with elevated PM expression were concomitantly associated with frequent interactions with CAFs as well as high expression of ligand-receptor pairs and transcription factors. Spatial data further corroborated this finding. Furthermore, the newly constructed PM disorder criteria indicated that patients with high PM levels were associated with a lack of response to immunotherapy and an immunosuppressive microenvironment. Finally, this study identified the singular role of NT5E in the immunosuppression resulting from PM reprogramming in PC. CCK8 and invasion experiments following the co-culture model demonstrated that intervention targeting NT5E could reverse the augmented malignancy of PC induced by co-cultured CAFs. NT5E is potentially a key target for reversing the "stiff-cancer" characteristics of PC. Conclusion This study demonstrates that PM metabolic disorders could impinge upon tumor immunotherapy and exacerbate the immunosuppression engendered by the progression of PC fibrosis. Therapeutic strategies targeting PM or NT5E may offer a ray of hope for patients with advanced PDAC.
Collapse
Affiliation(s)
- Junqian Zhang
- Henan Key Laboratory of Cancer Epigenetics; Cancer Institute, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xiaobo Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Ruixin Wu
- Preclinical Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chang-Sheng Dong
- Cancer Institute of Traditional Chinese Medicine/Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
48
|
Liu S, Chen Y, Zhou G, Sun C, Ma M, Huang R, Li X, Liang X, Shi C, Wu W, Yan X, Wang L, Han J. Uniform and controllable surface nano-structure on polyetheretherketone implants can regulate mechanical property to enhance soft tissue integration through Piezo1/TGF-β1 signaling axis. Mater Today Bio 2025; 31:101645. [PMID: 40151615 PMCID: PMC11946874 DOI: 10.1016/j.mtbio.2025.101645] [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: 12/17/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Polyetheretherketone (PEEK) has potential to repair the orbital floor bone defects following craniofacial trauma and orbital surgery. However, the inertness of the material impedes the soft tissue integration of implants, leading to complications such as implant migration and infection. Surface patterning modification on PEEK can promote the surface hydrophily to enhance better soft tissue integration, but it is difficult to obtain the uniform and controllable nano-structure. In this study, hot-pressing technology on PEEK implant was used to produce surface nanopores with a uniform diameter of 200, 500, 800 nm. Depending on the controllable craft, the surficial mechanical properties of PEEK can be regulated and assessed by finite element analysis. Furthermore, 500 nm interface has better mechanical properties to promote the proliferation, migration, and fibrosis of fibroblasts and achieved optimal integration effects in animal implantation experiments. To explore the mechanism of biological responses, transcriptomics and molecular biology experiments revealed that Piezo1/TGF-β1 axis played a critical role in the response of soft tissue cells to the mechanical stimulation of PEEK. Our study has established a novel modification technology for constructing uniform and controllable nanostructures on the surface of PEEK, thereby promoting the soft tissues integration with implants and improving the anchoring effect.
Collapse
Affiliation(s)
- Sida Liu
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
- The 940 Hospital of the Joint Logistic Support Force, 730050, Lanzhou, Gansu Province, China
| | - Yixuan Chen
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Gandong Zhou
- School of Advanced Materials and Nanotechnology, Xidian University, 710126, Xi'an, Shaanxi Province, China
| | - Changning Sun
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054, Xi'an, Shaanxi Province, China
| | - Minghai Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Rou Huang
- School of Advanced Materials and Nanotechnology, Xidian University, 710126, Xi'an, Shaanxi Province, China
| | - Xing Li
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Xiao Liang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Changquan Shi
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054, Xi'an, Shaanxi Province, China
| | - Weiwei Wu
- School of Advanced Materials and Nanotechnology, Xidian University, 710126, Xi'an, Shaanxi Province, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Lei Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| |
Collapse
|
49
|
Ning A, Xiao N, Yu X, Wang H, Guan C, Guo C, Dong Y, Ma X, Xia H. Dimethyloxallyl Glycine Preconditioning Promotes the Anti-inflammatory and Anti-fibrotic Effects of Human Umbilical Cord Mesenchymal Stem Cells on Kidney Damage in Systemic Lupus Erythematosus Related to TGF-β/Smad Signaling Pathway. Inflammation 2025; 48:839-854. [PMID: 39044003 DOI: 10.1007/s10753-024-02092-5] [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: 03/11/2024] [Revised: 06/03/2024] [Accepted: 06/27/2024] [Indexed: 07/25/2024]
Abstract
Systemic lupus erythematosus (SLE) is a chronic inflammatory autoimmune disease lacking effective treatments without adverse effects. Dimethyloxallyl glycine (DMOG) enhanced mesenchymal stem cells (MSC) capabilities, but it remains unclear how DMOG-pretreatment of MSCs augments their SLE treatment. Here, we explore the therapeutic potential of DMOG-pretreated human umbilical cord MSCs (hUC-MSCs) in a mouse lupus nephritis (LN) model. In vitro experiments showed that DMOG could alleviate the mRNA levels of tumor necrosis factor (TNF)-α, interferon (IFN)-γ, and interleukin (IL)-6 and increase the mRNA level of IL-13 in lipopolysaccharide (LPS)-induced inflammation in hUC-MSCs. DMOG enhanced the migratory and invasive abilities of the hUC-MSCs. In vivo animal studies revealed that DMOG-pretreated hUC-MSCs exhibited more pronounced inhibition of lymphadenectasis and reduced kidney weight and urinary protein content than MSCs alone. DMOG-pretreated hUC-MSCs improved renal morphological structure and alleviated inflammatory cell infiltration and renal fibrosis, evidenced by the reduced mRNA levels of fibrosis markers, including fibronectin (Fn), collagen alpha-1 chain (Colα1), collagen alpha-3 chain (Colα3), and TNF-α, IFN-γ, and IL-6 cytokines. Further investigation revealed that DMOG-pretreated hUC-MSCs down-regulated the expressions of transforming growth factor (Tgf)-β1 and its downstream effectors Smad2 and Smad3, recognized as central mediators in renal fibrosis (P < 0.05). The findings suggest that DMOG-pretreated hUC-MSCs can augment the therapeutic efficacy of hUC-MSCs in LN by enhancing their anti-inflammatory and antifibrotic effects, and the TGF-β/Smad signaling pathway may be involved in this process.
Collapse
Affiliation(s)
- Anfeng Ning
- Reproductive and Genetic Center & NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning (NRIFP), Beijing, 100081, China
- Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Nansong Xiao
- Reproductive and Genetic Center & NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning (NRIFP), Beijing, 100081, China
- Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiaoqin Yu
- Reproductive and Genetic Center & NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning (NRIFP), Beijing, 100081, China
- Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Hu Wang
- Reproductive and Genetic Center & NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning (NRIFP), Beijing, 100081, China
- Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Chunyi Guan
- Reproductive and Genetic Center & NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning (NRIFP), Beijing, 100081, China
- Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Changlong Guo
- National Human Genetic Resources Center, National Research Institute for Family Planning (NRIFP), Beijing, 100081, China
| | - Yichao Dong
- National Human Genetic Resources Center, National Research Institute for Family Planning (NRIFP), Beijing, 100081, China
| | - Xu Ma
- Reproductive and Genetic Center & NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning (NRIFP), Beijing, 100081, China.
- Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Hongfei Xia
- Reproductive and Genetic Center & NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning (NRIFP), Beijing, 100081, China.
- Graduate Schools, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
50
|
Du H, Yang K, He Z, Su L, Tan X, Li Z, Song W, Cao L, Ma Y. Tianjihuang compound alleviates aflatoxin B 1-induced hepatic steatosis and fibrosis by targeting PPARα-TGF-β pathway in ducklings. Poult Sci 2025; 104:105006. [PMID: 40073684 PMCID: PMC11932684 DOI: 10.1016/j.psj.2025.105006] [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: 01/09/2025] [Revised: 02/21/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
Aflatoxin B1 (AFB1), a potent mycotoxin, poses a significant threat to the poultry industry, particularly affecting the health and growth of ducklings. The present study aimed to investigate the therapeutic effects and mechanisms of the Tianjihuang compound (HRS), a traditional Chinese medicine formulation, on AFB1-induced chronic toxicity in ducklings. Firstly, 30 ingredients, including neochlorogenic acid, kaempferol 3-alpha-D-galactoside, quercetin, hispidulin, caffeic acid, and myricetin, were identified from HRS with UPLC-MS/MS method. Then, over a 25-day experimental period, a total of 100 one-day-old Sichuan Sheldrakes were randomly divided into five groups: control, AFB1 model, and HRS high (4 g/kg), medium (2 g/kg), and low dosage (1 g/kg) groups. Results indicated that HRS effectively mitigated the negative impact on the productivity, reduced the levels of liver index, AST, ALT, and AST/ALT in serum, increased the levels of serum TP content, and obviously alleviated inflammatory cell infiltration, liver fibrosis, and liver steatosis induced by AFB1. Additionally, HRS enhanced the levels of GST, CAT, and T-AOC, and decreased the levels of MDA and AFB1-DNA, thereby alleviating oxidative stress and AFB1-DNA generation caused by AFB1. Transcriptome analysis revealed that HRS may improve liver injury in AFB1-chronically poisoned ducklings by regulating the ECM receptor interaction, fatty acid metabolism, cell adhesion molecules, TGF-β signaling pathway, and PPAR signaling pathway. Further RT-qPCR analysis revealed that HRS might downregulate the expression of ASCL4 gene by promoting the activation of PPARα, thereby inhibiting the activation of the TGF-β signaling pathway and improving liver steatosis and fibrosis caused by AFB1 in ducklings. In conclusion, the HRS exhibits hepatoprotective effects against AFB1-induced chronic toxicity in ducklings by restoring liver function, enhancing antioxidant capacity, and its mechanism of damage resistance may be related to the improvement of liver steatosis and fibrosis in ducklings by inhibiting the PPARα-TGF-β signaling pathway.
Collapse
Affiliation(s)
- Hongxu Du
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, PR China; Institute of Traditional Chinese Veterinary Medicine, Southwest University, Chongqing 402460, PR China; Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 402460, PR China.
| | - Kunzhao Yang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, PR China
| | - Zhengke He
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, PR China
| | - Lijuan Su
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, PR China
| | - Xiaoyan Tan
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, PR China
| | - Zhangxun Li
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, PR China
| | - Weijie Song
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, PR China
| | - Liting Cao
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, PR China; Institute of Traditional Chinese Veterinary Medicine, Southwest University, Chongqing 402460, PR China; Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 402460, PR China
| | - Yue Ma
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, PR China; Institute of Traditional Chinese Veterinary Medicine, Southwest University, Chongqing 402460, PR China; Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 402460, PR China
| |
Collapse
|