1
|
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
|
2
|
Xiang J, Lv N, Yin S, Zhao T, Liu F, Cheng L, Liu F, Kuang J. TGF-β induces EMT in thyroid cancer cells by regulating transcription factors. Thyroid Res 2025; 18:25. [PMID: 40490818 DOI: 10.1186/s13044-025-00243-w] [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] [Received: 02/09/2025] [Accepted: 04/28/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Transforming growth factor-β (TGF-β) plays well-established roles in cancer cell invasion and epithelial-mesenchymal transition (EMT); however, its role in thyroid carcinoma (TC) remains unclear. This study aimed to evaluate the effects of TGF-β on EMT in TC and determine its underlying mechanisms. METHODS Treatment of TC cell lines with TGF-β the morphology of thyroid cancer cells changed, Immunofluorescence staining revealed that the localization of E-cadherin shifted from the cell membrane to the cytoplasm, and the fluorescence intensity decreases. Wound-healing assay in BCPAP and TPC-1 revealed that migration ability was significantly higher in the TGF-β (5 ng/mL) treatment group than in the control group (P < 0.01). RESULTS Transwell assays showed that the invasive abilities of TGF-β-treated BCPAP, TPC-1, and K1 cells were 7-, 10-, and 6-fold higher than those of the control group, respectively (P < 0.05). After TGF-β treatment, mRNA levels of SNAI1 significantly increased in TPC-1 and BCPAP cell lines. Treatment of TC cell lines with TGF-β downregulated the epithelial marker E-cadherin and upregulated the mesenchymal markers N-cadherin and vimentin, at the mRNA level. Western blotting indicated similar results at the protein level, TSH could enhance this process. CONCLUSIONS TGF-β promotes EMT-like phenotypic changes in thyroid cancer cells, accompanied by upregulation of SNAI1 and EMT-related markers, which is enhanced by TSH. Overall, this study provides a basis for the development of therapeutic strategies for TC targeting the EMT.
Collapse
Affiliation(s)
- Jianjian Xiang
- Department of Ultrasound Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Nannan Lv
- Department of Endocrinology, The Fourth People's Hospital of Shenyang, China Medical University, 20 Huanghe South Street, Huanggu District, Shenyang, 10032, China.
| | - Shanyu Yin
- Department of Ultrasound Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Tong Zhao
- Department of Endocrinology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Fei Liu
- Department of Endocrinology, The Fourth People's Hospital of Shenyang, China Medical University, 20 Huanghe South Street, Huanggu District, Shenyang, 10032, China
| | - Lan Cheng
- Department of Endocrinology, The Fourth People's Hospital of Shenyang, China Medical University, 20 Huanghe South Street, Huanggu District, Shenyang, 10032, China
| | - Feng Liu
- Department of Endocrinology, The Fourth People's Hospital of Shenyang, China Medical University, 20 Huanghe South Street, Huanggu District, Shenyang, 10032, China
| | - Jinsong Kuang
- Department of Endocrinology, The Fourth People's Hospital of Shenyang, China Medical University, 20 Huanghe South Street, Huanggu District, Shenyang, 10032, China.
| |
Collapse
|
3
|
Zhuang X, Wang C, Ge Z, Wu M, Chen M, Chen Z, Hu J. MICAL1 Mediates TGF-β1-Induced Epithelial-to-Mesenchymal Transition and Metastasis of Hepatocellular Carcinoma by Activating Smad2/3. Cell Biochem Biophys 2025; 83:2589-2606. [PMID: 39954154 DOI: 10.1007/s12013-025-01668-8] [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] [Accepted: 01/03/2025] [Indexed: 02/17/2025]
Abstract
Epithelial-mesenchymal transition (EMT) induced by transforming growth factor-β (TGF-β) is involved in hepatocellular carcinoma (HCC) growth and metastasis. Our study aimed to investigate the role of molecules interacting with CasL 1 (MICAL1) in regulating TGF-β-triggered EMT in HCC and the related mechanisms. After detecting MICAL1 expression and prognostic value in HCC, in vitro assays including CCK-8 assay, EdU staining, flow cytometry assay, Transwell assay, western blotting, and RT-qPCR and in vivo metastasis assay was conducted to evaluate the influence of MICAL1 knockdown on the proliferation and apoptosis as well as TGF-β-induced EMT and metastasis of Huh7 and MHCC97H cells. MICAL1 was highly expressed in HCC, and its high expression was related to histological grade, TNM stage, and shorter overall survival of HCC patients. MICAL1 silencing suppressed proliferation, promoted apoptosis, and curbed TGF-β1-triggered cytoskeletal remodeling, EMT, and metastasis of HCC cells. MICAL1 knockdown impeded TGF-β1-induced upregulation in phosphorylated-Smad2/3 protein levels and reduced Smad2/3 mRNA levels in HCC cells. MICAL1 downregulation enhanced the polyubiquitination and proteasomal degradation of TβRI. Additionally, MICAL1 silencing suppressed tumor growth and lung metastasis in Huh7-derived xenograft mouse models. Collectively, MICAL1 knockdown impairs TGF-β1-stimulated EMT and metastasis of HCC cells by restraining Smad2/3 phosphorylation and activation.
Collapse
Affiliation(s)
- Xun Zhuang
- Department of Gastroenterology, The People's Hospital of Dan Yang, Zhenjiang, Jiangsu, PR China
| | - Chunrong Wang
- Department of Gastroenterology, The People's Hospital of Dan Yang, Zhenjiang, Jiangsu, PR China
| | - Zhenghui Ge
- Department of Gastroenterology, The People's Hospital of Dan Yang, Zhenjiang, Jiangsu, PR China
| | - Mengjie Wu
- Department of Gastroenterology, The People's Hospital of Dan Yang, Zhenjiang, Jiangsu, PR China
| | - Mengjiao Chen
- Department of Gastroenterology, The People's Hospital of Dan Yang, Zhenjiang, Jiangsu, PR China
| | - Zhen Chen
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| | - Jianghong Hu
- Department of Gastroenterology, The People's Hospital of Dan Yang, Zhenjiang, Jiangsu, PR China.
| |
Collapse
|
4
|
Cheng W, Zheng Y, Tang Q, Qi L, Shi Z, Yu Q, Li M, Wei X, Zhou Y, Jiang X. Discovery of Novel Cyclic Peptides as SMAD2-SMAD4 Interaction Inhibitors for the Treatment of Hepatic Fibrosis. J Med Chem 2025; 68:9958-9972. [PMID: 40320643 DOI: 10.1021/acs.jmedchem.4c02938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Hepatic fibrosis, characterized by the excessive deposition of the extracellular matrix, represents a common consequence of various chronic liver disorders. However, no specific drugs are available for antifibrotic therapy to date. SMAD2 is phosphorylated by transforming growth factor-β and subsequently binds to SMAD4 to generate a heteromeric complex, which then translocates into the nucleus and aggravates liver fibrosis. Herein, based on molecular docking simulation and structure-activity relationship study, we report the discovery of a novel cyclic peptide CMF9 that targets SMAD2 and potently interferes with the SMAD2-SMAD4 interaction. The subsequent in vivo and in vitro pharmacological studies demonstrated that CMF9 dramatically suppressed hepatic stellate cells activation and collagen synthesis, alleviating CCl4-induced hepatic inflammation and fibrosis. Overall, we first demonstrated that the novel cyclic peptide CMF9 could efficiently block the SMAD2-SMAD4 interaction via selectively inhibiting SMAD2 phosphorylation, providing a promising therapeutic strategy for targeting SMAD2 and an alternative candidate for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Wei Cheng
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yawen Zheng
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Qinglin Tang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Liang Qi
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zihan Shi
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Qihong Yu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Mingmin Li
- College of Life Science, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Xianzhi Wei
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yifeng Zhou
- College of Life Science, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Xianxing Jiang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
5
|
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
|
6
|
Shen R, Xia P, Guo Y, Ji P, Yuan X, Wang L, Shuang S, Zhou L, Tong R, Zhang L, Liu D, Wang D. Effects of polystyrene microparticles exposures on spermatogenic cell differentiation and reproductive endpoints in male mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 373:126200. [PMID: 40185193 DOI: 10.1016/j.envpol.2025.126200] [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: 11/28/2024] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
The widespread distribution of microplastics in the environment has raised concerns about their potential implications for human health. Microplastics accumulate in animals and humans, but the risks associated with these pollutants are not fully understood. This study aimed to investigate the effects of polystyrene microplastics on the male reproductive system. The 0.1 μm polystyrene (PS) could accumulate in the testicular tissue and spermatogonia GC-1, while 1 μm PS was not easy to enter and accumulate in the testicular tissue and cells. Mice continuously exposed for 3-months to 0.1 μm PS demonstrated lower fertility and inhibited spermatogonium differentiation compared to control mice. The 0.1 μm PS were dispersed throughout the seminiferous tubule of the testis. Metabolic reprogramming was found to be involved in these processes. Histone methylation and autophagy-related pathways showed significant differences following PS treatment in testis tissue and GC-1 cells. Our findings suggest that chronic exposure to 0.1 μm PS inhibited spermatogenic cell differentiation and impaired fertility in male mice. We propose that abnormal epigenetic modifications in 0.1 μm PS exposed mice contributed to the dysregulation of glycolytic enzymes, and that the impaired autophagic pathway exacerbated the accumulation of glycolytic enzymes further. Glycolysis plays a critical role in the regulation of spermatogenic cell differentiation, and its regulation partially alleviated the impairments associated with PS exposure. In conclusion, our findings suggest that chronic exposure to nanoplastics PS inhibited spermatogenic cell differentiation and impaired fertility in male mice via disrupted epigenetic modification and metabolic dysregulation.
Collapse
Affiliation(s)
- Rong Shen
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China
| | - Peng Xia
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China
| | - Yanan Guo
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China
| | - Pengfei Ji
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China
| | - Xinyi Yuan
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China
| | - Lu Wang
- The First Hospital of Lanzhou University, Lanzhou University, Gansu, 730000, China
| | - Si Shuang
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China
| | - Liwei Zhou
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China
| | - Ruizhi Tong
- The First Hospital of Lanzhou University, Lanzhou University, Gansu, 730000, China
| | - Lijuan Zhang
- Medical Experimental Center, Lanzhou University, Gansu, 730000, China
| | - Disheng Liu
- The First Hospital of Lanzhou University, Lanzhou University, Gansu, 730000, China.
| | - Degui Wang
- School of Basic Medical Sciences, Lanzhou University, Gansu, 730000, China.
| |
Collapse
|
7
|
Fang L, Peng H, Tan Z, Deng N, Peng X. The Role of Gut Microbiota on Intestinal Fibrosis in Inflammatory Bowel Disease and Traditional Chinese Medicine Intervention. J Inflamm Res 2025; 18:5951-5967. [PMID: 40357383 PMCID: PMC12067688 DOI: 10.2147/jir.s504827] [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: 11/13/2024] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, relapsing inflammatory disorder of the intestine, frequently complicated by intestinal fibrosis. As fibrosis progresses, it can result in luminal stricture and compromised intestinal function, significantly diminishing patients' quality of life. Emerging evidence suggests that gut microbiota and their metabolites contribute to the pathogenesis of IBD-associated intestinal fibrosis by influencing inflammation and modulating immune responses. This review systematically explores the mechanistic link between gut microbiota and intestinal fibrosis in IBD and evaluates the therapeutic potential of traditional Chinese medicine (TCM) interventions. Relevant studies were retrieved from PubMed, Web of Science, Embase, Scopus, CNKI, Wanfang, and VIP databases. Findings indicate that TCM, including Chinese herbal prescriptions and bioactive constituents, can modulate gut microbiota composition and microbial metabolites, ultimately alleviating intestinal fibrosis through anti-inflammatory, immunemodulatory, and anti-fibrotic mechanisms. These insights highlight the potential of TCM as a promising strategy for targeting gut microbiota in the management of IBD-associated fibrosis.
Collapse
Affiliation(s)
- Leyao Fang
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Huiyi Peng
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Zhoujin Tan
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Na Deng
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Xinxin Peng
- The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| |
Collapse
|
8
|
Jin H, Liang G, Huang W, Wang Z, Wu L, Li Y. AGR2 activates the TGF-β/Smad signaling pathway to promote epithelial-mesenchymal transition, invasion, and metastasis in nasopharyngeal carcinoma. Eur Arch Otorhinolaryngol 2025; 282:2411-2418. [PMID: 40119906 DOI: 10.1007/s00405-025-09328-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/03/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Anterior gradient 2 protein (AGR2) is associated with tumorigenesis and metastasis in different cancers. However, its role in nasopharyngeal carcinoma (NPC) remains unknown. This study aimed to explore the effect of AGR2 on epithelial-mesenchymal transition (EMT) in NPC and its underlying mechanisms. METHODS AGR2 expression was analyzed in cancerous and para-cancerous tissues from ten NPC patients using RT-qPCR. Western blotting was used to determine the AGR2 protein levels in two NPC cell lines and a nasopharyngeal epithelial cell line. AGR2 was overexpressed or knocked out in NPC cells and its effects on cell viability, migration, invasion, and EMT markers were evaluated in vitro. RESULT AGR2 expression was significantly higher in NPC tissues compared to adjacent normal tissues. Similarly, NPC cell lines exhibited increased AGR2 levels compared to the nasopharyngeal epithelial cell line. AGR2 knockout significantly reduced cell viability, migration, and invasion. It also decreased N-cadherin protein levels while increasing E-cadherin, α-SMA, and vimentin expression. Conversely, AGR2 overexpression produced the opposite effects. Furthermore, AGR2 deletion inactivated the TGF-β/Smad signaling pathway. CONCLUSION AGR2 promotes tumor progression and EMT in NPC through activation of the TGF-β/Smad signaling pathway. These findings suggest that AGR2 may serve as a potential biomarker and therapeutic target for NPC.
Collapse
Affiliation(s)
- Hui Jin
- Department of Otolaryngology, Wuhan Third Hospital, No. 241 Pengliuyang Road, Wuchang District, Wuhan City, 430000, Hubei Province, China
| | - Gengtian Liang
- Department of Otolaryngology, Wuhan Third Hospital, No. 241 Pengliuyang Road, Wuchang District, Wuhan City, 430000, Hubei Province, China.
| | - Wenxia Huang
- Department of Otolaryngology, Wuhan Third Hospital, No. 241 Pengliuyang Road, Wuchang District, Wuhan City, 430000, Hubei Province, China
| | - Zhen Wang
- Department of Otolaryngology, Wuhan Third Hospital, No. 241 Pengliuyang Road, Wuchang District, Wuhan City, 430000, Hubei Province, China
| | - Longjun Wu
- Department of Otolaryngology, Wuhan Third Hospital, No. 241 Pengliuyang Road, Wuchang District, Wuhan City, 430000, Hubei Province, China
| | - Yaping Li
- Department of Cardiology, Wuhan Third Hospital, Wuhan, 430000, Hubei Province, China
| |
Collapse
|
9
|
Jing H, Gao Y, Jing L, Yang H, Liu S. Recent advances in therapeutic use of transforming growth factor-beta inhibitors in cancer and fibrosis. Front Oncol 2025; 15:1489701. [PMID: 40352593 PMCID: PMC12061708 DOI: 10.3389/fonc.2025.1489701] [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: 09/01/2024] [Accepted: 04/03/2025] [Indexed: 05/14/2025] Open
Abstract
Transforming growth factor-beta (TGF-β) has long been known to be associated with early embryonic development and organogenesis, immune supervision, and tissue repair and homeostasis in adults. TGF-β has complex roles in fibrosis and cancer that may be opposing at different stages of these diseases. Under pathological conditions, overexpression of TGF-β causes epithelial-mesenchymal transition, deposition of extracellular matrix, and formation of cancer-associated fibroblasts, leading to fibrotic disease or cancer. Fibroblasts, epithelial cells, and immune cells are the most common targets of TGF-β, while fibrosis and cancer are the most common TGF-β-associated diseases. Given the critical role of TGF-β and its downstream molecules in fibrosis and progression of cancer, therapies targeting TGF-β signaling appear to be a promising strategy. Preclinical and clinical studies have investigated therapies targeting TGF-β, including antisense oligonucleotides, monoclonal antibodies, and ligand traps. However, development of targeted TGF-β therapy has been hindered by systemic cytotoxicity. This review discusses the molecular mechanisms of TGF-β signaling and highlights targeted TGF-β therapy for cancer and fibrosis as a therapeutic strategy for related diseases.
Collapse
Affiliation(s)
- Hanhui Jing
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yan Gao
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Linyuan Jing
- Department of Integrated Chinese and Western Medicine, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Hanyu Yang
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shanglong Liu
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
10
|
Luo L, Yang X, Zhao H, Wang L, Li W, Zhang Y. High expression of ITGB3 ameliorates asthma by inhibiting epithelial-mesenchymal transformation through suppressing the activation of NF-kB pathway. Sci Rep 2025; 15:13837. [PMID: 40263524 PMCID: PMC12015361 DOI: 10.1038/s41598-025-98842-y] [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: 12/16/2024] [Accepted: 04/15/2025] [Indexed: 04/24/2025] Open
Abstract
Integrin β3 (ITGB3) has been identified as an asthma-associated gene; however, its molecular mechanisms remain poorly understood. Epithelial-mesenchymal transition (EMT) is a critical driver of airway remodeling in asthma, which underpins disease progression. This study aimed to elucidate the role of ITGB3 in asthma pathogenesis by investigating its regulation of EMT. Asthma models were established in vivo using C57BL/6 mice and in vitro with A549 cells, both exposed to house dust mite (HDM) extract. The effects of HDM and ITGB3 modulation on cellular viability, apoptosis, and inflammatory cytokines (IL-4, IL-5, IL-13) were assessed in cultured cells and murine lungs. EMT was evaluated via western blot analysis of E-cadherin, N-cadherin, and vimentin expression. The NF-κB pathway was examined by quantifying phosphorylated p65 and IkBa levels. Lung tissue pathology and ITGB3 expression were assessed using hematoxylin and eosin (H&E) staining and immunohistochemistry. Results demonstrated that HDM exposure reduced A549 cell viability, increased cytotoxicity, apoptosis, and pro-inflammatory cytokine production, while promoting EMT. ITGB3 knockdown exacerbated these effects, whereas ITGB3 overexpression mitigated them. Furthermore, HDM activated the NF-κB pathway, an effect reversed by ITGB3 overexpression. In HDM-challenged cells, NF-κB activation via an agonist counteracted the protective effects of ITGB3 overexpression on apoptosis, inflammation, and EMT. Notably, ITGB3 overexpression suppressed inflammation, EMT, and pathological remodeling in asthmatic mice. Collectively, our findings reveal that ITGB3 exerts protective effects in asthma by inhibiting EMT through suppression of the NF-κB signaling pathway, thereby identifying ITGB3 as a potential therapeutic target for asthma management.
Collapse
Affiliation(s)
- Lu Luo
- Department of Emergency, Jinan Children's Hospital, No.23976, Jingshi Road, Jinan, 250022, Shandong, China
| | - Xiaoshan Yang
- Department of Rheumatology And Immunology, Binzhou People's Hospital, Binzhou, China
| | - Haitao Zhao
- Department of Hematology, Binzhou People's Hospital, Binzhou, China
| | - Lingling Wang
- Department of Emergency, Jinan Children's Hospital, No.23976, Jingshi Road, Jinan, 250022, Shandong, China
| | - Wengang Li
- Department of Emergency, Jinan Children's Hospital, No.23976, Jingshi Road, Jinan, 250022, Shandong, China
| | - Yan Zhang
- Department of Emergency, Jinan Children's Hospital, No.23976, Jingshi Road, Jinan, 250022, Shandong, China.
| |
Collapse
|
11
|
Chen ZY, Ma MM, Wang R, Zhang QQ, Xie ML, Wang YL, Guo YX, Liu K, Cao LF, He FL, Fu L, Jiang YL. Gui-zhi-fu-ling-wan alleviates bleomycin-induced pulmonary fibrosis through inhibiting epithelial-mesenchymal transition and ferroptosis. Front Pharmacol 2025; 16:1552251. [PMID: 40308766 PMCID: PMC12041222 DOI: 10.3389/fphar.2025.1552251] [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: 12/27/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) has a higher morbidity and poor prognosis. Gui-Zhi-Fu-Ling-Wan (GFW) is a traditional Chinese herbal formula which exerts anti-inflammatory and anti-oxidative effects. The goal was to determine the protective effect of GFW on bleomycin (BLM)-induced pulmonary fibrosis. Methods One hundred and twenty-four mice were randomly divided into eight groups, and orally supplemented with GFW (1 g/kg) in 1 week ago and continuing to 1 week later of single BLM intratracheal injection (5.0 mg/kg). Lung tissues were collected in 7 days and 21 days after BLM injection. BEAS-2B cells were pretreated with GFW (100 μg/mL) for three consecutive days before BLM (10 μg/mL) exposure. Cells were harvested in 12 or 24 h after BLM co-culture. Results GFW supplementation alleviated BLM-induced alveolar structure destruction and inflammatory cell infiltration in mice lungs. BLM-incurred collagen deposition was attenuated by GFW. In addition, GFW pretreatment repressed BLM-evoked downregulation of E-cadherin, and elevation of N-cadherin and Vimentin in mouse lungs. Besides, BLM-excited GPX4 reduction, ferritin increases, lipid peroxidation, and free iron overload were significantly relieved by GFW pretreatment in mouse lungs and BEAS-2B cells. Notably, BLM-provoked mitochondrial reactive oxygen species (mtROS) excessive production, elevation of mitochondrial stress markers, such as HSP70 and CLPP, and mitochondrial injury, were all abolished in mouse lungs and BEAS-2B cells by GFW pretreatment. Conclusion GFW supplementation attenuated BLM-evoked lung injury and pulmonary fibrosis partially through repressing EMT and mtROS-mediated ferroptosis in pulmonary epithelial cells.
Collapse
Affiliation(s)
- Zi-Yong Chen
- The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
| | - Meng-Meng Ma
- The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
| | - Rui Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qing-Qing Zhang
- The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
| | - Mei-Ling Xie
- Department of Respiratory and Critical Care Medicine, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
| | - Ying-Li Wang
- Department of Respiratory and Critical Care Medicine, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
| | - Yong-Xia Guo
- The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
| | - Kui Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
| | - Li-Fang Cao
- Department of Respiratory and Critical Care Medicine, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
| | - Feng-Lian He
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lin Fu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ya-Lin Jiang
- Department of Respiratory and Critical Care Medicine, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
| |
Collapse
|
12
|
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
|
13
|
Khalili-Tanha G, Radisky ES, Radisky DC, Shoari A. Matrix metalloproteinase-driven epithelial-mesenchymal transition: implications in health and disease. J Transl Med 2025; 23:436. [PMID: 40217300 PMCID: PMC11992850 DOI: 10.1186/s12967-025-06447-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a process in which epithelial cells, defined by apical-basal polarity and tight intercellular junctions, acquire migratory and invasive properties characteristic of mesenchymal cells. Under normal conditions, EMT directs essential morphogenetic events in embryogenesis and supports tissue repair. When dysregulated, EMT contributes to pathological processes such as organ fibrosis, chronic inflammation, and cancer progression and metastasis. Matrix metalloproteinases (MMPs)-a family of zinc-dependent proteases that degrade structural components of the extracellular matrix-sit at the nexus of this transition by dismantling basement membranes, activating pro-EMT signaling pathways, and cleaving adhesion molecules. When normally regulated, MMPs promote balanced ECM turnover and support the cyclical remodeling necessary for proper development, wound healing, and tissue homeostasis. When abnormally regulated, MMPs drive excessive ECM turnover, thereby promoting EMT-related pathologies, including tumor progression and fibrotic disease. This review provides an integrated overview of the molecular mechanisms by which MMPs both initiate and sustain EMT under physiological and disease conditions. It discusses how MMPs can potentiate EMT through TGF-β and Wnt/β-catenin signaling, disrupt cell-cell junction proteins, and potentiate the action of hypoxia-inducible factors in the tumor microenvironment. It discusses how these pathologic processes remodel tissues during fibrosis, and fuel cancer cell invasion, metastasis, and resistance to therapy. Finally, the review explores emerging therapeutic strategies that selectively target MMPs and EMT, ranging from CRISPR/Cas-mediated interventions to engineered tissue inhibitors of metalloproteinases (TIMPs), and demonstrates how such approaches may suppress pathological EMT without compromising its indispensable roles in normal biology.
Collapse
Affiliation(s)
- Ghazaleh Khalili-Tanha
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Alireza Shoari
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
14
|
Li L, Sun S, Lai X, Li Q, Chen R, Zhang Z, Hao M, Zhang S, Sun L, Li D. Mechanism of Green Tea Peptides in Lowering Blood Pressure and Alleviating Renal Injury Induced by Hypertension Through the Ang II/TGF-β1/SMAD Signaling Pathway. Nutrients 2025; 17:1300. [PMID: 40284165 PMCID: PMC12030635 DOI: 10.3390/nu17081300] [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: 03/08/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: The kidney plays a crucial role in regulating normal blood pressure and is one of the major organs affected by hypertension. The present study aimed to investigate the hypotensive and renoprotective effects of four specific green tea peptides extracted from green tea dregs on spontaneously hypertensive rats (SHRs) and to investigate the underlying mechanisms. Methods: Four specific green tea peptides (40 mg/kg) were gavaged to SHRs for 4 weeks, and blood pressure, renal function, renal pathological changes, renal tissue fibrosis indexes, and inflammation indexes were examined in SHRs to analyze the role of the four green tea peptides in alleviating hypertension and its renal injury. Results: The results showed that the four TPs significantly reduced systolic and diastolic blood pressure (20-24% and 18-28%) in SHR compared to the model group. Meanwhile, gene levels and protein expression of renal fibrosis-related targets such as phospho-Smad2/3 (p-Smad2/3) (26-47%), Sma- and Mad-related proteins 2/3 (Smad2/3) (19-38%), transforming growth factor-β1 (TGF-β1) (36-63%), and alpha-smooth muscle actin (alpha-SMA) (58-86%) were also significantly reduced. In addition, the reduced expression levels of medullary differentiation factor 88 (MyD88) (14-36%), inducible nitric oxide synthase (iNOS) (58-73%), and nuclear factor-κB p65 (NF-kB p65) (35-78%) in kidneys also confirmed that TPs attenuated renal inflammation in SHR. Therefore, green tea peptides could attenuate the fibrosis and inflammatory responses occurring in hypertensive kidneys by inhibiting the Ang II/TGF-β1/SMAD signaling pathway and MyD88/NF-κB p65/iNOS signaling pathway. Conclusions: The results showed that green tea peptides may be effective candidates for lowering blood pressure and attenuating kidney injury.
Collapse
Affiliation(s)
- Lulu Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China;
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Key Laboratory of Tea Resources Innovation & Utilization, Guangzhou 510640, China; (S.S.); (X.L.); (Q.L.); (R.C.); (Z.Z.); (M.H.); (S.Z.)
| | - Shili Sun
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Key Laboratory of Tea Resources Innovation & Utilization, Guangzhou 510640, China; (S.S.); (X.L.); (Q.L.); (R.C.); (Z.Z.); (M.H.); (S.Z.)
| | - Xingfei Lai
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Key Laboratory of Tea Resources Innovation & Utilization, Guangzhou 510640, China; (S.S.); (X.L.); (Q.L.); (R.C.); (Z.Z.); (M.H.); (S.Z.)
| | - Qiuhua Li
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Key Laboratory of Tea Resources Innovation & Utilization, Guangzhou 510640, China; (S.S.); (X.L.); (Q.L.); (R.C.); (Z.Z.); (M.H.); (S.Z.)
| | - Ruohong Chen
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Key Laboratory of Tea Resources Innovation & Utilization, Guangzhou 510640, China; (S.S.); (X.L.); (Q.L.); (R.C.); (Z.Z.); (M.H.); (S.Z.)
| | - Zhenbiao Zhang
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Key Laboratory of Tea Resources Innovation & Utilization, Guangzhou 510640, China; (S.S.); (X.L.); (Q.L.); (R.C.); (Z.Z.); (M.H.); (S.Z.)
| | - Mengjiao Hao
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Key Laboratory of Tea Resources Innovation & Utilization, Guangzhou 510640, China; (S.S.); (X.L.); (Q.L.); (R.C.); (Z.Z.); (M.H.); (S.Z.)
| | - Suwan Zhang
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Key Laboratory of Tea Resources Innovation & Utilization, Guangzhou 510640, China; (S.S.); (X.L.); (Q.L.); (R.C.); (Z.Z.); (M.H.); (S.Z.)
| | - Lingli Sun
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Key Laboratory of Tea Resources Innovation & Utilization, Guangzhou 510640, China; (S.S.); (X.L.); (Q.L.); (R.C.); (Z.Z.); (M.H.); (S.Z.)
| | - Dongli Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China;
- International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
| |
Collapse
|
15
|
Park BS, Bang E, Lee H, Kim GY, Choi YH. Tagetes erecta Linn flower extract inhibits particulate matter 2.5-promoted epithelial-mesenchymal transition by attenuating reactive oxygen species generation in human retinal pigment epithelial ARPE-19 cells. Nutr Res Pract 2025; 19:170-185. [PMID: 40226757 PMCID: PMC11982690 DOI: 10.4162/nrp.2025.19.2.170] [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: 09/04/2024] [Revised: 10/15/2024] [Accepted: 11/12/2024] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Particulate matter 2.5 (PM2.5) exposure can promote epithelial-mesenchymal transition (EMT) in human retinal pigment epithelial (RPE) cells. The flowers of Tagetes erecta Linn, commonly known as marigold, are rich in diverse flavonoids and carotenoids and play a significant role in preventing cellular damage induced by oxidative stress, but the role of their extracts in RPE cells has not been reported. This study aimed to evaluate the influence of an ethanol extract of T. erecta Linn flower (TE) on PM2.5-induced EMT processes in RPE ARPE-19 cells. MATERIALS/METHODS To investigate the protective effect of TE against ARPE-19 cell damage following PM2.5 treatment, cells were exposed to TE for 1 h before exposure to PM2.5 for 24 h. We investigated whether the efficacy of TE on suppressing PM2.5-induced EMT was related to antioxidant activity and the effect on the expression changes of factors involved in EMT regulation. Additionally, we further explored the role of intracellular signaling pathways associated with EMT inhibition. RESULTS TE significantly blocked PM2.5-induced cytotoxicity while effectively preventing mitochondrial dysfunction, increased reactive oxygen species (ROS) generation, and mitochondrial membrane potential disruption. TE inhibited PM2.5-induced EMT and inflammatory response by suppressing the ROS-mediated transforming growth factor-β/suppressor of mothers against decapentaplegic/mitogen-activated protein kinases signaling pathway. CONCLUSION Our results suggest that marigold extract is a highly effective in protection against PM2.5-induced eye damage.
Collapse
Affiliation(s)
- Beom Su Park
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Korea
| | - EunJin Bang
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Korea
| | - Hyesook Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju 63243, Korea
| | - Yung Hyun Choi
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Korea
| |
Collapse
|
16
|
Lu Q, Liu J, Xiong Y, Jian J, Wang J, Chen Z, Wan S, Liu X, Wang L. Cyanidin-3-glucoside upregulated NDRG2 through the PI3K/AKT pathway to alleviate EMT and ECM in renal fibrosis. Sci Rep 2025; 15:10695. [PMID: 40155416 PMCID: PMC11953473 DOI: 10.1038/s41598-025-94918-x] [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: 11/09/2024] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
Renal fibrosis is a critical progression of chronic kidney disease, and epithelial-to-mesenchymal transition (EMT) and extracellular matrix(ECM) deposition are crucial pathologic change of renal fibrosis, which still lacks of effective treatment. In this study, it was found that cyanidin-3-O-glucoside (C3G) could inhibit EMT and ECM activated by unilateral ureteral obstruction (UUO) and transforming growth factor-β1 (TGF-β1) stimulation. Moreover, N-Myc downstream-regulated gene 2(NDRG2), which involved in the progression of renal fibrosis, was down-regulated in vivo and in vitro model. However, C3G pretreatment could reverse the reductive expression of NDRG2. Furthermore, we found that the combined treatment of C3G and si-NDRG2 could reverse the decreased EMT and ECM, which induced by C3G treatment only. And the activation of Phosphatidylinositol 3-kinase (PI3K)/ Protein Kinase B (AKT) pathway significantly enhanced EMT and ECM, which was decreased by C3G treatment only in TGF-β1 induced Human Kidney 2 (HK-2) cells. In conclusion, our results demonstrated that C3G alleviated EMT and ECM by elevating NDRG2 expression through the PI3K/AKT pathway, indicating that C3G could be a potential treatment against renal fibrosis.
Collapse
Affiliation(s)
- Qianxue Lu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jin Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yufeng Xiong
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jun Jian
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jingsong Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Shanshan Wan
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
17
|
Zhang J, Xie X, Li Y, Wang H, Zhang L, Shi P, Wei J, Zhang L, Lu Y, Cui L, Liu X, Liang X. Emo@KP MBs Modulates the TGF-β1/Smad Signaling Pathway by in situ Micro-Nano Conversion to Reduce Renal Inflammation and Fibrosis Caused by Unilateral Ureteral Obstruction. Int J Nanomedicine 2025; 20:3731-3747. [PMID: 40162332 PMCID: PMC11951927 DOI: 10.2147/ijn.s499550] [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: 10/07/2024] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Emodin alleviates renal interstitial fibrosis (RIF) and reduces inflammation by inhibiting the TGF-β1/Smad pathway, thus improving CKD outcomes. However, its clinical use is limited due to poor solubility and side effects. This study developed a targeted drug delivery system using αKIM-1 modified microbubbles carrying Emodin to enhance accumulation in renal tissues with high KIM-1 expression. Methods Emo@KP MBs were characterized by TEM and DLS, and their drug loading and encapsulation rates were measured by UV-VIS-NIR spectroscopy. Biocompatibility was assessed in vitro with HK-2 cells and in vivo via hematological and pathological markers. Contrast-enhanced ultrasound (CEUS) and fluorescence imaging were used for real-time visualization of treatment. Therapeutic experiments were performed on a unilateral ureteral obstruction (UUO) mouse model treated with Emo@KP MBs + US on days 1 and 3 post-surgery. Renal function, cytokine levels, and histological analysis were detected to evaluate therapeutic effects. Results Emo@KP MBs exhibited spherical structures (2 ~ 4 μm) with good stability. Ultrasound targeted microbubble destruction (UTMD) enabled controlled release of Emodin. CEUS and fluorescence imaging showed enhanced drug accumulation in diseased kidneys. In the UUO + Emo@KP MBs/US group, renal function was improved, inflammatory cytokines (IL-1β, TNF-α) were decreased, and renal lesions and collagen deposition were reduced. Immunohistochemistry revealed the downregulation of TGF-β, Smad2/3, and α-SMA, and upregulation of E-cadherin. Conclusion Emo@KP MBs enhanced drug delivery efficiency and therapeutic efficacy through αKIM-1 targeting and UTMD, while providing real-time imaging capabilities, suggesting good potential as a therapeutic approach to reduce renal inflammation and fibrosis in UUO.
Collapse
Affiliation(s)
- Jinxia Zhang
- Department of Ultrasound, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Xinxin Xie
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yuanjing Li
- Department of Ultrasound, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
| | - Haonan Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Lijuan Zhang
- Department of Ultrasound, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
| | - Peiqi Shi
- Department of Ultrasound, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
| | - Jing Wei
- Department of Ultrasound, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
| | - Ling Zhang
- Department of Ultrasound, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
| | - Yingdong Lu
- Department of Ultrasound, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
| | - Ligang Cui
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Xiaoning Liu
- Department of Ultrasound, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| |
Collapse
|
18
|
Zhao Z, Zhu Y, Wan D. Exercise and tissue fibrosis: recent advances in therapeutic potential and molecular mechanisms. Front Endocrinol (Lausanne) 2025; 16:1557797. [PMID: 40182630 PMCID: PMC11965137 DOI: 10.3389/fendo.2025.1557797] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Tissue fibrosis represents an aberrant repair process, occurring because of prolonged injury, sustained inflammatory response, or metabolic disorders. It is characterized by an excessive accumulation of extracellular matrix (ECM), resulting in tissue hardening, structural remodeling, and loss of function. This pathological phenomenon is a common feature in the end stage of numerous chronic diseases. Despite the advent of novel therapeutic modalities, including antifibrotic agents, these have only modest efficacy in reversing established fibrosis and are associated with adverse effects. In recent years, a growing body of research has demonstrated that exercise has significant benefits and potential in the treatment of tissue fibrosis. The anti-fibrotic effects of exercise are mediated by multiple mechanisms, including direct inhibition of fibroblast activation, reduction in the expression of pro-fibrotic factors such as transforming growth factor-β (TGF-β) and slowing of collagen deposition. Furthermore, exercise has been demonstrated to assist in maintaining the dynamic equilibrium of tissue repair, thereby indirectly reducing tissue damage and fibrosis. It can also help maintain the dynamic balance of tissue repair by improving metabolic disorders, exerting anti-inflammatory and antioxidant effects, regulating cellular autophagy, restoring mitochondrial function, activating stem cell activity, and reducing cell apoptosis, thereby indirectly alleviating tissue. This paper presents a review of the therapeutic potential of exercise and its underlying mechanisms for the treatment of a range of tissue fibrosis, including cardiac, pulmonary, renal, hepatic, and skeletal muscle. It offers a valuable reference point for non-pharmacological intervention strategies for the comprehensive treatment of fibrotic diseases.
Collapse
Affiliation(s)
- Zheng Zhao
- School of Physical Education, Anyang Normal University, Anyang, Henan, China
| | - Yongjia Zhu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Dongfeng Wan
- School of Health, Shanghai Normal University Tianhua College, Shanghai, China
| |
Collapse
|
19
|
Choudhury J, Richardson LS, Urrabaz-Garza R, Jacob J, Kammala AK, Menon R. Chorionic trophoblast cells demonstrate functionally different phenotypes from placental trophoblasts†. Biol Reprod 2025; 112:530-539. [PMID: 39756436 DOI: 10.1093/biolre/ioaf003] [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: 08/01/2024] [Revised: 10/09/2024] [Accepted: 01/03/2025] [Indexed: 01/07/2025] Open
Abstract
Chorionic trophoblast cells are one of the principal components of the fetal membrane and join with the decidua to form a feto-maternal interface. Recent success in isolating chorionic trophoblast cells dealt with two separate questions: (i) the necessity of highly enriched and defined media with inhibitors of oxidative stress and cell transition and their impact on growth and trophoblast phenotype, (ii) the functional differences between chorionic trophoblast cells and other placental trophoblast lineages of cells (placental cytotrophoblast cells, and extravillous trophoblast). Chorionic trophoblast cells were cultured either in defined media with various inhibitors or in media from which inhibitors were removed individually. Cellular morphology and growth (microscopy and crystal violet staining) and cellular and molecular biological features (immunofluorescence staining for GATA-binding protein 3, cytokeratin 7, and vimentin) were assessed. Syncytialization of cells (forskolin treatment) and invasive properties of chorionic trophoblast cells (cell invasion assay) were tested and compared with placental cytotrophoblast cells and extravillous trophoblasts (HTR8/SVneo), respectively. Removal of various growth-supporting agents from the media delayed cell growth and inclined towards cellular transition (increase in vimentin compared to cytokeratin 7 or GATA-binding protein 3) compared to chorionic trophoblast cells grown in complete and enriched media. The chorionic trophoblast cells failed to syncytialize, contrasting with the high levels of membrane fusion observed in placental cytotrophoblast cells. Although chorionic trophoblast cells express human leukocyte antigen G like extravillous trophoblasts, they do not invade. Chorionic trophoblast cells require several specific constituents for in vitro growth and phenotype maintenance. Chorionic trophoblast cells are trophoblast lineage cells that barricade immune cell-enriched decidua without invading them. These properties support their location and function, which are distinct from placental cytotrophoblast cells and extravillous trophoblasts.
Collapse
Affiliation(s)
- Jaganmoy Choudhury
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States of America
| | - Lauren S Richardson
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States of America
| | - Rheanna Urrabaz-Garza
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States of America
| | - Jeena Jacob
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States of America
| | - Ananth Kumar Kammala
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States of America
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States of America
| |
Collapse
|
20
|
Liu J, Wang S, Tan G, Tong B, Wu Y, Zhang L, Jiang B. Chitosan-Artesunate nanoparticles: A dual anti-fibrotic and anti-inflammatory strategy for preventing bleb fibrosis post-glaucoma filtration surgery. Drug Deliv Transl Res 2025:10.1007/s13346-025-01819-7. [PMID: 40019651 DOI: 10.1007/s13346-025-01819-7] [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] [Accepted: 02/13/2025] [Indexed: 03/01/2025]
Abstract
Glaucoma filtration surgery (GFS) effectively lowers intraocular pressure in glaucoma patients, but postoperative bleb fibrosis often leads to surgical failure. Artesunate (ART) has demonstrated antifibrotic potential; however, its clinical use is limited by poor solubility and rapid degradation. This study aimed to develop chitosan-ART nanoparticles (CS@ART NPs) to improve ART's therapeutic efficacy in preventing bleb fibrosis. CS@ART NPs were synthesized using an ionic gelation method for chitosan encapsulation. Their characterization, including analyses of morphology, hydrodynamic properties, surface charge, encapsulation efficiency, drug release kinetics, stability, chemical structure, and mucoadhesive interactions, was carried out using various techniques such as TEM, DLS, zeta potential analysis, HPLC, FT-IR, 1H-NMR, and adhesion assays. The antifibrotic effects were evaluated in a rabbit GFS model through subconjunctival injection. Histological analysis as well as immunohistochemistry for fibrosis markers α-SMA and fibronectin were detected. In vitro studies were conducted using human primary ocular fibroblasts stimulated with TGF-β1 to assess anti-inflammatory and anti-proliferative effects, measured by EdU incorporation, Western blot for signaling pathway components, and cytokine expression. CS@ART NPs exhibited a uniform size distribution (135.73 ± 0.90 nm), stable dispersion, high encapsulation efficiency (86.4%), and sustained drug release. In the GFS model, a single subconjunctival injection of CS@ART significantly reduced collagen deposition, as well as α-SMA and fibronectin expression at the surgical site. In vitro, CS@ART demonstrated superior antifibrotic effects with a significantly lower IC50 for inhibiting fibroblast proliferation compared to ART alone. Mechanically, CS@ART suppressed the Cyclin D1-CDK4/6, TGF-β1/SMAD, and PI3K/Akt signaling pathways. Additionally, CS@ART showed marked anti-inflammatory effects, reducing inflammatory cell infiltration and IL-6 expression. CS@ART NPs play a dual role both alleviate bleb fibrosis and inflammation after GFS as a promising therapeutic strategy for improving surgical outcomes in glaucoma patients.
Collapse
Affiliation(s)
- Jingyuan Liu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China
| | - Shutong Wang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China
| | - Guangshuang Tan
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China
| | - Boding Tong
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China
| | - Ying Wu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China
| | - Lusi Zhang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China.
| | - Bing Jiang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China.
| |
Collapse
|
21
|
JIANG DONG, QI ZHI, XU ZHIYING, LI YIRAN. CYB5D2 inhibits the malignant progression of hepatocellular carcinoma by inhibiting TGF-β expression and epithelial-mesenchymal transition. Oncol Res 2025; 33:709-722. [PMID: 40109873 PMCID: PMC11915040 DOI: 10.32604/or.2024.050125] [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: 01/28/2024] [Accepted: 06/19/2024] [Indexed: 03/22/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a prevalent liver malignancy. This study examined the roles of transforming growth factor beta (TGF-β) and cytochrome b5 domain containing 2 (CYB5D2) in HCC etiology and their prognostic biomarker potential. Methods Key modules and prognostic genes were identified by analyzing the GSE101685 dataset by weighted gene co-expression network analysis (WGCNA) and Least absolute shrinkage and selection operator (LASSO) Cox regression. The expression levels of CYB5D2 and TGF-β in HCC cell lines were quantified using Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blotting (WB) assays. Effects of CYB5D2 overexpression on cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) marker regulation were assessed in vitro, while in vivo tumorigenicity was evaluated using a xenograft model of HCC in nude mice. Results In this study, WGCNA identified the turquoise module as significantly associated with HCC, containing 452 DEGs. LASSO Cox regression analysis revealed 9 key prognostic genes, with CYB5D2 being underexpressed in HCC cells and tissues. TGF-β was negatively correlated with CYB5D2 expression, resulting in poor patient prognosis. Functional assays demonstrated that CYB5D2 overexpression inhibited proliferation, migration, and invasion of HCC cell lines, and altered EMT marker expression. Furthermore, the addition of TGF-β partially reversed the suppressive effects caused by CYB5D2 overexpression. In vivo, CYB5D2 overexpression significantly reduced tumor growth, indicating its potential as a therapeutic target for HCC. Conclusion The tumor suppressor function of CYB5D2 in HCC and its interaction with TGF-β offered fresh information on the molecular pathophysiology of HCC and possible treatment avenues.
Collapse
Affiliation(s)
- DONG JIANG
- Department of Ultrasound, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - ZHI QI
- Department of Neurology, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - ZHIYING XU
- Department of Hepatic Surgery IV, Shanghai Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - YIRAN LI
- Department of Ultrasound, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
22
|
Wu N, Xue C, Yu S, Wang Y, Sun D, Ye Q. CC-chemokine ligand 18, CXC motif chemokine 13 and osteopontin as biomarkers of silicosis and asbestosis: a prospective observational study. Sci Rep 2025; 15:6819. [PMID: 40000810 PMCID: PMC11861896 DOI: 10.1038/s41598-025-91423-z] [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: 12/18/2024] [Accepted: 02/20/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Silicosis and asbestosis, distinct forms of pneumoconiosis, manifest progressive interstitial fibrosis due to exposure to silica dust or asbestos fibers. This study aimed to identify potential biomarkers for diagnosing silicosis and asbestosis, while also evaluating disease severity and prognosis. We undertook an prospective observational study involving patients with silicosis or asbestosis. The correlation between baseline CC-chemokine ligand 18 (CCL18), CXC motif chemokine 13 (CXCL13), osteopontin (OPN), periostin, and fibulin-3 and clinical variables was analyzed. Diagnostic sensitivity was evaluated using receiver operating characteristic curves, and correlations between baseline biomarker levels and disease severity were analyzed. Multivariable Cox regression assessed the baseline concentrations' strength in predicting all-cause mortality for silicosis and asbestosis. Of 231 silicosis and 163 asbestosis included in the study, 29 silicosis (12.6%) and 28 (17.2%) asbestosis died within the five years follow-up period. Elevated baseline concentrations of CCL18, CXCL13, and OPN were observed in 231 silicosis patients and 163 asbestosis patients compared to 118 HCs. Diagnostic accuracy for silicosis or asbestosis, in order, was CCL18, OPN, and CXCL13. Combining CCL18, OPN, and CXCL13 enhanced diagnostic accuracy. In silicosis patients, these concentrations were significantly associated with lung function values. However, these biomarkers were not the risk factor for all-cause mortality. CCL18, CXCL13, and OPN stand out as promising biomarkers for diagnosing silicosis and asbestosis. Meanwhile, CCL18, CXCL13, and OPN may be used for the evaluation of silicosis conditions.
Collapse
Affiliation(s)
- Na Wu
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chaoyang District, Beijing, 100020, China
| | - Changjiang Xue
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chaoyang District, Beijing, 100020, China
| | - Shiwen Yu
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chaoyang District, Beijing, 100020, China
| | - Yuanying Wang
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chaoyang District, Beijing, 100020, China
| | - Di Sun
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chaoyang District, Beijing, 100020, China
| | - Qiao Ye
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Workers' Stadium South Road, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
23
|
Luo P, Hong H, Zhang B, Li J, Zhang S, Yue C, Cao J, Wang J, Dai Y, Liao Q, Xu P, Yang B, Liu X, Lin X, Yu Y, Feng XH. ERBB4 selectively amplifies TGF-β pro-metastatic responses. Cell Rep 2025; 44:115210. [PMID: 39854208 DOI: 10.1016/j.celrep.2024.115210] [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: 06/19/2024] [Revised: 11/19/2024] [Accepted: 12/24/2024] [Indexed: 01/26/2025] Open
Abstract
Transforming growth factor β (TGF-β) is well known to play paradoxical roles in tumorigenesis as it has both growth-inhibitory and pro-metastatic effects. However, the underlying mechanisms of how TGF-β drives the opposing responses remain largely unknown. Here, we report that ERBB4, a member of the ERBB receptor tyrosine kinase family, specifically promotes TGF-β's metastatic response but not its anti-growth response. ERBB4 directly phosphorylates Tyr162 in the linker region of SMAD4, which enables SMAD4 to achieve a higher DNA-binding ability and potentiates TGF-β-induced gene transcription associated with epithelial-to-mesenchymal transition (EMT), cell migration, and invasion without affecting the genes involved in growth inhibition. These selective effects facilitate lung cancer metastasis in mouse models. This discovery sheds light on the previously unrecognized role of SMAD4 as a substrate of ERBB4 and highlights the selective involvement of the ERBB4-SMAD4 regulatory axis in tumor metastasis.
Collapse
Affiliation(s)
- Peihong Luo
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang 321000, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Huanyu Hong
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang 321000, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Baoling Zhang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang 321000, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jie Li
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang 321000, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shuyi Zhang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang 321000, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Chaomin Yue
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jin Cao
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang 321000, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jia Wang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang 321000, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yuhan Dai
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang 321000, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qingqing Liao
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Pinglong Xu
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Bing Yang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xia Liu
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, Zhejiang 311215, China
| | - Xia Lin
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yi Yu
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang 321000, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Xin-Hua Feng
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang 321000, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China; The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
24
|
Yu J, Li Y, Li Y, Liu X, Huo Q, Wu N, Zhang Y, Zeng T, Zhang Y, Li HY, Lian J, Zhou J, Moses EJ, Geng J, Lin J, Li W, Zhu X. Phosphorylation of FOXN3 by NEK6 promotes pulmonary fibrosis through Smad signaling. Nat Commun 2025; 16:1865. [PMID: 39984467 PMCID: PMC11845461 DOI: 10.1038/s41467-025-56922-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025] Open
Abstract
The transcriptional repressor FOXN3 plays a key role in regulating pulmonary inflammatory responses, which are crucial in the development of pulmonary fibrosis. However, its specific regulatory function in lung fibrosis remains unclear. Here, we show that FOXN3 suppresses pulmonary fibrosis by inhibiting Smad transcriptional activity. FOXN3 targets a substantial number of Smad response gene promoters, facilitating Smad4 ubiquitination, which disrupts the association of the Smad2/3/4 complex with chromatin and abolishes its transcriptional response. In response to pro-fibrotic stimuli, NEK6 phosphorylates FOXN3 at S412 and S416, leading to its degradation. The loss of FOXN3 inhibits β-TrCP-mediated ubiquitination of Smad4, stabilizing the Smad complex's association with its responsive elements and promoting transcriptional activation, thus contributing to the development of pulmonary fibrosis. Notably, we found a significant inverse expression pattern between FOXN3 and Smad4 in clinical pulmonary fibrosis cases, underscoring the importance of the NEK6-FOXN3-Smad axis in the pathological process of pulmonary fibrosis.
Collapse
Affiliation(s)
- Jinjin Yu
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Bengbu Medical University, Bengbu, China
- Regenerative Medicine Sciences Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
- Molecular Diagnosis Center, First Affiliated Hospital, Bengbu Medical University, Bengbu, China
| | - Yingke Li
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Yiming Li
- Research Center of Clinical Laboratory Science, School of Laboratory Medicine, Bengbu Medical University, Bengbu, China
| | - Xiaotian Liu
- Research Center of Clinical Laboratory Science, School of Laboratory Medicine, Bengbu Medical University, Bengbu, China
| | - Qingyang Huo
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Nan Wu
- Molecular Diagnosis Center, First Affiliated Hospital, Bengbu Medical University, Bengbu, China
| | - Yangxia Zhang
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Taoling Zeng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Yong Zhang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Bengbu Medical University, Bengbu, China
| | - Henry You Li
- Biomolecular Interaction Centre, University of Canterbury, Christchurch, New Zealand
| | - Jie Lian
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Jihong Zhou
- Research Center of Clinical Laboratory Science, School of Laboratory Medicine, Bengbu Medical University, Bengbu, China
| | - Emmanuel Jairaj Moses
- Regenerative Medicine Sciences Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia.
| | - Jian Geng
- Research Center of Clinical Laboratory Science, School of Laboratory Medicine, Bengbu Medical University, Bengbu, China.
| | - Juntang Lin
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, China.
| | - Wei Li
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Bengbu Medical University, Bengbu, China.
| | - Xinxing Zhu
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, Department of Respiratory and Critical Care Medicine, First Affiliated Hospital, Bengbu Medical University, Bengbu, China.
| |
Collapse
|
25
|
Zhao WW, Gao Y, Zhu YT, Zhong FL, Luo XG. SMYD3 plays a pivotal role in mediating the epithelial-mesenchymal transition process in breast cancer. Biochem Biophys Res Commun 2025; 749:151363. [PMID: 39864383 DOI: 10.1016/j.bbrc.2025.151363] [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: 01/17/2025] [Accepted: 01/17/2025] [Indexed: 01/28/2025]
Abstract
In previous reports, we highlighted the significant involvement of SMYD3, a histone methyltransferase (HMT), in various aspects of cancer progression, including cell adhesion, migration, and invasion. In this study, we delved deeper into understanding the relationship between SMYD3 and epithelial-mesenchymal transition (EMT) both in cell lines and clinical samples. Our investigation uncovered a notable correlation between heightened SMYD3 expression and the presence of EMT markers in human breast cancer tissues. We found that the induction of SMYD3 expression is facilitated by transforming growth factor beta 1 (TGF-β1), which achieves this by suppressing miR-124, an inhibitor that targets SMYD3, through alterations in DNA methylation. Conversely, our experiments demonstrated that reducing SMYD3 levels through RNA interference impeded TGF-β1-induced EMT in breast cancer cells. Furthermore, our results revealed that SMYD3 alone has the capability to modulate the expression of markers associated with EMT. An intriguing aspect of our study is the revelation that SMYD3 influences the activation of vimentin by binding to its response elements within the core promoter region. Notably, this effect is independent of SMYD3's histone methyltransferase activity. These findings collectively underscore the pivotal role of SMYD3 in driving EMT, both in cell lines and primary cancer tissues, particularly emphasizing its significance in TGF-β1-induced EMT in breast cancer.
Collapse
Affiliation(s)
- Wen-Wen Zhao
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China; Shijiazhuang Finance & Trade School, Hebei, 050800, China
| | - Yuan Gao
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yu-Ting Zhu
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Fei-Liang Zhong
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China.
| | - Xue-Gang Luo
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China.
| |
Collapse
|
26
|
Xu Y, Li J, Qiu Y, Wu F, Xue Z, Liu B, Fan H, Zhou Y, Wu Q. USC-Derived Small Extracellular Vesicles-Functionalized Scaffolds Promote Scarless Vaginal Defect Repair via Delivery of Decorin and DUSP3 Proteins. Int J Nanomedicine 2025; 20:1615-1634. [PMID: 39931530 PMCID: PMC11808217 DOI: 10.2147/ijn.s499856] [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: 10/18/2024] [Accepted: 01/23/2025] [Indexed: 02/13/2025] Open
Abstract
Background Scar formation following large-area vaginal defects post-vaginoplasty is a major clinical challenge. Compared to skin scars, vaginal scars can lead to pain during intercourse and urinary difficulties, severely impacting quality of life. Small extracellular vesicles (sEVs) encapsulate diverse bioactive components, making them potential therapeutic agents. Designing functional scaffolds that incorporate sEVs is a promising approach for scarless vaginal defect repair. Methods sEVs-loaded scaffolds were developed through electrostatic interactions between negatively charged sEVs secreted by urine-derived stem cells (USC-sEVs) and positively charged human acellular amniotic membranes. The efficacy of sEVs-loaded scaffolds in the treatment of vaginal defects in rabbits was assessed by histological analysis. Immunofluorescence staining, Western blot, qRT-PCR and collagen gel contraction analyses were conducted to evaluate the antifibrotic effects of USC-sEVs. RNA sequencing was employed to elucidate the underlying mechanisms involved. LC‒MS/MS analysis was used to identify candidate upstream proteins in USC-sEVs. Results In vivo experiments demonstrated that the sEVs-loaded scaffolds promoted scarless healing of vaginal defects in rabbits by modulating collagen deposition, reducing fibrosis, and diminishing inflammation. In vitro experiments revealed that USC-sEVs significantly inhibited the proliferation, collagen production, and activation of fibroblasts with a fibrotic phenotype, indicating the antifibrotic properties of USC-sEVs. Transcriptome and Western blot analyses revealed that USC-sEVs treatment inhibited fibrosis by downregulating the TGF-β and p38 MAPK signaling pathways. LC‒MS/MS analysis identified 2653 proteins encapsulated in USC-sEVs. Western blot analysis revealed that decorin, an inhibitor of the TGF-β signaling pathway, and DUSP3, a negative regulator of p38 phosphorylation, were enriched in USC-sEVs and could be transferred to fibroblasts. Conclusion USC-sEVs inhibited fibrosis and promoted scarless healing by delivering decorin and DUSP3 proteins, which regulate the TGF-β and p38 MAPK signaling pathways, respectively. This study highlights the potential of sEVs-loaded scaffolds as a promising strategy for scarless vaginal repair following vaginoplasty, offering a novel approach for regenerative medicine with significant translational potential for clinical application.
Collapse
Affiliation(s)
- Yiyun Xu
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Jie Li
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Yu Qiu
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Fuyue Wu
- Organoid Regeneration Research Center, ReMed Regenerative Medicine Clinical Application Institute, Shanghai, 201114, People’s Republic of China
| | - Zhuowei Xue
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Bin Liu
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Hongjie Fan
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Yuedi Zhou
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Qingkai Wu
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| |
Collapse
|
27
|
Zhao JH, Li S, Du SL, Han GZ, Li H, Shao B, Liu X, Zhou Y, Zhang ZQ. miR-207 Suppresses the Progression of SiO 2-Induced Pulmonary Fibrosis by Targeting Smad3 to Regulate the TGF-β1/Smad3 Signaling Pathway in C57BL/6 Mice. J Biochem Mol Toxicol 2025; 39:e70170. [PMID: 39945717 DOI: 10.1002/jbt.70170] [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: 08/19/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 05/09/2025]
Abstract
Silicosis is a worldwide occupational disease characterized by irreversible pulmonary fibrosis. Recent studies have showed that microRNAs (miRNAs) may play a crucial role in silicosis progression by modulating fibrosis-related gene express. In this study, we selected miR-207 as our research subject because we found that miR-207 can be match with Smad3 using bioinformatic techniques, which might silence the key fibrosis-related TGF-β1/Smad3 signal pathway. In this study, the mice were given silica suspension (20 µg/µL, 80 µL) via nostril once a day for 16 days to establish silicosis models, and then were transfected with miR-207 mimic or inhibitor. The mice which were given phosphate-buffered saline (PBS) (80 µL) via nostril were used as control. All mice were killed on Day 45 after the first exposure to dust, after which their lungs were removed for pathological observation and to measure the hydroxyproline content. Then, real-time polymerase chain reaction and Western blot analysis were applied to detect the relative expression levels of TGF-β1/Smad3 signaling pathway indicators (TGF-β1, TGF-βR, and Smad3), and myofibroblast transformation indicators (α-SMA and Fn). Results showed that the lung pathological images of silicosis model group mice showed significant fibrosis, and TGF-β1, TGF-βR, Smad3, α-SMA, and Fn were all highly upregulated compared with the control group mice. Intervention with miR-207 mimics significantly inhibited pulmonary fibrosis in silicosis mice by downregulation of TGF-β1/Smad3 and inhibiting of myofibroblast formation. Whereas these phenomena were not observed in silicosis mice treated with miR-207 inhibitor. The results demonstrated that miR-207 can block the progression of SiO2-induced pulmonary fibrosis by targeting the TGF-β/Smad3 signaling pathway.
Collapse
Affiliation(s)
- Jia-Hui Zhao
- School of Public Health, Jining Medical University, Jining, China
- School of Public Health, Shandong Second Medical University, Weifang, China
| | - Shuang Li
- School of Public Health, Jining Medical University, Jining, China
| | - Shu-Ling Du
- School of Public Health, Jining Medical University, Jining, China
- School of Public Health, Shandong Second Medical University, Weifang, China
| | - Gui-Zhi Han
- School of Public Health, Jining Medical University, Jining, China
| | - Huan Li
- School of Public Health, Jining Medical University, Jining, China
| | - Bo Shao
- School of Public Health, Jining Medical University, Jining, China
| | - Xia Liu
- School of Public Health, Jining Medical University, Jining, China
| | - Yuting Zhou
- School of Public Health, Jining Medical University, Jining, China
| | - Zhao-Qiang Zhang
- School of Public Health, Jining Medical University, Jining, China
| |
Collapse
|
28
|
Majidpour M, Azizi SG, Davodabadi F, Sabeti Akbar-Abad M, Abdollahi Z, Sargazi S, Shahriari H. Recent advances in TGF-β signaling pathway in COVID-19 pathogenesis: A review. Microb Pathog 2025; 199:107236. [PMID: 39701478 DOI: 10.1016/j.micpath.2024.107236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/21/2024]
Abstract
The coronavirus disease 2019 (COVID-19) has resulted in approximately 7.0 million fatalities between 2019 and 2022, underscoring a pressing need for comprehensive research into its underlying mechanisms and therapeutic avenues. A distinctive feature of severe COVID-19 is the dysregulated immune response characterized by excessive activation of immune cells and the consequent cytokine storms. Recent advancements in our understanding of cellular signaling pathways have illuminated the role of Transforming Growth Factor Beta (TGF-β) as a pivotal signaling molecule with significant implications for the pathogenesis of infectious diseases, including COVID-19. Emerging evidence reveals that TGF-β signaling, when activated by viral components or secondary pathways, adversely affects diverse cell types, particularly immune cells, and lung tissue, leading to complications such as pulmonary fibrosis. In our review article, we critically evaluate recent literature on the involvement of TGF-β signaling in the progression of COVID-19. We discuss a range of pharmacological interventions, including nintedanib, pirfenidone, corticosteroids, proton pump inhibitors, and histone deacetylase inhibitors, and their potential to modulate the TGF-β pathway in the context of COVID-19 treatment. Additionally, we explore ongoing clinical trials involving mesenchymal stem cells, low-dose radiation therapy, and artemisinin derivatives to assess their impact on TGF-β levels and subsequent clinical outcomes in COVID-19 patients. This review is particularly relevant at this juncture as the global health community continues to grapple with the ramifications of the COVID-19 pandemic, highlighting the urgent need for targeted therapeutic strategies aimed at TGF-β modulation to mitigate disease severity and improve patient outcomes.
Collapse
Affiliation(s)
- Mahdi Majidpour
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Seyed Ghader Azizi
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Fatemeh Davodabadi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahboobeh Sabeti Akbar-Abad
- Department of Clinical Biochemistry, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Zahra Abdollahi
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran.
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Hossein Shahriari
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| |
Collapse
|
29
|
Yao Y, Niu Y, Zhou H, Yong M. KAT2B inhibits proliferation and invasion via inactivating TGF-β/Smad3 pathway-medicated autophagy and EMT in epithelial ovarian cancer. Sci Rep 2025; 15:3417. [PMID: 39870682 PMCID: PMC11772695 DOI: 10.1038/s41598-024-83977-1] [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: 08/28/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025] Open
Abstract
Lysine acetyltransferase 2B (KAT2B) plays a crucial role in epigenetic regulation and tumor pathogenesis. Our study investigates KAT2B's function in epithelial ovarian cancer (EOC) using in vivo and in vitro methods. Immunohistochemistry showed the KAT2B expression in EOC tissues. RNA sequencing (RNA-seq) further identified altered gene expression profiles following KAT2B silencing in EOC cells. Western blot and qRT-PCR were employed to assess the protein and mRNA expression, respectively. KAT2B downregulated in EOC tissues and correlated with both FIGO stage and grade. KAT2B silencing induced autophagy, enhancing cell proliferation and invasion, while overexpression had opposite effects. In vivo, KAT2B silencing increased tumor volume and weight, mitigated by autophagy inhibitor chloroquine. Bioinformatics and co-immunoprecipitation assays identified a KAT2B-SMAD7 interaction. Mechanistic investigations suggested that KAT2B knockdown enhanced autophagy via activation of the TGF-β/Smad3/7 signaling pathway, thereby driving epithelial-mesenchymal transition (EMT), proliferation, and invasion in EOC. Additionally, our data hint at a potential role for the AKT/mTOR pathway. KAT2B acts as a putative tumor suppressor in EOC, where its reduced expression correlates with advanced disease stages. It is implicated in the regulation of autophagy, proliferation, and invasion via the TGF-β/Smad3/7 pathway, positioning KAT2B as a candidate therapeutic target for EOC interventions.
Collapse
Affiliation(s)
- Yuqin Yao
- Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, People's Republic of China
- Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, People's Republic of China
| | - Yuna Niu
- Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, People's Republic of China
| | - Honggui Zhou
- Department of Obstetrics and Gynecology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, People's Republic of China.
| | - Min Yong
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, People's Republic of China.
| |
Collapse
|
30
|
Ze Y, Wu Y, Tan Z, Li R, Li R, Gao W, Zhao Q. Signaling pathway mechanisms of circadian clock gene Bmal1 regulating bone and cartilage metabolism: a review. Bone Res 2025; 13:19. [PMID: 39870641 PMCID: PMC11772753 DOI: 10.1038/s41413-025-00403-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/17/2024] [Accepted: 12/31/2024] [Indexed: 01/29/2025] Open
Abstract
Circadian rhythm is ubiquitous in nature. Circadian clock genes such as Bmal1 and Clock form a multi-level transcription-translation feedback network, and regulate a variety of physiological and pathological processes, including bone and cartilage metabolism. Deletion of the core clock gene Bmal1 leads to pathological bone alterations, while the phenotypes are not consistent. Studies have shown that multiple signaling pathways are involved in the process of Bmal1 regulating bone and cartilage metabolism, but the exact regulatory mechanisms remain unclear. This paper reviews the signaling pathways by which Bmal1 regulates bone/cartilage metabolism, the upstream regulatory factors that control Bmal1, and the current Bmal1 knockout mouse models for research. We hope to provide new insights for the prevention and treatment of bone/cartilage diseases related to circadian rhythms.
Collapse
Affiliation(s)
- Yiting Ze
- Department of Orthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yongyao Wu
- Department of Orthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhen Tan
- Department of Implant Dentistry, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Rui Li
- Department of Orthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Rong Li
- Department of Orthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Wenzhen Gao
- Department of Orthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Qing Zhao
- Department of Orthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
31
|
Chatterjee A, Tumarin J, Prabhakar S. Cellular cross-talk drives mesenchymal transdifferentiation in diabetic kidney disease. Front Med (Lausanne) 2025; 11:1499473. [PMID: 39839616 PMCID: PMC11747801 DOI: 10.3389/fmed.2024.1499473] [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: 09/20/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
While changes in glomerular function and structure may herald diabetic kidney disease (DKD), many studies have underscored the significance of tubule-interstitial changes in the progression of DKD. Indeed, tubule-interstitial fibrosis may be the most important determinant of progression of DKD as in many forms of chronic glomerulopathies. The mechanisms underlying the effects of tubular changes on glomerular function in DKD have intrigued many investigators, and therefore, the signaling mechanisms underlying the cross-talk between tubular cells and glomerular cells have been the focus of investigation in many recent studies. Additionally, the observations of slowing of glomerular filtration rate (GFR) decline and reduction of proteinuria by recent drugs such as SGLT-2 blockers, whose primary mechanism of action is on proximal tubules, further strengthen the concept of cross-talk between the tubular and glomerular cells. Recently, the focus of research on the pathogenesis of DKD has primarily centered around exploring the cross-talk between various signaling pathways in the diabetic kidney as well as cross-talk between tubular and glomerular endothelial cells and podocytes with special relevance to epithelial-to-mesenchymal transition (EMT) and endothelial-to-mesenchymal transition (EndoMT). The focus of this review is to provide a general description of cell-to-cell cross-talk in the diabetic kidney and to highlight these concepts with evidence in relation to the physiology and pathophysiology of DKD.
Collapse
Affiliation(s)
| | | | - Sharma Prabhakar
- Department of Internal Medicine, Texas Tech University Health Sciences Centre, Lubbock, TX, United States
| |
Collapse
|
32
|
Avelar LE, Nabhani S, Wüst S. Unveiling the Mechanism: Injectable Poly-L-Lactic Acid's Evolving Role-Insights From Recent Studies. J Cosmet Dermatol 2025; 24:e16635. [PMID: 39412038 PMCID: PMC11743305 DOI: 10.1111/jocd.16635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/14/2024] [Accepted: 10/03/2024] [Indexed: 01/21/2025]
Affiliation(s)
- Luiz Eduardo Avelar
- Private practiceBelo HorizonteBrazil
- Police Department of Minas Gerais StateBelo HorizonteBrazil
- Clinic DomaniBelo HorizonteBrazil
| | | | - Stas Wüst
- Z282 Medical Affairs ConsultingBeverlyMassachusettsUSA
| |
Collapse
|
33
|
Zhang Z, Ma C, Gao X, Wang C, Li Y, Yang C, Ma E, Cheng M. Design, synthesis, and biological evaluation of novel 3-naphthylthiophene derivatives as potent SIRT2 inhibitors for the treatment of myocardial fibrosis. Bioorg Chem 2025; 154:108033. [PMID: 39672075 DOI: 10.1016/j.bioorg.2024.108033] [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: 11/06/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
SIRT2 (sirtuin2) is a NAD+-dependent deacetylase implicated in fibrosis and inflammation of the liver, kidney, and heart. In this study, we designed and synthesized a series of 3-naphthylthiophene derivatives and evaluated their inhibitory activity against the SIRT2 enzyme. Among them, Z18 demonstrated outstanding SIRT2 inhibitory activity and selectivity. It significantly inhibited both the proliferation of cardiac fibroblasts (CFs) and the activity and expression of SIRT2 in CFs. Moreover, compound Z18 effectively suppressed TGF-β1-induced increases in α-SMA and CoL-1A1 protein expression, as well as hydroxyproline levels. Pharmacological mechanism tests demonstrated that Z18 inhibited SIRT2, thereby suppressing the TGF-β1-induced autocrine production of TGF-β1 and the phosphorylation of Smad2/3 in CFs. In MTT assays, Z18 exhibited a significant inhibitory effect on the proliferation of CFs induced by TGF-β1. In vivo, Z18 effectively ameliorated TAC- and ISO-induced declines in cardiac function, histopathological morphological changes, and collagen deposition. It also inhibited SIRT2 activity and reduced the expression of α-SMA and p-Smad2/3. In hepatorenal toxicity assays, Z18 exhibited an excellent safety profile. These results support the further development of the selective SIRT2 inhibitor Z18 as a potential lead compound for the treatment of myocardial fibrosis.
Collapse
Affiliation(s)
- Zhuo Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Chao Ma
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Xiong Gao
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Chuncheng Wang
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Yanchun Li
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Chen Yang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China
| | - Enlong Ma
- Department of Pharmacology, School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China.
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, People's Republic of China.
| |
Collapse
|
34
|
Hua S, Chi J, Zhang N, Yang X, Zhang P, Jiang C, Feng Y, Hong X, Feng Z, Yan Y. WHAMM Inhibits Type II Alveolar Epithelial Cell EMT by Mediating Autophagic Degradation of TGF-β1 in Bronchopulmonary Dysplasia. J Cell Physiol 2025; 240:e31486. [PMID: 39564703 DOI: 10.1002/jcp.31486] [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: 06/12/2024] [Revised: 10/11/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is one of the most prevalent complication in preterm infants, primarily characterized by arrested alveolar growth. The involvement of epithelial-mesenchymal transition (EMT) of AECII cells is proposed to have a crucial role in the pathogenesis of BPD; however, the underlying mechanism remains unclear. The present study reveals a significant reduction of WHAMM (WASP homolog associated with actin, membranes, and microtubules) in hyperoxia-induced BPD mice, highlighting its crucial role in suppressing the progression of BPD through the inhibition of EMT in AECIIs. We demonstrated that hyperoxia-induced downregulation of WHAMM leads to the accumulation of TGF-β1 primarily through its mediation of the autophagic degradation pathway. Mechanistically, WHAMM enhanced the autophagosomal localization of TGF-β1 and concurrently promoted the process of autophagy, thereby comprehensively facilitating the autophagic degradation of TGF-β1. These findings reveal the important role of WHAMM in the development of BPD, and the proposed WHAMM/autophagy/TGF-β1/EMT pathway may represent a potential therapeutic strategy for BPD treatment.
Collapse
Affiliation(s)
- Shaodong Hua
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Jinghan Chi
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Ning Zhang
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Xiao Yang
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Pan Zhang
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Chenyang Jiang
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Yao Feng
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Xiaoyang Hong
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Zhichun Feng
- General Internal Medicine, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Yurou Yan
- Department of Pediatrics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
35
|
Zhou H, Chen Y, Jiang N, Ren Y, Zhuang J, Ren Y, Shen L, Li C. Epoxymicheliolide Reduces Radiation-Induced Senescence and Extracellular Matrix Formation by Disrupting NF-κB and TGF-β/SMAD Pathways in Lung Cancer. Phytother Res 2025; 39:51-63. [PMID: 39506320 DOI: 10.1002/ptr.8352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/02/2024] [Accepted: 09/14/2024] [Indexed: 11/08/2024]
Abstract
Lung cancer is a major cause of cancer-related mortality, and radiotherapy is often limited by tumor resistance and side effects. This study explores whether epoxymicheliolide (ECL), a compound from feverfew, can enhance radiotherapy efficacy in lung cancer. We tested ECL on A549 and PC-9 lung cancer cell lines to evaluate its effect on x-ray irradiation. We measured apoptosis, NF-κB pathway inhibition, TGF-β secretion reduction, and epithelial-mesenchymal transition suppression. In vivo, C57BL/6 mice with lung tumors received ECL and radiotherapy. ECL enhanced the antiproliferative effects of x-ray irradiation, induced apoptosis in senescent cells, inhibited the NF-κB pathway, reduced TGF-β levels, and suppressed epithelial-mesenchymal transition. ECL also inhibited tumor growth and improved survival in mice. ECL is a promising adjunct to radiotherapy for lung cancer, improving treatment outcomes by targeting multiple tumor progression mechanisms. It offers potential for enhanced management of lung cancer.
Collapse
Affiliation(s)
- Heng Zhou
- School of Public Health, Yangzhou University, Yangzhou, China
- Department of Radio-Chemotherapy, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yong Chen
- Department of Radio-Chemotherapy, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Medical College, Yangzhou University, Yangzhou, China
| | - Ningzu Jiang
- School of Public Health, Yangzhou University, Yangzhou, China
- The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Yanxian Ren
- School of Public Health, Yangzhou University, Yangzhou, China
- The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Jiayuan Zhuang
- School of Public Health, Yangzhou University, Yangzhou, China
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yue Ren
- Department of Radio-Chemotherapy, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Medical College, Yangzhou University, Yangzhou, China
| | - Lin Shen
- Department of Radio-Chemotherapy, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Medical College, Yangzhou University, Yangzhou, China
| | - Chenghao Li
- Department of Radio-Chemotherapy, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
36
|
Lu Y, Huangfu S, Ma C, Ding Y, Zhang Y, Zhou C, Liao L, Li M, You J, Chen Y, Wang D, Chen A, Jiang B. Exosomes derived from umbilical cord mesenchymal stem cells promote healing of complex perianal fistulas in rats. Stem Cell Res Ther 2024; 15:414. [PMID: 39732731 DOI: 10.1186/s13287-024-04028-0] [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: 07/23/2024] [Accepted: 10/28/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Complex perianal fistulas, challenging to treat and prone to recurrence, often require surgical intervention that may cause fecal incontinence and lower quality of life due to large surgical wounds and potential sphincter damage. Human umbilical cord-derived MSCs (hUC-MSCs) and their exosomes (hUCMSCs-Exo) may promote wound healing. METHODS This study assessed the efficacy, mechanisms, and safety of these exosomes in treating complex perianal fistulas in SD rats. We established a rat model, divided rats with fistulas into the control and the exosome groups. We assessed treatment efficacy through ultrasound, clinical observations, and histopathological analysis. We also evaluated the activation of the HIF-1α/TGF-β/Smad signaling pathway via PCR and Western blot and assessed serological markers for HIF-1α and inflammatory indices through ELISA. We analyzed gut microbiota and the systemic metabolic environment via untargeted metabolomics. RESULTS The hUCMSCs-Exo effectively promoted healing of wound, regulated the immune balance enhanced collagen synthesis and angiogenesis in the perianal fistulas model of rats, and regulated the gut microbiota and metabolomic profiles. Results of PCR and Western blot analyses indicated that the exosomes activated HIF-1α/TGF-β/Smad signaling pathways. To the dosages tested, the 10ug/100ul concentration (medium dose) was found to be the most effective to the treatment of complex perianal fistulas. CONCLUSIONS The hUCMSCs-Exo significantly promoted the healing of wound in perianal fistulas of rats and demonstrated higher safety. The underlying mechanism facilitating the healing process was likely associated with the activation of the HIF-1α/TGF-β/Smad signaling pathway.
Collapse
Affiliation(s)
- Yafei Lu
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Shaohua Huangfu
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Chuanxue Ma
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Yan Ding
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Yajie Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
- Department of Biobank, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Chungen Zhou
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Lianming Liao
- Center of Laboratory Medicine, Union Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China
| | - Ming Li
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui, People's Republic of China
| | - Jia You
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Yuting Chen
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Dawei Wang
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Ao Chen
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China
| | - Bin Jiang
- National Colorectal Disease CenterNanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, People's Republic of China.
| |
Collapse
|
37
|
Kuburich NA, Kiselka JM, den Hollander P, Karam AA, Mani SA. The Cancer Chimera: Impact of Vimentin and Cytokeratin Co-Expression in Hybrid Epithelial/Mesenchymal Cancer Cells on Tumor Plasticity and Metastasis. Cancers (Basel) 2024; 16:4158. [PMID: 39766058 PMCID: PMC11674825 DOI: 10.3390/cancers16244158] [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: 10/01/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The epithelial-mesenchymal transition (EMT) program is critical to metastatic cancer progression. EMT results in the expression of mesenchymal proteins and enhances migratory and invasive capabilities. In a small percentage of cells, EMT results in the expression of stemness-associated genes that provide a metastatic advantage. Although EMT had been viewed as a binary event, it has recently become clear that the program leads to a spectrum of phenotypes, including hybrid epithelial/mesenchymal (E/M) cells that have significantly greater metastatic capability than cells on the epithelial or mesenchymal ends of the spectrum. As hybrid E/M cells are rarely observed in physiological, non-diseased states in the adult human body, these cells are potential biomarkers and drug targets. Hybrid E/M cells are distinguished by the co-expression of epithelial and mesenchymal proteins, such as the intermediate filament proteins cytokeratin (CK; epithelial) and vimentin (VIM; mesenchymal). Although these intermediate filaments have been extensively used for pathological characterization and detection of aggressive carcinomas, little is known regarding the interactions between CK and VIM when co-expressed in hybrid E/M cells. This review describes the characteristics of hybrid E/M cells with a focus on the unique co-expression of VIM and CK. We will discuss the structures and functions of these two intermediate filament proteins and how they may interact when co-expressed in hybrid E/M cells. Additionally, we review what is known about cell-surface expression of these intermediate filament proteins and discuss their potential as predictive biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Nick A. Kuburich
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (N.A.K.); (J.M.K.); (P.d.H.); (A.A.K.)
- Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Julia M. Kiselka
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (N.A.K.); (J.M.K.); (P.d.H.); (A.A.K.)
- Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Petra den Hollander
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (N.A.K.); (J.M.K.); (P.d.H.); (A.A.K.)
- Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Andrew A. Karam
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (N.A.K.); (J.M.K.); (P.d.H.); (A.A.K.)
- Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Sendurai A. Mani
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (N.A.K.); (J.M.K.); (P.d.H.); (A.A.K.)
- Department of Pathology and Lab Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| |
Collapse
|
38
|
Napoli M, Bauer J, Bonod C, Vadon-Le Goff S, Moali C. PCPE-2 (procollagen C-proteinase enhancer-2): The non-identical twin of PCPE-1. Matrix Biol 2024; 134:59-78. [PMID: 39251075 DOI: 10.1016/j.matbio.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
PCPE-2 was discovered at the beginning of this century, and was soon identified as a close homolog of PCPE-1 (procollagen C-proteinase enhancer 1). After the demonstration that it could also stimulate the proteolytic maturation of fibrillar procollagens by BMP-1/tolloid-like proteinases (BTPs), PCPE-2 did not attract much attention as it was thought to fulfill the same functions as PCPE-1 which was already well-described. However, the tissue distribution of PCPE-2 shows both common points and significant differences with PCPE-1, suggesting that their activities are not fully overlapping. Also, the recently established connections between PCPE-2 (gene name PCOLCE2) and several important diseases such as atherosclerosis, inflammatory diseases and cancer have highlighted the need for a thorough reappraisal of the in vivo roles of this regulatory protein. In this context, the recent finding that, while retaining the ability to bind fibrillar procollagens and to activate their C-terminal maturation, PCPE-2 can also bind BTPs and inhibit their activity has substantially extended its potential functions. In this review, we describe the current knowledge about PCPE-2 with a focus on collagen fibrillogenesis, lipid metabolism and inflammation, and discuss how we could further advance our understanding of PCPE-2-dependent biological processes.
Collapse
Affiliation(s)
- Manon Napoli
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Julien Bauer
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Christelle Bonod
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Sandrine Vadon-Le Goff
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Catherine Moali
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France.
| |
Collapse
|
39
|
Wang S, Sha P, Zhao X, Tao Z, Liu S. Peritendinous adhesion: Therapeutic targets and progress of drug therapy. Comput Struct Biotechnol J 2024; 23:251-263. [PMID: 38173878 PMCID: PMC10762322 DOI: 10.1016/j.csbj.2023.11.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
Peritendinous adhesion (PA) is one of the most common complications following hand surgery and characterized with abnormal hyperplasia of connective tissue and excessive deposition of extracellular matrix. Subsequently, various clinical symptoms such as chronic pain, limb dyskinesia and even joint stiffness occur and patients are always involved in the vicious cycle of "adhesion - release - re-adhesion", which seriously compromise the quality of life. Until present, the underlying mechanism remains controversial and lack of specific treatment, with symptomatic treatment being the only option to relieve symptoms, but not contributing no more to the fundamentally rehabilitation of basic structure and function. Recently, novel strategies have been proposed to inhibit the formation of adhesion tissues including implantation of anti-adhesion barriers, anti-inflammation, restraint of myofibroblast transformation and regulation of collagen overproduction. Furthermore, gene therapy has also been considered as a promising anti-adhesion treatment. In this review, we provide an overview of anti-adhesion targets and relevant drugs to summarize the potential pharmacological roles and present subsequent challenges and prospects of anti-adhesion drugs.
Collapse
Affiliation(s)
| | | | | | - Zaijin Tao
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Hanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Shen Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Hanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| |
Collapse
|
40
|
Chen P, Lin X, Zhang C, Xie Y, Guo Z, Ren F. Fusobacterium nucleatum-infected periodontitis promotes renal interstitial fibrosis in rats through the TGF-β/SMAD2/3 and β-catenin signaling pathways. Gene 2024; 927:148729. [PMID: 38936784 DOI: 10.1016/j.gene.2024.148729] [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: 03/21/2024] [Revised: 06/03/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
OBJECTIVES Periodontitis is associated with Fusobacterium nucleatum (F.n) infection. Although the colonization of renal tissue by F.n is well documented, its specific role in kidney disease has yet to be determined. This study aimed to investigate the potential association between F.n-induced periodontitis and renal interstitial fibrosis. METHODS The rat gingival sulcus was injected with F.n suspension, while the control group (NC) was injected with PBS. The levels of total protein (TP), albumin (ALB), creatinine, and urea nitrogen (BUN) in rat serum and/or urine were quantified using the appropriate kits. Renal interstitial fibrosis and epithelial-mesenchymal transition (EMT) were evaluated in rats using Masson staining, Periodic Schiff-Methenamine (PASM) staining, and immunohistochemical staining. The levels of fibrosis- and EMT-related proteins and the TGF-β/SMAD2/3 and β-catenin signaling pathways were determined using Western blot analysis. F.n in the kidney tissues was quantitatively determined using bacterial 16S rRNA technology. RESULTS Serum levels of TP, ALB, creatinine, and BUN were not significantly decreased in F.n-infected rats with periodontitis. The levels of creatinine and ALB in the urine were not statistically different between two groups. Masson and PASM staining showed that F.n-induced periodontitis could promote renal interstitial fibrosis in rats. The levels of collagen I, fibronectin (FN), vimentin, and α-SMA were upregulated in the kidney tissues of rats with F.n-induced periodontitis and in F.n-treated HK-2 cells. However, E-cadherin levels were reduced. F.n promoted renal interstitial and HK-2 cell fibrosis in rats by modulating the TGF-β/SMAD2/3 and β-catenin signaling pathways. F.n colonization increased renal interstitial fibrosis in rats. CONCLUSION F.n-induced periodontitis promoted EMT by activating the TGF-β/SMAD2/3 and β-catenin signaling pathways, thus promoting renal interstitial fibrosis in rats.
Collapse
Affiliation(s)
- Pei Chen
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Xi Lin
- Center of Oral Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Caimei Zhang
- Second Department of endodontics, Haizhu Square Branch of Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510120, China
| | - Yu Xie
- Department of prosthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Zehong Guo
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China.
| | - Fei Ren
- VIP clinic, Panfu Branch of Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510180, China.
| |
Collapse
|
41
|
Lee JH, Sánchez-Rivera FJ, He L, Basnet H, Chen FX, Spina E, Li L, Torner C, Chan JE, Yarlagadda DVK, Park JS, Sussman C, Rudin CM, Lowe SW, Tammela T, Macias MJ, Koche RP, Massagué J. TGF-β and RAS jointly unmask primed enhancers to drive metastasis. Cell 2024; 187:6182-6199.e29. [PMID: 39243762 PMCID: PMC12035776 DOI: 10.1016/j.cell.2024.08.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/08/2024] [Accepted: 08/07/2024] [Indexed: 09/09/2024]
Abstract
Epithelial-to-mesenchymal transitions (EMTs) and extracellular matrix (ECM) remodeling are distinct yet important processes during carcinoma invasion and metastasis. Transforming growth factor β (TGF-β) and RAS, signaling through SMAD and RAS-responsive element-binding protein 1 (RREB1), jointly trigger expression of EMT and fibrogenic factors as two discrete arms of a common transcriptional response in carcinoma cells. Here, we demonstrate that both arms come together to form a program for lung adenocarcinoma metastasis and identify chromatin determinants tying the expression of the constituent genes to TGF-β and RAS inputs. RREB1 localizes to H4K16acK20ac marks in histone H2A.Z-loaded nucleosomes at enhancers in the fibrogenic genes interleukin-11 (IL11), platelet-derived growth factor-B (PDGFB), and hyaluronan synthase 2 (HAS2), as well as the EMT transcription factor SNAI1, priming these enhancers for activation by a SMAD4-INO80 nucleosome remodeling complex in response to TGF-β. These regulatory properties segregate the fibrogenic EMT program from RAS-independent TGF-β gene responses and illuminate the operation and vulnerabilities of a bifunctional program that promotes metastatic outgrowth.
Collapse
Affiliation(s)
- Jun Ho Lee
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Francisco J Sánchez-Rivera
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Lan He
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Harihar Basnet
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Fei Xavier Chen
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elena Spina
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Liangji Li
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carles Torner
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Jason E Chan
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dig Vijay Kumar Yarlagadda
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Tri-Institutional Graduate Program in Computational Biology and Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jin Suk Park
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carleigh Sussman
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tuomas Tammela
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maria J Macias
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona 08028, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
42
|
Wen Y, Zheng Y, Hua S, Li T, Bi X, Lu Q, Li M, Sun S. Mechanisms of Bone Morphogenetic Protein 2 in Respiratory Diseases. Curr Allergy Asthma Rep 2024; 25:1. [PMID: 39466470 DOI: 10.1007/s11882-024-01181-7] [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: 10/17/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE OF REVIEW Bone morphogenetic protein 2 (BMP2) belongs to the transforming growth factor-β (TGF-β) superfamily and plays an important role in regulating embryonic development, angiogenesis, osteogenic differentiation, tissue homeostasis, and cancer invasion. Increasing studies suggest BMP2 is involved in several respiratory diseases. This study aimed to review the role and mechanisms of BMP2 in respiratory diseases. RECENT FINDINGS BMP2 signaling pathway includes the canonical and non-canonical signaling pathway. The canonical signaling pathway is the BMP2-SMAD pathway, and the non-canonical signaling pathway includes mitogen-activated protein kinase (MAPK) pathway and phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway. The BMP2 is related to pulmonary hypertension (PH), lung cancer, pulmonary fibrosis (PF), asthma, and chronic obstructive pulmonary disease (COPD). BMP2 inhibits the proliferation of pulmonary artery smooth muscle cells (PASMCs), promotes the apoptosis of PASMCs to reduce pulmonary vascular remodeling in PH, which is closely related to the canonical and non-canonical pathway. In addition, BMP2 stimulates the proliferation and migration of cells to promote the occurrence, colonization, and metastasis of lung cancer through the canonical and the non-canonical pathway. Meanwhile, BMP2 exert anti-fibrotic function in PF through canonical signaling pathway. Moreover, BMP2 inhibits airway inflammation to maintain airway homeostasis in asthma. However, the signaling pathways involved in asthma are poorly understood. BMP2 inhibits the expression of ciliary protein and promotes squamous metaplasia of airway epithelial cells to accelerate the development of COPD. In conclusion, BMP2 may be a therapeutic target for several respiratory diseases.
Collapse
Affiliation(s)
- Yiqiong Wen
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Yuanyuan Zheng
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Shu Hua
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Tongfen Li
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Xiaoqing Bi
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Qiongfen Lu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Min Li
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming, China.
| |
Collapse
|
43
|
Cui X, Fu J. Reinitiating lung development: a novel approach in the management of bronchopulmonary dysplasia. Respir Res 2024; 25:384. [PMID: 39449014 PMCID: PMC11515458 DOI: 10.1186/s12931-024-02996-8] [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: 06/12/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the predominant chronic lung disease in preterm infants, linked with various adverse long-term outcomes. Multiple prenatal and postnatal risk factors can impede lung development, leading to BPD. Current management of BPD relies heavily on pharmacotherapies and alterations in ventilatory strategies. However, these interventions only mitigate BPD symptoms without addressing underlying alveolar, vascular, structural, and functional deficiencies. Given the retarded lung development in infants with BPD and the limitations of existing modalities, new therapeutic approaches are imperative. The induced differentiation of stem/progenitor cells and the spatiotemporal expression patterns of growth factors associated with lung developmental processes are critical for lung development reactivation in BPD, which focuses on stimulating pulmonary vasculogenesis and alveolarization. This review summarizes the process of lung development and offers a comprehensive overview of advancements in therapies designed to reinitiate lung development in BPD. Furthermore, we assessed the potential of these therapies for maintaining lung homeostasis and effectively restoring pulmonary structure and function through stem/progenitor cells and growth factors, which have been widely researched.
Collapse
Affiliation(s)
- Xuewei Cui
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, China.
| |
Collapse
|
44
|
Chen H, Xu X, Li J, Xue Y, Li X, Zhang K, Jiang H, Liu X, Li M. Decoding tumor-fibrosis interplay: mechanisms, impact on progression, and innovative therapeutic strategies. Front Pharmacol 2024; 15:1491400. [PMID: 39534084 PMCID: PMC11555290 DOI: 10.3389/fphar.2024.1491400] [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: 09/04/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Malignant tumors are a category of diseases that possess invasive and metastatic capabilities, with global incidence and mortality rates remaining high. In recent years, the pivotal role of fibrosis in tumor progression, drug resistance, and immune evasion has increasingly been acknowledged. Fibrosis enhances the proliferation, migration, and invasion of tumor cells by modifying the composition and structure of the extracellular matrix, thereby offering protection for immune evasion by tumor cells. The activation of cancer-associated fibroblasts (CAFs) plays a significant role in this process, as they further exacerbate the malignant traits of tumors by secreting a variety of cytokines and growth factors. Anti-fibrotic tumor treatment strategies, including the use of anti-fibrotic drugs and inhibition of fibrosis-related signaling pathways such as Transforming Growth Factor-β (TGF-β), have demonstrated potential in delaying tumor progression and improving the effectiveness of chemotherapy, targeted therapy, and immunotherapy. In the future, by developing novel drugs that target the fibrotic microenvironment, new therapeutic options may be available for patients with various refractory tumors.
Collapse
Affiliation(s)
- Huiguang Chen
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xuexin Xu
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Jingxian Li
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Yu Xue
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xin Li
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Kaiyu Zhang
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Haihui Jiang
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaoliu Liu
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
- Department of Anatomy, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Mingzhe Li
- Department of Anatomy, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
45
|
Zhou S, Cheng W, Liu Y, Gao H, Yu L, Zeng Y. MiR-125b-5p alleviates pulmonary fibrosis by inhibiting TGFβ1-mediated epithelial-mesenchymal transition via targeting BAK1. Respir Res 2024; 25:382. [PMID: 39427175 PMCID: PMC11491022 DOI: 10.1186/s12931-024-03011-w] [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: 06/10/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024] Open
Abstract
This study explores the role and potential mechanisms of microRNA-125b-5p (miR-125b-5p) in pulmonary fibrosis (PF). PF is a typical outcome of many chronic lung diseases, with poor prognosis and the lack of appropriate medical treatment because PF's molecular mechanisms remain poorly understood. In this study, using in vitro and in vivo analyses, we find that miR-125b-5p is likely a potent regulator of lung fibrosis. The findings reveal that, on the one hand, miR-125b-5p not only specifically decreases in the epithelial-mesenchymal transition (EMT) of lung epithelial cells, but also shows a downregulation trend in the lung tissues of mice with PF. On the other hand, overexpression of miR-125b-5p on the cellular and animal levels downregulates EMT and fibrotic phenotypes, respectively. To clarify the molecular mechanism of the "therapeutic" effect of miR-125b-5p, we use the target prediction tool combined with a dual luciferase assay and complete a rescue experiment by constructing the overexpression vector of the target gene Bcl-2 homologous antagonist/ killer (BAK1), thus confirming that miR-125b-5p can effectively inhibit EMT and fibrosis process by targeting BAK1 gene. MiR-125b-5p inhibits the EMT in lung epithelial cells by negatively regulating BAK1, while overexpression of miR-125b-5p can alleviate lung fibrosis. The findings suggest that MiR-125b-5p/BAK1 can serve as a potential treatment target for PF.
Collapse
Affiliation(s)
- Shuang Zhou
- The Second Clinical Medical School of Fujian Medical University, Quanzhou, Fujian Province, China
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Respirology Medicine Centre of Fujian Province, Quanzhou, China
| | - Wenzhao Cheng
- Fujian Provincial Key Laboratory of Lung Stem Cells, Stem Cell Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Yifei Liu
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Hongzhi Gao
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Liying Yu
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.
| | - Yiming Zeng
- Fujian Provincial Key Laboratory of Lung Stem Cells, Stem Cell Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Respirology Medicine Centre of Fujian Province, Quanzhou, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province, China.
| |
Collapse
|
46
|
Wang Z, Elbanna Y, Godet I, Peters L, Lampe G, Chen Y, Xavier J, Huse M, Massagué J. TGF-β induces an atypical EMT to evade immune mechanosurveillance in lung adenocarcinoma dormant metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618357. [PMID: 39463937 PMCID: PMC11507679 DOI: 10.1101/2024.10.15.618357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The heterogeneity of epithelial-to-mesenchymal transition (EMT) programs is manifest in the diverse EMT-like phenotypes occurring during tumor progression. However, little is known about the mechanistic basis and functional role of specific forms of EMT in cancer. Here we address this question in lung adenocarcinoma (LUAD) cells that enter a dormancy period in response to TGF-β upon disseminating to distant sites. LUAD cells with the capacity to enter dormancy are characterized by expression of SOX2 and NKX2-1 primitive progenitor markers. In these cells, TGF-β induces growth inhibition accompanied by a full EMT response that subsequently transitions into an atypical mesenchymal state of round morphology and lacking actin stress fibers. TGF-β induces this transition by driving the expression of the actin-depolymerizing factor gelsolin, which changes a migratory, stress fiber-rich mesenchymal phenotype into a cortical actin-rich, spheroidal state. This transition lowers the biomechanical stiffness of metastatic progenitors, protecting them from killing by mechanosensitive cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells. Inhibiting this actin depolymerization process clears tissues of dormant metastatic cells. Thus, LUAD primitive progenitors undergo an atypical EMT as part of a strategy to evade immune-mediated elimination during dormancy. Our results provide a mechanistic basis and functional role of this atypical EMT response of LUAD metastatic progenitors and further illuminate the role of TGF-β as a crucial driver of immune evasive metastatic dormancy.
Collapse
Affiliation(s)
- Zhenghan Wang
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Yassmin Elbanna
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Gerstner Sloan Kettering Graduate School, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Inês Godet
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lila Peters
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Gerstner Sloan Kettering Graduate School, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - George Lampe
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Current affiliation: Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Yanyan Chen
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Current affiliation: Specialized Microscopy Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, 10032, USA
| | - Joao Xavier
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Morgan Huse
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- The Alan and Sandra Gerry Metastasis and Tumor Ecosystems Center, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
47
|
Chi WY, Hu Y, Huang HC, Kuo HH, Lin SH, Kuo CTJ, Tao J, Fan D, Huang YM, Wu AA, Hung CF, Wu TC. Molecular targets and strategies in the development of nucleic acid cancer vaccines: from shared to personalized antigens. J Biomed Sci 2024; 31:94. [PMID: 39379923 PMCID: PMC11463125 DOI: 10.1186/s12929-024-01082-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/01/2024] [Indexed: 10/10/2024] Open
Abstract
Recent breakthroughs in cancer immunotherapies have emphasized the importance of harnessing the immune system for treating cancer. Vaccines, which have traditionally been used to promote protective immunity against pathogens, are now being explored as a method to target cancer neoantigens. Over the past few years, extensive preclinical research and more than a hundred clinical trials have been dedicated to investigating various approaches to neoantigen discovery and vaccine formulations, encouraging development of personalized medicine. Nucleic acids (DNA and mRNA) have become particularly promising platform for the development of these cancer immunotherapies. This shift towards nucleic acid-based personalized vaccines has been facilitated by advancements in molecular techniques for identifying neoantigens, antigen prediction methodologies, and the development of new vaccine platforms. Generating these personalized vaccines involves a comprehensive pipeline that includes sequencing of patient tumor samples, data analysis for antigen prediction, and tailored vaccine manufacturing. In this review, we will discuss the various shared and personalized antigens used for cancer vaccine development and introduce strategies for identifying neoantigens through the characterization of gene mutation, transcription, translation and post translational modifications associated with oncogenesis. In addition, we will focus on the most up-to-date nucleic acid vaccine platforms, discuss the limitations of cancer vaccines as well as provide potential solutions, and raise key clinical and technical considerations in vaccine development.
Collapse
Affiliation(s)
- Wei-Yu Chi
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Yingying Hu
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hsin-Che Huang
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hui-Hsuan Kuo
- Pharmacology PhD Program, Weill Cornell Medicine, New York, NY, USA
| | - Shu-Hong Lin
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas Graduate School of Biomedical Sciences at Houston and MD Anderson Cancer Center, Houston, TX, USA
| | - Chun-Tien Jimmy Kuo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Julia Tao
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Darrell Fan
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Yi-Min Huang
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Annie A Wu
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Obstetrics and Gynecology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - T-C Wu
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA.
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Obstetrics and Gynecology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
48
|
Bakalenko N, Kuznetsova E, Malashicheva A. The Complex Interplay of TGF-β and Notch Signaling in the Pathogenesis of Fibrosis. Int J Mol Sci 2024; 25:10803. [PMID: 39409132 PMCID: PMC11477142 DOI: 10.3390/ijms251910803] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Fibrosis is a major medical challenge, as it leads to irreversible tissue remodeling and organ dysfunction. Its progression contributes significantly to morbidity and mortality worldwide, with limited therapeutic options available. Extensive research on the molecular mechanisms of fibrosis has revealed numerous factors and signaling pathways involved. However, the interactions between these pathways remain unclear. A comprehensive understanding of the entire signaling network that drives fibrosis is still missing. The TGF-β and Notch signaling pathways play a key role in fibrogenesis, and this review focuses on their functional interplay and molecular mechanisms. Studies have shown synergy between TGF-β and Notch cascades in fibrosis, but antagonistic interactions can also occur, especially in cardiac fibrosis. The molecular mechanisms of these interactions vary depending on the cell context. Understanding these complex and context-dependent interactions is crucial for developing effective strategies for treating fibrosis.
Collapse
Affiliation(s)
| | | | - Anna Malashicheva
- Institute of Cytology, Russian Academy of Sciences, St-Petersburg 194064, Russia; (N.B.); (E.K.)
| |
Collapse
|
49
|
Han Y, Liu C, Chen S, Sun H, Jia Z, Shi J, Wang L, Du K, Chang Y. Columbianadin ameliorates rheumatoid arthritis by attenuating synoviocyte hyperplasia through targeted vimentin to inhibit the VAV2/Rac-1 signaling pathway. J Adv Res 2024:S2090-1232(24)00432-6. [PMID: 39369957 DOI: 10.1016/j.jare.2024.09.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/13/2024] [Accepted: 09/29/2024] [Indexed: 10/08/2024] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is an autoimmune disease pathologically characterized by synovial inflammation. The abnormal activation of synoviocytes seems to accompany the progression of RA. The role and exact molecular mechanism in RA of columbianadin (CBN) which is a natural coumarin is still unclear. OBJECTIVES The present research aimed to investigate the effect of vimentin on the abnormal growth characteristics of RA synoviocytes and the targeted regulatory role of CBN. METHODS Cell migration and invasion were detected using the wound healing and transwell method. Mechanistically, the direct molecular targets of CBN were screened and identified by activity-based protein profiling. The expression of relevant proteins and mRNA in cells and mouse synovium was detected by western blotting and qRT-PCR. Changes in the degree of paw swelling and body weight of mice were recorded. H&E staining, toluidine blue staining, and micro-CT were used to visualize the degree of pathological damage in the ankle joints of mice. Small interfering RNA and plasmid overexpression of vimentin were used to observe their effects on MH7A cell proliferation, migration, apoptosis, and downstream molecular signaling. RESULTS The TNF-α-induced proliferation and migration of MH7A cells could be significantly repressed by CBN (25,50 μM), and the expression of apoptosis and autophagy-associated proteins could be modulated. Furthermore, CBN could directly bind to vimentin and inhibit its expression and function in synoviocytes, thereby ameliorating foot and paw swelling and joint damage in CIA mice. Silencing and overexpression of vimentin might be involved in developing RA synovial hyperplasia and invasive cartilage by activating VAV2 phosphorylation-mediated expression of Rac-1, which affects abnormal growth characteristics, such as synoviocyte invasion and migration. CONCLUSION CBN-targeted vimentin restrains the overactivation of RA synoviocytes thereby delaying the pathological process in CIA mice, which provides valuable targets and insights for understanding the pathological mechanisms of RA synovial hyperplasia.
Collapse
Affiliation(s)
- Yuli Han
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Changqing Liu
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shujing Chen
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Huihui Sun
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Zhaoyu Jia
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jiaxin Shi
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lirong Wang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Kunze Du
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yanxu Chang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
50
|
Zheng X, Wang Y, Qiu X. Comprehensive analysis of MAPK genes in the prognosis, immune characteristics, and drug treatment of renal clear cell carcinoma using bioinformatic analysis and Mendelian randomization. Eur J Pharmacol 2024; 980:176840. [PMID: 39038636 DOI: 10.1016/j.ejphar.2024.176840] [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: 01/23/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/24/2024]
Abstract
Mitogen-activated protein kinase (MAPK) signalling is vitally important in tumour development and progression. This study is the first to comprehensively analyse the role of MAPK-family genes in the progression, prognosis, immune-cell infiltration, methylation, and potential therapeutic value drug candidates in ccRCC. We identified a novel prognostic panel of six MAPK-signature genes (MAP3K12, MAP3K1, MAP3K5, MAPK1, MAPK8, MAPK9), and introduced a robust MAPK-signature risk model for predicting ccRCC prognosis. Model construction, evaluation, and external validation using datasets from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database demonstrated its stability, as well as high sensitivity and specificity. Enrichment analysis suggested the participation of immune-mediated mechanism in MAPK dysregulation in ccRCC. Immune-infiltration analysis confirmed the relationship and revealed that the MAPK-signature risk model might stratify immunotherapy response in ccRCC, which was verified in drug sensitivity analysis and validated in external ccRCC immunotherapy dataset (GSE67501). Potential therapeutic drug predictions for key MAPKs using DSigDB, Network Analyst, CTD, and DGIdb were subsequently verified by molecular docking with AutoDock Vina and PyMol. Mendelian randomization further demonstrated the possibilities of the MAPK-signature genes as targets for therapeutic drugs in ccRCC. Methylation analysis using UALCAN and MethSurv revealed the participation of epigenetic modifications in dysregulation and survival difference of MAPK pathway in ccRCC. Among the key MAPKs, MAP3K12 exhibited the highest significance, indicating its independent prognostic value as single gene in ccRCC. Knockout and overexpression validation experiments in vitro and in vivo found that MAP3K12 acted as a promoter of tumour progression in RCC, suggesting a pivotal role for MAP3K12 in the proliferation, migration, and invasion of RCC cells. Our findings proposed the potential of MAPK-signature genes as biomarkers for prognosis and therapy response, as well as targets for therapeutic drugs in ccRCC.
Collapse
Affiliation(s)
- Xinyi Zheng
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Yiqiu Wang
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; State Key Laboratory of Oncogenes and Related Genes, Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Xiaoyan Qiu
- Department of Pharmacy, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China.
| |
Collapse
|