1
|
Chu C, Huang Y, Cao L, Ji S, Zhu B, Shen Q. Role of macrophages in peritoneal dialysis-associated peritoneal fibrosis. Ren Fail 2025; 47:2474203. [PMID: 40044628 PMCID: PMC11884102 DOI: 10.1080/0886022x.2025.2474203] [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: 10/11/2024] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/09/2025] Open
Abstract
Peritoneal dialysis (PD) can be used as renal replacement therapy when chronic kidney disease (CKD) progresses to end-stage renal disease. However, peritoneal fibrosis (PF) is a major cause of PD failure. Studies have demonstrated that PD fluid contains a significantly larger numbers of macrophages compared with the healthy individuals. During PD, macrophages can secrete cytokines to keep peritoneal tissue in sustained low-grade inflammation, and participate in the regulation of fibrosis-related signaling pathways, such as NF-κB, TGF-β/Smad, IL4/STAT6, and PI3K/AKT. A series of basic pathological changes occurs in peritoneal tissues, including epithelial mesenchymal transformation, overgeneration of neovasculature, and abnormal deposition of extracellular matrix. This review focuses on the role of macrophages in promoting PF during PD, summarizes the targets of macrophage-related inhibition of fibrosis, and provides new ideas for clinical research on delaying PF, maintaining the function and integrity of peritoneum, prolonging duration of PD as a renal replacement modality, and achieving longer survival in CKD patients.
Collapse
Affiliation(s)
- Chenling Chu
- Department of Clinical Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Ying Huang
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
- Department of Public Health and Preventive Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Luxi Cao
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Shuiyu Ji
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Bin Zhu
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Quanquan Shen
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
- Department of Nephrology, Zhejiang Provincial People’s Hospital Bijie Hospital, Bijie, Guizhou, China
| |
Collapse
|
2
|
Liang L, Dong Z, Shen Z, Zang Y, Yang W, Wu L, Bao L. Inhibitory effects of umbelliferone on carbon tetrachloride-induced hepatic fibrosis in rats through the TGF‑β1‑Smad signaling pathway. Mol Med Rep 2025; 32:171. [PMID: 40242963 PMCID: PMC12020354 DOI: 10.3892/mmr.2025.13536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Hepatic fibrosis (HF) is a critical marker of advanced‑stage chronic liver disease and involves pivotal contributions from hepatic stellate cells (HSCs). Currently, there are no effective treatments for HF. Umbelliferone (7‑hydroxycoumarin; UMB) is a natural compound with significant anti‑inflammatory, antioxidant and anti‑tumor activities. However, its potential efficacy in treating HF has not been studied. The present study explored the protective effects of UMB against HF, targeting the TGF‑β1‑Smad signaling pathway to explore the underlying mechanisms of UMB. Carbon tetrachloride (CCl4) was injected intraperitoneally to induce HF in rats and primary HSCs were treated in vitro with UMB to investigate the improvement effect of UMB on HF. The levels of fibrosis markers, inflammation, oxidative stress and TGF‑β1‑Smad signaling pathway in the rat liver tissue and HSCs were detected using hematoxylin and eosin staining, enzyme‑linked immunosorbent assay, reverse transcription‑quantitative PCR, Cell Counting Kit‑8 and western blotting. The improvement in liver histopathology, liver function indexes and fibrosis markers demonstrated that UMB markedly inhibited the CCl4‑induced HF and inflammation in the rats. Additionally, UMB prominently reduced the pro‑inflammatory factors and oxidative stress levels. In vitro, UMB markedly inhibited primary HSC activation and decreased alpha‑smooth muscle actin and collagen I expression. The mechanism experiment proved that UMB inhibited the TGF‑β1‑Smad signaling pathway and ameliorated HF. The present study was the first to demonstrate, to the best of the authors' knowledge, that UMB might be a promising natural active compound for treating HF. Its therapeutic effect is associated with its modulation of the TGF‑β1‑Smad signaling pathway.
Collapse
Affiliation(s)
- Lijuan Liang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010110, P.R. China
| | - Zhiheng Dong
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010030, P.R. China
| | - Ziqing Shen
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010110, P.R. China
| | - Yifan Zang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010110, P.R. China
| | - Wenlong Yang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010110, P.R. China
| | - Lan Wu
- Mongolia Medical School, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010110, P.R. China
| | - Lidao Bao
- Department of Pharmacy, Hohhot First Hospital, Hohhot, Inner Mongolia Autonomous Region 010030, P.R. China
| |
Collapse
|
3
|
Zhao S, Zhang Y, Zhao Y, Lu X. Cellular senescence as a key player in chronic heart failure pathogenesis: Unraveling mechanisms and therapeutic opportunities. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:8-18. [PMID: 39961550 DOI: 10.1016/j.pbiomolbio.2025.02.002] [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: 05/12/2024] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Chronic heart failure (CHF) is the final stage of heart disease and is caused by various factors. Unfortunately, CHF has a poor prognosis and a high mortality rate. Recent studies have found that aging is a significant risk factor for the development of CHF and that cellular senescence plays a vital role in its development. This article reviews different types of cellular senescence, mitochondrial dysfunction in senescent cells, autophagy in senescent cells, and senescence-associated secretory phenotype (SASP), and epigenetic regulation, to provide new perspectives on the research and treatment of CHF.
Collapse
Affiliation(s)
- Shuqing Zhao
- The First Clinical College of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Zhang
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying Zhao
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Xiaohui Lu
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
4
|
Zheng Y, Lan T, Zhang Z, He X, Yang M, Li X, You J, Gu H, Nashun B, Guo J. Gestational Exposure to Phenanthrene Induces Superfluous Fibrosis and Calcification and Metabolic Imbalance of the Placenta in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12949-12960. [PMID: 40359265 DOI: 10.1021/acs.jafc.4c12387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
The placenta is a vital organ that facilitates maternal-fetal circulation, ensuring proper fetal development. Phenanthrene (Phe), a typical low-molecular-weight polycyclic aromatic hydrocarbon, is widely present in the environment and food. In this study, pregnant mice were exposed to Phe (0, 0.6, 6, and 60 μg/kg of body weight) via gavage every 3 days from pregnancy day 0.5 (PGD 0.5) for a total of six exposures during pregnancy. Placentas were collected on PGD of 18.5 for analysis. The results showed that Phe exposure altered placental structure and function, inducing trophoblast thickening at low doses (0.6 μg/kg) but thinning at higher doses (6 and 60 μg/kg), reducing blood cell density in the placental labyrinth, disrupting metabolite composition, causing oxidative damage, and leading to excessive fibrosis and calcification. Molecular analysis revealed that PCNA was significantly upregulated in the 0.6 μg/kg group and downregulated in 6 and 60 μg/kg groups, indicating an initial compensatory proliferative response at low doses and impaired placental proliferation at higher doses, while Bad was abnormally accumulated in trophoblasts and dose-dependently upregulated, along with decline in antioxidant capacity. Meanwhile, increases in protein levels of TGF-β1, Smad2, p-Smad2, Smad1/5/9, p-Smad1/5/9, BMP2, TIMP1, Runx2, Collagen I, and SMA, and a decrease in MMP1 level was observed. These findings suggested that Phe exposure during pregnancy induced activation of the TGF-β/Smad2 pathway and BMP2/Smad1/5/9/Runx2 pathway, which might further lead to excessive fibrosis and calcification. The abnormally increased fibrosis and calcification, together with the oxidative damage, further elevated cellular apoptosis, destroyed the structure of the placenta, and reduced blood cell counts, impairing placental exchange efficiency and leading to systemic metabolic imbalances, which might further impair the health of their offspring.
Collapse
Affiliation(s)
- Yajie Zheng
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences and Inner Mongolia Engineering Technology Research Center of Germplasm Resources Conservation and Utilization, School of Life Sciences, Inner Mongolia University, No. 49, Xilin South Road, Yuquan District, Hohhot 010000, China
| | - Tian Lan
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences and Inner Mongolia Engineering Technology Research Center of Germplasm Resources Conservation and Utilization, School of Life Sciences, Inner Mongolia University, No. 49, Xilin South Road, Yuquan District, Hohhot 010000, China
| | - Zixuan Zhang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences and State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, No. 49, Xilin South Road, Yuquan District, Hohhot 010000, China
| | - Xige He
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences and State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, No. 49, Xilin South Road, Yuquan District, Hohhot 010000, China
| | - Minhui Yang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences and State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, No. 49, Xilin South Road, Yuquan District, Hohhot 010000, China
| | - Xianghui Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences and Inner Mongolia Engineering Technology Research Center of Germplasm Resources Conservation and Utilization, School of Life Sciences, Inner Mongolia University, No. 49, Xilin South Road, Yuquan District, Hohhot 010000, China
| | - Jun You
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences and Inner Mongolia Engineering Technology Research Center of Germplasm Resources Conservation and Utilization, School of Life Sciences, Inner Mongolia University, No. 49, Xilin South Road, Yuquan District, Hohhot 010000, China
| | - Haotian Gu
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences and State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, No. 49, Xilin South Road, Yuquan District, Hohhot 010000, China
| | - Buhe Nashun
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences and State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, No. 49, Xilin South Road, Yuquan District, Hohhot 010000, China
| | - Jiaojiao Guo
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences and Inner Mongolia Engineering Technology Research Center of Germplasm Resources Conservation and Utilization, School of Life Sciences, Inner Mongolia University, No. 49, Xilin South Road, Yuquan District, Hohhot 010000, China
| |
Collapse
|
5
|
Guo H, Zhao Z, Liu L. HIF-1α modulates pancreatic cancer ECM proteins via the TGF-β1/Smad signaling pathway introduction. Front Oncol 2025; 15:1564655. [PMID: 40406267 PMCID: PMC12094911 DOI: 10.3389/fonc.2025.1564655] [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: 01/21/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Introduction Pancreatic cancer is characterized by its aggressive nature and poor prognosis, ranking among the most lethal malignancies. The tumor microenvironment, particularly the extracellular matrix (ECM), plays a crucial role in cancer progression. This study investigated the relationship between hypoxia-inducible factor-1α (HIF-1α) and transforming growth factor-β1 (TGF-β1) in regulating ECM protein expression in pancreatic cancer. Methods PANC-1 cells were cultured under both normoxic and hypoxic conditions. Pharmacological inhibition of HIF-1α and TGF-β1, as well as TGF-β1 stimulation, were employed to evaluate ECM protein expression. HIF-1α knockdown experiments and co-immunoprecipitation were performed to assess molecular interactions. Clinical specimens were analyzed for HIF-1α and TGF-β1 expression. Results HIF-1α was found to modulate ECM protein expression through the TGF-β1/Smad signaling pathway. Pharmacological inhibition of either HIF-1α or TGF-β1 significantly decreased the expression of ECM proteins, while TGF-β1 stimulation enhanced their production. HIF-1α knockdown abolished TGF-β1-induced ECM protein expression, indicating that HIF-1α is essential for TGF-β1-mediated ECM regulation. Co-immunoprecipitation experiments revealed a physical interaction between HIF-1α and TGF-β1. Clinical specimens showed significantly elevated expression of both HIF-1α and TGF-β1 in pancreatic cancer tissues compared to adjacent normal tissues, correlating with advanced disease stages. Discussion These findings elucidate a novel mechanism where HIF-1α and TGF-β1 cooperatively regulate ECM production in pancreatic cancer, providing potential therapeutic targets for intervention.
Collapse
Affiliation(s)
| | | | - Linxun Liu
- Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| |
Collapse
|
6
|
Hu S, Ai Y, Hu C, Cassim Bawa FN, Xu Y. Transcription factors, metabolic dysfunction-associated fatty liver disease, and therapeutic implications. Genes Dis 2025; 12:101372. [PMID: 39911797 PMCID: PMC11795806 DOI: 10.1016/j.gendis.2024.101372] [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: 09/08/2023] [Revised: 03/27/2024] [Accepted: 06/21/2024] [Indexed: 02/07/2025] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) encompasses a spectrum of liver diseases ranging from metabolic dysfunction-associated fatty liver to metabolic dysfunction-associated steatohepatitis, which may progress to liver cirrhosis and hepatocellular carcinoma. Several mechanisms, including obesity, insulin resistance, dyslipidemia, inflammation, apoptosis, mitochondrial dysfunction, and reactive oxygen species, have been proposed to underlie the progression of MAFLD. Transcription factors are proteins that specifically bind to DNA sequences to regulate the transcription of target genes. Numerous transcription factors regulate MAFLD by modulating the transcription of genes involved in steatosis, inflammation, apoptosis, and fibrosis. Here, we review the pathological factors associated with MAFLD, with a particular emphasis on the transcription factors that contribute to the progression of MAFLD and their therapeutic implications.
Collapse
Affiliation(s)
- Shuwei Hu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Yingjie Ai
- Department of Pathology of School of Basic Medical Sciences, Department of Gastroenterology and Hepatology of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chencheng Hu
- Department of Pathology of School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Fathima N. Cassim Bawa
- Institute of Diabetes, Obesity and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Yanyong Xu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Frontier Innovation Center, Department of Pathology of School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
7
|
Joly R, Tasnim F, Krutsinger K, Li Z, Pullen NA, Han Y. Cannabigerol Alleviates Liver Damage in Metabolic Dysfunction-Associated Steatohepatitis Female Mice via Inhibition of Transforming Growth Factor Beta 1. Nutrients 2025; 17:1524. [PMID: 40362835 PMCID: PMC12073672 DOI: 10.3390/nu17091524] [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: 03/04/2025] [Revised: 04/09/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Background and Aims: Metabolic dysfunction-associated steatohepatitis (MASH), a progressive form of metabolic dysfunction-associated steatotic liver disease (MASLD), involves inflammation, fibrosis, steatosis, and oxidative stress. Previous research from our lab shows that cannabigerol (CBG) reduces inflammation and fibrosis in male MASH mice, but its effects in females remain unknown. Given immune cell population changes in MASLD patients, this study examines CBG's impact on methionine-choline deficient (MCD) diet-induced MASH in female mice. Methods: MCD-fed female mice are supplemented with two different doses for three weeks. Liver fibrosis, steatosis, oxidative stress, ductular reaction, and inflammation are assessed via Sirius Red, Oil Red O, immunohistochemistry, and immunofluorescence staining. Immune cell changes in non-parenchymal cells (NPCs) are analyzed via flow cytometry. Results: CBG treatment improves liver health by reducing leukocyte infiltration. Both CBG doses significantly decrease fibrosis, oxidative stress, ductular proliferation, and inflammation in MCD-fed mice, including monocyte and T lymphocyte reductions. Additionally, CBG downregulates mast cell activation, inhibiting transforming growth factor (TGF)-β1 release, thereby suppressing hepatic stellate cell activation. This reduces collagen deposition, fibrosis, and ductular proliferation. Conclusions: Our findings provide insights for pre-clinical and clinical research, highlighting CBG's potential therapeutic role and dosage considerations in mitigating liver fibrosis and inflammation in female patients.
Collapse
Affiliation(s)
- Raznin Joly
- College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA;
| | - Fariha Tasnim
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA; (F.T.); (K.K.); (N.A.P.)
| | - Kelsey Krutsinger
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA; (F.T.); (K.K.); (N.A.P.)
| | - Zhuorui Li
- College of Biology, China Agricultural University, Beijing 100107, China;
| | - Nicholas A. Pullen
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA; (F.T.); (K.K.); (N.A.P.)
| | - Yuyan Han
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA; (F.T.); (K.K.); (N.A.P.)
| |
Collapse
|
8
|
Liang Z, Li S, Wang Z, Zhou J, Huang Z, Li J, Bao H, Yam JWP, Xu Y. Unraveling the Role of the Wnt Pathway in Hepatocellular Carcinoma: From Molecular Mechanisms to Therapeutic Implications. J Clin Transl Hepatol 2025; 13:315-326. [PMID: 40206274 PMCID: PMC11976435 DOI: 10.14218/jcth.2024.00401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 04/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest malignant tumors in the world, and its incidence and mortality have increased year by year. HCC research has increasingly focused on understanding its pathogenesis and developing treatments.The Wnt signaling pathway, a complex and evolutionarily conserved signal transduction system, has been extensively studied in the genesis and treatment of several malignant tumors. Recent investigations suggest that the pathogenesis of HCC may be significantly influenced by dysregulated Wnt/β-catenin signaling. This article aimed to examine the pathway that controls Wnt signaling in HCC and its mechanisms. In addition, we highlighted the role of this pathway in HCC etiology and targeted treatment.
Collapse
Affiliation(s)
- Zixin Liang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shanshan Li
- School of Pharmacy, Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, Anhui, China
| | - Zhiyu Wang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Junting Zhou
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ziyue Huang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jiehan Li
- Department of Urology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haolin Bao
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- School of Pharmacy, Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, Anhui, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
9
|
Wang W, Gao X, Niu W, Yin J, He K. Targeting Metabolism: Innovative Therapies for MASLD Unveiled. Int J Mol Sci 2025; 26:4077. [PMID: 40362316 PMCID: PMC12071536 DOI: 10.3390/ijms26094077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/01/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
The recent introduction of the term metabolic-dysfunction-associated steatotic liver disease (MASLD) has highlighted the critical role of metabolism in the disease's pathophysiology. This innovative nomenclature signifies a shift from the previous designation of non-alcoholic fatty liver disease (NAFLD), emphasizing the condition's progressive nature. Simultaneously, MASLD has become one of the most prevalent liver diseases worldwide, highlighting the urgent need for research to elucidate its etiology and develop effective treatment strategies. This review examines and delineates the revised definition of MASLD, exploring its epidemiology and the pathological changes occurring at various stages of the disease. Additionally, it identifies metabolically relevant targets within MASLD and provides a summary of the latest metabolically targeted drugs under development, including those in clinical and some preclinical stages. The review finishes with a look ahead to the future of targeted therapy for MASLD, with the goal of summarizing and providing fresh ideas and insights.
Collapse
Affiliation(s)
- Weixin Wang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.W.); (W.N.)
| | - Xin Gao
- School of Public Health, Jilin University, Changchun 130021, China;
| | - Wentong Niu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.W.); (W.N.)
| | - Jinping Yin
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130041, China;
| | - Kan He
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.W.); (W.N.)
| |
Collapse
|
10
|
Xu Y, Chen L, Liu D, Xue B, Li C, Khan AJ, Li X, Shi R. The inhibitory effect of Osthole on A549 lung adenocarcinoma cells and its biomarker. Sci Rep 2025; 15:12948. [PMID: 40234644 PMCID: PMC12000571 DOI: 10.1038/s41598-025-97305-8] [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: 05/17/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025] Open
Abstract
Some natural compounds derived from medicinal plants show anti-tumor activity with high efficacy and safety, low toxicity and residual levels etc. The aim of this study was to select natural compounds and biomarkers having high inhibitory effects against A549 adenocarcinoma cells. A total of eight natural compounds having pure plant origin were initially screened, purchased, and their potential anti-cancer activities were comprehensively and systematically evaluated against A549 lung adenocarcinoma cells. The maximum non-cytotoxic concentration (MNTC) and 50% cytotoxic concentration (CC50) of the eight compounds against A549 cells were obtained by cytopathological and MTT assays, respectively. Using Cisplatin as a positive control, the effect of selected compounds were elucidated on the proliferation, migration and invasion of A549 cells by MTT, wound healing and invasion assays, respectively. AnnexinV-FITC/PI, JC-1, ROS and Cell Cycle Kits were used to detect the pro-apoptotic mechanism of A549 cells induced by the tested compounds. qRT-PCR and RNA-seq were used to investigate the effective biomarkers involved in the inhibition process. The results showed that Curcumin, Osthole, Paeonol, Cepharanthine and Cisplatin significantly reduced the proliferation, migration and invasion abilities of A549 cells in a dose-dependent manner. Post 48 h of treatment, Osthole inhibited the metastatic ability of A549 cells by regulating mitochondrial apoptosis, arresting A549 cell in G1-phase and inhibiting release of ROS, while Curcumin, Paeonol and Cepharanthine did not showed the same response. It was therefore elucidated that Osthole was the optimal natural compound showing powerful anti-inhibitory properties against A549 cells. Moreover, the expressions of EGF, IL-2 and IL-10 genes were significantly decreased in Osthole treated group, while IL-6 gene was significantly increased. This study suggested that EGF gene has the potential to be used as a biomarker for Osthole treatment against A549 cells, involved in mitochondrial apoptosis and ROS down-regulation, inhibiting proliferation and epithelial mesenchymal transition (EMT), inflammation and immune processes in A549 cells providing a foundation to develop Osthole as a potential target drug to prevent the occurrence and development of lung adenocarcinoma.
Collapse
Affiliation(s)
- Yinlan Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| | - Lulu Chen
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Dong Liu
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Bo Xue
- School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Chaoying Li
- School of Foreign Languages, Xinxiang Medical University, Xinxiang, 453003, China
| | - AJab Khan
- Faculty of Veterinary and Animal Sciences, The University of Agriculture, Dera Ismail Khan , 29050, Khyber Pakhtunkhwa, Pakistan
| | - Xuehua Li
- Henan Engineering Laboratory for Molecular Diagnosis of Animal Diseases, School of Life Sciences & Basic Medicine, Xinxiang University, Xinxiang, 453003, Henan, China
| | - Ruling Shi
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| |
Collapse
|
11
|
Zhang L, Liu S, Zhao Q, Liu X, Zhang Q, Liu M, Zhao W. The role of ubiquitination and deubiquitination in the pathogenesis of non-alcoholic fatty liver disease. Front Immunol 2025; 16:1535362. [PMID: 40292292 PMCID: PMC12021615 DOI: 10.3389/fimmu.2025.1535362] [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: 11/27/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases and is closely associated with metabolic abnormalities. The causes of NAFLD are exceedingly complicated, and it is known that a variety of signaling pathways, endoplasmic reticulum stress, and mitochondrial dysfunction play a role in the pathogenesis of NAFLD. Recent studies have shown that ubiquitination and deubiquitination are involved in the regulation of the NAFLD pathophysiology. Protein ubiquitination is a dynamic and diverse post-translational alteration that affects various cellular biological processes. Numerous disorders, including NAFLD, exhibit imbalances in ubiquitination and deubiquitination. To highlight the significance of this post-translational modification in the pathogenesis of NAFLD and to aid in the development of new therapeutic approaches for the disease, we will discuss the role of enzymes involved in the processes of ubiquitination and deubiquitination, specifically E3 ubiquitin ligases and deubiquitinating enzymes that are important in the regulation of NAFLD.
Collapse
Affiliation(s)
- Lihui Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Sutong Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Qing Zhao
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaoyan Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Qiang Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Minghao Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Wenxiao Zhao
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| |
Collapse
|
12
|
Ding Z, Wang L, Sun J, Zheng L, Tang Y, Tang H. Hepatocellular carcinoma: pathogenesis, molecular mechanisms, and treatment advances. Front Oncol 2025; 15:1526206. [PMID: 40265012 PMCID: PMC12011620 DOI: 10.3389/fonc.2025.1526206] [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: 11/11/2024] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
Hepatocellular Carcinoma (HCC), a highly prevalent malignancy, poses a significant global health challenge. Its pathogenesis is intricate and multifactorial, involving a complex interplay of environmental and genetic factors. Viral hepatitis, excessive alcohol consumption, and cirrhosis are known to significantly elevate the risk of developing HCC. The underlying biological processes driving HCC are equally complex, encompassing aberrant activation of molecular signaling pathways, dysregulation of hepatocellular differentiation and angiogenesis, and immune dysfunction. This review delves into the multifaceted nature of HCC, exploring its etiology and the intricate molecular signaling pathways involved in its development. We examine the role of immune dysregulation in HCC progression and discuss the potential of emerging therapeutic strategies, including immune-targeted therapy and tumor-associated macrophage interventions. Additionally, we explore the potential of traditional Chinese medicine (TCM) monomers in inhibiting tumor growth. By elucidating the complex interplay of factors contributing to HCC, this review aims to provide a comprehensive understanding of the disease and highlight promising avenues for future research and therapeutic development.
Collapse
Affiliation(s)
- Zhixian Ding
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Lusheng Wang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Jiting Sun
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Lijie Zheng
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Yu Tang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Heng Tang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| |
Collapse
|
13
|
Xu Y, Yang X, Lu Y, Liu J. Hepatotoxicity from long-term administration of hepatoprotective low doses of oleanolic acid in mice. Toxicol Appl Pharmacol 2025; 497:117277. [PMID: 39984133 DOI: 10.1016/j.taap.2025.117277] [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: 11/21/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
Oleanolic acid is a triterpenoid existed in many medicinal herbs/plants. Oleanolic acid at low doses are hepatoprotective but at high doses produce cholestasis. This study examined hepatotoxicity potential of low doses of oleanolic acid after log-term administration. Male Kunming mice were orally given oleanolic acid at 100, 200 and 300 μmol/kg daily for 14 weeks. Body weights were monitored, and liver injury was determined via blood biochemistry. Histopathology was examined via H&E, Masson, and Sirius red stains. Oleanolic acid accumulation in plasma and liver was determined by LC-MS and hepatic gene expression by qPCR. Oleanolic acid at low doses did not affect animal body weights, but increased liver index. Serum alanine aminotransferase and alkaline phosphatase were increased, while total bilirubin was unchanged. Chronic oleanolic acid produced hepatocyte degeneration, spot necrosis, and fibrosis. Plasma oleanolic acid was increased more than that in the liver. Oleanolic acid increased hepatic expression of Nrf2, Nqo1, Gclc and Mgst1; Expression of bile acid synthesis genes (Cyp7a1, Cyp8b1, Cyp27a1, Cyp7b1, FXR, SHP) was also suppressed at higher doses. The expression of TGF-β1 and Smad3 was increased, while Smad7 decreased, suggesting the progression to liver fibrosis. High dose of oleanolic acid was less effective in producing these changes, probably due to increased liver injury. Overall, oral administration of low doses of oleanolic acid for 14 weeks produced liver injury and fibrosis. These harmful effects were associated with increased oleanolic acid in plasma and liver, and the disruptions of bile acid metabolism, the Nrf2 and TGF-β/Smad signaling pathways.
Collapse
Affiliation(s)
- Yasha Xu
- Key Laboratory for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China
| | - Xi Yang
- Key Laboratory for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China
| | - Yuanfu Lu
- Key Laboratory for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China; Department of Pharmacy, Zunyi Medical and Pharmaceutical College, China.
| | - Jie Liu
- Key Laboratory for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
14
|
Arafa SS, Elnoury HA, Badr El-Din S, Sakr MA, Hendawi FF, Masoud RAE, Barghash SS, Elbehairy DS, Hemeda AA, Farrag IM, Abdelrahman DS, Elsadek AM, Ghanem SK, AboShabaan HS, Atwa AM, Nour El Din M, Radwan AF, Al-Zahrani M, Alhomodi AF, Abdulfattah AM, Abdelkader A. Acetamiprid-induced pulmonary toxicity via oxidative stress, epithelial-mesenchymal transition, apoptosis, and extracellular matrix accumulation in human lung epithelial cells and fibroblasts: Protective role of heat-killed Lactobacilli. Food Chem Toxicol 2025; 198:115322. [PMID: 39961414 DOI: 10.1016/j.fct.2025.115322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
Acetamiprid (ACE) is a neonicotinoid insecticide with widespread global application, resulting in persistent human exposure. The current research examined the toxicological implications of ACE exposure on human lung fibroblasts (MRC-5 cells) and bronchial epithelial cells (BEAS-2B cells). The following implications were explored: oxidative stress, epithelial-mesenchymal transition, apoptosis, cellular proliferation, and extracellular matrix accumulation. The prospective protective properties of heat-killed Lactobacillus fermentum and Lactobacillus delbrueckii (HKL) were further studied. The 14-day exposure to ACE at 4 μM triggered oxidative stress and inflammation. ACE promoted epithelial-mesenchymal transition, as evidenced by the decline of protein and mRNA abundances of E-cadherin alongside increased protein and mRNA quantities of α-SMA and N-cadherin in BEAS-2B cells. Additionally, it elicited apoptosis in BEAS-2B cells and stimulated the cellular growth of MRC-5 cells. The TGF-β1/Smad pathway was activated upon ACE exposure, leading to the accumulation of extracellular matrix. HKL demonstrated antioxidant, anti-apoptotic, anti-proliferative, and anti-fibrotic properties, mitigating ACE-induced toxicity. Our findings delineate the molecular mechanisms underlying epithelial-mesenchymal transition, inflammation, oxidative stress, and extracellular matrix accumulation in ACE-induced pulmonary fibrosis, which provides new insights into pulmonary injury. Additionally, this investigation would offer us an approach to mitigate lung deterioration induced by ACE through utilizing heat-killed probiotic supplementation.
Collapse
Affiliation(s)
- Samah S Arafa
- Department of Pesticides, Faculty of Agriculture, Menoufia University, Shibin Elkom, Egypt.
| | - Heba A Elnoury
- Department of Pharmacology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Sahar Badr El-Din
- Department of Pharmacology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Mohamed A Sakr
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Suez University, Suez, Egypt
| | - Fatma Fawzi Hendawi
- Department of Pharmacology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Rehab Ali Elsayed Masoud
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Samia Soliman Barghash
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt; Department of Pharmacology and Toxicology, Pharmacy College, Qassim University, Saudi Arabia
| | - Doaa Sabry Elbehairy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Ayat Abdelaty Hemeda
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Islam Mostafa Farrag
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Doaa Sayed Abdelrahman
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Amira Mohammad Elsadek
- Department of Chest Diseases, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Sahar K Ghanem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sohag University, Sohag, Egypt
| | - Hind S AboShabaan
- Department of Clinical Pathology, National Liver Institute Hospital, Menoufia University, Shibin Elkom, Egypt
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Ayen Iraqi University, Thi-Qar, Iraq
| | - Mahmoud Nour El Din
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Sadat City, Cairo, Egypt
| | - Abdullah F Radwan
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt; Department of Pharmacy, Kut University College, Al Kut, Wasit, Iraq
| | - Majid Al-Zahrani
- Department of Biological Sciences, College of Sciences and Art, King Abdulaziz University, Rabigh, Saudi Arabia
| | - Ahmad F Alhomodi
- Department of Biology, College of Science and Arts, Najran University, Saudi Arabia
| | - Ahmed M Abdulfattah
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia; Embryonic Stem Cell Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Afaf Abdelkader
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha, Egypt
| |
Collapse
|
15
|
Yoon J, Choi WI, Lee WH, Lee GB, Choi BW, Kim P, Heo Y, Kim DG, Kim HA, Bae MA, Kim SS, Lee EY, Oh CM, Lee HJ, Kim HW, Namkung W, Kim H, Ahn JH. Synthesis and Biological Evaluation of Peripheral 5HT 2B Antagonists for Liver Fibrosis. J Med Chem 2025; 68:6493-6506. [PMID: 40048549 DOI: 10.1021/acs.jmedchem.4c03003] [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: 03/28/2025]
Abstract
Liver fibrosis is characterized by an excessive accumulation of extracellular matrix components, leading to the distortion of liver architecture and function. Recent studies have shown that antagonizing 5-hydroxytryptamine receptor 2B (5HT2B) stimulates the apoptosis of activated hepatic stellate cells and inhibits their proliferation while concurrently regressing hepatocyte proliferation. In this study, we present compound 19c, which demonstrates promising efficacy both in vitro and in vivo. 19c showed robust in vitro activity with an IC50 value of 1.09 nM and limited blood-brain barrier penetration. Furthermore, 19c did not significantly inhibit hERG and cytochrome P450 enzymes. 19c markedly reduced fibrotic deposition, with a decrease in fibrosis stage and area in the CCl4-induced liver fibrosis mouse model. Additionally, treatment with 19c led to downregulation of key fibrosis-related genes, including α-SMA, Timp1, Col1a1, and Col3a1. Taken together, these results suggest that 19c has the potential to be a novel antifibrotic agent.
Collapse
Affiliation(s)
- Jihyeon Yoon
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Won-Il Choi
- Department of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Won Hee Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Biomedical Research Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Gwi Bin Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Byeong Wook Choi
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Pyeongkeun Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Yerim Heo
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Dong Gun Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hyeon Ah Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Myung Ae Bae
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Seong Soon Kim
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Eun Young Lee
- JD Bioscience Inc., TJS Knowledge Industrial Center, Suite 801, 208 Beon-gil Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hyeok Jae Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hyun Woo Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- Center for Quantum Information, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Wan Namkung
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Biomedical Research Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jin Hee Ahn
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- JD Bioscience Inc., TJS Knowledge Industrial Center, Suite 801, 208 Beon-gil Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| |
Collapse
|
16
|
Li Q, Xiao N, Zhang H, Liang G, Lin Y, Qian Z, Yang X, Yang J, Fu Y, Zhang C, Liu A. Systemic aging and aging-related diseases. FASEB J 2025; 39:e70430. [PMID: 40022602 DOI: 10.1096/fj.202402479rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/07/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Aging is a biological process along with systemic and multiple organ dysfunction. It is more and more recognized that aging is a systemic disease instead of a single-organ functional disorder. Systemic aging plays a profound role in multiple diseases including neurodegenerative diseases, cardiovascular diseases, and malignant diseases. Aged organs communicate with other organs and accelerate aging. Skeletal muscle, heart, bone marrow, skin, and liver communicate with each other through organ-organ crosstalk. The crosstalk can be mediated by metabolites including lipids, glucose, short-chain fatty acids (SCFA), inflammatory cytokines, and exosomes. Metabolic disorders including hyperglycemia, hyperinsulinemia, and hypercholesterolemia caused by chronic diseases accelerate hallmarks of aging. Systemic aging leads to the destruction of systemic hemostasis, causes the release of inflammatory cytokines, senescence-associated secretory phenotype (SASP), and the imbalance of microbiota composition. Released inflammatory factors further aggregate senescence, which promotes the aging of multiple solid organs. Targeting senescence or delaying aging is emerging as a critical health strategy for solving age-related diseases, especially in the old population. In the current review, we will delineate the mechanisms of organ crosstalk in systemic aging and age-related diseases to provide therapeutic targets for delaying aging.
Collapse
Affiliation(s)
- Qiao Li
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Nanyin Xiao
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Heng Zhang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Guangyu Liang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yan Lin
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zonghao Qian
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiao Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jiankun Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yanguang Fu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Cuntai Zhang
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
17
|
Airola C, Varca S, Del Gaudio A, Pizzolante F. The Covert Side of Ascites in Cirrhosis: Cellular and Molecular Aspects. Biomedicines 2025; 13:680. [PMID: 40149656 PMCID: PMC11940454 DOI: 10.3390/biomedicines13030680] [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: 12/21/2024] [Revised: 02/25/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Ascites, a common complication of portal hypertension in cirrhosis, is characterized by the accumulation of fluid within the peritoneal cavity. While traditional theories focus on hemodynamic alterations and renin-angiotensin-aldosterone system (RAAS) activation, recent research highlights the intricate interplay of molecular and cellular mechanisms. Inflammation, mediated by cytokines (interleukin-1, interleukin-4, interleukin-6, tumor necrosis factor-α), chemokines (chemokine ligand 21, C-X-C motif chemokine ligand 12), and reactive oxygen species (ROS), plays a pivotal role. Besides pro-inflammatory cytokines, hepatic stellate cells (HSCs), sinusoidal endothelial cells (SECs), and smooth muscle cells (SMCs) contribute to the process through their activation and altered functions. Once activated, these cell types can worsen ascites accumulationthrough extracellular matrix (ECM) deposition and paracrine signals. Besides this, macrophages, both resident and infiltrating, through their plasticity, participate in this complex crosstalk by promoting inflammation and dysregulating lymphatic system reabsorption. Indeed, the lymphatic system and lymphangiogenesis, essential for fluid reabsorption, is dysregulated in cirrhosis, exacerbating ascites. The gut microbiota and intestinal barrier alterations which occur in cirrhosis and portal hypertension also play a role by inducing inflammation, creating a vicious circle which worsens portal hypertension and fluid accumulation. This review aims to gather these aspects of ascites pathophysiology which are usually less considered and to date have not been addressed using specific therapy. Nonetheless, it emphasizes the need for further research to understand the complex interactions among these mechanisms, ultimately leading to targeted interventions in specific molecular pathways, aiming towards the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Carlo Airola
- CEMAD Centro Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.V.); (A.D.G.)
- Facoltà di Medicina e Chirurgia, Università Cattolica Sacro Cuore, Largo Agostino Gemelli, 8, 00168 Rome, Italy
| | - Simone Varca
- CEMAD Centro Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.V.); (A.D.G.)
- Facoltà di Medicina e Chirurgia, Università Cattolica Sacro Cuore, Largo Agostino Gemelli, 8, 00168 Rome, Italy
| | - Angelo Del Gaudio
- CEMAD Centro Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.V.); (A.D.G.)
- Facoltà di Medicina e Chirurgia, Università Cattolica Sacro Cuore, Largo Agostino Gemelli, 8, 00168 Rome, Italy
| | - Fabrizio Pizzolante
- CEMAD Centro Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.V.); (A.D.G.)
| |
Collapse
|
18
|
Boel F, Akimov V, Teuchler M, Terkelsen MK, Wernberg CW, Larsen FT, Hallenborg P, Lauridsen MM, Krag A, Mandrup S, Ravnskjær K, Blagoev B. Deep proteome profiling of metabolic dysfunction-associated steatotic liver disease. COMMUNICATIONS MEDICINE 2025; 5:56. [PMID: 40032974 PMCID: PMC11876662 DOI: 10.1038/s43856-025-00780-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) affects roughly 1 in 3 adults and is a leading cause of liver transplants and liver related mortality. A deeper understanding of disease pathogenesis is essential to assist in developing blood-based biomarkers. METHODS Here, we use data-independent acquisition mass spectrometry to assess disease-state associated protein profiles in human liver, blood plasma, and white adipose tissue (WAT). RESULTS In liver, we find that MASLD is associated with an increased abundance of proteins involved in immune response and extracellular matrix (ECM) and a decrease in proteins involved in metabolism. Cell type deconvolution of the proteome indicates liver endothelial and hepatic stellate cells are the main source of ECM rearrangements, and hepatocytes are the major contributor to the changes in liver metabolism. In the blood, profiles of several MASLD-associated proteins correlate with expression in WAT rather than liver and so could serve as suitable liver disease predictors in a multi-protein panel marker. Moreover, our proteomics-based logistic regression models perform better than existing methods for predicting MASLD and liver fibrosis from human blood samples. CONCLUSIONS Our comprehensive proteomic analysis deepens the understanding of liver function and MASLD pathology by elucidating key cellular mechanisms and multi-organ interactions, and demonstrates the robustness of a proteomics-based biomarker panel to enhance diagnosis of MASLD and significant fibrosis.
Collapse
Affiliation(s)
- Felix Boel
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, Odense M, Denmark
| | - Vyacheslav Akimov
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, Odense M, Denmark
| | - Mathias Teuchler
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, Odense M, Denmark
| | - Mike Krogh Terkelsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, Odense M, Denmark
| | - Charlotte Wilhelmina Wernberg
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, Odense M, Denmark
- Department of Gastroenterology and Hepatology, University Hospital of Southern Denmark, Esbjerg, Denmark
| | - Frederik Tibert Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, Odense M, Denmark
| | - Philip Hallenborg
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Mette Munk Lauridsen
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, Odense M, Denmark
- Department of Gastroenterology and Hepatology, University Hospital of Southern Denmark, Esbjerg, Denmark
| | - Aleksander Krag
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, Odense M, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense M, Denmark
| | - Susanne Mandrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, Odense M, Denmark
| | - Kim Ravnskjær
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, Odense M, Denmark
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark.
- Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, Odense M, Denmark.
| |
Collapse
|
19
|
Wang T, Xia G, Li X, Gong M, Lv X. Endoplasmic reticulum stress in liver fibrosis: Mechanisms and therapeutic potential. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167695. [PMID: 39864668 DOI: 10.1016/j.bbadis.2025.167695] [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: 09/29/2024] [Revised: 01/07/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
This paper reviews the important role of endoplasmic reticulum stress in the patho mechanism of liver fibrosis and its potential as a potential target for the treatment of liver fibrosis. Liver fibrosis is the result of sustained inflammation and injury to the liver due to a variety of factors, triggering excessive deposition of extracellular matrix and fibrous scar formation, which in turn leads to loss of liver function and a variety of related complications. Endoplasmic reticulum stress is one of the characteristics of chronic liver disease and is closely related to the pathological process of chronic liver disease, including alcohol-related liver disease, viral hepatitis, and liver fibrosis. The unfolded protein response is one of the important response mechanisms to endoplasmic reticulum stress. It is associated with several pathological aspects of liver fibrosis and the maintenance of endoplasmic reticulum homeostasis. Interventions targeting endoplasmic reticulum stress for the treatment of liver fibrosis have potential research and application value. An in-depth understanding of the biological basis of endoplasmic reticulum stress is also needed in the treatment of liver fibrosis, as well as the development of more effective drugs and interventions to accurately regulate the endoplasmic reticulum signaling network, to achieve the restoration and maintenance of endoplasmic reticulum homeostasis at the cellular and organ levels, and to further promote the reversal of the pathological process of liver fibrosis.
Collapse
Affiliation(s)
- Tiantian Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Hefei, China; School of Pharmacy, Anhui Medical University, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Guoqing Xia
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Hefei, China; School of Pharmacy, Anhui Medical University, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xue Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Hefei, China; School of Pharmacy, Anhui Medical University, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Mingxu Gong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Hefei, China; School of Pharmacy, Anhui Medical University, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiongwen Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Hefei, China; School of Pharmacy, Anhui Medical University, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China.
| |
Collapse
|
20
|
Mao Y, Xie Z, Zhang X, Fu Y, Yu X, Deng L, Zhang X, Hou B, Wang X, Ma M, Ren F. Ergothioneine Ameliorates Liver Fibrosis by Inhibiting Glycerophospholipids Metabolism and TGF-β/Smads Signaling Pathway: Based on Metabonomics and Network Pharmacology. J Appl Toxicol 2025; 45:514-530. [PMID: 39579000 DOI: 10.1002/jat.4728] [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: 09/22/2024] [Revised: 10/23/2024] [Accepted: 11/06/2024] [Indexed: 11/24/2024]
Abstract
Ergothioneine (EGT) is a diet-derived natural sulfur-containing amino acid that exhibits strong anti-oxidant and anti-inflammation activities. Oxidative stress and chronic inflammatory injury are predominant pro-fibrogenic factors. Therefore, EGT may have therapeutic potential against liver fibrosis; however, its underlying mechanism is incompletely understood. This study aimed at investigating the protective effects of EGT on liver fibrosis based on metabonomics and network pharmacology. A mouse model of liver fibrosis was established by intraperitoneal injection with 40% CCl4 solution (2 mL/kg, twice a week) and intragastric administration with EGT (5, 10 mg/kg/d) for six weeks. Results showed that EGT improved liver function by reducing serum levels of ALT (alanine aminotransferase), AST (aspartate aminotransferase), and TBIL (total bilirubin), and alleviated liver fibrosis by reducing LN (laminin) and HyP (hydroxyproline) levels, decreasing expressions of α-SMA (α-smooth muscle actin), Col-I (collagen type I), and Col-III (collagen type III), and improving pathological changes. EGT also significantly inhibited CCl4-induced hepatic inflammation and TGF-β/Smads signaling pathway. Metabolomics identified six key metabolic pathways, such as purine metabolism, glycerophospholipid metabolism, and sphingolipid metabolism, and eight key metabolites, such as xanthine, guanine, ATP, phosphatidylcholine, and sphingosine. Network pharmacology analysis showed that IL-17, cAMP and NF-κB signaling pathways were potential key mechanisms. Integrated analysis revealed that PLA2G2A might be a potential target of EGT against liver fibrosis. EGT may inhibit the glycerophospholipid metabolism through PLA2G2A to inhibit the TGF-β/Smads signaling pathway, thereby alleviating fibrosis. The present study indicates that EGT may be considered a valid therapeutic strategy to regress liver fibrosis, and provides novel insights into the pharmacological mechanism of EGT against liver fibrosis.
Collapse
Affiliation(s)
- Yaping Mao
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Zhenghui Xie
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Xiangxia Zhang
- Department of Anatomy, School of Basic Medicine, Shenyang Medical College, Shenyang, China
- Department of Morphology, School of Nursing and Health, Qingdao Huanghai University, Qingdao, China
| | - Yu Fu
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Xiaotong Yu
- Department of Anatomy, School of Basic Medicine, Shenyang Medical College, Shenyang, China
| | - Lili Deng
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Xiu Zhang
- Department of Anatomy, School of Basic Medicine, Shenyang Medical College, Shenyang, China
| | - Bo Hou
- Department of Morphology, School of Nursing and Health, Qingdao Huanghai University, Qingdao, China
| | - Xiao Wang
- Department of Gastroenterology, Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| | - Mingyue Ma
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Fu Ren
- Department of Anatomy, School of Basic Medicine, Shenyang Medical College, Shenyang, China
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang, China
| |
Collapse
|
21
|
Meng X, Wang D, Zhang H, Kang T, Meng X, Liang S. Portulaca oleracea L. extract relieve mice liver fibrosis by inhibiting TLR-4/NF-κB, Bcl-2/Bax and TGF-β1/Smad2 signalling transduction. Nat Prod Res 2025; 39:1435-1443. [PMID: 38164691 DOI: 10.1080/14786419.2023.2300034] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Portulaca oleracea L. are annual herb, which has various pharmacological effects including hepatoprotective property. However, the effect of Portulaca oleracea L. (POL-1) in mice with carbon tetrachloride (CCl4)-induced liver fibrosis and its mechanism of action have not been clarified. POL-1 ameliorated the CCl4-induced liver fibrosis in mice, as shown by decreased collagen deposition and the decreased expression of liver fibrosis marker collagen I and α-smooth muscle actin (α-SMA) mRNA. In addition, treatment with POL-1 suppressed the proliferation of activated human hepatic stellate cell line (LX-2). POL-1 inhibited the oxidative stress and inflammation in fibrotic livers of mice. Mechanistically, POL-1 inhibited the CCl4-induced expression of toll-like receptor-4 (TLR4), myeloid differentiation factor 88 (MyD88), nuclear factor kappa-B (NF-κBp65) p65, Bcl2-associated X (Bax), transforming growth factor-β1 (TGF-β1) and drosophila mothers against decapentaplegic 2 (Smad2) proteins, upregulated B-cell lymphoma -2 (Bcl-2) proteins in livers of mice. These findings suggested that POL-1 attenuated liver fibrosis.
Collapse
Affiliation(s)
- Xianqun Meng
- Department of Traditional Chinese Medicine Identification, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Dan Wang
- Department of Traditional Chinese Medicine Identification, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Hui Zhang
- Department of Traditional Chinese Medicine Identification, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Tingguo Kang
- Department of Traditional Chinese Medicine Identification, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Xiansheng Meng
- Department of Traditional Chinese Medicine Identification, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Shanshan Liang
- Plant Polysaccharide Research Center, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
22
|
Zhang J, Wang H, Wang Q, Mo J, Fu L, Peng S. EEF1A2 identified as a hub gene associated with the severity of metabolic dysfunction-associated steatotic liver disease. Mamm Genome 2025; 36:93-105. [PMID: 39414652 DOI: 10.1007/s00335-024-10078-9] [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: 07/18/2024] [Accepted: 10/10/2024] [Indexed: 10/18/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent chronic liver disease that ranges from metabolic dysfunction-associated steatotic liver (MASL) to metabolic dysfunction-associated steatohepatitis (MASH), and may eventually progress to cirrhosis and hepatocellular carcinoma (HCC). The underlying mechanism of MASLD remains incompletely understood. This study aimed to identify key gene implicated in MASLD pathogenesis and validate its correlation with disease severity through an integration of bioinformatics and experimental approaches. Liver transcriptome data from MASLD patients were obtained from the Gene Expression Omnibus (GEO) database. A diet-induced MASLD mouse model was developed, and liver RNA-sequencing was performed. Liver specimens and clinical data from patients were collected for further analysis. A total of 120 differentially expressed genes (DEGs) were shared between datasets GSE89632 and GSE213621, with functional enrichment in inflammatory, metabolic, and cell cycle-related pathways. Protein-protein interaction (PPI) network analysis identified three modules associated with MASLD, with the cell cycle-related module being the most notable. EEF1A2 was identified as a novel hub gene and revealed to be elevated with MASLD progression through dataset analysis. EEF1A2 was confirmed to be highly expressed in the livers of both MASLD mouse models and patients. Moreover, the increased expression of EEF1A2 in MASH was positively correlated with higher serum alanine aminotransferase (ALT), alanine aminotransferase (AST), total cholesterol (TC), and body mass index (BMI). In conclusion, EEF1A2 is a novel hub gene significantly associated with MASLD severity and is a promising biomarker and therapeutic target for MASLD.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huiwen Wang
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qianbing Wang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan Mo
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Fu
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Shifang Peng
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
23
|
Liao Z, Tang X, Yang B, Yang J. Dopamine receptors and organ fibrosis. Biochem Biophys Rep 2025; 41:101910. [PMID: 39867679 PMCID: PMC11761258 DOI: 10.1016/j.bbrep.2024.101910] [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: 08/12/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 01/28/2025] Open
Abstract
Organ fibrosis, considered as a major global health concern, is a pathological condition often occurring after tissue injury in various organs. The pathogenesis of fibrosis involves multiple phases and multiple cell types. Dopamine is involved in various life activities by activating five receptors (D1, D2, D3, D4, D5). Activation or loss of function of dopamine receptors has been reported to be associated with the fibrosis of several organs, such as ocular, lung, liver, heart, and kidney. In this paper, we review dopamine receptors' potential roles in organ fibrosis and mechanisms by which organ fibrosis develops or decreases when dopamine receptors function is activated or perturbed.
Collapse
Affiliation(s)
- ZhongLi Liao
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 400030, China
- Department of Gastroenterology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - XueFeng Tang
- Department of Pathology, Chongqing General Hospital, Chongqing University, Chongqing, 400030, China
| | - Bin Yang
- Department of Anesthesiology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Fujian, 361000, China
| | - Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 400030, China
| |
Collapse
|
24
|
Gu XY, Gu SL, Chen ZY, Tong JL, Li XY, Dong H, Zhang CY, Qian WX, Ma XC, Yi CH, Yi YX. Uncovering immune cell heterogeneity in hepatocellular carcinoma by combining single-cell RNA sequencing with T-cell receptor sequencing. World J Hepatol 2025; 17:99046. [PMID: 40027555 PMCID: PMC11866147 DOI: 10.4254/wjh.v17.i2.99046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/13/2024] [Accepted: 12/31/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Understanding the status and function of tumor-infiltrating immune cells is essential for improving immunotherapeutic effects and predicting the clinical response in human patients with carcinoma. However, little is known about tumor-infiltrating immune cells, and the corresponding research results in hepatocellular carcinoma (HCC) are limited. AIM To investigate potential biomarker genes that are important for the development of HCC and to understand how immune cell subsets react throughout this process. METHODS Using single-cell RNA sequencing and T-cell receptor sequencing, the heterogeneity and potential functions of immune cell subpopulations from HCC tissue and normal tissue adjacent to carcinoma, as well as their possible interactions, were analyzed. RESULTS Eight T-cell clusters from patients were analyzed and identified using bioinformatics, including six typical major T-cell clusters and two newly identified T-cell clusters, among which Fc epsilon receptor 1G+ T cells were characterized by the upregulation of Fc epsilon receptor 1G, tyrosine kinase binding protein, and T cell receptor delta constant, whereas metallothionein 1E+ T cells proliferated significantly in tumors. Differentially expressed genes, such as regulator of cell cycle, cysteine and serine rich nuclear protein 1, SMAD7 and metallothionein 1E, were identified as significantly upregulated in tumors and have potential as biomarkers. In association with T-cell receptor analysis, we inferred the clonal expansion characteristics of each T-cell cluster in HCC patients. CONCLUSION We identified lymphocyte subpopulations and potential biomarker genes critical for HCC development and revealed the clonal amplification of infiltrating T cells. These data provide valuable resources for understanding the response of immune cell subsets in HCC.
Collapse
Affiliation(s)
- Xin-Yu Gu
- Department of Infectious Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, Jiangsu Province, China
- Department of General Surgery, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu 215500, Jiangsu Province, China
| | - Shuang-Lin Gu
- Department of Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, Jiangsu Province, China
| | - Zi-Yi Chen
- Genetic Center, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410078, Hunan Province, China
| | - Jin-Long Tong
- Department of Infectious Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, Jiangsu Province, China
| | - Xiao-Yue Li
- Department of Infectious Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, Jiangsu Province, China
| | - Hui Dong
- Department of Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, Jiangsu Province, China
| | - Cai-Yun Zhang
- Department of Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, Jiangsu Province, China
| | - Wen-Xian Qian
- Department of Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, Jiangsu Province, China
| | - Xiu-Chang Ma
- Department of Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, Jiangsu Province, China
| | - Chang-Hua Yi
- Department of Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, Jiangsu Province, China
- College of Medical Technology, Shaoyang University, Shaoyang 422000, Hunan Province, China
| | - Yong-Xiang Yi
- Department of Infectious Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, Jiangsu Province, China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China.
| |
Collapse
|
25
|
Bourganou MV, Chondrogianni ME, Kyrou I, Flessa CM, Chatzigeorgiou A, Oikonomou E, Lambadiari V, Randeva HS, Kassi E. Unraveling Metabolic Dysfunction-Associated Steatotic Liver Disease Through the Use of Omics Technologies. Int J Mol Sci 2025; 26:1589. [PMID: 40004054 PMCID: PMC11855544 DOI: 10.3390/ijms26041589] [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: 01/05/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), now referred to as metabolic dysfunction-associated steatotic liver disease (MASLD), is the most prevalent liver disorder globally, linked to obesity, type 2 diabetes, and cardiovascular risk. Understanding its potential progression from simple steatosis to cirrhosis and hepatocellular carcinoma (HCC) is crucial for patient management and treatment strategies. The disease's complexity requires innovative approaches for early detection and personalized care. Omics technologies-such as genomics, transcriptomics, proteomics, metabolomics, and exposomics-are revolutionizing the study of MASLD. These high-throughput techniques allow for a deeper exploration of the molecular mechanisms driving disease progression. Genomics can identify genetic predispositions, whilst transcriptomics and proteomics reveal changes in gene expression and protein profiles during disease evolution. Metabolomics offers insights into the metabolic alterations associated with MASLD, while exposomics links environmental exposures to MASLD progression and pathology. By integrating data from various omics platforms, researchers can map out the intricate biochemical pathways involved in liver disease progression. This review discusses the roles of omics technologies in enhancing the understanding of disease progression and highlights potential diagnostic and therapeutic targets within the MASLD spectrum, emphasizing the need for non-invasive tools in disease staging and treatment development.
Collapse
Affiliation(s)
- Maria V. Bourganou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
| | - Maria Eleni Chondrogianni
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioannis Kyrou
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- College of Health, Psychology and Social Care, University of Derby, Derby DE22 IGB, UK
| | - Christina-Maria Flessa
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, “Sotiria” Thoracic Diseases Hospital of Athens, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vaia Lambadiari
- 2nd Department of Internal-Medicine, Diabetes Centre, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
26
|
Sun SH, Fan HH, Wang XW, Bing BD, Hu YJ. Platelet-rich fibrin attenuates inflammation and fibrosis in vulvar lichen sclerosus via the TGF-β/SMAD3 pathway. Arch Dermatol Res 2025; 317:360. [PMID: 39918612 DOI: 10.1007/s00403-025-03811-0] [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: 10/30/2024] [Revised: 12/14/2024] [Accepted: 01/03/2025] [Indexed: 02/09/2025]
Abstract
Vulvar lichen sclerosus (VLS) is a chronic, inflammatory, and progressive skin disease mainly involved in the anogenital area. Platelet-rich fibrin (PRF) is a fibrin adhesive-concentrated platelet-rich plasma (PRP) used for tissue repair and angiogenesis. In this study, we explored the effects of PRF on VLS patients, further to utilize the established VLS animal model, to confirm the therapeutic effect of PRF and regulation on the TGF-β/SMAD3 pathway. Among the 46 VLS patients included in the analysis, injectable RPF (i-PRF) treatment improved the symptoms of VLS. The immunohistochemical analysis showed that i-PRF decreased the local blurring of the dermal-epidermal boundary and increased the number of basal keratinocytes. I-PRF increased positive PGD9.5, CD34, and Melan A cell numbers, and decreases positive IL-17 and INF-γ cell numbers in VLS tissues. In the VLS rat model, i-PRF reduced inflammatory factors IL-17 and INF-γ via inhibition of NF-κB and increased CD31 and VEGF expression in external genital tissue. The i-PRF decreases fibronectin and collagen-I by inhibiting TGF-β/SMAD3 in VLS, which is the main factor that triggers inflammation and fibrosis of the external genital skin.
Collapse
Affiliation(s)
- Shu-Hong Sun
- Clinical School of Obstetrics and Gynecology Center, Tianjin Medical University, Tianjin, China
- Department of Obstetrics and Gynecology, Chengde Maternal and Child Health Care Hospital, Chengde, Hebei, China
| | - Hong-He Fan
- Department of Obstetrics and Gynecology, Chengde Maternal and Child Health Care Hospital, Chengde, Hebei, China
| | - Xiao-Wei Wang
- Department of Obstetrics and Gynecology, Chengde Maternal and Child Health Care Hospital, Chengde, Hebei, China
| | - Bo-Dong Bing
- Department of Obstetrics and Gynecology, Chengde Maternal and Child Health Care Hospital, Chengde, Hebei, China
| | - Yuan-Jing Hu
- Department of Gynecology Oncology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China.
| |
Collapse
|
27
|
Li Y, Xing Y, Liu N, Liu B, Wang Z. SOX9: a key transcriptional regulator in organ fibrosis. Front Pharmacol 2025; 16:1507282. [PMID: 39974732 PMCID: PMC11835943 DOI: 10.3389/fphar.2025.1507282] [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: 10/07/2024] [Accepted: 01/17/2025] [Indexed: 02/21/2025] Open
Abstract
The SOX9 gene locus is not only extensive but also intricate, and it could promote fibrosis in different organs or tissues, including cardiac fibrosis, liver fibrosis, kidney fibrosis, pulmonary fibrosis, as well as other organ fibrosis. Many disorders are associated with the process of fibrosis; moreover, fibrosis is a common symptom of chronic inflammatory diseases, characterized by the accumulation of excessive components in the extracellular matrix through different signaling pathways. The advanced stage of the fibrotic process leads to organ dysfunction and, ultimately, death. In this review, we first give an overview of the original structure and functions of SOX9. Second, we will discuss the role of SOX9 in fibrosis in various organs or tissues. Third, we describe and reveal the possibility of SOX9 as an antifibrotic treatment target. Finally, we will focus on the application of novel technologies for SOX9 and the subsequent investigation of fibrosis.
Collapse
Affiliation(s)
| | | | | | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Zhihui Wang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
28
|
Xu J, Li Y, Feng Z, Chen H. Cigarette Smoke Contributes to the Progression of MASLD: From the Molecular Mechanisms to Therapy. Cells 2025; 14:221. [PMID: 39937012 PMCID: PMC11816580 DOI: 10.3390/cells14030221] [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: 12/11/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
Cigarette smoke (CS), an intricate blend comprising over 4000 compounds, induces abnormal cellular reactions that harm multiple tissues. Non-alcoholic fatty liver disease (NAFLD) is a prevalent chronic liver disease (CLD), encompassing non-alcoholic fatty liver (NAFL), non-alcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma (HCC). Recently, the term NAFLD has been changed to metabolic dysfunction-associated steatotic liver disease (MASLD), and NASH has been renamed metabolic dysfunction-associated steatohepatitis (MASH). A multitude of experiments have confirmed the association between CS and the incidence and progression of MASLD. However, the specific signaling pathways involved need to be updated with new scientific discoveries. CS exposure can disrupt lipid metabolism, induce inflammation and apoptosis, and stimulate liver fibrosis through multiple signaling pathways that promote the progression of MASLD. Currently, there is no officially approved efficacious pharmaceutical intervention in clinical practice. Therefore, lifestyle modifications have emerged as the primary therapeutic approach for managing MASLD. Smoking cessation and the application of a series of natural ingredients have been shown to ameliorate pathological changes in the liver induced by CS, potentially serving as an effective approach to decelerating MASLD development. This article aims to elucidate the specific signaling pathways through which smoking promotes MASLD, while summarizing the reversal factors identified in recent studies, thereby offering novel insights for future research on and the treatment of MASLD.
Collapse
Affiliation(s)
- Jiatong Xu
- Queen Mary School, Medical College, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (Z.F.)
| | - Yifan Li
- Queen Mary School, Medical College, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (Z.F.)
| | - Zixuan Feng
- Queen Mary School, Medical College, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (Z.F.)
| | - Hongping Chen
- Department of Histology and Embryology, Jiangxi Medical College, Nanchang University, Nanchang 330019, China
| |
Collapse
|
29
|
Hou JP, Yang LY, Liu LB, Han EK, Han CQ, Yang LP. Correlation of local and serum CircHIPK3 expressions with the progression of liver fibrosis/cirrhosis. Arab J Gastroenterol 2025; 26:71-77. [PMID: 39765392 DOI: 10.1016/j.ajg.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/24/2024] [Accepted: 11/24/2024] [Indexed: 03/16/2025]
Abstract
BACKGROUND AND STUDY AIMS This study was aimed to validate the correlation of circular RNA HIPK3 (CircHIPK3) expression in serum and tissues with the progression of liver fibrosis (LF) and liver cirrhosis (LC). PATIENTS AND METHODS Serum CircHIPK3 expressions were detected in 120 patients with LF/LC and 120 healthy controls (HCs). CircHIPK3 expression in tissues was detected in 120 fibrotic liver tissues and compared to 57 healthy liver tissues from patients with hepatic hemangioma. The expressions of CircHIPK3, TGF-β1, and CollA1 mRNAs were assessed by qRT-PCR. The Child-Pugh (CP) classification was used to evaluate disease severity. The Ishak score was applied to assess LF/LC in liver biopsy samples. The levels of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were also investigated. Receiver operating characteristic (ROC) analysis was conducted to assess the diagnostic value of CircHIPK3 expressions in serum and tissues. RESULTS CircHIPK3 expressions in serum and tissues were upregulated in patients with LF/LC compared to HCs. The patient group comprised 39 with CP class A (CP-A), 45 with CP class B (CP-B), and 36 with CP class B (CP-C). Patients with CP-C had markedly increased serum and local CircHIPK3 levels compared to those with CP-B and CP-A. Patients with CP-B showed upregulated CircHIPK3 expressions in serum and tissues compared to CP-A with statistical significance. ROC curve analysis indicated that CircHIPK3 expressions in both serum and tissues may serve as potential diagnostic indicators for the progression of LF/LC. Moreover, serum CircHIPK3 expressions were positively associated with serum ALT and AST levels. Tissue CircHIPK3 expressions were positively correlated with tissue TGF-β1 and CollA1 mRNA expressions. In addition, both serum and tissue CircHIPK3 expressions were positively associated with the Ishak score. CONCLUSIONS For the first time, this study demonstrated the positive correlation of CircHIPK3 expressions in both serum and tissues with the progression of LF/LC, regardless of etiology. CircHIPK3 might play a significant role in the development of LF/LC and act as a potential therapeutic target for these conditions.
Collapse
Affiliation(s)
- Ji-Ping Hou
- Department of General Surgery, Tianjin Medical University Baodi Hospital, China
| | - Lian-Ying Yang
- Department of General Surgery, Tianjin Medical University Baodi Hospital, China
| | - Li-Bin Liu
- Department of General Surgery, Tianjin Medical University Baodi Hospital, China
| | - En-Kun Han
- Department of General Surgery, Tianjin Medical University Baodi Hospital, China
| | - Chun-Qi Han
- Department of General Surgery, Tianjin Medical University Baodi Hospital, China
| | - Li-Ping Yang
- Department of Medical Imaging, Tianjin Medical University Baodi Hospital, China.
| |
Collapse
|
30
|
Yu NR, Huang S, Deng ZT, Wang J, Shen Y, Leng Y, Zhao QS. Design, synthesis and biological evaluation of Alisol B derivatives for potential treatment of non-alcoholic steatohepatitis. Bioorg Chem 2025; 155:108132. [PMID: 39813949 DOI: 10.1016/j.bioorg.2025.108132] [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: 09/11/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/18/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD), also known as metabolic dysfunction- associated with fatty liver disease (MAFLD), is one of the most prevalent chronic liver diseases globally. NAFLD is characterized by the accumulation of liver fat unrelated to excessive alcohol consumption. Non-alcoholic steatohepatitis (NASH) is the disease progression of NAFLD and could develop into cirrhosis and hepatocellular carcinoma. In previous studies, the preclinical efficacy of alisol B and alisol B 23-acetate against NASH and metabolic syndrome was identified. However, there is a paucity of literature pertaining to the specialized structural optimization of alisol B for the treatment of NASH. In this study, a series of alisol B derivatives (1-21) were designed, synthesized and evaluated in order to improve the activity of alisol B and to obtain candidate compounds for treating NASH. The effects of the synthesized compounds on de novo lipogenesis and α-SMA gene expression were tested to explore the preliminary structure-activity relationship (SAR). Compounds 14 and 21 were selected for further in vivo investigation. The high-fat diet plus carbon tetrachloride (HFD + CCl4)-induced NASH mice model was employed for biological evaluation in vivo. Compounds 14 and 21 effectively improved hepatic steatosis, ballooning, inflammatory infiltration, and hepatic fibrosis in the livers of HFD + CCl4 mice. Thus, 14 and 21 are promising lead compounds for the treatment of NASH.
Collapse
Affiliation(s)
- Nai-Rong Yu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201 China; University of Chinese Academy of Sciences, Beijing 100049 China
| | - Suling Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203 China
| | - Zhen-Tao Deng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201 China
| | - Jiayue Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203 China; University of Chinese Academy of Sciences, Beijing 100049 China
| | - Yu Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203 China
| | - Ying Leng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203 China; University of Chinese Academy of Sciences, Beijing 100049 China.
| | - Qin-Shi Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201 China; University of Chinese Academy of Sciences, Beijing 100049 China.
| |
Collapse
|
31
|
Chen SY, Chen X, Zhu S, Xu JJ, Li XF, Yin NN, Xiao YY, Huang C, Li J. miR-324-3p Suppresses Hepatic Stellate Cell Activation and Hepatic Fibrosis Via Regulating SMAD4 Signaling Pathway. Mol Biotechnol 2025; 67:673-688. [PMID: 38407690 PMCID: PMC11711260 DOI: 10.1007/s12033-024-01078-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: 09/12/2023] [Accepted: 01/15/2024] [Indexed: 02/27/2024]
Abstract
In hepatic fibrosis (HF), hepatic stellate cells (HSCs) form the extracellular matrix (ECM), and the pathological accumulation of ECM in the liver leads to inflammation. Our previous research found that miR-324-3p was down-regulated in culture-activated human HSCs. However, the precise effect of miR-324-3p on HF has not been elucidated. In this study, the HF mouse models were induced through directly injecting carbon tetrachloride (CCl4) into mice; the HF cell models were constructed using TGF-β1-treated LX-2 cells. Next, real-time-quantitative polymerase chain reaction (RT-qPCR), western blot (WB) and immunohistochemistry (IHC) were applied to assess the expression levels of miR-324-3p, α-smooth muscle actin (α-SMA), Vimentin or SMAD4; hematoxylin and eosin (H&E), Masson' s trichrome and Sirius red staining to evaluate the liver injury; luciferase reporter assay to verify the targeting relationship between miR-324-3p and SMAD4; enzyme-linked immunosorbent assay (ELISA) to determine the levels of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST); and cell counting kit-8 (CCK-8) and flow cytometry to evaluate the effects of miR-324-3p on cell proliferation and cycle/apoptosis, respectively. The experimental results showed a reduction in miR-324-3p level in CCl4-induced HF mice as well as transforming growth factor (TGF)-β1-activated HSCs. Interestingly, the miR-324-3p level was rescued following the HF recovery process. In HF mice induced by CCl4, miR-324-3p overexpression inhibited liver tissue damage, decreased serum ALT and AST levels, and inhibited fibrosis-related biomarkers (α-SMA, Vimentin) expression, thereby inhibiting HF. Similarly, miR-324-3p overexpression up-regulated α-SMA and Vimentin levels in HF cells, while knockdown of miR-324-3p had the opposite effect. Besides, miR-324-3p played an antifibrotic role through inhibiting the proliferation of hepatocytes. Further experiments confirmed that miR-324-3p targeted and down-regulated SMAD4 expression. SMAD4 was highly expressed in HF cells, and silencing SMAD4 significantly decreased the α-SMA and Vimentin levels in HF cells. Collectively, the miR-324-3p may suppress the activation of HSCs and HF by targeting SMAD4. Therefore, miR-324-3p is identified as a potential and novel therapeutic target for HF.
Collapse
Affiliation(s)
- Si-Yu Chen
- Department of Pharmacy, Hefei BOE Hospital, Intersection of Dongfang Avenue and Wenzhong Road, Hefei, China
| | - Xin Chen
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Sai Zhu
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Jin-Jin Xu
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Xiao-Feng Li
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Na-Na Yin
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Yan-Yan Xiao
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Cheng Huang
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Jun Li
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
32
|
Xie J, Chen S, Chen Y, Tong J, Huang H, Liao J, Sun J, Cong L, Zeng Y. FFA intervention on LO2 cells mediates SNX-10 synthesis and regulates MMP9 secretion in LX2 cells via TGF-β1. Arch Biochem Biophys 2025; 764:110255. [PMID: 39662717 DOI: 10.1016/j.abb.2024.110255] [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: 05/03/2024] [Revised: 11/10/2024] [Accepted: 11/29/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Metabolic-associated fatty liver disease (MAFLD) is a public health concern. Transforming growth factor-β1(TGF-β1) plays an important regulatory role in multiple MAFLD stages, as it can promote the expression of matrix metalloproteinase-9 (MMP9) and promote liver fibrosis. Sorting nexin protein-10 (SNX-10) may be involved in the occurrence and development of fatty liver disease. METHODS Free fatty acids (FFA) treatment was used to simulate the cellular lipid deposition stage of MAFLD and the interactions between cells were simulated via LX2 and LO2 coculture. The molecular interaction between the two cell types was studied via ELISA, immunoprecipitation, qPCR, and western blotting. RESULTS In FFA-treated LO2 cells, intracellular TGF-β1 expression increased as lipid deposition increased. FFA-treated LO2 cells promoted the expression and secretion of MMP9 by LX2 cells through paracrine pathways. MMP9 secretion decreased with decreasing SNX-10 expression in LX2 cells. The interaction between MMP9 and SNX-10 was confirmed by coimmunoprecipitation. TGF-β1 promoted the synthesis of SNX-10 through the p38 MAPK pathway, and SNX-10 affected the secretion of MMP9 through protein interactions, thereby affecting the development of liver fibrosis. CONCLUSIONS FFA induced lipid deposition in LO2 cells, and TGF-β1 mediated the p38 MAPK pathway to promote SNX-10 synthesis and stimulate MMP9 secretion, thereby regulating the involvement of LX2 in the process of liver fibrosis.
Collapse
Affiliation(s)
- Jianhui Xie
- Department of Endocrinology and Metabolic Diseases, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Shiyan Chen
- Department of Endocrinology and Metabolic Diseases, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yangli Chen
- Department of Endocrinology and Metabolic Diseases, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Junlu Tong
- Department of Endocrinology and Metabolic Diseases, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, Anhui, China
| | - Huijie Huang
- Department of Endocrinology and Metabolic Diseases, The First Huizhou Affiliated Hospital of Guangdong Medical University, Huizhou, Guangdong, China
| | - Jingwen Liao
- Department of Endocrinology and Metabolic Diseases, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jixin Sun
- Department of Endocrinology and Metabolic Diseases, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Li Cong
- Department of Endocrinology and Metabolic Diseases, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China.
| | - Yingjuan Zeng
- Department of Endocrinology and Metabolic Diseases, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China.
| |
Collapse
|
33
|
Liu S, He F, Jin C, Li Q, Zhao G, Ding K. Design and Synthesis of Dual Galectin-3 and EGFR Inhibitors Against Liver Fibrosis. Chem Asian J 2025; 20:e202401078. [PMID: 39504308 DOI: 10.1002/asia.202401078] [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] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/08/2024]
Abstract
Liver fibrosis, mainly arising from chronic viral or metabolic liver diseases, is a significant global health concern. There is currently only one FDA-approved drug (Resmetirom) in the market to combat liver fibrosis. Both galectin-3 and epidermal growth factor receptor (EGFR) play important roles in liver fibrosis, while galectin-3 may interact with EGFR. Galectin-3 inhibitors, typically lactose or galactose derivatives may inhibit liver fibrosis. We hypothesized that targeting both galectin-3 and EGFR may have better effect against liver fibrosis. Here, EGFR inhibitor erlotinib was used in a series of designed galectin-3 inhibitors after hybridization with the pharmacophore structure in reported galectin-3 inhibitors to impede hepatic stellate cells (HSCs) activation by a typical method of click chemistry. Bioactivity test results showed that compound 29 suppressed TGF-β-induced upregulation of fibrotic markers (α-SMA, fibronectin-1, and collagen I). The preferred compound 29 displayed better binding to galectin-3 (KD=52.29 μM) and EGFR protein (KD=3.31 μM) by SPR assay. Further docking studies were performed to clarify the possible binding mode of compound 29 with galectin-3 and EGFR. Taken together, these results suggested that compound 29 could be a potential dual galectin-3 and EGFR inhibitor as leading compound for anti-liver fibrosis new drug development.
Collapse
Affiliation(s)
- Shuanglin Liu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
- Henan Polysaccharide Research Center, Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, SSIP Healthcare and Medicine Demonstration Zone, Zhongshan, Guangdong, Tsuihang New District, 528400, China
| | - Fei He
- Glycochemistry and Glycobiology Lab, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Science, No.19 A Yuquan Road, Beijing, 100049, China
| | - Can Jin
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, SSIP Healthcare and Medicine Demonstration Zone, Zhongshan, Guangdong, Tsuihang New District, 528400, China
- Glycochemistry and Glycobiology Lab, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Science, No.19 A Yuquan Road, Beijing, 100049, China
| | - Qing Li
- Glycochemistry and Glycobiology Lab, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Science, No.19 A Yuquan Road, Beijing, 100049, China
- School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Guilong Zhao
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, SSIP Healthcare and Medicine Demonstration Zone, Zhongshan, Guangdong, Tsuihang New District, 528400, China
- Glycochemistry and Glycobiology Lab, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Science, No.19 A Yuquan Road, Beijing, 100049, China
| | - Kan Ding
- Henan Polysaccharide Research Center, Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, SSIP Healthcare and Medicine Demonstration Zone, Zhongshan, Guangdong, Tsuihang New District, 528400, China
- Glycochemistry and Glycobiology Lab, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Science, No.19 A Yuquan Road, Beijing, 100049, China
| |
Collapse
|
34
|
Noroozi F, Asle-Rousta M, Amini R, Sahraeian Z. Alpha-pinene ameliorates liver fibrosis by suppressing oxidative stress, inflammation, and the TGF-β/Smad3 signaling pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2025; 28:451-460. [PMID: 39968080 PMCID: PMC11831751 DOI: 10.22038/ijbms.2025.81693.17678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/18/2024] [Indexed: 02/20/2025]
Abstract
Objectives A monoterpene alpha-pinene possesses anti-oxidant, anti-inflammatory, and anti-apoptotic properties. Here, we investigated the effect of alpha-pinene on molecular, biochemical, and histological changes induced by carbon tetrachloride (CCl4) in the liver of male Wistar rats. Materials and Methods Animals were divided into four groups: Control, Pinene, CCl4, and CCl4.Pinene. Pinene and CCl4.Pinene groups were given alpha-pinene (50 mg/kg/day) through intraperitoneal (IP) injections for six consecutive weeks. CCl4 and CCl4.Pinene groups received IP injections of CCl4 (2 ml/kg twice weekly for six consecutive weeks). Results The results revealed that alpha-pinene inhibited enhancing liver enzyme AST (P<0.001), ALT (P<0.001), ALP (P<0.01), and GGT (P<0.001) activity in CCl4.Pinene rats. It reduced malondialdehyde (P<0.05) and nitric oxide (P<0.05) levels and increased the catalase enzyme activity (P<0.05) and glutathione levels (P<0.01) in the liver. Likewise, alpha-pinene suppressed proinflammatory and profibrotic gene expression and prevented significant histological damage and collagen deposition in the liver of these animals. Also, alpha-pinene reduced the expression of TLR4 (P<0.01), NF-κB (P<0.05), PI3K (P<0.05), Akt (P<0.05), mTOR (P<0.01), TGF-β1 (P<0.01), and Smad3 (P<0.01) in the liver of rats receiving CCl4. Conclusion We concluded that alpha-pinene reduced CCl4-induced liver fibrosis by lowering oxidative stress, suppressing liver inflammation, and inhibiting TLR4/NF-κB, TGF-β/Smad3, and PI3K/Akt/mTOR signaling pathways. Consequently, alpha-pinene may have potential therapeutic value in treating liver diseases.
Collapse
Affiliation(s)
- Fatemeh Noroozi
- Department of Physiology, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | | | - Rahim Amini
- Department of Biology, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | - Zeinab Sahraeian
- Nanobiotechnology Research Center, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| |
Collapse
|
35
|
Abolfazli S, Butler AE, Jamialahmadi T, Sahebkar A. A Golden Shield: The Protective Role of Curcumin against Liver Fibrosis. Curr Med Chem 2025; 32:1987-2004. [PMID: 37605399 DOI: 10.2174/0929867331666230821095329] [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: 04/16/2023] [Revised: 05/28/2023] [Accepted: 07/13/2023] [Indexed: 08/23/2023]
Abstract
Several chronic liver injuries can result in liver fibrosis, a wound-healing response defined by an excessive buildup of diffuse extracellular matrix (ECM). Liver fibrosis may progress to liver cirrhosis, liver failure, or hepatocellular carcinoma. Many cellular routes are implicated in the fibrosis process; however, hepatic stellate cells appear to be the main cell type involved. Curcumin, a polyphenolic substance extracted from the Curcuma longa plant, has a diversity of pharmacologic impacts, including anti- inflammatory, antioxidant, antiproliferative and antiangiogenic actions. The anti-fibrotic property of curcumin is less clear, but curcumin's ability to influence inflammatory cytokines, inflammatory pathways, the expression of pro-apoptotic (up-regulated) and anti- apoptotic (down-regulated) proteins, and its ability to lower oxidative stress likely underlie its anti-fibrotic properties. In this review, we investigate and analyze the impact of curcumin on several disorders that lead to liver fibrosis, and discuss the therapeutic applications of curcumin for these disorders.
Collapse
Affiliation(s)
- Sajad Abolfazli
- Student Research Committee, School of Pharmacy, Mazandaran University of Medical Science, Sari, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland-Bahrain, Adliya, Bahrain
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
36
|
Di X, Li Y, Wei J, Li T, Liao B. Targeting Fibrosis: From Molecular Mechanisms to Advanced Therapies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410416. [PMID: 39665319 PMCID: PMC11744640 DOI: 10.1002/advs.202410416] [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] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/27/2024] [Indexed: 12/13/2024]
Abstract
As the final stage of disease-related tissue injury and repair, fibrosis is characterized by excessive accumulation of the extracellular matrix. Unrestricted accumulation of stromal cells and matrix during fibrosis impairs the structure and function of organs, ultimately leading to organ failure. The major etiology of fibrosis is an injury caused by genetic heterogeneity, trauma, virus infection, alcohol, mechanical stimuli, and drug. Persistent abnormal activation of "quiescent" fibroblasts that interact with or do not interact with the immune system via complicated signaling cascades, in which parenchymal cells are also triggered, is identified as the main mechanism involved in the initiation and progression of fibrosis. Although the mechanisms of fibrosis are still largely unknown, multiple therapeutic strategies targeting identified molecular mechanisms have greatly attenuated fibrotic lesions in clinical trials. In this review, the organ-specific molecular mechanisms of fibrosis is systematically summarized, including cardiac fibrosis, hepatic fibrosis, renal fibrosis, and pulmonary fibrosis. Some important signaling pathways associated with fibrosis are also introduced. Finally, the current antifibrotic strategies based on therapeutic targets and clinical trials are discussed. A comprehensive interpretation of the current mechanisms and therapeutic strategies targeting fibrosis will provide the fundamental theoretical basis not only for fibrosis but also for the development of antifibrotic therapies.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Ya Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jingwen Wei
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Tianyue Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Banghua Liao
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| |
Collapse
|
37
|
Gao Y, Wu H, Luo Y, Deng X, Chen J, Wu T. Mechanisms of Dihydromyricetin for Improving Hepatic Fibrosis through the Integration of Metabolomics and Gut Microbiota. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:889-908. [PMID: 40374379 DOI: 10.1142/s0192415x25500338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
It is crucial to prevent and treat liver fibrosis in patients with chronic liver disease. Dihydromyricetin (DMY) is a natural flavonoid compound from traditional Chinese medicine, known to alleviate chronic liver injury. However, its role in regulating inflammatory responses through gut microbiota and metabolic changes remains unclear. In this study, a mouse model of liver fibrosis was induced with carbon tetrachloride (CCl4), and DMY was administered via gavage. Histopathology, immunohistochemistry, Reverse Transcription Polymerase Chain Reaction (RT-PCR), 16S rRNA sequencing, and untargeted metabolomics were employed to evaluate DMY's pharmacological effects on CCl4-induced liver fibrosis and explore its underlying mechanisms. Our results show that DMY reduced the aspartate transaminase (AST) and alanine transaminase (ALT) serum levels in liver fibrosis model mice, and lowered the mRNA expression of pro-inflammatory cytokines and fibrosis markers. Additionally, DMY restored the richness and diversity of the gut microbiota, with several microbiota taxa significantly correlating with inflammatory markers. Metabolomic analysis of serum and liver tissue revealed that DMY significantly altered the liver metabolite disturbances induced by CCl4. Pearson correlation analysis demonstrated a strong relationship between microbial composition and liver metabolites. These results suggest that DMY alleviates liver fibrosis in mice by reshaping the gut microbiota and host metabolism, thereby improving the inflammatory response.
Collapse
Affiliation(s)
- Ying Gao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hao Wu
- Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Yanqun Luo
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaoliang Deng
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Junming Chen
- Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
38
|
Dai W, Wu J, Li K, Xu Y, Wang W, Xiao W. Andrographolide: A promising therapeutic agent against organ fibrosis. Eur J Med Chem 2024; 280:116992. [PMID: 39454221 DOI: 10.1016/j.ejmech.2024.116992] [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: 08/18/2024] [Revised: 09/07/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024]
Abstract
Fibrosis is the terminal pathology of chronic illness in many organs, marked by excessive accumulation of extracellular matrix proteins. These changes influence organ function, ultimately resulting in organ failure. Although significant progress has been achieved in comprehending the molecular pathways responsible for fibrosis in the last decades, effective and approved clinical therapies for the condition are still lacking. Andrographolide is a diterpenoid isolated and purified mainly from the aboveground parts of the Andrographis paniculata plant, which possesses good effects of purging heat, detoxifying, antibacterial and anti-inflammatory. In-depth research has gradually confirmed the anticancer, antioxidant, antiviral and other effects of Andro so that it can play a preventive and therapeutic role in various diseases. Over the past few years, an increasing number of research findings have indicated that Andro exerts antifibrotic effects in various organs by acting on transforming growth factor-β/small mother against decapentaplegic protein, mitogen-activated protein kinases, nuclear factor-E2-related factor 2, nuclear factor kappa-B and other signalling molecules to inhibit inflammation, oxidative stress, epithelial-mesenchymal transition, fibroblast activation and collagen buildup. This review presents a compilation of findings regarding the antifibrotic impact of Andro in tissue and cell models in vitro and in vivo. Emphasis is placed on the potential therapeutic benefits of Andro in diseases related to organ fibrosis. Existing studies and cutting-edge technologies on Andro pharmacokinetics, toxicity and bioavailability are briefly discussed to provide evidence for accelerating its clinical conversion and adoption.
Collapse
Affiliation(s)
- Wei Dai
- Shanghai Key Lab of Human Performance(Shanghai University of Sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Jiabin Wu
- Shanghai Key Lab of Human Performance(Shanghai University of Sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Ke Li
- Shanghai Key Lab of Human Performance(Shanghai University of Sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Yingying Xu
- Shanghai Key Lab of Human Performance(Shanghai University of Sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Wenhong Wang
- Shanghai Key Lab of Human Performance(Shanghai University of Sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; Research Institute for Biology and Medicine, Hunan University of Medicine, Huaihua 418000, China.
| | - Weihua Xiao
- Shanghai Key Lab of Human Performance(Shanghai University of Sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
39
|
Hosseini Farzad S, Lashkarboloki M, Mowla SJ, Soltani BM. LncRNA DANCR-V1 is a novel regulator of Wnt/β-catenin and TGF-β1/SMAD signaling pathways in colorectal cancer: an in vitro and in silico study. Mol Biol Rep 2024; 52:36. [PMID: 39643825 DOI: 10.1007/s11033-024-10128-0] [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: 08/26/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND DANCR is an oncogenic lncRNA associated with advanced colorectal cancer, one of the most common malignancies worldwide. This lncRNA has a new variant, DANCR-V1, whose function is not yet understood. In this study, we aimed to evaluate the expression pattern of DANCR-V1 and its regulatory mechanism in colorectal cancer. METHOD AND RESULT Bioinformatics analysis and RT-qPCR showed that DANCR-V1 expression was higher in colorectal cancer tissues than in normal pairs obtained from microarray data and 20 samples, respectively. LncRNA subcellular localization and hsa-miR-222 binding sites were predicted using bioinformatics tools. Dual luciferase assays confirmed that miR-222-mediated downregulation of DANCR-V1 through its targeting, and RT-qPCR showed that overexpression of miR-222 decreased the level of DANCR-V1. Functionally, Wnt/β-catenin and TGF-β1/SMAD-related genes changed under DANCR-V1 overexpression in the SW480 cell line, while their expression was reversed following miR-222 overexpression. Finally, at the cellular level, overexpression of DANCR-V1 elevated the proliferation and migration rates of SW480 cells, as determined using flow cytometry, western blotting and scratch assays. CONCLUSION Our data suggest that DANCR-V1 is a novel transcript variant that has crucial crosstalk with miR-222 via negative feedback and plays a critical role in colorectal cancer progression through Wnt/β-catenin and TGF-β1/SMAD signaling modulation.
Collapse
Affiliation(s)
- Sana Hosseini Farzad
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mina Lashkarboloki
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Javad Mowla
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahram M Soltani
- Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
40
|
Guo H, Hu Z, Yang X, Yuan Z, Wang M, Chen C, Xie L, Gao Y, Li W, Bai Y, Lin C. Smad4 regulates TGF-β1-mediated hedgehog activation to promote epithelial-to-mesenchymal transition in pancreatic cancer cells by suppressing Gli1 activity. Comput Struct Biotechnol J 2024; 23:1189-1200. [PMID: 38525105 PMCID: PMC10957521 DOI: 10.1016/j.csbj.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 03/10/2024] [Accepted: 03/10/2024] [Indexed: 03/26/2024] Open
Abstract
Pancreatic cancer (PC) is an aggressive and metastatic gastrointestinal tumor with a poor prognosis. Persistent activation of the TGF-β/Smad signaling induces PC cell (PCC) invasion and infiltration via epithelial-to-mesenchymal transition (EMT). Hedgehog signaling is a crucial pathway for the development of PC via the transcription factors Gli1/2/3. This study aimed to investigate the underlying molecular mechanisms of action of hedgehog activation in TGF-β1-triggered EMT in PCCs (PANC-1 and BxPc-3). In addition, overexpression and shRNA techniques were used to evaluate the role of Smad4 in TGF-β1-treated PCCs. Our data showed that TGF-β1 promoted PCC invasion and infiltration via Smad2/3-dependent EMT. Hedgehog-Gli signaling axis in PCCs was activated upon TGF-β1 stimulation. Inhibition of hedgehog with cyclopamine effectively antagonized TGF-β1-induced EMT, thereby suggesting that the hedgehog signaling may act as a downstream cascade signaling of TGF-β1. As a key protein that assists the nuclear translocation of Smad2/3, Smad4 was highly expressed in PANC-1 cells, but not in BxPc-3 cells. Conversely, Gli1 expression was low in PANC-1 cells, but high in BxPc-3 cells. Furthermore, knockdown of Smad4 in PANC-1 cells by shRNA inhibited TGF-β1-mediated EMT and collagen deposition. Overexpression of Smad4 did not affect TGF-β1-mediated EMT due to the lack of significant increase in nuclear expression of Smad4. Importantly, Gli1 activity was upregulated by Smad4 knockdown in PANC-1 cells and downregulated by Smad4 overexpression in BxPc-3 cells, indicating that Gli1 may be a negative target protein downstream of Smad4. Thus, Smad4 regulates TGF-β1-mediated hedgehog activation to promote EMT in PCCs by suppressing Gli1 activity.
Collapse
Affiliation(s)
- Hangcheng Guo
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
- Sichuan Mianyang 404 Hospital, Mianyang 621000, China
| | - Zujian Hu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xuejia Yang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Ziwei Yuan
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Mengsi Wang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Chaoyue Chen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Lili Xie
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Yuanyuan Gao
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Wangjian Li
- Department of Urology, The Central Hospital Affiliated to Shaoxing University, Shaoxing 312030, China
| | - Yongheng Bai
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
- National Key Clinical Specialty (General Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Chunjing Lin
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Medicine and Health Care Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
41
|
Corbalan JJ, Jagadeesan P, Frietze KK, Taylor R, Gao GL, Gallagher G, Nickels JT. Humanized monoacylglycerol acyltransferase 2 mice develop metabolic dysfunction-associated steatohepatitis. J Lipid Res 2024; 65:100695. [PMID: 39505262 DOI: 10.1016/j.jlr.2024.100695] [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: 08/19/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Mice lacking monoacylglycerol acyltransferase 2 (mMGAT21) are resistant to diet-induced fatty liver, suggesting hMOGAT2 inhibition is a viable option for treating metabolic dysfunction-associated steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH). We generated humanized hMOGAT2 mice (HuMgat2) for use in pre-clinical studies testing the efficacy of hMOGAT2 inhibitors for treating MASLD/MASH. HuMgat2 mice developed MASH when fed a steatotic diet. Computer-aided histology revealed the presence of hepatocyte cell ballooning, immune cell infiltration, and fibrosis. Hepatocytes accumulated Mallory-Denk bodies containing phosphorylated p62/sequestosome-1-ubiquitinated protein aggregates likely caused by defects in autophagy. Metainflammation and apoptotic cell death were seen in the livers of HuMgat2 mice. Treating HuMgat2 mice with elafibranor reduced several MASH phenotypes. RNASeq analysis predicted changes in bile acid transporter expression that correlated with altered bile acid metabolism indicative of cholestasis. Our results suggest that HuMgat2 mice will serve as a pre-clinical model for testing hMOGAT2 inhibitor efficacy and toxicity and allow for the study of hMOGAT2 in the context of MASH.
Collapse
Affiliation(s)
- J Jose Corbalan
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Pranavi Jagadeesan
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Karla K Frietze
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Rulaiha Taylor
- Department of Pharmacology and Toxicology, Earnest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Grace L Gao
- Department of Pharmacology and Toxicology, Earnest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, USA
| | - Grant Gallagher
- Oncoveda, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Joseph T Nickels
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
42
|
Ouyang S, Shi S, Ding W, Ge Y, Su Y, Mo J, Peng K, Zhang Q, Liu G, Xiao W, Yue P, Lu J, Wang Y, Xiong X, Zhang X. Neuropeptide Precursor VGF Promotes Liver Metastatic Colonization of Gαq Mutant Uveal Melanoma by Facilitating Tumor Microenvironment via Paracrine Loops. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407967. [PMID: 39422674 PMCID: PMC11633529 DOI: 10.1002/advs.202407967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Uveal melanoma (UM), the predominant primary ocular malignancy, often progresses to liver metastasis with limited therapeutic options. The interplay of the tumor microenvironment, encompassing secreted soluble factors, plays a crucial role in facilitating liver metastasis. In this study, the role is elucidated of the neural growth factor-inducible gene (VGF), a secreted neuropeptide precursor, in Gαq mutant UM. Employing a multiomics approach, encompassing transcriptomic and secretomic analyses, the intricate involvement of VGF in UM progression is unveiled. VGF is upregulated in Gαq mutant UM cells and associated with poor prognosis of UM patients. Targeting VGF significantly suppressed the growth of UM in vitro and in vivo. Further evidence shows that VGF is regulated by Gαq through MAPK/CREB pathway. Mechanistically, CREB modulates VGF expression by directly binding to consensus DNA response elements in the promoters of the VGF gene. Combined inhibition of Gαq and MEK remarkably reduces tumor burden in the UM xenograft model. Notably, VGF triggers liver metastatic colonization of UM and activates the fibrosis of hepatic stellate cells (HSCs), creating a favorable microenvironment, through an autocrine and paracrine loop. Furthermore, VGF directly binds to TGFBR2 and regulates TGF-β-SMAD signaling pathway, thereby regulating genes associated with endothelial-mesenchymal transition (EMT) to promote metastasis. Taken together, these findings identify VGF as a pivotal driver in the progression and metastasis of Gαq mutant UM and confers a promising therapeutic target and strategy for UM patients.
Collapse
Affiliation(s)
- Shumin Ouyang
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Shuo Shi
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Wen Ding
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Yang Ge
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Yingxue Su
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhou510060China
| | - Jianshan Mo
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Keren Peng
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Qiyi Zhang
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Guopin Liu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhou510060China
| | - Wei Xiao
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhou510060China
| | - Peibin Yue
- Department of MedicineDivision of Hematology‐Oncologyand Samuel Oschin Comprehensive Cancer InstituteCedars‐Sinai Medical CenterLos AngelesCA90048USA
| | - Jinjian Lu
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauMacao999078China
| | - Yandong Wang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhou510060China
| | - Xiaofeng Xiong
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Xiaolei Zhang
- National‐Local Joint Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Key Laboratory of Chiral Molecule and Drug DiscoverySchool of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| |
Collapse
|
43
|
Liu Q, Li J, Li X, Zhang L, Yao S, Wang Y, Tuo B, Jin H. Advances in the understanding of the role and mechanism of action of PFKFB3‑mediated glycolysis in liver fibrosis (Review). Int J Mol Med 2024; 54:105. [PMID: 39301662 PMCID: PMC11448561 DOI: 10.3892/ijmm.2024.5429] [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: 07/02/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
Liver fibrosis is a pathophysiologic manifestation of chronic liver disease and a precursor to cirrhosis and hepatocellular carcinoma. Glycolysis provides intermediate metabolites as well as energy support for cell proliferation and phenotypic transformation in liver fibers. 6‑Phosphofructo‑2‑kinase/fructose‑2,6‑bisphosphatase 3 (PFKFB3) is a key activator of glycolysis and plays an important role in the process of glycolysis. The role of PFKFB3‑mediated glycolysis in myocardial fibrosis, renal fibrosis and pulmonary fibrosis has been demonstrated, and the role of PFKFB3 in the activation of hepatic stellate cells by aerobic glycolysis has been proven by relevant experiments. The present study reviews the research progress on the role and mechanism of action of PFKFB3‑mediated glycolysis in the progression of hepatic fibrosis to discuss the role of PFKFB3‑mediated glycolysis in hepatic fibrosis and to provide new ideas for research on PFKFB3 as a target for the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Qian Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jiajia Li
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Xin Li
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yongfeng Wang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
44
|
Shen C, Jiang Y, Lin J, Guo Q, Fang D. METTL3 silencing inhibits ferroptosis to suppress ovarian fibrosis in PCOS by upregulating m6A modification of GPX4. J Mol Histol 2024; 55:1163-1175. [PMID: 39261364 DOI: 10.1007/s10735-024-10257-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Methyltransferase-like 3 (METTL3) is extensively reported to be involved in organ fibrosis. Ovarian fibrosis is a main characteristic of polycystic ovary syndrome (PCOS). However, the reaction mechanism of METTL3 in PCOS is poorly investigated. This paper was intended to reveal the role and the mechanism of METTL3 in PCOS. Animal and cell models of PCOS were induced by dehydroepiandrosterone (DHEA). H&E staining was performed to detect the pathological alterations in ovary tissues. Masson staining, immunofluorescence, along with western blot measured fibrosis both in vitro and in vivo. To evaluate estrous cycle, vaginal smear was performed. Lipid peroxidation and ferroptosis were evaluated by MDA assay kits, GSH assay kits, immunohistochemistry, Prussian blue staining and western blot. qRT-PCR and western blot were adopted to estimate METTL3 and GPX4 expression. The m6A and hormone secretion levels were respectively assessed by m6A RNA Methylation Quantitative Kit and corresponding kits. The interaction between METTL3 and GPX4 was testified by immunoprecipitation. The fibrosis and ferroptosis were aggravated and m6A and METTL3 expression were increased in ovarian tissues of DHEA-induced PCOS mice. METTL3 silencing alleviated pathological changes, affected hormone secretion level, and repressed fibrosis, lipid peroxidation and ferroptosis in the ovarian tissues of PCOS mice. In vitro, DHEA stimulation increased m6A and METTL3 expression and induced ferroptosis and fibrosis. METTL3 knockdown promoted GPX4 expression in DHEA-induced granulosa cells by m6A modification and restrained DHEA-induced fibrosis, lipid peroxidation and ferroptosis in granulosa cells via elevating GPX4. METTL3 silence inhibited ovarian fibrosis in PCOS, which was mediated through suppressing ferroptosis by upregulating GPX4 in m6A-dependent manner.
Collapse
Affiliation(s)
- Chuan Shen
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, No. 17, Section 3, Ren Min Nan Lu, Chengdu, 610041, Sichuan, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yongmei Jiang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Jia Lin
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, No. 17, Section 3, Ren Min Nan Lu, Chengdu, 610041, Sichuan, People's Republic of China
| | - Qiwei Guo
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, No. 17, Section 3, Ren Min Nan Lu, Chengdu, 610041, Sichuan, People's Republic of China
| | - Dingzhi Fang
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, No. 17, Section 3, Ren Min Nan Lu, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
45
|
Liu T, Cheng Z, Song D, Zhu E, Li H, Lin R, Wan Z, Liu S, Gong Z, Shan C. Arbutin alleviates Mycoplasma gallinarum-induced damage caused by pulmonary fibrosis via the JAK2/STAT3 pathway. Poult Sci 2024; 103:104434. [PMID: 39467406 PMCID: PMC11550161 DOI: 10.1016/j.psj.2024.104434] [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: 07/12/2024] [Revised: 09/18/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024] Open
Abstract
Mycoplasma gallinarum (MG) can cause infectious respiratory diseases in poultry that are chronic. Arbutin (AR) possesses anti-inflammatory, bacteriostatic, antitussive, and expectorant pharmacological effects, but whether it exerts regulatory effects on MG-induced pneumonia and fibrosis remains unclear. The study results unveiled that pulmonary connective tissue hyperplasia, pulmonary capillary congestion, and inflammatory cell infiltration, as well as serum levels of cytokines (i.e., TNF-α, IL-1β, IL-6, and IL-10), were elevated after MG infection. Collagen fibers were significantly deposited in the lung tissue from MG-infected chicks. Furthermore, the expression levels of key factors in the JAK2/STAT3 and TGF-β/Smad pathways markedly increased. AR intervention significantly alleviated MG-induced pneumonic injury, and reduced collagen deposition and the expression of fibrosis markers in the lung tissue. AR reduced the degree of pulmonary fibrosis by regulating key factors of the JAK2/STAT3 signaling pathway in the MG-infected HD11 cells. Thus, AR effectively reduced the expression of inflammatory factors by regulating the JAK2/STAT3 signaling pathway, thereby improving lung inflammation and fibrosis.
Collapse
Affiliation(s)
- Ting Liu
- College of Animal Science, Guizhou University, Guiyang, 550000, PR China
| | - Zhentao Cheng
- College of Animal Science, Guizhou University, Guiyang, 550000, PR China
| | - Derong Song
- Bijie Institute of Animal Husbandry and Veterinary Sciences, Bijie, 551700, PR China
| | - Erpeng Zhu
- College of Animal Science, Guizhou University, Guiyang, 550000, PR China
| | - Hui Li
- College of Animal Science, Guizhou University, Guiyang, 550000, PR China
| | - Rutao Lin
- College of Animal Science, Guizhou University, Guiyang, 550000, PR China
| | - Zhiling Wan
- College of Animal Science, Guizhou University, Guiyang, 550000, PR China
| | - Shunxing Liu
- College of Animal Science, Guizhou University, Guiyang, 550000, PR China
| | - Zeguang Gong
- College of Animal Science, Guizhou University, Guiyang, 550000, PR China
| | - Chunlan Shan
- College of Animal Science, Guizhou University, Guiyang, 550000, PR China.
| |
Collapse
|
46
|
Xing D, Xia G, Tang X, Zhuang Z, Shan J, Fang X, Qiu L, Zha X, Chen XL. A Multifunctional Nanocomposite Hydrogel Delivery System Based on Dual-Loaded Liposomes for Scarless Wound Healing. Adv Healthc Mater 2024; 13:e2401619. [PMID: 39011810 DOI: 10.1002/adhm.202401619] [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: 05/03/2024] [Revised: 06/21/2024] [Indexed: 07/17/2024]
Abstract
Increased inflammatory responses and oxidative stress at the wound site following skin trauma impair healing. Furthermore, skin scarring places fibroblasts under severe mechanical stress and aggravates pathological fibrosis. A novel liposomal composite hydrogel is engineered for wound microenvironment remodeling, incorporating dual-loaded liposomes into gelatin methacrylate to create a nanocomposite hydrogel. Notably, tetrahydrocurcumin (THC) and hepatocyte growth factor (HGF) are encapsulated in the hydrophobic and hydrophilic layers of liposomes, respectively. The composite hydrogel maintains porous nanoarchitecture, demonstrating sustainable THC and HGF release and enhanced mechanical properties and biocompatibility. This system effectively promotes cell proliferation and angiogenesis and attenuates apoptosis. It decreases the expression of the inflammatory factors by inhibiting the high-mobility group box /receptor for advanced glycation end product/NF-κB (HMGB1/RAGE/NF-κB)pathway and increases macrophage polarization from M1 to M2 in vitro, effectively controlling inflammatory responses. It exhibits remarkable antioxidant properties by scavenging excess reactive oxygen species and free radicals. Most importantly, it effectively prevents scar formation by restraining the transforming growth factor beta (TGF-β)/Smads pathway that downregulates associated fibrotic factors. It demonstrates strong therapeutic effects against inflammation and fibrosis in a rat skin wound model with biosafety, advancing the development of innovative hydrogel-based therapeutic delivery strategies for clinical scarless wound therapy.
Collapse
Affiliation(s)
- Danlei Xing
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Guoqing Xia
- Institute for Liver Diseases of Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230022, P. R. China
| | - Xudong Tang
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Zhiwei Zhuang
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Jie Shan
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Xiao Fang
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Le Qiu
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230022, P. R. China
| | - Xu-Lin Chen
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| |
Collapse
|
47
|
Ding C, Liu B, Yu T, Wang Z, Peng J, Gu Y, Li Z. SIRT7 protects against liver fibrosis by suppressing stellate cell activation via TGF-β/SMAD2/3 pathway. Biomed Pharmacother 2024; 180:117477. [PMID: 39316972 DOI: 10.1016/j.biopha.2024.117477] [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: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND SIRT7 is a class III HDACs deacetylase which plays critical roles in various biological processes. Aberrant SIRT7 expression is associated with tumorigenesis and disease progression while role of SIRT7 in hepatic fibrosis remain elusive. METHODS SIRT7 expression was examined in fibrotic liver sample via WB and IHC. Myeloid cell-specific knockout (SIRT7MKO) mice were generated by crossing SIRT7flox/flox mice with LysM-Cre mice. Primary hepatic stellate cells (HSCs) was isolated to examine stellate cells activation. SIRT7 and SMAD2/3 interaction were analyzed by immunoprecipitation. SB525334 was used to prevent SMAD2/3 phosphorylation. RESULTS SIRT7 expression was decreased during chronic liver disease progression but was increased in liver cancer. IHC staining indicated that SIRT7 was primarily expressed in non-parenchymal cells in both fibrotic and cirrhotic liver. Knockout SIRT7 in myeloid cells resulted in significant elevation of serum ALT and liver fibrosis, but mildly affected hepatic inflammation after CCl4 treatment. We further observed significant elevation of elevation of stellate cell activation and SMAD2/3 activation in SIRT7MKO mice. By using primary HSCs and stellate cell line, we confirmed that SIRT7 interacted with SMAD2/3, induced its deacetylation and was critical in regulation of SMAD2/3 activation and stellate cell activation upon TGF-β stimulation. Pharmacological inhibition of SMAD2/3 reversed the hyperactivation of SIRT7MKO HSCs after TGF-β stimulation, and abolished stellate cell activation and liver fibrosis in SIRT7MKO mice. CONCLUSION Our findings revealed previously unidentified role of SIRT7 in regulating HSCs activation via modulating TGF-β/SMAD2/3 signaling pathway. Targeting SIRT7 might offer novel therapeutic option against liver fibrosis.
Collapse
Affiliation(s)
- Cong Ding
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, and Institute of Interdisciplinary Studies, Hunan Normal University School of Medicine, Changsha, Hunan 410013, China
| | - Bohao Liu
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, and Institute of Interdisciplinary Studies, Hunan Normal University School of Medicine, Changsha, Hunan 410013, China
| | - Tingzi Yu
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, and Institute of Interdisciplinary Studies, Hunan Normal University School of Medicine, Changsha, Hunan 410013, China
| | - Zhiqiang Wang
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, and Institute of Interdisciplinary Studies, Hunan Normal University School of Medicine, Changsha, Hunan 410013, China
| | - Jinying Peng
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, and Institute of Interdisciplinary Studies, Hunan Normal University School of Medicine, Changsha, Hunan 410013, China
| | - Yiying Gu
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, and Institute of Interdisciplinary Studies, Hunan Normal University School of Medicine, Changsha, Hunan 410013, China
| | - Zhuan Li
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, and Institute of Interdisciplinary Studies, Hunan Normal University School of Medicine, Changsha, Hunan 410013, China.
| |
Collapse
|
48
|
Zhang Q, Liang Q, Wang G, Xie X, Cao Y, Sheng N, Zeng Z, Ren C. Highly Selective Artificial K + Transporters Reverse Liver Fibrosis In Vivo. JACS AU 2024; 4:3869-3883. [PMID: 39483224 PMCID: PMC11522913 DOI: 10.1021/jacsau.4c00521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 11/03/2024]
Abstract
Liver fibrosis is a life-threatening disease that currently lacks clinically effective therapeutic agents. Given the close correlation between dysregulated intracellular K+ homeostasis and the progression of liver fibrosis, developing artificial K+ transporters mimicking the essential function of their natural counterparts in regulating intracellular K+ levels might offer an appealing yet unexplored treatment strategy. Here, we present an unconventional class of artificial K+ transporters involving the "motional" collaboration between two K+ transporter molecules. In particular, 6C6 exhibits an impressive EC50 value of 0.28 μM (i.e., 0.28 mol % relative to lipid) toward K+ and an exceptionally high K+/Na+ selectivity of 15.5, representing one of the most selective artificial K+ transporters reported to date. Most importantly, our study demonstrates, for the first time, the potential therapeutic effect of K+-selective artificial ion transporters in reversing liver fibrosis both in vitro and in vivo.
Collapse
Affiliation(s)
- Qiuping Zhang
- State
Key Laboratory of Cellular Stress Biology and Fujian Provincial Key
Laboratory of Innovative Drug Target Research, School of Pharmaceutical
Sciences, Xiamen University, Xiamen, Fujian 361102, China
- Shenzhen
Research Institute of Xiamen University, Shenzhen, Guangdong 518057, China
| | - Qinghong Liang
- State
Key Laboratory of Cellular Stress Biology and Fujian Provincial Key
Laboratory of Innovative Drug Target Research, School of Pharmaceutical
Sciences, Xiamen University, Xiamen, Fujian 361102, China
- Shenzhen
Research Institute of Xiamen University, Shenzhen, Guangdong 518057, China
| | - Guijiang Wang
- State
Key Laboratory of Cellular Stress Biology and Fujian Provincial Key
Laboratory of Innovative Drug Target Research, School of Pharmaceutical
Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiaopan Xie
- State
Key Laboratory of Cellular Stress Biology and Fujian Provincial Key
Laboratory of Innovative Drug Target Research, School of Pharmaceutical
Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yin Cao
- State
Key Laboratory of Cellular Stress Biology and Fujian Provincial Key
Laboratory of Innovative Drug Target Research, School of Pharmaceutical
Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Nan Sheng
- State
Key Laboratory of Cellular Stress Biology and Fujian Provincial Key
Laboratory of Innovative Drug Target Research, School of Pharmaceutical
Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhiping Zeng
- State
Key Laboratory of Cellular Stress Biology and Fujian Provincial Key
Laboratory of Innovative Drug Target Research, School of Pharmaceutical
Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Changliang Ren
- State
Key Laboratory of Cellular Stress Biology and Fujian Provincial Key
Laboratory of Innovative Drug Target Research, School of Pharmaceutical
Sciences, Xiamen University, Xiamen, Fujian 361102, China
- Shenzhen
Research Institute of Xiamen University, Shenzhen, Guangdong 518057, China
| |
Collapse
|
49
|
Weber F, Utpatel K, Evert K, Weiss TS, Buechler C. Hepatic Bone Morphogenetic Protein and Activin Membrane-Bound Inhibitor Levels Decline in Hepatitis C but Are Not Associated with Progression of Hepatocellular Carcinoma. Biomedicines 2024; 12:2397. [PMID: 39457709 PMCID: PMC11504530 DOI: 10.3390/biomedicines12102397] [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: 09/24/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) is an antagonist of transforming growth factor (TGF)-β type 1 signaling. BAMBI functions as an anti-fibrotic protein and exerts pro- as well as anti-cancerogenic activities. Our study aimed to correlate hepatocyte BAMBI protein levels in hepatocellular carcinoma (HCC) with T stage, lymph node invasion, vessel invasion, grading, tumor size and Union for International Cancer Control (UICC) stage, as well as with liver inflammation and fibrosis stages. METHODS Hepatocyte BAMBI protein expression was assessed by immunohistochemistry in HCC tissues of 320 patients and non-tumor tissues of 51 patients. RESULTS In the HCC tissues of the whole cohort and sex-specific analysis, BAMBI protein was not related to T stage, vessel invasion, lymph node invasion, histologic grade, UICC stage and tumor size. Accordingly, BAMBI was not associated with overall survival, recurrence-free and metastasis-free survival. BAMBI protein levels in tumor and non-tumor tissues were not related to inflammation and fibrosis grade. BAMBI protein levels in HCC tissues and non-tumor tissues from HCC patients, which were analyzed by immunoblot in a small cohort and by immunohistochemistry in the tissues of patients described above, were similar. Notably, BAMBI protein was low-abundant in HCC tissues of hepatitis C virus (HCV) compared to hepatitis B virus (HBV)-infected patients with comparable disease severity. Immunoblot analysis revealed reduced BAMBI protein in non-tumor tissues of patients with HCV in comparison to patients with HBV and normal human liver tissues. CONCLUSIONS In summary, this analysis showed that hepatocyte BAMBI protein levels of patients with HCC are related to HCV infection rather than the severity of the underlying liver disease and cancer staging.
Collapse
Affiliation(s)
- Florian Weber
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (F.W.); (K.U.); (K.E.)
| | - Kirsten Utpatel
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (F.W.); (K.U.); (K.E.)
| | - Katja Evert
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (F.W.); (K.U.); (K.E.)
| | - Thomas S. Weiss
- Children’s University Hospital (KUNO), Regensburg University Hospital, 93053 Regensburg, Germany;
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93053 Regensburg, Germany
| |
Collapse
|
50
|
Rong Y, Kang Y, Wen J, Gong Q, Zhang W, Sun K, Shuang W. Time-dependent arachidonic acid metabolism and functional changes in rats bladder tissue after suprasacral spinal cord injury. Exp Neurol 2024; 383:114989. [PMID: 39424042 DOI: 10.1016/j.expneurol.2024.114989] [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: 05/08/2024] [Revised: 09/04/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND A critical aspect affecting the quality of life in Traumatic spinal cord injury (TSCI) patients is bladder dysfunction. Metabolities in arachidonic acid are crucial lipid signaling molecules involved innumerous physiological processes. In this study, We are the first use eicosanoid metabolomics detrusor contraction examine, to assess the effect of the arachidonic acid metabolic in bladder dysfunction following TSCI. In additon, we explore the time of inflammatory and function changes in bladder tissue. METHODS Adult male Sprague-Dawley rats were subjected to improved Weight Drop method surgeries. Detrusor contraction examination, urodynamic examination, eicosanoid metabolomics, transmission electron microscopy, Elisa and histological staining were performed to assess the change of inflammatory, metabolic and function variation over time after TSCI. RESULTS Following TSCI, before the variations of bladder function, inflammatory changes including the increase of inflammatory factors, mitochondrial damage, and slight lipid peroxidation, occurred in bladder tissue. And the inflammatory changes gradually decreases over time. However, From the third day after TSCI, secondary lesions appeared in bladder tissue. Not only did inflammation-related indexes increase again, the degree of mitochondrial damage and lipid peroxidation increased, but also the contractility of detrusor began to change significantly. We also found that the content of metabolites in arachidonic acid metabolic pathway and the degree of detrusor contractility change showed a strong correlation. In addition, we found that rats had moved beyond the spinal shock stage on the seventh day after TSCI. CONCLUSION Altogether, we are the first to demonstrate that abnormal arachidonic acid metabolism plays an important role in bladder dysfunction after TSCI. We also demonstrate that 3d is a critical juncture for changes in rat bladder tissue, which indicates it is an important juncture in the treatment of neurogenic bladder.
Collapse
Affiliation(s)
- Yi Rong
- The First Hospital of Shanxi Medical University, Jiefang Rd, Shanxi 030000, China; First Clinical Medical College, Shanxi Medical University, Jiefang Rd, Shanxi 030000, China
| | - Yinbo Kang
- The First Hospital of Shanxi Medical University, Jiefang Rd, Shanxi 030000, China; First Clinical Medical College, Shanxi Medical University, Jiefang Rd, Shanxi 030000, China
| | - Jie Wen
- The First Hospital of Shanxi Medical University, Jiefang Rd, Shanxi 030000, China; First Clinical Medical College, Shanxi Medical University, Jiefang Rd, Shanxi 030000, China
| | - Qian Gong
- The First Hospital of Shanxi Medical University, Jiefang Rd, Shanxi 030000, China; First Clinical Medical College, Shanxi Medical University, Jiefang Rd, Shanxi 030000, China
| | - Wenlong Zhang
- The First Hospital of Shanxi Medical University, Jiefang Rd, Shanxi 030000, China; First Clinical Medical College, Shanxi Medical University, Jiefang Rd, Shanxi 030000, China
| | - Ke Sun
- The First Hospital of Shanxi Medical University, Jiefang Rd, Shanxi 030000, China; First Clinical Medical College, Shanxi Medical University, Jiefang Rd, Shanxi 030000, China
| | - Weibing Shuang
- The First Hospital of Shanxi Medical University, Jiefang Rd, Shanxi 030000, China.
| |
Collapse
|