1
|
Liu Y, Feng LL, Han B, Cai LJ, Liu RY, Tang S, Yang Q. Exploring the molecular mechanisms through which overexpression of TET3 alleviates liver fibrosis in mice via ferroptosis in hepatic stellate cells. Cell Signal 2025; 131:111747. [PMID: 40096933 DOI: 10.1016/j.cellsig.2025.111747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/19/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
Hepatic stellate cell (HSC) activation is crucial in the onset and progression of liver fibrosis, and inhibiting or eliminating activated HSCs is a key therapeutic strategy. Ferroptosis may help eliminate activated HSCs; however, its role and regulatory pathways in liver fibrosis remain unclear. As a DNA demethylase, TET3 regulates gene expression via DNA demethylation. We previously demonstrated that TET3 overexpression alleviates CCL4-induced liver fibrosis in mice; however, the specific mechanisms, including whether TET3 affects ferroptosis in HSCs, remain unexplored. Thus, we aimed to explore the molecular mechanisms wherein TET3 overexpression improves liver fibrosis in mice via ferroptosis in HSCs. Our in vivo observations showed that overexpression of TET3 ameliorate liver fibrosis in mice, and is associated with increased levels of malondialdehyde (MDA) and Fe2+ in liver tissue, as well as decreased protein expression of SLC7A11, GPX4, and FTH1. Further in vitro studies on HSCs showed that TET3 overexpression inhibits the expression of SLC7A11, GPX4, and FTH1, and reduces intracellular GSH levels, leading to accumulation of MDA and iron ions. This induces ferroptosis in HSC-LX2 cells, while simultaneously decreasing ECM accumulation in HSCs. Furthermore, hMeDIP-SEQ and ChIP-qPCR analyses revealed that TET3 directly interacts with the promoter regions of GPX4 and FTH1 to regulate their transcriptional expression. We propose that overexpression of TET3 modulates the gene methylation status of ferroptosis-related proteins, thereby regulating HSC ferroptosis, reducing activated HSCs, and decreasing ECM deposition in the liver. This may represent one of the molecular mechanisms wherein TET3 overexpression ameliorates liver fibrosis in mice.
Collapse
Affiliation(s)
- Yin Liu
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, 550025 Guiyang, Guizhou, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, 550025 Guiyang, Guizhou, China
| | - Lin-Lin Feng
- Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, 550025 Guiyang, Guizhou, China
| | - Bing Han
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, 550025 Guiyang, Guizhou, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, 550025 Guiyang, Guizhou, China
| | - Li-Jun Cai
- Department of Rehabilitation Medicine, The Affiliated Hospital of Guizhou Medical University, 550025 Guiyang, Guizhou, China
| | - Ran-Yang Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Shuang Tang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, 550025 Guiyang, Guizhou, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, 550025 Guiyang, Guizhou, China
| | - Qin Yang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, 550025 Guiyang, Guizhou, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, 550025 Guiyang, Guizhou, China.
| |
Collapse
|
2
|
Liu F, Lv J, Chen Y, Wang L, Liu Z, Li X. Lipoic acid in metabolic dysfunction-associated steatotic liver disease: a review. Nutr Metab (Lond) 2025; 22:56. [PMID: 40483465 PMCID: PMC12145643 DOI: 10.1186/s12986-025-00954-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
The incidence and prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD), a chronic liver disease characterized by hepatic steatosis without substantial alcohol consumption, are rapidly increasing worldwide. Liver cirrhosis and cancer are relatively common in MASLD patients. Therefore, it is essential to take proactive measures in preventing its onset or initiating prompt treatment. However, there is a lack of approved medications for effectively treating this ailment. Lipoic acid (LA), a compound with antioxidant, insulin-sensitization, anti-inflammatory, and prooxidant activities, has been proven to inhibit lipid deposition. Many studies have shown that supplementation of LA can alleviate MASLD. Therefore, the latest evidence on the relationship between LA and MASLD is presented in this review. The effect of LA on the accumulation of fat in the liver is emphasized following different diet models (normal, high fat, high fructose, choline deficiency) and other models (gene mutation, diabetes), with the main mechanisms from mitochondrial function to inflammation and oxidative stress being summarized. LA possesses excellent preventive effects on MASLD, which can provide new opportunities for clinical research.
Collapse
Affiliation(s)
- Fangli Liu
- School of Nursing and Health, Henan University, Kaifeng, 475004, China
| | - Jingjing Lv
- School of Nursing and Health, Henan University, Kaifeng, 475004, China
| | - Yuanyuan Chen
- College of Life Sciences Institute of Biological Engineering, Henan University, Kaifeng, 475004, China
- Engineering Research Center for Applied Microbiology of Henan Province, Kaifeng, 475004, China
| | - Linfeng Wang
- College of Life Sciences Institute of Biological Engineering, Henan University, Kaifeng, 475004, China
- Engineering Research Center for Applied Microbiology of Henan Province, Kaifeng, 475004, China
| | - Zhuoxin Liu
- School of Medical Sciences, Xinjiang Medical University, Urumqi, 830000, China
| | - Xingke Li
- College of Life Sciences Institute of Biological Engineering, Henan University, Kaifeng, 475004, China.
- Engineering Research Center for Applied Microbiology of Henan Province, Kaifeng, 475004, China.
| |
Collapse
|
3
|
Waxman S, Villafranca-Baughman D, Phillippi J, Jakobs TC, Alarcon-Martinez L, Di Polo A, Sigal IA. Pericytes in the Optic Nerve Head. Prog Retin Eye Res 2025:101375. [PMID: 40449651 DOI: 10.1016/j.preteyeres.2025.101375] [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: 01/04/2025] [Revised: 05/28/2025] [Accepted: 05/29/2025] [Indexed: 06/03/2025]
Abstract
Pericytes are a unique population of contractile mural cells and an essential part of the microvasculature. In the retina and brain, pericytes play crucial roles in regulating blood flow, maintaining the blood-brain barrier, signaling with neighboring cells, and depositing extracellular matrix. Pericyte dysfunction is an early process in a variety of neurodegenerative conditions. However, remarkably little is known about pericytes at an early site of neurodegeneration in glaucoma, the optic nerve head (ONH). This work summarizes the current understanding of pericyte contributions to ONH physiology, identifies potential roles in glaucomatous pathophysiology, and uncovers open questions at the intersection of these areas. We surveyed the literature to identify the roles of ONH pericytes in the context of health and glaucoma. Additionally, we probed for the presence of pericytes along microvasculature in mouse, nonhuman primate, and human donor ONH tissues. We identified an association between factors influencing ONH dysfunction in glaucoma and factors influencing pericyte dysfunction in other neurodegenerative conditions. Pericytes exist in the mouse, nonhuman primate, and human ONH, implicating their capacity for local function. ONH pericytes represent a promising but underexplored target for treating microvascular impairment in glaucoma. Investigating the contribution of pericytes in both healthy and disease states can help inform mechanisms of dysfunction in glaucomatous pathology, paving the way for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Susannah Waxman
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA
| | - Deborah Villafranca-Baughman
- Department of Neuroscience, Université de Montréal, Montréal, QC; Canada Neuroscience Division, Centre de Recherche du Centre Hospitalier, Université de Montréal, Montréal, QC, Canada
| | - Julie Phillippi
- Division of Cardiac Surgery, Department of Cardiothoracic Surgery, University of Pittsburgh
| | - Tatjana C Jakobs
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston, MA, USA
| | - Luis Alarcon-Martinez
- Centre for Eye Research Australia, Department of Ophthalmology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Adriana Di Polo
- Department of Neuroscience, Université de Montréal, Montréal, QC; Canada Neuroscience Division, Centre de Recherche du Centre Hospitalier, Université de Montréal, Montréal, QC, Canada
| | - Ian A Sigal
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh PA, USA.
| |
Collapse
|
4
|
Chi KY, Kim G, Son JS, Han J, Kim JH. Recent Advances in Three-Dimensional In Vitro Models for Studies of Liver Fibrosis. Tissue Eng Regen Med 2025:10.1007/s13770-025-00719-8. [PMID: 40358834 DOI: 10.1007/s13770-025-00719-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/02/2025] [Accepted: 03/11/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Liver fibrosis is a reversible but complex pathological condition associated with chronic liver diseases, affecting over 1.5 billion people worldwide. It is characterized by excessive extracellular matrix deposition resulting from sustained liver injury, often advancing to cirrhosis and cancer. As its progression involves various cell types and pathogenic factors, understanding the intricate mechanisms is essential for the development of effective therapies. In this context, extensive efforts have been made to establish three-dimensional (3D) in vitro platforms that mimic the progression of liver fibrosis. METHODS This review outlines the pathophysiology of liver fibrosis and highlights recent advancements in 3D in vitro liver models, including spheroids, organoids, assembloids, bioprinted constructs, and microfluidic systems. It further assesses their biological relevance, with particular focus on their capacity to reproduce fibrosis-related characteristics. RESULTS 3D in vitro liver models offer significant advantages over conventional two-dimensional cultures. Although each model exhibits unique strengths, they collectively recapitulate key fibrotic features, such as extracellular matrix remodeling, hepatic stellate cell activation, and collagen deposition, in a physiologically relevant 3D setting. In particular, multilineage liver organoids and assembloids integrate architectural complexity with scalability, enabling deeper mechanistic insights and supporting therapeutic evaluation with improved translational relevance. CONCLUSION 3D in vitro liver models represent a promising strategy to bridge the gap between in vitro studies and in vivo realities by faithfully replicating liver-specific architecture and microenvironments. With enhanced reproducibility through standardized protocols, these models hold great potential for advancing drug discovery and facilitating the development of personalized therapies for liver fibrosis.
Collapse
Affiliation(s)
- Kyun Yoo Chi
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Gyeongmin Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Jeong Sang Son
- User Convenience Technology R&D Department, Korea Institute of Industrial Technology (KITECH), Ansan, 15588, South Korea
| | - Jiyou Han
- Department of Biomedical and Chemical Sciences, Hyupsung University, Hwasung-Si, 18330, South Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
5
|
Feng W, Yang K, Zou Y, Xiao Z, Qian R, Qian R. Progress of ursolic acid on the regulation of macrophage: summary and prospect. Front Immunol 2025; 16:1576771. [PMID: 40421013 PMCID: PMC12104263 DOI: 10.3389/fimmu.2025.1576771] [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: 02/14/2025] [Accepted: 04/15/2025] [Indexed: 05/28/2025] Open
Abstract
Ursolic acid (UA), a prevalent pentacyclic triterpenoid found in numerous fruits and herbs, has garnered significant attention for its vital role in anti-inflammatory processes and immune regulation. The study of immune cells has consistently been a focal point, particularly regarding macrophages, which play crucial roles in antigen presentation, immunomodulation, the inflammatory response, and pathogen phagocytosis. This paper reveals the underlying regulatory effects of UA on the function of macrophages and the specific therapeutic effects of UA on a variety of diseases. Owing to the superior effect of UA on macrophages, different types of macrophages in different tissues have been described. Through the multifaceted regulation of macrophage function, UA may provide new ideas for the development of novel anti-inflammatory and immunomodulatory drugs. However, to facilitate its translation into actual medical means, the specific mechanism of UA in macrophages and its clinical application still need to be further studied.
Collapse
Affiliation(s)
- Wenjing Feng
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Kehong Yang
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Ying Zou
- Department of Anatomy, Anatomy Teaching Center of Hunan University of Chinese Medicine, Changsha, China
| | - Zhaohua Xiao
- Xiangya Hospital, Central South University, Changsha, China
| | - Rongkang Qian
- Department of Integrated Traditional Chinese and Western Medicine, Qian Rongkang Clinic, Loudi, China
| | - Ronghua Qian
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
6
|
Wang J, Shi K, Xu Q, Wang H, Wang Y, Liu S, Jiang W, Chen R, Chen Y, Zhang Y, Wu M, Li X, Li C. Aldose reductase -mediated HUR ubiquitination enhances exosome release and hepatic fibrosis via ROS/PI3K/AKT pathway. Free Radic Biol Med 2025; 236:1-16. [PMID: 40334999 DOI: 10.1016/j.freeradbiomed.2025.04.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/18/2025] [Accepted: 04/26/2025] [Indexed: 05/09/2025]
Abstract
INTRODUCTION Liver fibrosis is caused by the activation of hepatic stellate cells due to various reasons. Our previous research has shown that aldose reductase (AR) played an important role in liver ischemia-reperfusion injury and liver regeneration. OBJECTIVES Here, we aimed to investigate the role and mechanism of AR in the progression of liver fibrosis induced by various factors. METHODS AR expression was detected in liver tissue of fibrosis patients and mouse models. The role and mechanism of AR in fibrosis progression were investigated in AR knockout mice and cell lines. RESULTS AR expression was increased in liver from patients with fibrosis and mouse models. The knockout of AR protected against CCL4 or HFD induced liver injury and development of fibrosis. Furthermore, AR promoted ubiquitization degradation of HUR through competitive binding with OTUB1, thereby exacerbating the accumulation of ROS, and ultimately activating PI3K/AKT pathway. The impaired autophagolysosome resulted in the massive release of exosomes, which activated stellate cells by regulating PTP4a1/SMAD3 pathway. The hepatocyte specific recovery of AR in AR knockout mice aggravated ROS damage and fibrosis, while recovery of HUR in wild-type mice reduced ROS damage and fibrosis. CONCLUSIONS In conclusion, these findings suggest that AR might be a promising therapeutic target for treating liver fibrosis.
Collapse
Affiliation(s)
- Jifei Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China; Gusu School, Nanjing Medical University, Soochow, China
| | - Kuangheng Shi
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Qingqiao Xu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | | | - Yirui Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shuochen Liu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Wangjie Jiang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ruixiang Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yananlan Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yaodong Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Mingyu Wu
- Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, China
| | - Xiangcheng Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China; Gusu School, Nanjing Medical University, Soochow, China.
| | - Changxian Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China.
| |
Collapse
|
7
|
Ramandi A, Diehl AM, Sanyal AJ, de Jong YP. Experimental Models to Investigate PNPLA3 in Liver Steatosis. Liver Int 2025; 45:e70091. [PMID: 40231787 PMCID: PMC12147532 DOI: 10.1111/liv.70091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/16/2025]
Abstract
Patatin-like phospholipase domain-containing 3 (PNPLA3) was the first gene identified through genome-wide association studies to be linked to hepatic fat accumulation. A missense variant, encoding the PNPLA3-148M allele, has since been shown to increase the risk for the full spectrum of steatotic liver disease (SLD), from simple steatosis to steatohepatitis, cirrhosis, and hepatocellular carcinoma. Despite extensive validation of this association and ongoing research into its pathogenic role, the precise mechanisms by which PNPLA3-148M contributes to the progression of SLD remain poorly understood. In this review, we evaluate preclinical in vitro and in vivo models used to investigate PNPLA3 and its involvement in SLD, with particular emphasis on metabolic dysfunction-associated steatotic liver disease. We assess the strengths and limitations of these models, as well as the challenges arising from species differences in PNPLA3 expression and function between human and murine systems.
Collapse
Affiliation(s)
- Alireza Ramandi
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, New York, USA
| | - Anna-Mae Diehl
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Ype P de Jong
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
8
|
Jiao X, Lai L, Qian Y, Sun B, Yang W. Identification of MEOX1 as a potential target in metabolic dysfunction-associated steatohepatitis-related liver fibrosis. Int J Biol Markers 2025:3936155251335975. [PMID: 40270091 DOI: 10.1177/03936155251335975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
BackgroundThe mechanisms underlying the occurrence and progression of metabolic dysfunction-associated steatohepatitis (MASH)-related liver fibrosis remains poorly understood. This study aims to identify key transcription factors involved in the development of liver fibrosis in MASH patients, thereby providing potential targets for drug discovery.MethodsMicroarray data were retrieved from liver biopsy specimens of MASH patients exhibiting varying stages of fibrosis via the Gene Expression Omnibus database. Differentially expressed transcription factors (DETFs) were identified through the application of Weighted Gene Co-expression Network Analysis. A set of in vitro and in vivo experiments were conducted to investigate the role of MEOX1 in MASH-related fibrosis. To delineate the potential mechanisms, the transcriptomic RNA sequencing (RNA-seq), Alphafold, and PyMOL were used.ResultsA total of six DETFs (MEOX1, SOX4, LEF1, SOX9, MYC, and CBX2) were identified as being positively correlated with the progression of MASH-related fibrosis. MEOX1 was increased in mouse model of MASH diet-induced liver fibrosis and hepatic stellate cells (HSCs) stimulated by transforming growth factor-β1. Knockdown of the MEOX1 markedly suppressed the activation, proliferation, and migration of HSCs. RNA-Seq analysis identified serine protease inhibitor family E member 1 (SERPINE1) as the critical target of MEOX1 within HSCs. The protein interaction sites of MEOX1 and SERPINE1 were predicted using Alphafold and PyMOL.ConclusionIn summary, as a pivotal transcription factor, MEOX1 activates HSCs via SERPINE1, thereby promoting liver fibrosis associated with MASH. Inhibition of the MEOX1-SERPINE1 pathway could offer a novel therapeutic avenue for treating MASH-related fibrosis.
Collapse
Affiliation(s)
- Xiaoxiao Jiao
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| | - Linying Lai
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| | - Yiting Qian
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| | - Bo Sun
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| | - Wenzhuo Yang
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
| |
Collapse
|
9
|
Puri M, Sonawane S. Liver Sinusoidal Endothelial Cells in the Regulation of Immune Responses and Fibrosis in Metabolic Dysfunction-Associated Fatty Liver Disease. Int J Mol Sci 2025; 26:3988. [PMID: 40362227 PMCID: PMC12071881 DOI: 10.3390/ijms26093988] [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: 03/12/2025] [Revised: 04/08/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Liver Sinusoidal Endothelial Cells (LSECs) play a crucial role in maintaining liver homeostasis, regulating immune responses, and fibrosis in liver diseases. This review explores the unique functions of LSECs in liver pathology, particularly their roles in immune tolerance, antigen presentation, and the modulation of hepatic stellate cells (HSCs) during fibrosis. LSECs act as key regulators of immune balance in the liver by preventing excessive immune activation while also filtering antigens and interacting with immune cells, including Kupffer cells and T cells. Metabolic Dysfunction-Associated Fatty Liver Disease(MAFLD) is significant because it can lead to advanced liver dysfunction, such as cirrhosis and liver cancer. The prevalence of Metabolic Associated Steatohepatitis (MASH) is increasing globally, particularly in the United States, and is closely linked to rising rates of obesity and type 2 diabetes. Early diagnosis and intervention are vital to prevent severe outcomes, highlighting the importance of studying LSECs in liver disease. However, during chronic liver diseases, LSECs undergo dysfunction, leading to their capillarization, loss of fenestrations, and promotion of pro-fibrotic signaling pathways such as Transforming growth factor-beta (TGF-β), which subsequently activates HSCs and contributes to the progression of liver fibrosis. The review also discusses the dynamic interaction between LSECs, HSCs, and other hepatic cells during the progression of liver diseases, emphasizing how changes in LSEC phenotype contribute to liver scarring and fibrosis. Furthermore, it highlights the potential of LSECs as therapeutic targets for modulating immune responses and preventing fibrosis in liver diseases. By restoring LSECs' function and targeting pathways associated with their dysfunction, novel therapies could be developed to halt or reverse liver disease progression. The findings of this review reinforce the importance of LSECs in liver pathology and suggest that they hold significant promises as targets for future treatment strategies aimed at addressing chronic liver diseases.
Collapse
Affiliation(s)
- Munish Puri
- Onco-Immunology, Magnit Global, Folsom, CA 95630, USA
| | - Snehal Sonawane
- Department of Pathology, University of Illinois, Chicago, IL 60612, USA;
| |
Collapse
|
10
|
Liu X, Mu X, Wang Y, Liu Z, Li Y, Lan J, Feng S, Wang S, Zhao Q. Metal-based mesoporous polydopamine with dual enzyme-like activity as biomimetic nanodrug for alleviating liver fibrosis. J Colloid Interface Sci 2025; 684:586-599. [PMID: 39809020 DOI: 10.1016/j.jcis.2025.01.081] [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: 10/16/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Liver fibrosis is a common pathological stage in the development of several chronic liver diseases, and early intervention can effectively reverse the developing process. Excessive reactive oxygen species (ROS) can promote the activation of hepatic stellate cells (HSCs), but existing treatments have not addressed this problem. In this study, different metal-based mesoporous polydopamine (MPDA) was prepared by the soft template method, and their free radical scavenging abilities, as well as the efficacy and safety of the carriers were investigated, so as to select Cu2+-coordinated MPDA (CMP) as the optimal nanocarrier. CMP exhibited superior SOD- and CAT-like activities compared to MPDA. Subsequently, a novel liver-targeted nanodrug delivery system (Cur/CMPH) with biosafety was constructed. Moreover, Cur/CMPH consisted of CMP loaded with the antifibrotic drug curcumin (Cur/CMP) and coated hyaluronic acid (HA) with liver-targeting properties on the surface of Cur/CMP, thus effectively intervening in the progression of liver fibrosis. Cur/CMPH possessed uniform particle size, negative Zeta potential, excellent antioxidant capacity, and pH-responsive drug release. Furthermore, Cur/CMPH in vitro studies demonstrated efficient cellular uptake, inhibition of the proliferation of HSCs, and excellent intracellular ROS scavenging without cytotoxicity. Besides, Cur/CMPH had specific targeting effect on fibrotic liver as well as good accumulation ability. In vivo studies, Cur/CMPH showcased the combined therapeutic effect of Cur and CMP, which significantly decreased the deposition of collagen fibers and alleviated the degree of liver fibrosis with good biosafety. In summary, the construction of Cur/CMPH opens up a novel idea in the field of nanodrug delivery systems for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Xinlin Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Xiaoyang Mu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Ying Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Zhu Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Yian Li
- School of Libra Arts of Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Jiaru Lan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Shuaipeng Feng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|
11
|
Khaliq A, Badshah H, Shah Y. Combination therapy with vitamin E and ertugliflozin in patients with non-alcoholic fatty liver disease and type 2 diabetes mellitus: a randomized clinical trial. Ir J Med Sci 2025:10.1007/s11845-025-03945-0. [PMID: 40202706 DOI: 10.1007/s11845-025-03945-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a common comorbidity in type 2 diabetes mellitus (T2DM), with shared pathophysiological mechanisms, including insulin resistance, oxidative stress, and inflammation. OBJECTIVES This study evaluates the effects of vitamin E and ertugliflozin, individually and in combination, alongside standard pioglitazone therapy, on hepatic and metabolic parameters in patients with NAFLD and T2DM. METHODS A 24-week, double-blind, randomized, controlled clinical trial on 173 patients with NAFLD and T2DM was assigned into four groups: vitamin E (n = 42), pioglitazone (n = 43), ertugliflozin (n = 44), and vitamin E + ertugliflozin (n = 44) combination therapy. The primary outcome was to monitor changes in liver steatosis assessed via ultrasound. Secondary outcomes included evaluation of liver enzymes, glycemic control, fibrosis markers, and lipid profiles. RESULTS Combination therapy of vitamin E + ertugliflozin showed the highest decrease in liver fat content, with 11 participants achieving successful Grade 0 (p < 0.001). Significant improvements were also observed in glycemic control, HbA1c, triglycerides, and liver enzymes. Ertugliflozin monotherapy showed significant efficacy in improving liver enzymes, glycemic parameters, and fibrosis markers. Pioglitazone improved the initial stage of NAFLD but had a limited impact on advanced fibrosis. Ertugliflozin, in combination with vitamin E, decreases oxidative stress; however, vitamin E by itself has no impact on the metabolic and fibrosis index. CONCLUSION The ertugliflozin and vitamin E combination is a very effective treatment for patients with NAFLD and T2DM. It improves hepatic steatosis and metabolic indicators. Exploration is required for combination therapy in order to assess the prolonged efficacy and safety of the treatment.
Collapse
Affiliation(s)
- Adil Khaliq
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Haroon Badshah
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan.
| | - Yasar Shah
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| |
Collapse
|
12
|
Luo YY, Guan YP, Zhan HF, Sun CY, Cai LY, Tao KG, Lin Y, Zeng X. Circ_0098181 binds PKM2 to attenuate liver fibrosis. Front Pharmacol 2025; 16:1517250. [PMID: 40248098 PMCID: PMC12003362 DOI: 10.3389/fphar.2025.1517250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/14/2025] [Indexed: 04/19/2025] Open
Abstract
Background Liver cirrhosis seriously harms human health and fibrosis is the essential pathological process of cirrhosis. Recently, circular RNAs (circRNAs) were found to play critical roles in liver fibrosis, but the key circRNAs and precise mechanisms remained unclear. This study aimed to investigate the effect of circ_0098181 in fibrogenesis and explore its mechanism. Methods RNA sequencing was conducted to identify circRNA signatures in human liver cirrhotic tissues. Hepatic stellate cells (HSCs) (including primary rat HSCs, LX2, HSC-T6) and carbon tetrachloride (CCl4) induced liver cirrhosis model were used to explore the role of circ_0098181 on HSC activation and liver fibrogenesis in vitro and in vivo. RNA sequencing, RNA pull-down, mass spectrometry, and RNA immunoprecipitation (RIP) experiments were performed to elucidate the mechanism. Results Circ_0098181 was obviously reduced in human fibrotic liver tissues and activated HSCs. Exogenous administration of circ_0098181 blocked the activation, proliferation, and migration of HSCs in vitro and mitigated the progression of CCl4-induced liver fibrosis in vivo. Mechanistically, adenosine deaminase acting on RNA1 (ADAR1) combined with the intronic complementary sequences (ICSs) in the flanking regions, thereby regulating the biogenesis of circ_0098181. RNA sequencing and qRT-PCR revealed the suppression of circ_0098181 on pro-inflammation cytokines expression (TNFα, Fas, Cxcl11, etc.). RNA pull-down, mass spectrometry, and RIP experiments indicated that pyruvate kinase M2 (PKM2) was the direct target of circ_0098181. Circ_0098181 bound to PKM2, restrained its nuclear translocation and phosphorylation. Conclusion In conclusion, circ_0098181 exerts a significant anti-fibrotic effect by binding PKM2 to repress its nuclear translocation and inhibiting hepatic inflammation, suggesting the promising therapeutic merit in liver cirrhosis.
Collapse
Affiliation(s)
- Yuan-Yuan Luo
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ya-Ping Guan
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hong-Fei Zhan
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chun-Yan Sun
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ling-Yan Cai
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ke-Gong Tao
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yong Lin
- Department of Gastroenterology, Shanghai Changzheng Hospital, Navy Military Medical University, Shanghai, China
| | - Xin Zeng
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Jiao X, Lai L, Sun B, Qian Y, Yang W. The transcription factor mesenchyme homeobox 1 exacerbates hepatic fibrosis by transcriptional activation of connective tissue growth factor. Exp Cell Res 2025; 447:114513. [PMID: 40073959 DOI: 10.1016/j.yexcr.2025.114513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/26/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
As a chronic condition, liver fibrosis is characterized by diverse etiological factors, and the pivotal event to its pathogenesis is the activation of quiescent hepatic stellate cells (HSCs) into myofibroblasts. Mesenchyme homeobox 1 (MEOX1) is a transcription factors central to cellular development and differentiation. However, the role of MEOX1 signaling in hepatic fibrosis still remains largely unknown. In this study, we investigated the potential role and mechanism of MEOX1 in liver fibrosis using different models in vivo and in vitro. The hepatic expression of MEOX1 exhibited a positive correlation with the degree of fibrosis in patients diagnosed with non-alcoholic steatohepatitis (NASH), as determined through bioinformatics analysis. Furthermore, MEOX1 demonstrated high expression levels in activated HSCs and fibrotic liver tissues induced by methionine and choline-deficient diet (MCD), thioacetamide (TAA), or carbon tetrachloride (CCl4) treatment in C57/BL6 mice. Mechanistically, MEOX1 facilitated HSC activation, proliferation, and migration. The comprehensive analysis of transcriptome sequencing and chromatin immunoprecipitation sequencing data revealed that connective tissue growth factor (CTGF) served as a target gene for MEOX1 in HSCs. Specifically, MEOX1 bound to the promoter region of CTGF and enhanced its transcriptional activity, thereby mediating the exacerbating effect of MEOX1 on hepatic fibrosis. In conclusion, our current findings elucidate the role of MEOX1 in exacerbating hepatic fibrosis progression through transcriptional activation of CTGF. Our findings provide valuable insights into the therapeutic potential of targeting MEOX1 for the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Xiaoxiao Jiao
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, PR China
| | - Linying Lai
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, PR China
| | - Bo Sun
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, PR China
| | - Yiting Qian
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, PR China
| | - Wenzhuo Yang
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, PR China.
| |
Collapse
|
14
|
Chaulagain RP, Padder AM, Shrestha H, Gupta R, Bhandari R, Shrestha Y, Qasem Moqbel A, Gautam S, Lal N, Jin S. Deciphering the Matrisome: Extracellular Matrix Remodeling in Liver Cirrhosis and Hepatocellular Carcinoma. Cureus 2025; 17:e82171. [PMID: 40370880 PMCID: PMC12076258 DOI: 10.7759/cureus.82171] [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] [Accepted: 04/13/2025] [Indexed: 05/16/2025] Open
Abstract
Liver cirrhosis and hepatocellular carcinoma (HCC) are major public health concerns due to their high morbidity and mortality rates. The liver, a vital organ for metabolism, detoxification, and homeostasis, depends on the matrisome, a complex and dynamic network of extracellular matrix (ECM) components for maintaining structural and functional integrity. Chronic liver inflammation, induced by factors such as alcohol abuse, viral hepatitis, and non-alcoholic fatty liver disease, leads to fibrosis and cirrhosis, progressing to HCC. The matrisome, composed of ECM proteins including collagen, fibronectin, and laminin, plays a critical role in regulating tissue homeostasis, cell signaling, and tissue repair. Dysregulation of ECM components contributes to the pathogenesis of both liver cirrhosis and cancer. In cirrhosis, matrisome alterations are characterized by excessive ECM deposition and fibrosis, which disrupt the liver's architecture and impair its function. Activated hepatic stellate cells (HSCs) are the principal mediators of fibrosis, producing large quantities of ECM components. In liver cancer, matrisome remodeling facilitates tumorigenesis by promoting cancer cell proliferation, invasion, and metastasis. The tumor microenvironment, shaped by ECM alterations, further supports tumor growth and dissemination. Matrix metalloproteinases (MMPs) play a pivotal role in ECM degradation, fibrosis progression, and tumor invasion, while tissue inhibitors of metalloproteinases (TIMPs) modulate MMP activity. A comprehensive understanding of the molecular mechanisms that link matrisome alterations with the progression from cirrhosis to liver cancer is essential for identifying novel diagnostic and therapeutic targets. This review highlights the dynamic responses of the hepatic matrisome to both acute and chronic insults, emphasizing the complex interplay between ECM components, cellular behavior, and disease progression. Elucidating these interactions may inform strategies aimed at improving clinical outcomes for patients with liver cirrhosis and HCC.
Collapse
Affiliation(s)
- Ram Prasad Chaulagain
- Internal Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin, CHN
| | - Aadil Mushtaq Padder
- Gastroenterology and Hepatology, Second Affiliated Hospital of Harbin Medical University, Harbin, CHN
| | | | - Radheshyam Gupta
- Urology Surgery, Cancer Hospital, Harbin Medical University, Harbin, CHN
| | - Rameshor Bhandari
- Surgical Gastroenterology, Grande International Hospital, Kathmandu, NPL
| | - Yelona Shrestha
- Dermatology, First Affiliated Hospital of Xinjiang Medical University, Xinxiang, CHN
| | | | - Smriti Gautam
- Dermatology, Kathmandu Medical College, Kathmandu, NPL
| | - Nand Lal
- Physiology, School of Biomedical Sciences, Harbin Medical University, Harbin, CHN
| | - Shizhu Jin
- Gastroenterology and Hepatology, Second Affiliated Hospital of Harbin Medical University, Harbin, CHN
| |
Collapse
|
15
|
Xu F, Gao Y, Li T, Jiang T, Wu X, Yu Z, Zhang J, Hu Y, Cao J. Single-Cell Sequencing Reveals the Heterogeneity of Hepatic Natural Killer Cells and Identifies the Cytotoxic Natural Killer Subset in Schistosomiasis Mice. Int J Mol Sci 2025; 26:3211. [PMID: 40244063 PMCID: PMC11989782 DOI: 10.3390/ijms26073211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/08/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
Schistosoma japonicum eggs in the host liver form granuloma and liver fibrosis and then lead to portal hypertension and cirrhosis, seriously threatening human health. Natural killer (NK) cells can kill activated hepatic stellate cells (HSCs) against hepatic fibrosis. We used single-cell sequencing to screen hepatic NK cell subsets against schistosomiasis liver fibrosis. Hepatic NK cells were isolated from uninfected mice and mice infected for four and six weeks. The NK cells underwent single-cell sequencing. The markers' expression in the NK subsets was detected through Reverse Transcription-Quantitative PCR (RT-qPCR). The proportion and granzyme B (Gzmb) expression of the total NK and Thy1+NK were detected. NK cells overexpressing Thy1 (Thy1-OE) were constructed, and functions were detected. The results revealed that the hepatic NK cells could be divided into mature, immature, regulatory-like, and memory-like NK cells and re-clustered into ten subsets. C3 (Cx3cr1+NK) and C4 (Thy1+NK) increased at week four post-infection, and other subsets decreased continuously. The successfully constructed Thy1-OE NK cells had significantly higher effector molecules and induced greater HSC apoptosis than the control NK cells. It revealed a pattern of hepatic NK cells in a mouse model of schistosomiasis. The Thy1+NK cells could be used as target cells against hepatic fibrosis.
Collapse
Affiliation(s)
- Fangfang Xu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 200025, China; (F.X.); (Y.G.); (T.L.); (T.J.); (X.W.); (Z.Y.); (J.Z.)
| | - Yuan Gao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 200025, China; (F.X.); (Y.G.); (T.L.); (T.J.); (X.W.); (Z.Y.); (J.Z.)
| | - Teng Li
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 200025, China; (F.X.); (Y.G.); (T.L.); (T.J.); (X.W.); (Z.Y.); (J.Z.)
| | - Tingting Jiang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 200025, China; (F.X.); (Y.G.); (T.L.); (T.J.); (X.W.); (Z.Y.); (J.Z.)
| | - Xiaoying Wu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 200025, China; (F.X.); (Y.G.); (T.L.); (T.J.); (X.W.); (Z.Y.); (J.Z.)
| | - Zhihao Yu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 200025, China; (F.X.); (Y.G.); (T.L.); (T.J.); (X.W.); (Z.Y.); (J.Z.)
| | - Jing Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 200025, China; (F.X.); (Y.G.); (T.L.); (T.J.); (X.W.); (Z.Y.); (J.Z.)
| | - Yuan Hu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 200025, China; (F.X.); (Y.G.); (T.L.); (T.J.); (X.W.); (Z.Y.); (J.Z.)
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai 200025, China; (F.X.); (Y.G.); (T.L.); (T.J.); (X.W.); (Z.Y.); (J.Z.)
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
16
|
Qian B, Zhao Y, Zhang X, Zhao C, Cui X, Wang F, Jing X, Ge L, Yao Z, Gao X, Yang J. Tudor staphylococcal nuclease (Tudor-SN) regulates activation of quiescent hepatic stellate cells. FEBS J 2025. [PMID: 40098321 DOI: 10.1111/febs.70073] [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: 07/07/2024] [Revised: 09/26/2024] [Accepted: 03/11/2025] [Indexed: 03/19/2025]
Abstract
Several liver diseases have been associated with the Tudor staphylococcal nuclease (Tudor-SN) protein. Our previous results demonstrated that, in comparison to wild-type (WT) mice, systemic overexpression of Tudor-SN in transgenic (Tg) mice (Tudor-SN-Tg) ameliorates obesity-induced insulin resistance and hepatic steatosis. In this study, we observed an inverse correlation in the expression levels of Tudor-SN and profibrogenic factors, such as alpha-smooth muscle actin (α-SMA) and collagen alpha-1(I) chain (COL1A1), in liver tissue samples between Tudor-SN-Tg and WT mice. The correlation was further validated in hepatic fibrotic tissues from patients with cirrhosis and fibrosis. Utilizing a carbon tetrachloride (CCl4)-induced hepatic fibrosis model, we observed that Tudor-SN attenuated hepatic fibrosis in mice. Tudor-SN was abundantly expressed in hepatic stellate cells (HSCs). In the Tudor-SN-Tg group, primary HSCs showed stellate-like morphology as well as reduced in vitro proliferation and chemotactic ability compared to the WT group. Pseudotime series analysis of HSCs further showed the role of Tudor-SN during the dynamic evolution of HSC activation. Reduced Tudor-SN expression facilitated the in vitro activation of LX-2 cells. Furthermore, primary HSC cells from WT and Tudor-SN knockout (KO) mice were isolated for RNA-sequencing analysis. The findings suggested that Tudor-SN may regulate the activation of primary HSCs by influencing lipid metabolism, translation initiation, immune response, and the extracellular matrix. In summary, we identified Tudor-SN as a newly identified regulator involved in the transition of quiescent HSCs to activated states, shedding light on the antifibrotic impact of Tudor-SN expression in the development of hepatic fibrosis.
Collapse
Affiliation(s)
- Baoxin Qian
- State Key Laboratory of Experimental Hematology, Key Laboratory of Cellular and Molecular Immunology in Tianjin, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Tianjin Medical University, China
- Department of Immunology, School of Basic Medical Science, Tianjin Medical University, China
- Department of Gastroenterology and Hepatology, Nankai University Affinity the Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, China
| | - Yan Zhao
- State Key Laboratory of Experimental Hematology, Key Laboratory of Cellular and Molecular Immunology in Tianjin, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Tianjin Medical University, China
- Department of Immunology, School of Basic Medical Science, Tianjin Medical University, China
| | - Xinxin Zhang
- State Key Laboratory of Experimental Hematology, Key Laboratory of Cellular and Molecular Immunology in Tianjin, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Tianjin Medical University, China
- Department of Immunology, School of Basic Medical Science, Tianjin Medical University, China
| | - Chunyan Zhao
- State Key Laboratory of Experimental Hematology, Key Laboratory of Cellular and Molecular Immunology in Tianjin, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Tianjin Medical University, China
- Department of Immunology, School of Basic Medical Science, Tianjin Medical University, China
- Department of Medical Laboratory, Tianjin Hospital, Tianjin University, China
| | - Xiaoteng Cui
- State Key Laboratory of Experimental Hematology, Key Laboratory of Cellular and Molecular Immunology in Tianjin, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Tianjin Medical University, China
- Department of Immunology, School of Basic Medical Science, Tianjin Medical University, China
| | - Fengmei Wang
- Department of Gastroenterology and Hepatology, Nankai University Affinity the Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, China
- Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin First Central Hospital, China
| | - Xiang Jing
- Department of Ultrasound, Nankai University Affinity the Third Central Hospital, Tianjin, China
| | - Lin Ge
- State Key Laboratory of Experimental Hematology, Key Laboratory of Cellular and Molecular Immunology in Tianjin, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Tianjin Medical University, China
- Department of Immunology, School of Basic Medical Science, Tianjin Medical University, China
| | - Zhi Yao
- State Key Laboratory of Experimental Hematology, Key Laboratory of Cellular and Molecular Immunology in Tianjin, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Tianjin Medical University, China
- Department of Immunology, School of Basic Medical Science, Tianjin Medical University, China
| | - Xingjie Gao
- State Key Laboratory of Experimental Hematology, Key Laboratory of Cellular and Molecular Immunology in Tianjin, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Tianjin Medical University, China
- Department of Immunology, School of Basic Medical Science, Tianjin Medical University, China
| | - Jie Yang
- State Key Laboratory of Experimental Hematology, Key Laboratory of Cellular and Molecular Immunology in Tianjin, and Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Tianjin Medical University, China
- Department of Immunology, School of Basic Medical Science, Tianjin Medical University, China
| |
Collapse
|
17
|
Hassan MAM, Fahmy MI, Azzam HN, Ebrahim YM, El-Shiekh RA, Aboulmagd YM. Multifaceted therapeutic potentials of catalpol, an iridoid glycoside: an updated comprehensive review. Inflammopharmacology 2025:10.1007/s10787-025-01694-1. [PMID: 40097877 DOI: 10.1007/s10787-025-01694-1] [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: 12/04/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
Catalpol, classified as an iridoid glucoside, is recognized for its significant role in medicine, particularly in the treatment of various conditions such as diabetes mellitus, neuronal disorders, and inflammatory diseases. This review aims to evaluate the biological implications of catalpol and the mechanisms underlying its diverse pharmacological effects. A thorough exploration of existing literature was conducted utilizing the keyword "Catalpol" across prominent public domains like Google Scholar, PubMed, and EKB. Catalpol has demonstrated a diverse array of pharmacological effects in experimental models, showcasing its anti-diabetic, cardiovascular-protective, neuroprotective, anticancer, hepatoprotective, anti-inflammatory, and antioxidant properties. In summary, catalpol manifests a spectrum of biological effects through a myriad of mechanisms, prominently featuring its anti-inflammatory and antioxidant capabilities. Its diverse pharmacological profile underscores its potential for therapeutic applications across a range of conditions. Further research is warranted to fully elucidate the clinical implications of catalpol and optimize its use in medical practice.
Collapse
Affiliation(s)
- Mennat-Allah M Hassan
- Department of Pharmacology & Toxicology Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Mohamed I Fahmy
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| | - Hany N Azzam
- Pharmacy Practice Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Yasmina M Ebrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Yara M Aboulmagd
- Department of Pharmacology & Toxicology Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| |
Collapse
|
18
|
Lee W, Bates EA, Kipp ZA, Pauss SN, Martinez GJ, Blair CA, Hinds TD. Insulin receptor responsiveness governs TGFβ-induced hepatic stellate cell activation: Insulin resistance instigates liver fibrosis. FASEB J 2025; 39:e70427. [PMID: 40022609 PMCID: PMC11871568 DOI: 10.1096/fj.202402169r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/21/2024] [Accepted: 02/19/2025] [Indexed: 03/03/2025]
Abstract
The insulin receptor (INSR) has been shown to be hyperactive in hepatic stellate cells (HSCs) in humans and rodents with liver fibrosis. To explore HSC cellular mechanisms that INSR regulates during pro-fibrotic stimulation, we used CRISPR-Cas9 technology. We knocked out a portion of the INSR gene in human LX2 HSC cells (INSRe5-8 KO) that regulates insulin responsiveness but not the insulin-like growth factor (IGF) or transforming growth factor-β (TGFβ) signaling. The INSRe5-8 KO HSCs had significantly higher cell growth, BrdU incorporation, and lower TP53 expression that suppresses growth, and they also exhibited increased migration compared to the Scramble control. We treated the scramble control and INSRe5-8 KO HSCs with insulin or TGFβ and profiled hundreds of kinase activities using the PamGene PamStation kinome technology. Our analysis showed that serine/threonine kinase (STK) activities were reduced, and most of the protein-tyrosine kinase (PTK) activities were increased in the INSRe5-8 KO compared to the Scramble control HSCs. To study gene transcripts altered in activated Scramble control and INSRe5-8 KO HSCs, we treated them with TGFβ for 24 h. We isolated RNA for sequencing and found that the INSRe5-8 KO cells, compared to control HSCs, had altered transcriptional responsiveness to TGFβ stimulation, collagen-activated signaling, smooth muscle cell differentiation pathways, SMAD protein signaling, collagen metabolic process, integrin-mediated cell adhesion, and notch signaling. This study demonstrates that reduced INSR responsiveness enhances HSC growth and selectively mediates TGFβ-induced HSC activation. These findings provide new insights into the development of more effective treatments for liver fibrosis.
Collapse
Grants
- R01DK121797 HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
- R01DA058933 HHS | NIH | National Institute on Drug Abuse (NIDA)
- F31HL170972 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- F31HL175979 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 25PRE1374495 American Heart Association (AHA)
- HHS | NIH | NIDDK | Division of Diabetes, Endocrinology, and Metabolic Diseases (DEM)
- HHS | NIH | National Institute on Drug Abuse (NIDA)
- HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- American Heart Association (AHA)
Collapse
Affiliation(s)
- Wang‐Hsin Lee
- Drug & Disease Discovery D3 Research CenterUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Evelyn A. Bates
- Drug & Disease Discovery D3 Research CenterUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Zachary A. Kipp
- Drug & Disease Discovery D3 Research CenterUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Sally N. Pauss
- Drug & Disease Discovery D3 Research CenterUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Genesee J. Martinez
- Drug & Disease Discovery D3 Research CenterUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Cheavar A. Blair
- Drug & Disease Discovery D3 Research CenterUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Markey Cancer CenterUniversity of KentuckyLexingtonKentuckyUSA
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Terry D. Hinds
- Drug & Disease Discovery D3 Research CenterUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Markey Cancer CenterUniversity of KentuckyLexingtonKentuckyUSA
- Barnstable Brown Diabetes CenterUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| |
Collapse
|
19
|
Liu X, Fang C, Yu H, Huang L, Feng J, Luo S, Song L, Wu M, Tan Y, Dong J, Gong T, Xiao P. Chondroitin Sulfate-Based Imatinib Nanoparticles Targeting Activated Hepatic Stellate Cells Against Hepatic Fibrosis. Pharmaceutics 2025; 17:351. [PMID: 40143016 PMCID: PMC11944399 DOI: 10.3390/pharmaceutics17030351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/16/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Activated hepatic stellate cells (aHSCs) play a significant role during the onset of hepatic fibrosis, ultimately leading to excessive deposition of extracellular matrix (ECM) and other typical pathological features, and thus have become a popular target for the treatment of hepatic fibrosis. However, current aHSC-centric therapy strategies achieve unsatisfactory results, mainly due to the lack of approved anti-fibrosis drugs and sufficiently efficient aHSC-targeted delivery systems. In this study, our aim was to develop an Imatinib-loaded nanoparticle delivery system based on a chondroitin sulfate derivative to enhance aHSC targeting efficiency, improve the therapeutic effect for hepatic fibrosis, and investigate the underlying mechanism. Methods: The carboxyl group of chondroitin sulfate and the amino group of 1-hexadecylamine were linked by an amide bond in this study to produce the amphiphilic carrier CS-HDA. Then, the Imatinib-loaded nanoparticles (IM-CS NPs) were designed to efficiently target aHSCs through CD44-mediated endocytosis and effectively inhibit HSC overactivation via PDGF and TGF-β signaling pathways. Results: Both in vitro cellular uptake experiments and in vivo distribution experiments demonstrated that CS-HDA-modified nanoparticles (IM-CS NPs) exhibited a better targeting ability for aHSCs, which were subsequently utilized to treat carbon tetrachloride-induced hepatic fibrosis mouse models. Finally, significant fibrosis resolution was observed in the carbon tetrachloride-induced hepatic fibrosis mouse models after tail vein injection of the IM-CS NPs, along with their outstanding biocompatibility and biological safety. Conclusions: IM-loaded NPs based on an amphiphilic CS derivative have remarkable antifibrotic effects, providing a promising avenue for the clinical treatment of advanced hepatic fibrosis.
Collapse
Affiliation(s)
- Xunzhi Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (X.L.); (H.Y.); (L.H.); (J.F.); (S.L.); (M.W.); (Y.T.); (T.G.)
| | - Changlong Fang
- Department of Pharmacy, Chongqing University Fuling Hospital, Chongqing University, Chongqing 408099, China;
| | - Hongling Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (X.L.); (H.Y.); (L.H.); (J.F.); (S.L.); (M.W.); (Y.T.); (T.G.)
| | - Lu Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (X.L.); (H.Y.); (L.H.); (J.F.); (S.L.); (M.W.); (Y.T.); (T.G.)
| | - Jiaxing Feng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (X.L.); (H.Y.); (L.H.); (J.F.); (S.L.); (M.W.); (Y.T.); (T.G.)
| | - Shiqin Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (X.L.); (H.Y.); (L.H.); (J.F.); (S.L.); (M.W.); (Y.T.); (T.G.)
| | - Li Song
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China;
| | - Mengying Wu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (X.L.); (H.Y.); (L.H.); (J.F.); (S.L.); (M.W.); (Y.T.); (T.G.)
| | - Yulu Tan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (X.L.); (H.Y.); (L.H.); (J.F.); (S.L.); (M.W.); (Y.T.); (T.G.)
| | - Jianxia Dong
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (X.L.); (H.Y.); (L.H.); (J.F.); (S.L.); (M.W.); (Y.T.); (T.G.)
| | - Peihong Xiao
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China;
| |
Collapse
|
20
|
Ran Q, Song D, Wang Q, Wang D, Chen X, Zhang A, Ma L. Resveratrol Alleviates Arsenic Exposure-Induced Liver Fibrosis in Rats by Inhibiting Hepatocyte Senescence. Biol Trace Elem Res 2025; 203:1528-1538. [PMID: 38831176 DOI: 10.1007/s12011-024-04255-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
Arsenic is an environmental pollutant that has garnered considerable attention from the World Health Organization. Liver fibrosis is an advanced pathological stage of liver injury that can be caused by chronic arsenic exposure and has the potential to be reversed to prevent cirrhosis and hepatic malignancies. However, effective treatment options are currently limited. Given the profibrogenic effect of hepatocyte senescence, we established a rat model of sub-chronic sodium arsenite exposure and investigated the ability of resveratrol (RSV), a potential anti-senescence agent, to ameliorate arsenic-induced liver fibrosis and elucidate the underlying mechanism from the perspective of hepatocyte senescence. The results demonstrated that RSV was capable of mitigating fibrosis phenotypes in rat livers, including the activation of hepatic stellate cell (HSC), the generation of extracellular matrix, and the deposition of collagen fibers in the liver vascular zone, which are all induced by arsenic exposure. Furthermore, as an activator of the longevity factor SIRT1, RSV antagonized the arsenic-induced inhibition of SIRT1 expression, thereby restoring the suppression of the senescence protein p16 by SIRT1. This prevented arsenic-induced hepatocyte senescence, manifesting as a decrease in telomere shortening and a reduction in the release of senescence-associated secretory phenotype (SASP)-related proteins. In conclusion, this study demonstrated that RSV counteracts arsenic-induced hepatocyte senescence and the release of SASP-related proteins by restoring the inhibitory effect of SIRT1 on p16, thereby suppressing the activation of fibrotic phenotypes and mitigating liver fibrosis. These findings provide new insights for understanding the mechanism of arsenic-induced liver fibrosis, and more importantly, they reveal novel potential interventional approaches.
Collapse
Affiliation(s)
- Qiming Ran
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
- Collaborative Innovation Center for Prevention, Control of Endemic and Ethnic Regional Diseases Co-Constructed By the Province and Ministry, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Dingyi Song
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
- Collaborative Innovation Center for Prevention, Control of Endemic and Ethnic Regional Diseases Co-Constructed By the Province and Ministry, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Qi Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
- Collaborative Innovation Center for Prevention, Control of Endemic and Ethnic Regional Diseases Co-Constructed By the Province and Ministry, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Dapeng Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
- Collaborative Innovation Center for Prevention, Control of Endemic and Ethnic Regional Diseases Co-Constructed By the Province and Ministry, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Xiong Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
- Collaborative Innovation Center for Prevention, Control of Endemic and Ethnic Regional Diseases Co-Constructed By the Province and Ministry, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China.
- Collaborative Innovation Center for Prevention, Control of Endemic and Ethnic Regional Diseases Co-Constructed By the Province and Ministry, Guizhou Medical University, Guiyang, 550025, Guizhou, China.
| | - Lu Ma
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China.
- Collaborative Innovation Center for Prevention, Control of Endemic and Ethnic Regional Diseases Co-Constructed By the Province and Ministry, Guizhou Medical University, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
21
|
Chen M, Song L, Zeng A. Harnessing nature's arsenal: Targeting the TGF-β/Smad Cascade with novel natural anti-fibrotic agents. Fitoterapia 2025; 181:106372. [PMID: 39778722 DOI: 10.1016/j.fitote.2024.106372] [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: 07/15/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Hepatic fibrosis is a wound healing response that leads to excessive deposition of extracellular matrix (ECM) due to sustained liver injury. Hepatic stellate cells (HSCs) are key players in ECM synthesis, with the TGF-β/Smad signaling pathway being central to their activation. Despite advances in understanding the pathogenesis of hepatic fibrosis, effective anti-fibrotic therapies are still lacking. METHODS This treatise conducts a comprehensive review of the literature on the hepatoprotective effects of natural products, including natural medicine compounds, herbal extracts, and polysaccharides. The focus is on their ability to modulate the TGF-β pathway, which is critical in the activation of HSCs and ECM synthesis in hepatic fibrosis. RESULTS The review identifies a variety of natural products that have shown promise in inhibiting the TGF-β/Smad signaling cascade, thereby reducing the activation of HSCs and ECM accumulation. These findings highlight the potential of these natural products as therapeutic agents in the treatment of hepatic fibrosis. CONCLUSIONS The exploration of natural products as modulators of the TGF-β pathway presents a novel avenue for both clinical and preclinical research into hepatic fibrosis. Further investigation is warranted to fully understand the mechanisms of action and to develop these compounds into effective anti-fibrotic pharmaceuticals.
Collapse
Affiliation(s)
- Maohua Chen
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine,Chengdu, Sichuan 610041, PR China; Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China. Chengdu, Sichuan 610072, PR China
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, PR China.
| | - Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine,Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
22
|
Kim DH, Go HS, Jeon EJ, Nguyen TQT, Kim DY, Park H, Eom HJ, Kim SY, Park SC, Cho KA. The Impact of Toll-Like Receptor 5 on Liver Function in Age-Related Metabolic Disorders. Aging Cell 2025:e70009. [PMID: 39957532 DOI: 10.1111/acel.70009] [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: 08/15/2024] [Revised: 12/09/2024] [Accepted: 01/10/2025] [Indexed: 02/18/2025] Open
Abstract
Toll-like receptor 5 (TLR5) plays a critical role beyond its traditional function in innate immunity, significantly impacting metabolic regulation and liver health. Previously, we reported that TLR5 activation extends the healthspan and lifespan of aging mice. This study demonstrates that TLR5 deficiency leads to pronounced metabolic abnormalities with age, primarily affecting liver metabolic functions rather than intestinal inflammation. Comprehensive RNA sequencing analysis revealed that TLR5 deficiency induces gene expression changes in liver tissue similar to those caused by the methionine-choline deficient (MCD) diet, particularly affecting lipid metabolism and circadian rhythm-related genes. TLR5 knockout (TLR5 KO) mice displayed an increased propensity for liver fibrosis and lipid accumulation under the MCD diet, exacerbating liver pathology. Both hepatocytes and hepatic stellate cells in TLR5 KO mice were functionally impacted, leading to metabolic dysfunction and fibrosis. These findings suggest that TLR5 could be a significant target for addressing metabolic diseases that arise and worsen with aging. Furthermore, understanding the mechanisms by which TLR5 activation extends healthspan could provide valuable insights into therapeutic strategies for enhancing longevity and managing age-related metabolic disorders.
Collapse
Affiliation(s)
- Dong-Hyun Kim
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, Republic of Korea
| | - Hye Sun Go
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, Republic of Korea
| | - Eun Jae Jeon
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, Republic of Korea
| | - Thi Quynh Trang Nguyen
- Department of Biochemistry, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, Republic of Korea
- Center for Creative Biomedical Scientists, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, Republic of Korea
| | - Da Yeon Kim
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, Republic of Korea
| | - Hansung Park
- Department of Biochemistry, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, Republic of Korea
| | - Hyo-Ji Eom
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, Republic of Korea
| | - Sung Young Kim
- Department of Biochemistry, Konkuk University School of Medicine, Seoul, South Korea
| | - Sang Chul Park
- Future Life and Society Research Center, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, Republic of Korea
| | - Kyung A Cho
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, Republic of Korea
- Department of Biochemistry, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, Republic of Korea
- Center for Creative Biomedical Scientists, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, Republic of Korea
| |
Collapse
|
23
|
Sharip A, Kunz J. Mechanosignaling via Integrins: Pivotal Players in Liver Fibrosis Progression and Therapy. Cells 2025; 14:266. [PMID: 39996739 PMCID: PMC11854242 DOI: 10.3390/cells14040266] [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: 12/09/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Liver fibrosis, a consequence of chronic liver injury, represents a major global health burden and is the leading cause of liver failure, morbidity, and mortality. The pathological hallmark of this condition is excessive extracellular matrix deposition, driven primarily by integrin-mediated mechanotransduction. Integrins, transmembrane heterodimeric proteins that serve as primary ECM receptors, orchestrate complex mechanosignaling networks that regulate the activation, differentiation, and proliferation of hepatic stellate cells and other ECM-secreting myofibroblasts. These mechanical signals create self-reinforcing feedback loops that perpetuate the fibrotic response. Recent advances have provided insight into the roles of specific integrin subtypes in liver fibrosis and revealed their regulation of key downstream effectors-including transforming growth factor beta, focal adhesion kinase, RhoA/Rho-associated, coiled-coil containing protein kinase, and the mechanosensitive Hippo pathway. Understanding these mechanotransduction networks has opened new therapeutic possibilities through pharmacological manipulation of integrin-dependent signaling.
Collapse
Affiliation(s)
- Aigul Sharip
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan;
- Laboratory of Bioinformatics and Systems Biology, National Laboratory Astana, Astana 020000, Kazakhstan
| | - Jeannette Kunz
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan;
| |
Collapse
|
24
|
Jain S. Can Schistosoma japonicum infection cause liver cancer? J Helminthol 2025; 99:e11. [PMID: 39924660 DOI: 10.1017/s0022149x24000762] [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] [Indexed: 02/11/2025]
Abstract
A co-relation between Schistosoma japonicum (Sj) and liver cancer (LC) in humans has been reported in the literature; however, this association is circumstantial. Due to the inconclusive nature of this association, the International Agency for Research on Cancer has placed Sj in Group 2B for LC, signifying it to be a 'possible carcinogen'. Many epidemiological, pathological and clinical studies have identified multiple factors, linked with Sj infection, which can lead to liver carcinogenesis. These factors include chronic inflammation in response to deposited eggs (which leads to fibrosis, cirrhosis and chromosomal instability at cellular level), hepatotoxic effects of egg-antigens, co-infection with hepatitis viruses, and up-regulation of glycolysis linked genes among others which predisposes hepatic tissue towards malignant transformation. The objective of this work is to present the current understanding on the association of Sj infection with LC. Mechanisms and factors linked with Sj infection that can lead to LC are emphasized, along with measures to diagnose and treat it. A comparison of liver carcinogenesis is also provided for cases linked with and independent of Sj infection. It appears that Sj, alone or with another carcinogen, is an important factor in liver carcinogenesis, but further studies are warranted to conclusively label 'infection with Sj alone' as a liver carcinogen.
Collapse
Affiliation(s)
- S Jain
- Independent Researcher, Institute for Globally Distributed Open Research and Education (IGDORE), Rewari, Haryana, India
| |
Collapse
|
25
|
Qiu X, Li W, Li X, Wu B, Dai M, Xia Y, Zhang G, Bian Y, Chen J, Wu K, Lu Y, Tang M, Lin H, Shang J. Discovery of Fluorescent Probe ABDS-2 for Farnesoid X Receptor Modulator Characterization and Cell-Based Imaging. Anal Chem 2025; 97:2019-2027. [PMID: 39841563 DOI: 10.1021/acs.analchem.4c03743] [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: 01/24/2025]
Abstract
The farnesoid X receptor (FXR) regulates key physiological processes, such as bile acid homeostasis and lipid metabolism, making it an important target for drug discovery. However, the overactivation of FXR often leads to adverse effects. This study presents the development of a novel fluorescent probe utilizing the computer-aided drug design (CADD) approach to optimize linkers between more potent warhead and FITC fluorescent groups. The probes were designed and assessed via molecular dynamics simulations, and four were selected for synthesis to be evaluated in in vitro biochemical assays. Among these, ABDS-2 exhibited high sensitivity and stability, which demonstrated satisfactory validation in high-throughput screening assays. Furthermore, ABDS-2 facilitated real-time bioimaging to monitor FXR homeostasis at the cellular level, providing spatially resolved insights into molecular interactions critical for cellular function studies. This research underscores the efficiency of CADD in probe design and positions ABDS-2 as a valuable chemical tool for in vitro assays and cellular-level bioimaging.
Collapse
Affiliation(s)
- Xianjie Qiu
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| | - Wenqi Li
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaoqin Li
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| | - Bin Wu
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Minxian Dai
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| | - Yi Xia
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| | - Gong Zhang
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Yizhou Bian
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Jiayi Chen
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Kunzhong Wu
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yongzhi Lu
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| | - Miru Tang
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| | - Hua Lin
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Jinsai Shang
- School of Basic Medical Sciences, Guangzhou Νational Laboratory, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
26
|
Huang WC, Li YC, Chen PX, Ma KSK, Wang LT. Mesenchymal stem cell therapy as a game-changer in liver diseases: review of current clinical trials. Stem Cell Res Ther 2025; 16:3. [PMID: 39762946 PMCID: PMC11705688 DOI: 10.1186/s13287-024-04127-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
Chronic liver diseases, including cirrhosis and liver failure, remain formidable challenges due to their complex progression and limited therapeutic options. Mesenchymal stem cell (MSC) therapy has emerged as a game-changing approach, leveraging its potent immunomodulatory, anti-fibrotic, and regenerative capabilities, along with the ability to transdifferentiate into hepatocytes. This review delves into the latest advances in MSC-based treatments for chronic and end-stage liver diseases, as highlighted in current clinical trials. MSCs derived from bone marrow and umbilical cord have shown remarkable promise in reversing liver damage, improving liver function, and providing hope for patients who do not respond to conventional therapies. When administered through hepatic, portal, or peripheral veins, MSCs have significantly improved liver histology, reduced fibrosis, and restored functional capacity. Furthermore, MSC-derived materials, such as extracellular vesicles and exosomes, are emerging as cutting-edge tools for treating liver failure and mitigating post-transplant complications. While autologous MSC-derived hepatocytes hold promise for non-fatal cirrhosis, allogeneic MSCs are being applied in more severe conditions, including liver failure and transplantation cases. Despite these promising early outcomes, larger trials and long-term studies are essential to fully harness MSCs as a transformative, off-the-shelf alternative to liver transplantation, heralding a new era in regenerative liver therapies.
Collapse
Affiliation(s)
- Wei-Chen Huang
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Laboratory of Clinical Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Chi Li
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, 10F., Teaching & Research Building, Shuang-Ho Campus, No. 301, Yuantong Rd., Zhonghe Dist., Taipei, 235, Taiwan
| | - Pin-Xuan Chen
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, 10F., Teaching & Research Building, Shuang-Ho Campus, No. 301, Yuantong Rd., Zhonghe Dist., Taipei, 235, Taiwan
| | - Kevin Sheng-Kai Ma
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Li-Tzu Wang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, 10F., Teaching & Research Building, Shuang-Ho Campus, No. 301, Yuantong Rd., Zhonghe Dist., Taipei, 235, Taiwan.
- Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
27
|
Zan Q, Fan L, Lu W, Zhang Y, Huang Y, Yu X, Han Y, Zhang R, Dong C, Shuang S. Early Diagnosis of Liver Injury and Real-Time Evaluation of Photothermal Therapy Efficacy with a Viscosity-Responsive NIR-II Smart Molecule. Adv Healthc Mater 2025; 14:e2402614. [PMID: 39440592 DOI: 10.1002/adhm.202402614] [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: 07/16/2024] [Revised: 10/14/2024] [Indexed: 10/25/2024]
Abstract
The early diagnosis of liver injury and in situ real-time monitoring of tumor therapy efficacy are important for the enhancement of personalized precision therapy but remain challenging due to the lack of reliable in vivo visualization tools with integrated diagnostic, therapeutic, and efficacy monitoring functions. Herein, a smart second near-infrared window (NIR-II) molecule (BITX-OH) is rationally designed for diagnosis and therapy by vinyl-bridging hydroxyl diphenyl xanthine unit and benzo[cd]indolium skeleton. BITX-OH exhibits high selectivity and sensitivity toward viscosity, exhibiting a significant enhancement (1167-fold) in NIR-II fluorescence at 962 nm. With the assistance of BITX-OH and NIR-II fluorescence imaging, early diagnosis and therapeutic evaluation of non-alcoholic fatty liver (NAFL), as well as in-site real-time monitoring of hepatic fibrosis (HF) in live mice have been successfully achieved, which is at least several hours earlier than the typical clinical test. Notably, BITX-OH displays excellent photothermal conversion efficiency when exposed to an 808 nm laser, which can induce tumor ablation and increase viscosity, thereby enhancing NIR-II fluorescence for the real-time evaluation of photothermal therapy (PTT). This viscosity-based "self-monitoring" strategy provides a convenient and reliable platform for timely obtaining therapeutic feedback to avoid over- or under-treatment, thus enabling personalized precision therapy.
Collapse
Affiliation(s)
- Qi Zan
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Li Fan
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Wenjing Lu
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Yuewei Zhang
- School of Chemistry and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin, 132022, China
| | - Yunong Huang
- School of Chemistry and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin, 132022, China
| | - Xue Yu
- School of Chemistry and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin, 132022, China
| | - Yahong Han
- Shanxi Medical University, Taiyuan, 030032, China
| | - Ruiping Zhang
- Shanxi Provincial People's Hospital, Taiyuan, 030001, China
| | - Chuan Dong
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Shaomin Shuang
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| |
Collapse
|
28
|
Gripshover TC, Treves RS, Rouchka EC, Chariker JH, Zheng S, Hudson E, Smith ML, Singal AK, McClain CJ, Hardesty JE. Visium spatial transcriptomics and proteomics identifies novel hepatic cell populations and transcriptomic signatures of alcohol-associated hepatitis. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:106-116. [PMID: 39592394 PMCID: PMC11747835 DOI: 10.1111/acer.15494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Alcohol-associated hepatitis (AH) is the clinical manifestation of alcohol-associated liver disease (ALD). AH is a complex disease encompassing the dysregulation of many cells and cell subpopulations. This study used a hepatic spatial transcriptomic and proteomic approach (10X Genomics Visium) to identify hepatic cell populations and their associated transcriptomic and proteomic alterations in human AH. METHODS Formalin-fixed paraffin-embedded liver tissue from AH patients (n = 2) and non-ALD controls (donors) (n = 2) were used for Visium spatial transcriptomic and proteomic analysis. RESULTS AH cell clusters and cell markers were drastically different in regard to tissue pattern and number of cell types compared to non-ALD controls. Cholangiocytes, endothelial cells, macrophages, and stellate cells were more profuse in AH relative to non-ALD controls. Transcriptionally, proliferating cell nuclear antigen-positive (PCNA+) hepatocytes in AH more closely resembled cholangiocytes suggesting they were non-functional hepatocytes derived from cholangiocytes. Furthermore, mitochondria protein-coding genes were reduced in AH versus non-ALD control hepatocytes, suggesting reduced functionality and loss of regenerative mechanisms. Macrophages in AH exhibited elevated gene expression involved in exosomes as compared to non-ALD controls. The most upregulated macrophage genes observed in AH were those involved in exosome trafficking. Gene and protein signatures of disease-associated hepatocytes (ANXA2+/CXCL1+/CEACAM8+) were elevated in AH and could visually identify a pre-malignant lesion. CONCLUSIONS This study identified global cell type alterations in AH and distinct transcriptomic changes between AH and non-ALD controls. These findings characterize cellular plasticity and profuse transcriptomic and proteomic changes that are apparent in AH and contribute to the identification of novel therapeutics.
Collapse
Affiliation(s)
- Tyler C. Gripshover
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Rui S. Treves
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Eric C. Rouchka
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
- Kentucky INBRE Bioinformatics Core, University of Louisville School of Medicine, Louisville, KY, USA
| | - Julia H. Chariker
- Kentucky INBRE Bioinformatics Core, University of Louisville School of Medicine, Louisville, KY, USA
| | - Shirong Zheng
- Sequencing Technology Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Elizabeth Hudson
- Sequencing Technology Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Melissa L. Smith
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
- Sequencing Technology Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Ashwani K. Singal
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA
- Robley Rex Veterans Medical Center, Louisville, KY, USA
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Craig J. McClain
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA
- Robley Rex Veterans Medical Center, Louisville, KY, USA
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
- University of Louisville Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Josiah E. Hardesty
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
29
|
Sotoudeheian M. Value of Mac-2 Binding Protein Glycosylation Isomer (M2BPGi) in Assessing Liver Fibrosis in Metabolic Dysfunction-Associated Liver Disease: A Comprehensive Review of its Serum Biomarker Role. Curr Protein Pept Sci 2025; 26:6-21. [PMID: 38982921 DOI: 10.2174/0113892037315931240618085529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 07/11/2024]
Abstract
Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD) is a broad condition characterized by lipid accumulation in the liver tissue, which can progress to fibrosis and cirrhosis if left untreated. Traditionally, liver biopsy is the gold standard for evaluating fibrosis. However, non-invasive biomarkers of liver fibrosis are developed to assess the fibrosis without the risk of biopsy complications. Novel serum biomarkers have emerged as a promising tool for non-invasive assessment of liver fibrosis in MAFLD patients. Several studies have shown that elevated levels of Mac-2 binding protein glycosylation isomer (M2BPGi) are associated with increased liver fibrosis severity in MAFLD patients. This suggests that M2BPGi could serve as a reliable marker for identifying individuals at higher risk of disease progression. Furthermore, the use of M2BPGi offers a non-invasive alternative to liver biopsy, which is invasive and prone to sampling errors. Overall, the usage of M2BPGi in assessing liver fibrosis in MAFLD holds great promise for improving risk stratification and monitoring disease progression in affected individuals. Further research is needed to validate its utility in clinical practice and establish standardized protocols for its implementation.
Collapse
|
30
|
Lu C, Liu Y, Ren F, Zhang H, Hou Y, Zhang H, Chen Z, Du X. HO-1: An emerging target in fibrosis. J Cell Physiol 2025; 240:e31465. [PMID: 39420552 DOI: 10.1002/jcp.31465] [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: 05/13/2024] [Revised: 09/08/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
Fibrosis, an aberrant reparative response to tissue injury, involves a disruption in the equilibrium between the synthesis and degradation of the extracellular matrix, leading to its excessive accumulation within normal tissues, and culminating in organ dysfunction. Manifesting in the terminal stages of nearly all chronic ailments, fibrosis carries a high mortality rate and poses a significant threat to human health. Heme oxygenase-1 (HO-1) emerges as an endogenous protective agent, mitigating tissue damage through its antioxidant, anti-inflammatory, and antiapoptotic properties. Numerous studies have corroborated HO-1's potential as a therapeutic target in anti-fibrosis treatment. This review delves into the structural and functional attributes, and the upstream and downstream pathways of HO-1. Additionally, the regulatory networks and mechanisms of HO-1 in cells associated with fibrosis are elucidated. The role of HO-1 in various fibrosis-related diseases is also explored. Collectively, this comprehensive information serves as a foundation for future research and augments the viability of HO-1 as a therapeutic target for fibrosis.
Collapse
Affiliation(s)
- Chenxi Lu
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Yuan Liu
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Feifei Ren
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Haoran Zhang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Yafang Hou
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Hong Zhang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Zhiyong Chen
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Xia Du
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| |
Collapse
|
31
|
Guan Y, Fang Z, Hu A, Roberts S, Wang M, Ren W, Johansson PK, Heilshorn SC, Enejder A, Peltz G. Live-cell imaging of human liver fibrosis using hepatic micro-organoids. JCI Insight 2024; 10:e187099. [PMID: 39656528 PMCID: PMC11790020 DOI: 10.1172/jci.insight.187099] [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: 09/18/2024] [Accepted: 12/03/2024] [Indexed: 01/24/2025] Open
Abstract
Due to the limitations of available in vitro systems and animal models, we lack a detailed understanding of the pathogenetic mechanisms of and have minimal treatment options for liver fibrosis. Therefore, we engineered a live-cell imaging system that assessed fibrosis in a human multilineage hepatic organoid in a microwell (i.e., microHOs). Transcriptomic analysis revealed that TGFB converted mesenchymal cells in microHOs into myofibroblast-like cells resembling those in fibrotic human liver tissue. When pro-fibrotic intracellular signaling pathways were examined, the antifibrotic effect of receptor-specific tyrosine kinase inhibitors was limited to the fibrosis induced by the corresponding growth factor, which indicates their antifibrotic efficacy would be limited to fibrotic diseases solely mediated by that growth factor. Based upon transcriptomic and transcription factor activation analyses in microHOs, glycogen synthase kinase 3β and p38 MAPK inhibitors were identified as potential new broad-spectrum therapies for liver fibrosis. Other new therapies could subsequently be identified using the microHO system.
Collapse
Affiliation(s)
- Yuan Guan
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Zhuoqing Fang
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Angelina Hu
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Sarah Roberts
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Meiyue Wang
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Wenlong Ren
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Patrik K. Johansson
- Department of Materials Science and Engineering, Stanford University, Stanford, California, USA
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California, USA
| | - Annika Enejder
- Department of Materials Science and Engineering, Stanford University, Stanford, California, USA
| | - Gary Peltz
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
32
|
Zhang G, Wu K, Jiang X, Gao Y, Ding D, Wang H, Yu C, Wang X, Jia N, Zhu L. The role of ferroptosis-related non-coding RNA in liver fibrosis. Front Cell Dev Biol 2024; 12:1517401. [PMID: 39717848 PMCID: PMC11663870 DOI: 10.3389/fcell.2024.1517401] [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: 10/26/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024] Open
Abstract
Liver fibrosis represents a reversible pathophysiological process, caused by chronic inflammation stemming from hepatocyte damage. It delineates the initial stage in the progression of chronic liver disease. This pathological progression is characterized by the excessive accumulation of the extracellular matrix (ECM), which leads to significant structural disruption and ultimately impairs liver function. To date, no specific antifibrotic drugs have been developed, and advanced liver fibrosis remains largely incurable. Liver transplantation remains the sole efficacious intervention for advanced liver fibrosis; nevertheless, it is constrained by exorbitant costs and the risk of postoperative immune rejection, underscoring the imperative for novel therapeutic strategies. Ferroptosis, an emergent form of regulated cell death, has been identified as a pivotal regulatory mechanism in the development of liver fibrosis and is intricately linked with the progression of liver diseases. Recent investigations have elucidated that a diverse array of non-coding RNAs (ncRNAs), including microRNAs, long non-coding RNAs, and circular RNAs, are involved in the ferroptosis pathway, thereby modulating the progression of various diseases, including liver fibrosis. In recent years, the roles of ferroptosis and ferroptosis-related ncRNAs in liver fibrosis have attracted escalating scholarly attention. This paper elucidates the pathophysiology of liver fibrosis, explores the mechanisms underlying ferroptosis, and delineates the involvement of ncRNA-mediated ferroptosis pathways in the pathology of liver fibrosis. It aims to propose novel strategies for the prevention and therapeutic intervention of liver fibrosis.
Collapse
Affiliation(s)
- Guozhu Zhang
- Department of Emergency Medicine, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Kejia Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaobo Jiang
- Kunshan Zhenchuan Community Health Service Center, Kunshan, Jiangsu, China
| | - Yuan Gao
- Department of Hepato-Biliary-Pancreatic Surgery, The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Dong Ding
- Department of Hepato-Biliary-Pancreatic Surgery, The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Hao Wang
- Department of Emergency Medicine, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Chongyuan Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiaozhong Wang
- Department of General Surgery, Wujin Affiliated Hospital of Jiangsu University and the Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Naixin Jia
- Department of Hepatobiliary Surgery, Kunshan First People’s Hospital affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Li Zhu
- Department of Emergency Medicine, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
33
|
Ceccherini E, Morlando A, Norelli F, Coco B, Bellini M, Brunetto MR, Cecchettini A, Rocchiccioli S. Cytoskeleton Remodeling-Related Proteins Represent a Specific Salivary Signature in PSC Patients. Molecules 2024; 29:5783. [PMID: 39683940 DOI: 10.3390/molecules29235783] [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: 10/02/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Primary sclerosing cholangitis (PSC) and Primary biliary cholangitis (PBC) are chronic inflammatory biliary diseases characterized by progressive damage of the bile ducts, resulting in hepatobiliary fibrosis and cirrhosis. Currently, specific biomarkers that allow to distinguish between PSC and PBC do not exist. In this study, we examined the salivary proteome by carrying out a comprehensive and non-invasive screening aimed at highlighting possible quali-quantitative protein deregulations that could be the starting point for the identification of effective biomarkers in future. Saliva samples collected from 6 PBC patients were analyzed using a liquid chromatography-tandem mass spectrometry technique, and the results were compared with those previously obtained in the PSC group. We identified 40 proteins as significantly deregulated in PSC patients compared to the PBC group. The Gene Ontology and pathway analyses highlighted that several proteins (e.g., small integral membrane protein 22, cofilin-1, macrophage-capping protein, plastin-2, and biliverdin reductase A) were linked to innate immune responses and actin cytoskeleton remodeling, which is a critical event in liver fibrosis and cancer progression. These findings provide new foundations for a deeper understanding of the pathophysiology of PSC and demonstrate that saliva is a suitable biological sample for obtaining proteomic fingerprints useful in the search for biomarkers capable of discriminating between the two cholestatic diseases.
Collapse
Affiliation(s)
- Elisa Ceccherini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| | - Antonio Morlando
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| | - Francesco Norelli
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| | - Barbara Coco
- Hepatology Unit, Reference Centre of the Tuscany Region for Chronic Liver Disease and Cancer, University Hospital of Pisa, 56124 Pisa, Italy
| | - Massimo Bellini
- Gastrointestinal Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy
| | - Maurizia Rossana Brunetto
- Hepatology Unit, Reference Centre of the Tuscany Region for Chronic Liver Disease and Cancer, University Hospital of Pisa, 56124 Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Antonella Cecchettini
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Silvia Rocchiccioli
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| |
Collapse
|
34
|
Zhao Y, Kong H, Li Y, Zhao Y, Zhang Y, Zhao Y, Qu H. Inhibitory effects of Curcumae Radix carbonisata-based carbon dots against liver fibrosis induced by carbon tetrachloride in mice. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:23-34. [PMID: 38035609 DOI: 10.1080/21691401.2023.2239522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 07/18/2023] [Indexed: 12/02/2023]
Abstract
As a processed product of traditional Chinese medicine Curcumae Radix, Curcumae Radix Carbonisata (CRC) has been widely used in the treatment of liver diseases in ancient medical books. In this study, novel carbon dots (CDs) extending from 1.0 to 4.5 nm were separated from fluid extricates of CRC. Meanwhile, a liver fibrosis model induced by carbon tetrachloride (CCl4) was utilized to determine the inhibitory effects of CRC-CDs against liver fibrosis. The results exhibited the CRC-CDs with a quantum yield of 1.34% have a significant inhibitory effect on CCl4-induced liver fibrosis, as demonstrated by improving hepatocyte degeneration and necrosis, inflammatory cell infiltration and fibrotic tissue hyperplasia, downregulating the levels of alanine transaminase (ALT), aspartate aminotransferase (AST), total bilirubin (TBIL), direct bilirubin (DBIL), total bile acid (TBA), triglyceride (TG), tumour necrosis factor-α (TNF-α), interleukin (IL)-6 and IL-1β in the serum, upregulating the contents of superoxide dismutase (SOD), reduced glutathione (GSH), and downregulating the concentration of malondialdehyde (MDA), which lays an important foundation for the development of CRC-CDs as a novel drug for the treatment of liver fibrosis, and provide a certain experimental basis for the clinical application of CRC-CDs in the future.
Collapse
Affiliation(s)
- Yusheng Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuru Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Yafang Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huihua Qu
- Centre of Scientific Experiment, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
35
|
Liu J, Li H, Chen H, Xiao X, Jin Z, Paerhati P, Bao W, Cui C, Zhu J, Yuan Y. An anti-RAGE chimeric antibody alleviates CCl 4-induced liver fibrosis via RAGE/NF-kB pathway in mice. Biomed Pharmacother 2024; 181:117737. [PMID: 39657505 DOI: 10.1016/j.biopha.2024.117737] [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: 08/31/2024] [Revised: 11/23/2024] [Accepted: 12/03/2024] [Indexed: 12/12/2024] Open
Abstract
Liver fibrosis is a progressive condition characterized by excessive deposition of extracellular matrix components, leading to organ dysfunction. Chronic inflammation and activation of hepatic stellate cells (HSCs) are two dominant events in all stages of fibrosis development. The receptor for advanced glycation end products (RAGE) pathway is involved in modulating liver injury and fibrosis, and preventing it, or deletion of Ager gene can protect the liver against fibrosis progression. To investigate functions and mechanism of chimeric anti-RAGE monoclonal antibody against liver fibrosis, murine-derived monoclonal anti-RAGE antibodies were used to construct murine-human chimeric antibodies. The properties of the chimeric antibody were characterized, and the biological functions of antibody A5 or its evolved humanized molecule, huA5, were investigated in cell or animal model. The data showed that blocking the RAGE pathway with huA5 robustly reduced liver injury and fibrosis. Furthermore, huA5 significantly suppressed the activation of HSCs and inhibited expression of fibrosis-associated genes, including COL1A1,TIMP1, and ACTA2. huA5 also interfered with RAGE downstream signal transduction and down-regulate both ERK and NF-κB phosphorylation, inhibited the RAGE/NF-kB pathway, leading to reduced expression of pro-inflammatory cytokines and profibrotic markers. Finally, RAGE silencing significantly decreased the expression of activation-related genes in HSCs, inhibiting HSCs proliferation and migration. These results clearly revealed that the anti-RAGE chimeric antibody exerted antifibrotic efficacy in vitro and attenuated liver fibrosis in vivo. HuA5 can be further developed as a lead molecule of drug to treat patients with liver fibrosis.
Collapse
Affiliation(s)
- Jing Liu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Huiyi Li
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Hui Chen
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Xinyi Xiao
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Zhedong Jin
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Pameila Paerhati
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Wenxin Bao
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Caixia Cui
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Jianwei Zhu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| | - Yunsheng Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 201100, China.
| |
Collapse
|
36
|
Gurgul A, Żurowski J, Szmatoła T, Kucharski M, Sawicki S, Semik-Gurgul E, Ocłoń E. Cannabidiol (CBD) modulates the transcriptional profile of ethanol-exposed human dermal fibroblast cells. J Appl Genet 2024; 65:773-796. [PMID: 39466591 PMCID: PMC11561130 DOI: 10.1007/s13353-024-00915-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/30/2024]
Abstract
Cannabidiol (CBD) is abundant in the Cannabis sativa plant and exhibits complex immunomodulatory, anxiolytic, antioxidant, and antiepileptic properties. Several studies suggest that CBD could be used for different purposes in alcohol use disorder (AUD) and alcohol-related injuries to the brain and the liver. In this study, we focused on analyzing transcriptional alterations in human dermal fibroblasts (HDFs) cell line challenged simultaneously with ethanol and CBD as an ethanol-protective agent. We aimed to expose the genes and pathways responsible for at least some of the CBD effects in those cells that can be related to the AUD. Transcriptome analysis was performed using HDFs cell line that expresses both cannabinoid receptors and can metabolize ethanol through alcohol dehydrogenase activity. Fibroblasts are also responsible for the progression of liver fibrosis, a common comorbidity in AUD. With the use of a cellular test, we found that CBD at the lowest applied concentration (0.75 μM) was able to stimulate depressed metabolism and reduce the level of apoptosis of cells treated with different concentrations of ethanol to the level observed in the control cells. Similar observations were made at the transcriptome level, in which cells treated with ethanol and CBD had similar expression profiles to the control cells. CBD also affects several genes connected with extracellular matrix formation (especially its collagen constituent), which can have potential implications for, e.g., fibrosis process.
Collapse
Affiliation(s)
- Artur Gurgul
- Faculty of Veterinary Medicine, Department of Basic Sciences, University of Agriculture in Kraków, Redzina 1C, 30-248, Krakow, Poland.
| | - Jakub Żurowski
- Faculty of Veterinary Medicine, Department of Basic Sciences, University of Agriculture in Kraków, Redzina 1C, 30-248, Krakow, Poland
| | - Tomasz Szmatoła
- Faculty of Veterinary Medicine, Department of Basic Sciences, University of Agriculture in Kraków, Redzina 1C, 30-248, Krakow, Poland
| | - Mirosław Kucharski
- Faculty of Animal Science, Department of Animal Physiology and Endocrinology, University of Agriculture in Kraków, Mickiewicza 24/28, 30‑059, Krakow, Poland
| | - Sebastian Sawicki
- Faculty of Animal Science, Department of Animal Reproduction, Anatomy and Genomics, University of Agriculture in Kraków, Mickiewicza 24/28, 30-059, Krakow, Poland
| | - Ewelina Semik-Gurgul
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1, 32-083, Balice, Poland
| | - Ewa Ocłoń
- Faculty of Veterinary Medicine, Laboratory of Recombinant Proteins Production, University of Agriculture in Kraków, Rędzina 1C, 30-248, Kraków, Poland
| |
Collapse
|
37
|
Li Y, Chen L, Li S, Song H, Chen Y, Wang S. The m6A reader IGF2BP1 contributes to the activation of hepatic stellate cells through facilitating TUBB4B mRNA stabilization. J Gastroenterol Hepatol 2024; 39:2916-2925. [PMID: 39403946 DOI: 10.1111/jgh.16765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/30/2024]
Abstract
The m6A reader insulin-like growth factor-2 mRNA-binding protein 1 (IGF2BP1) is involved in multiple pathophysiological processes through enhanced expression of the proteins encoded by their target mRNAs. However, the functional role of IGF2BP1-mediated m6A in liver fibrosis remains elusive. Here, we report that IGF2BP1 is highly expressed in activated hepatic stellate cells (HSCs), the major driver of fibrogenesis, and TUBB4B is identified as a potential target of IGF2BP1 by re-analysis of the RNA-seq, RIP-seq, and m6A-seq data. The relevant findings were subsequently demonstrated by a series of molecular and cellular evidences. The knockdown of IGF2BP1 or TUBB4B and pharmacological inhibition of TUBB4B by mebendazole treatments significantly suppress the proliferation, migration, and activation of HSCs. Mechanistically, IGF2BP1 upregulates TUBB4B expression through stabilizing TUBB4B in an m6A-dependent manner, and TUBB4B induces liver fibrosis by activating the FAK signaling pathway. Collectively, our results indicate that targeting IGF2BP1/TUBB4B/FAK axis in HSCs could be a promising therapeutic approach for liver fibrosis.
Collapse
Affiliation(s)
- Yanshan Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ling Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shuyi Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Haoxin Song
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yijun Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shuzhen Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
38
|
Lee J, Hong SW, Kim MJ, Lim YM, Moon SJ, Kwon H, Park SE, Rhee EJ, Lee WY. Sodium-glucose cotransporter 2 inhibitors ameliorate ER stress-induced pro-inflammatory cytokine expression by inhibiting CD36 in NAFLD progression in vitro. Biochem Biophys Res Commun 2024; 735:150620. [PMID: 39265364 DOI: 10.1016/j.bbrc.2024.150620] [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: 06/28/2024] [Revised: 08/18/2024] [Accepted: 08/28/2024] [Indexed: 09/14/2024]
Abstract
Sodium-dependent glucose cotransporter-2 (SGLT2) inhibitors, antidiabetic drugs that reduce blood sugar levels by inhibiting glucose reabsorption in the renal proximal tubules, also ameliorate nonalcoholic fatty liver disease (NAFLD). This study aimed to examine the effects of SGLT2 inhibition on hepatic steatosis and nonalcoholic steatohepatitis (NASH) using an in vitro model of NAFLD progression. HepG2 cells and a coculture of Hepa1c1c7 and Raw 264.7 cells were treated with 400 μM palmitic acid (PA), followed by treatment with or without 10 μM empagliflozin and dapagliflozin. In HepG2 cells, PA increased hepatic lipid accumulation, the expression of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β), exocytosis mediators (VAMP3 and SNAP23), and ER stress markers (GRP78, PERK, IRE1α, ATF6, ATF4, and CHOP), and the gene and protein expression of CD36. SGLT2 inhibitors reversed the effects of PA. SGLT2 inhibition via siRNA reduced proinflammatory-cytokine gene expression in thapsigargin-treated HepG2 cells. Transfection with CD36 siRNA reversed the elevated ATF4 and CHOP expression in PA-treated HepG2 cells. SGLT2 inhibition via an SGTL2 inhibitor and SGLT2 siRNA reduced CD36, Tnf-α, Il-6, Il-1β, Vamp2, Snap23, Atf4, and Chop expression in the PA-treated Hepa1c1c7-Raw 264.7 cell coculture and suppressed Tnf-α release in the Hepa1c1c7-Raw 264.7 cell coculture treated with lipopolysaccharide and PA. These findings indicate that SGLT2 inhibitors inhibited NAFLD progression by reducing hepatic lipid accumulation and inflammation.
Collapse
Affiliation(s)
- Jinmi Lee
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Seok-Woo Hong
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Min-Jeong Kim
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Yu-Mi Lim
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Sun Joon Moon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Hyemi Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Se Eun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Eun-Jung Rhee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea.
| | - Won-Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea.
| |
Collapse
|
39
|
Kouroumalis E, Tsomidis I, Voumvouraki A. Extracellular Vesicles in Viral Liver Diseases. Viruses 2024; 16:1785. [PMID: 39599900 PMCID: PMC11598962 DOI: 10.3390/v16111785] [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: 09/30/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Extracellular vesicles (EVs) are bilayer vesicles released by cells in the microenvironment of the liver including parenchymal and non-parenchymal cells. They are the third important mechanism in the communications between cells, besides the secretion of cytokines and chemokines and the direct cell-to-cell contact. The aim of this review is to discuss the important role of EVs in viral liver disease, as there is increasing evidence that the transportation of viral proteins, all types of RNA, and viral particles including complete virions is implicated in the pathogenesis of both viral cirrhosis and viral-related hepatocellular carcinoma. The biogenesis of EVs is discussed and their role in the pathogenesis of viral liver diseases is presented. Their use as diagnostic and prognostic biomarkers is also analyzed. Most importantly, the significance of possible novel treatment strategies for liver fibrosis and hepatocellular carcinoma is presented, although available data are based on experimental evidence and clinical trials have not been reported.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Ioannis Tsomidis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Greece;
| |
Collapse
|
40
|
Trillos-Almanza MC, Aguilar MM, Buist-Homan M, Bomer N, Gomez KA, de Meijer VE, van Vilsteren FGI, Blokzijl H, Moshage H. Branched-chain amino acids and their metabolites decrease human and rat hepatic stellate cell activation. Mol Biol Rep 2024; 51:1116. [PMID: 39495311 PMCID: PMC11534903 DOI: 10.1007/s11033-024-10027-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND End-stage liver diseases (ESLDs) are a significant global health challenge due to their high prevalence and severe health impacts. Despite the severe outcomes associated with ESLDs, therapeutic options remain limited. Targeting the activation of hepatic stellate cells (HSCs), key drivers of extracellular matrix accumulation during liver injury presents a novel therapeutic approach. In ESLDs patients, branched-chain amino acids (BCAAs, leucine, isoleucine and valine) levels are decreased, and supplementation has been proposed to attenuate liver fibrosis and improve regeneration. However, their effects on HSCs require further investigation. OBJECTIVE To evaluate the efficacy of BCAAs and their metabolites, branched-chain α-keto acids (BCKAs), in modulating HSCs activation in human and rat models. METHODS Primary HSCs from rats and cirrhotic and non-cirrhotic human livers, were cultured and treated with BCAAs or BCKAs to assess their effects on both preventing (from day 1 of isolation) and reversing (from day 7 of isolation) HSCs activation. RESULTS In rat HSCs, leucine and BCKAs significantly reduced fibrotic markers and cell proliferation. In human HSCs, the metabolite of isoleucine decreased cell proliferation around 85% and increased the expression of branched-chain ketoacid dehydrogenase. The other metabolites also showed antifibrotic effects in HSCs from non-cirrhotic human livers. CONCLUSION BCAAs and their respective metabolites inhibit HSC activation with species-specific responses. Further research is needed to understand how BCAAs influence liver fibrogenesis. BCKAs supplementation could be a strategic approach for managing ESLDs, considering the nutritional status and amino acid profiles of patients.
Collapse
Affiliation(s)
- Maria Camila Trillos-Almanza
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.
| | - Magnolia Martinez Aguilar
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Manon Buist-Homan
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Nils Bomer
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Karla Arevalo Gomez
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Vincent E de Meijer
- Division of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Frederike G I van Vilsteren
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Hans Blokzijl
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.
- Department of Laboratory Medicine, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
41
|
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
|
42
|
Hegde SS, Malashetty VB. Azoospermia and multi-organ damage in juvenile rats exposed to α-Terpineol from weaning to sexual maturity. Toxicol Appl Pharmacol 2024; 492:117106. [PMID: 39278549 DOI: 10.1016/j.taap.2024.117106] [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: 08/02/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
This study aimed to evaluate the repeated oral administration of α-terpineol in juvenile Wistar rats over a 70-day period. The objective was to assess the potential systemic and reproductive toxicity of α-terpineol when administered by gavage at doses of 75, 150, and 300 mg/kg/day to juvenile Wistar rats for 70 days from postnatal day 24. The control group received corn oil for 70 days. During the study, various parameters were evaluated, including clinical signs, body weight, food intake, neurobehavioral observations, haematology, serum biochemistry, organ weights, steroidogenic gene expression, and histopathological examination. No toxicity-related changes were observed in body weight, food intake, neurobehavioral observations, or steroidogenic gene expression. However, sperm evaluation revealed a complete absence of sperm and delayed sexual maturation. Total cholesterol was significantly elevated in both sexes, and serum testosterone was reduced at the 150 and 300 mg/kg doses. Microscopic examination showed severe pathological changes in the testes, epididymis, liver, and kidneys of both males and females. After the 14-day recovery period, total cholesterol levels returned to the normal range, but no recovery was observed in the other organs. The no-observed-adverse-effect level was 75 mg/kg/day for male rats based on the histopathological findings in the testes, liver, and kidneys, and for female rats based on the kidney and liver histopathology.
Collapse
Affiliation(s)
- Sneha Suma Hegde
- Reproductive Biology and Mechanistic Toxicology Lab, Department of Studies in Zoology Vijayanagara Sri Krishnadevaraya University, Ballari 58103, India
| | - Vijaykumar B Malashetty
- Reproductive Biology and Mechanistic Toxicology Lab, Department of Studies in Zoology Vijayanagara Sri Krishnadevaraya University, Ballari 58103, India.
| |
Collapse
|
43
|
Schenker RB, Machle CJ, Allayee H, Lurmann F, Patterson WB, Kohli R, Goran MI, Alderete TL. Ambient air pollution exposure is associated with liver fat and stiffness in Latino youth with a more pronounced effect in those with PNPLA3 genotype and more advanced liver disease. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117234. [PMID: 39454357 PMCID: PMC11578286 DOI: 10.1016/j.ecoenv.2024.117234] [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: 07/22/2024] [Revised: 10/10/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Exposure to ambient air pollutants has emerged as a risk for metabolic-dysfunction associated steatotic liver disease (MASLD). OBJECTIVES We sought to examine associations between short-term (prior month) and long-term (prior year) ambient air pollution exposure with hepatic fat fraction (HFF) and liver stiffness in Latino youth with obesity. A secondary aim was to investigate effect modification by patatin-like phospholipase domain-containing protein 3 (PNPLA3) genotype and liver disease severity. METHODS Data was analyzed from 113 Latino youth (age 11-19) with obesity in Southern California. Individual exposure to particulate matter with aerodynamic diameter ≤ 2.5μm (PM2.5), ≤ 10μm (PM10), nitrogen dioxide (NO2), 8-hour maximum ozone (8hrMax-O3), 24-hr O3, and redox-weighted oxidative capacity (Oxwt) were estimated using residential address histories and United States Environmental Protection Agency air quality observations. HFF and liver stiffness were measured using magnetic resonance imaging. Linear models were used to determine associations between short-term and long-term exposure to air pollutants with HFF and liver stiffness. Modification by PNPLA3 and liver disease severity was then examined. RESULTS Short-term exposure to 8hrMax-O3 was positively associated with HFF. Relationships between air pollution exposure and HFF were not impacted by PNPLA3 genotype or liver disease severity. Long-term exposure to 8hrMax-O3 and Oxwt were positively associated with liver stiffness. Associations between air pollution exposure and liver stiffness depended on PNPLA3 genotype, such that individuals with GG genotypes exhibited stronger, more positive relationships between short-term exposure to PM10, 8hrMax-O3, 24-hr O3, and Oxwt and liver stiffness than individuals with CC/CG genotypes. In addition, relationships between short-term exposure to NO2 and liver stiffness were stronger in those with severe liver disease. DISCUSSION Air pollution exposure may be a risk factor for liver disease among Latino youth with obesity, particularly in those with other preexisting risks for liver damage.
Collapse
Affiliation(s)
- Rachel B Schenker
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, CA, USA.
| | - Christopher J Machle
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA; Department of Psychology, University of Oregon, Eugene, OR, USA.
| | - Hooman Allayee
- Departments of Population & Public Health Sciences and Biochemistry & Molecular Medicine, University of Southern California, Los Angeles, CA, USA.
| | | | - William B Patterson
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, USA.
| | - Rohit Kohli
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, CA, USA.
| | - Michael I Goran
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA.
| | - Tanya L Alderete
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
44
|
Jarocki M, Turek K, Saczko J, Tarek M, Kulbacka J. Lipids associated with autophagy: mechanisms and therapeutic targets. Cell Death Discov 2024; 10:460. [PMID: 39477959 PMCID: PMC11525783 DOI: 10.1038/s41420-024-02224-8] [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: 04/24/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Autophagy is a molecular process essential for maintaining cellular homeostasis, with its impairment or dysregulation linked to the progression of various diseases in mammals. Specific lipids, including phosphoinositides, sphingolipids, and oxysterols, play pivotal roles in inducing and regulating autophagy, highlighting their significance in this intricate process. This review focuses on the critical involvement of these lipids in autophagy and lipophagy, providing a comprehensive overview of the current understanding of their functions. Moreover, we delve into how abnormalities in autophagy, influenced by these lipids, contribute to the pathogenesis of various diseases. These include age-related conditions such as cardiovascular diseases, neurodegenerative disorders, type 2 diabetes, and certain cancers, as well as inflammatory and liver diseases, skeletal muscle pathologies and age-related macular degeneration (AMD). This review aims to highlight function of lipids and their potential as therapeutic targets in treating diverse human pathologies by elucidating the specific roles of phosphoinositides, sphingolipids, and oxysterols in autophagy.
Collapse
Affiliation(s)
- Michał Jarocki
- University Clinical Hospital, Wroclaw Medical University, Wroclaw, Poland
| | | | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Mounir Tarek
- Université de Lorraine, CNRS, LPCT, Nancy, France
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland.
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
| |
Collapse
|
45
|
Jain S. Is Schistosoma mansoni playing a part in liver carcinogenesis? J Helminthol 2024; 98:e61. [PMID: 39469749 DOI: 10.1017/s0022149x24000506] [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] [Indexed: 10/30/2024]
Abstract
The relationship between Schistosoma mansoni (Sm) and hepatocellular carcinoma (HCC) has been evaluated by many studies that point towards a co-relation between schistosomal infection and HCC. While many such studies demonstrated that Sm infection in the presence of another carcinogenic factors leads to HCC, none of these studies could conclusively prove the cancer-inducing ability of Sm in humans, independent of other carcinogenic factors. The aim of this work is to present the current understanding on the association of Sm with HCC. Many epidemiological, pathological, and clinical studies have shown the role of multiple events like chronic inflammation and fibrosis as well as hepato-toxic agents like soluble egg antigens (SEAs), which help in creating a micro-environment which is suitable for HCC development. The role of Sm infection and deposited eggs in causing persistent inflammation, advanced fibrosis, and the role of SEAs, especially IPSE/alpha-1, is emphasised. This work concludes that Sm infection has the potential to induce cancer independently but the same has not been reported in humans to date. Extensive research is required to establish a causal relationship between Sm infection and HCC induction, or a complete lack thereof. However, Sm infection definitely acts along with other carcinogenic factors to induce HCC at a much faster pace and also leads to an aggressive form of liver cancer, which the other carcinogenic factor could not have achieved alone.
Collapse
Affiliation(s)
- S Jain
- Institute for Globally Distributed Open Research and Education (IGDORE), India
| |
Collapse
|
46
|
Al Ageeli E. Dual Roles of microRNA-122 in Hepatocellular Carcinoma and Breast Cancer Progression and Metastasis: A Comprehensive Review. Curr Issues Mol Biol 2024; 46:11975-11992. [PMID: 39590305 PMCID: PMC11592835 DOI: 10.3390/cimb46110711] [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: 09/27/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
microRNA-122 (miR-122) plays crucial yet contrasting roles in hepatocellular carcinoma (HCC) and breast cancer (BC), two prevalent and aggressive malignancies. This review synthesizes current research on miR-122's functions in these cancers, focusing on its potential as a diagnostic, prognostic, and therapeutic target. A comprehensive literature search was conducted using PubMed, Web of Science, and Scopus databases. In HCC, miR-122 is downregulated in most cases, suppressing oncogenic pathways and reducing tumor growth and metastasis. Restoring miR-122 levels has shown promising therapeutic potential, increasing sensitivity to treatments like sorafenib. In contrast, in BC, miR-122 plays a pro-metastatic role, especially in triple-negative breast cancer (TNBC) and metastatic lesions. miR-122's ability to influence key pathways, such as the Wnt/β-catenin and NF-κB pathways in HCC, and its role in enhancing the Warburg effect in BC underline its significance in cancer biology. miR-122, a key factor in breast cancer radioresistance, suppresses tumors in radiosensitive cells. Inhibiting miR-122 could reverse resistance and potentially overcome radiotherapy resistance. Given its context-dependent functions, miR-122 could serve as a potential therapeutic target, where restoring or inhibiting its expression may help in treating HCC and BC, respectively. The dual roles of miR-122 underscore its significance in cancer biology and its potential in precision medicine.
Collapse
Affiliation(s)
- Essam Al Ageeli
- Department of Basic Medical Sciences (Medical Genetics), Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
47
|
Diwan R, Gaytan SL, Bhatt HN, Pena-Zacarias J, Nurunnabi M. Liver fibrosis pathologies and potentials of RNA based therapeutics modalities. Drug Deliv Transl Res 2024; 14:2743-2770. [PMID: 38446352 DOI: 10.1007/s13346-024-01551-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2024] [Indexed: 03/07/2024]
Abstract
Liver fibrosis (LF) occurs when the liver tissue responds to injury or inflammation by producing excessive amounts of scar tissue, known as the extracellular matrix. This buildup stiffens the liver tissue, hinders blood flow, and ultimately impairs liver function. Various factors can trigger this process, including bloodborne pathogens, genetic predisposition, alcohol abuse, non-steroidal anti-inflammatory drugs, non-alcoholic steatohepatitis, and non-alcoholic fatty liver disease. While some existing small-molecule therapies offer limited benefits, there is a pressing need for more effective treatments that can truly cure LF. RNA therapeutics have emerged as a promising approach, as they can potentially downregulate cytokine levels in cells responsible for liver fibrosis. Researchers are actively exploring various RNA-based therapeutics, such as mRNA, siRNA, miRNA, lncRNA, and oligonucleotides, to assess their efficacy in animal models. Furthermore, targeted drug delivery systems hold immense potential in this field. By utilizing lipid nanoparticles, exosomes, nanocomplexes, micelles, and polymeric nanoparticles, researchers aim to deliver therapeutic agents directly to specific biomarkers or cytokines within the fibrotic liver, increasing their effectiveness and reducing side effects. In conclusion, this review highlights the complex nature of liver fibrosis, its underlying causes, and the promising potential of RNA-based therapeutics and targeted delivery systems. Continued research in these areas could lead to the development of more effective and personalized treatment options for LF patients.
Collapse
Affiliation(s)
- Rimpy Diwan
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA
- Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX, 79968, USA
| | - Samantha Lynn Gaytan
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA
- Department of Interdisciplinary Health Sciences, College of Health Sciences, The University of Texas El Paso, El Paso, Texas, 79968, USA
| | - Himanshu Narendrakumar Bhatt
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA
- Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX, 79968, USA
| | - Jacqueline Pena-Zacarias
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA
- Department of Biological Sciences, College of Science, The University of Texas El Paso, El Paso, Texas, 79968, USA
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA.
- Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX, 79968, USA.
- Department of Interdisciplinary Health Sciences, College of Health Sciences, The University of Texas El Paso, El Paso, Texas, 79968, USA.
- Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX, 79968, USA.
| |
Collapse
|
48
|
Jeong M, Shin S, Lee G, Lee Y, Park SB, Kang J, Lee YS, Seo W, Lee H. Engineered lipid nanoparticles enable therapeutic gene silencing of GTSE1 for the treatment of liver fibrosis. J Control Release 2024; 374:337-348. [PMID: 39154935 DOI: 10.1016/j.jconrel.2024.08.012] [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: 06/16/2024] [Revised: 07/26/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
Liver fibrosis is characterized by abnormal accumulation of extracellular matrix proteins, disrupting normal liver function. Despite its significant health impact, effective treatments remain limited. Here, we present the development of engineered lipid nanoparticles (LNPs) for targeted RNA therapeutic delivery in the liver. We investigated the therapeutic potential of modulating the G2 and S-phase expressed 1 (GTSE1) protein for treating liver fibrosis. Through screening, we identified P138Y LNP as a potent candidate with superior delivery efficiency and lower toxicity. Using these engineered LNPs, we successfully delivered siGTSE1 to hepatocytes, significantly reducing collagen accumulation and restoring liver function in a fibrosis animal model. Additionally, GTSE1 downregulation altered miRNA expression and upregulated hepatocyte nuclear factor 4 alpha (HNF4α). These findings suggest that therapeutic gene silencing of GTSE1 is a promising strategy for treating liver fibrosis by regenerating liver phenotypes and functions.
Collapse
Affiliation(s)
- Michaela Jeong
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Sumin Shin
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Gyeongseok Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yeji Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seo Bhin Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jisoo Kang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Young-Sun Lee
- Department of Internal Medicine, Korea University Medical Center, Seoul 08308, Republic of Korea
| | - Wonhyo Seo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Hyukjin Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
49
|
Zhao S, Zhuang H, Ji W, Cheng C, Liu Y. Identification of Disulfidptosis-Related Genes in Ischemic Stroke by Combining Single-Cell Sequencing, Machine Learning Algorithms, and In Vitro Experiments. Neuromolecular Med 2024; 26:39. [PMID: 39278970 PMCID: PMC11402847 DOI: 10.1007/s12017-024-08804-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/27/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Ischemic stroke (IS) is a severe neurological disorder with a pathogenesis that remains incompletely understood. Recently, a novel form of cell death known as disulfidptosis has garnered significant attention in the field of ischemic stroke research. This study aims to investigate the mechanistic roles of disulfidptosis-related genes (DRGs) in the context of IS and to examine their correlation with immunopathological features. METHODS To enhance our understanding of the mechanistic underpinnings of disulfidptosis in IS, we initially retrieved the expression profile of peripheral blood from human IS patients from the GEO database. We then utilized a suite of machine learning algorithms, including LASSO, random forest, and SVM-RFE, to identify and validate pivotal genes. Furthermore, we developed a predictive nomogram model, integrating multifactorial logistic regression analysis and calibration curves, to evaluate the risk of IS. For the analysis of single-cell sequencing data, we employed a range of analytical tools, such as "Monocle" and "CellChat," to assess the status of immune cell infiltration and to characterize intercellular communication networks. Additionally, we utilized an oxygen-glucose deprivation (OGD) model to investigate the effects of SLC7A11 overexpression on microglial polarization. RESULTS This study successfully identified key genes associated with disulfidptosis and developed a reliable nomogram model using machine learning algorithms to predict the risk of ischemic stroke. Examination of single-cell sequencing data showed a robust correlation between disulfidptosis levels and the infiltration of immune cells. Furthermore, "CellChat" analysis elucidated the intricate characteristics of intercellular communication networks. Notably, the TNF signaling pathway was found to be intimately linked with the disulfidptosis signature in ischemic stroke. In an intriguing finding, the OGD model demonstrated that SLC7A11 expression suppresses M1 polarization while promoting M2 polarization in microglia. CONCLUSION The significance of our findings lies in their potential to shed light on the pathogenesis of ischemic stroke, particularly by underscoring the pivotal role of disulfidptosis-related genes (DRGs). These insights could pave the way for novel therapeutic strategies targeting DRGs to mitigate the impact of ischemic stroke.
Collapse
Affiliation(s)
- Songyun Zhao
- Department of Neurosurgery, The Afliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
| | - Hao Zhuang
- Department of Neurosurgery, The Afliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
| | - Wei Ji
- Department of Neurosurgery, The Afliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China
| | - Chao Cheng
- Department of Neurosurgery, The Afliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China.
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China.
| | - Yuankun Liu
- Department of Neurosurgery, The Afliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China.
- Wuxi Medical Center of Nanjing Medical University, Wuxi, China.
| |
Collapse
|
50
|
Khatoon S, Das N, Chattopadhyay S, Joharapurkar A, Singh A, Patel V, Nirwan A, Kumar A, Mugale MN, Mishra DP, Kumaravelu J, Guha R, Jain MR, Chattopadhyay N, Sanyal S. Apigenin-6-C-glucoside ameliorates MASLD in rodent models via selective agonism of adiponectin receptor 2. Eur J Pharmacol 2024; 978:176800. [PMID: 38950835 DOI: 10.1016/j.ejphar.2024.176800] [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: 01/30/2024] [Revised: 06/13/2024] [Accepted: 06/28/2024] [Indexed: 07/03/2024]
Abstract
Adiponectin plays key roles in energy metabolism and ameliorates inflammation, oxidative stress, and mitochondrial dysfunction via its primary receptors, adiponectin receptors -1 and 2 (AdipoR1 and AdipoR2). Systemic depletion of adiponectin causes various metabolic disorders, including MASLD; however adiponectin supplementation is not yet achievable owing to its large size and oligomerization-associated complexities. Small-molecule AdipoR agonists, thus, may provide viable therapeutic options against metabolic disorders. Using a novel luciferase reporter-based assay here, we have identified Apigenin-6-C-glucoside (ACG), but not apigenin, as a specific agonist for the liver-rich AdipoR isoform, AdipoR2 (EC50: 384 pM) with >10000X preference over AdipoR1. Immunoblot analysis in HEK-293 overexpressing AdipoR2 or HepG2 and PLC/PRF/5 liver cell lines revealed rapid AMPK, p38 activation and induction of typical AdipoR targets PGC-1α and PPARα by ACG at a pharmacologically relevant concentration of 100 nM (reported cMax in mouse; 297 nM). ACG-mediated AdipoR2 activation culminated in a favorable modulation of key metabolic events, including decreased inflammation, oxidative stress, mitochondrial dysfunction, de novo lipogenesis, and increased fatty acid β-oxidation as determined by immunoblotting, QRT-PCR and extracellular flux analysis. AdipoR2 depletion or AMPK/p38 inhibition dampened these effects. The in vitro results were recapitulated in two different murine models of MASLD, where ACG at 10 mg/kg body weight robustly reduced hepatic steatosis, fibrosis, proinflammatory macrophage numbers, and increased hepatic glycogen content. Together, using in vitro experiments and rodent models, we demonstrate a proof-of-concept for AdipoR2 as a therapeutic target for MASLD and provide novel chemicobiological insights for the generation of translation-worthy pharmacological agents.
Collapse
Affiliation(s)
- Shamima Khatoon
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Nabanita Das
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Sourav Chattopadhyay
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | | | - Abhinav Singh
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Vishal Patel
- Zydus Research Center, Moraiya, Ahmedabad, 382213, Gujarat, India
| | - Abhishek Nirwan
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Akhilesh Kumar
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Madhav Nilakanth Mugale
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Durga Prasad Mishra
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Jagavelu Kumaravelu
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Rajdeep Guha
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Laboratory Animal Facility, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | | | - Naibedya Chattopadhyay
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Sabyasachi Sanyal
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|