1
|
Li MH, Yang Y, Dong QQ, Tao H, Lu C, Yang JJ. Novel epitranscriptomic and epigenetic therapeutic strategies and targets for ferroptosis in liver fibrosis. Eur J Pharmacol 2025; 996:177344. [PMID: 40015597 DOI: 10.1016/j.ejphar.2025.177344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 03/01/2025]
Abstract
Liver fibrosis is characterized by an excessive accumulation of extracellular matrix (ECM) and the activation of hepatic stellate cells (HSCs), which are influenced by epitranscriptomic and epigenetic factors. Recent advancements in epigenetic and epitranscriptomic research have revealed new opportunities for therapeutic interventions, particularly through the regulation of ferroptosis, a type of programmed cell death that is specifically linked to iron-dependent lipid peroxidation. In the context of liver fibrosis, a progressive scarring process that can progress to cirrhosis and ultimately end-stage liver disease, targeting these regulatory mechanisms to modulate ferroptosis presents a promising therapeutic strategy. This review aims to consolidate current knowledge on the epigenetic and epitranscriptomic control of ferroptosis and investigate its potential implications for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Ming-Hui Li
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yang Yang
- Department of General Surgery, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215153, China
| | - Qi-Qi Dong
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Chao Lu
- First Affiliated Hospital, Anhui University of Science & Technology, Huainan, 232001, China.
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
2
|
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:S0891-5849(25)00266-7. [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
|
3
|
Li J, Kemper T, Broering R, Lin Y, Wang X, Lu M. Amphisome plays a role in HBV production and release through the endosomal and autophagic pathways. Hepatol Commun 2025; 9:e0654. [PMID: 40079732 PMCID: PMC11908759 DOI: 10.1097/hc9.0000000000000654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 01/08/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Autophagic and endosomal pathways coordinately contribute to HBV virions and subviral particles (SVPs) production. To date, limited evidence supports that HBV and exosomes have a common pathway for their biogenesis and secretion. The final steps of HBV production and release have not yet been well studied. METHODS We examined the production and release of HBV virions and SVPs by using GW4869 (N,N'-Bis[4-(4,5-dihydro-1H-imidazol-2-yl)phenyl]-3,3'-pht hal amide dihydrochloride), a small molecule inhibiting ceramide-mediated inward membrane budding. Neutral sphingomyelinase, the target of GW4869, and RAB27A and -B, 2 small GTPases involved in exosome release control, were silenced using gene silencing to confirm the results obtained. Western blot, immunofluorescence staining, and confocal microscopy were applied. RESULTS GW4869 inhibited HBV virion release, causing their accumulation along with SVPs in hepatocytes. This triggered cellular endoplasmic reticulum stress, leading to protein kinase B-mechanistic target of rapamycin kinase signaling pathway inactivation. GW4869 treatment increased autophagosome formation and impaired autophagic degradation by blocking autophagosome-lysosome fusion. Consequently, HBsAg is increasingly localized to autophagosomes and late endosomes/multivesicular bodies. Silencing neutral sphingomyelinase yielded consistent results. Similarly, RAB27A silencing inhibited HBV virion and SVP secretion, causing their accumulation within hepatoma cells. Notably, GW4869 treatment, as well as RAB27A and -B silencing, increased the presence of LC3+CD63+HBsAg+ complexes. CONCLUSIONS Our results demonstrate the involvement of the autophagosome-late endosome/multivesicular bodies-exosome axis in regulating HBV production and release, highlighting amphisomes as a potential platform for HBV release.
Collapse
Affiliation(s)
- Jia Li
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Thekla Kemper
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ruth Broering
- Department of Gastroenterology, Hepatology and Transplant Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Yong Lin
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Xueyu Wang
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
4
|
Battaglia AM, Sacco A, Giorgio E, Petriaggi L, Elzanowska J, Cruz AR, Rocha L, Pereira CE, Strano Moraes MC, Palazzo L, De Vitis C, Costa-Silva B, Biamonte F. Expulsion of iron-rich ferritin via CD63-mediated exosome drives ferroptosis resistance in ovarian cancer cells. Front Cell Dev Biol 2025; 13:1532097. [PMID: 40177133 PMCID: PMC11962263 DOI: 10.3389/fcell.2025.1532097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/17/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Ferroptosis is a promising new target for ovarian cancer (OVCA) treatment. However, some OVCA cell types resist the induction of ferroptosis by limiting the intracellular accumulation of the labile iron pool (LIP). Methods HEY, COV318 and PEO4 were treated with erastin and assessed for cell viability by using PI flow cytometry assays. Erastin-affected iron metabolism was analysed by using FerroOrange assay, Western Blot (WB) analysis of ferritin heavy chain (FtH), transferrin receptor (CD71), and ferroportin (FPN). Mitochondrial reactive oxygen species (mitROS) and lipid peroxidation were quantified via MitoSOX and BODIPY-C11 flow cytometry assays, respectively. Exosomes (EVs) were collected from cell culture media through ultracentrifugation and then enumerated and analyzed by Nanoparticale Tracking Analysis (NTA) and transmission electron microscopy (TEM). CD63 protein expression in EVs was measured through WB by using CD9 as a loading control. Loss-of-function assays for FtH and CD63 were performed by using siRNA-mediated transient transfection. Results We demonstrate that erastin treatment (8 µM, 8 h) is accompanied by the release of iron-rich ferritin via EV pathway in COV318 and PEO4 OVCA cells, thus failing to exert cytotoxic effects. Mechanistically, erastin causes the upregulation of CD63, a tetraspanin involved in forming multivesicular bodies (MVBs) and EVs, and the increase of MBVs assessed by transmission electron microscopy. Consistent with these findings, EV isolation followed by nanoparticle tracking analysis revealed a significant increase in EVs/cell in erastin-treated COV318 and PEO4 cells. Notably, EVs harvested from these cells contained CD63 and FtH, a major iron-storage protein. Inhibition of EV biogenesis with GW4869 prevented FtH release and restored LIP accumulation, lipid peroxidation, and ferroptosis sensitivity in COV318 and PEO4 cells. Discussion Overall, our results indicate that OVCA cells can utilize CD63+ EVs to secrete iron-rich ferritin as a mechanism to evade erastin-induced ferroptosis. These findings suggest that combining erastin with EV inhibitors could offer promising strategy for overcoming ferroptosis resistance in OVCA.
Collapse
Affiliation(s)
- Anna Martina Battaglia
- Laboratory of Biochemistry and Cell Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Alessandro Sacco
- Laboratory of Biochemistry and Cell Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Emanuele Giorgio
- Laboratory of Biochemistry and Cell Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Lavinia Petriaggi
- Laboratory of Biochemistry and Cell Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Julia Elzanowska
- Systems Oncology Laboratory, Champalimaud Foundation, Lisbon, Portugal
| | - Ana Rita Cruz
- Systems Oncology Laboratory, Champalimaud Foundation, Lisbon, Portugal
| | - Luis Rocha
- Systems Oncology Laboratory, Champalimaud Foundation, Lisbon, Portugal
| | | | | | - Luca Palazzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - Claudia De Vitis
- Department of Clinical and Molecular Medicine, Sant’ Andrea Hospital-Sapienza University of Rome, Rome, Italy
| | - Bruno Costa-Silva
- Systems Oncology Laboratory, Champalimaud Foundation, Lisbon, Portugal
| | - Flavia Biamonte
- Laboratory of Biochemistry and Cell Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
5
|
Zhao X, Zhang Y, Luo B. Ferroptosis, from the virus point of view: opportunities and challenges. Crit Rev Microbiol 2025; 51:246-263. [PMID: 38588443 DOI: 10.1080/1040841x.2024.2340643] [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: 03/24/2023] [Revised: 12/21/2023] [Accepted: 04/01/2024] [Indexed: 04/10/2024]
Abstract
Ferroptosis is a new type of cell death, which is mainly dependent on the formation and accumulation of reactive oxygen species and lipid peroxides mediated by iron. It is distinct from other forms of regulation of cell death in morphology, immunology, biochemistry, and molecular biology. Various cell death mechanisms have been observed in many viral infections, and virus-induced cell death has long been considered as a double-edged sword that can inhibit or aggravate viral infections. However, understanding of the role of ferroptosis in various viral infections is limited. Special attention will be paid to the mechanisms of ferroptosis in mediating viral infection and antiviral treatment associated with ferroptosis. In this paper, we outlined the mechanism of ferroptosis. Additionally, this paper also review research on ferroptosis from the perspective of the virus, discussed the research status of ferroptosis in virus infection and classified and summarized research on the interaction between viral infections and ferroptosis.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yan Zhang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
- Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Bing Luo
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
6
|
Ge Y, Jiang L, Dong Q, Xu Y, Yam JWP, Zhong X. Exosome-mediated Crosstalk in the Tumor Immune Microenvironment: Critical Drivers of Hepatocellular Carcinoma Progression. J Clin Transl Hepatol 2025; 13:143-161. [PMID: 39917466 PMCID: PMC11797817 DOI: 10.14218/jcth.2024.00302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 02/09/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a significant global health issue, ranking as the sixth most prevalent malignancy and the fourth leading cause of cancer-related mortality worldwide. Despite advancements in therapeutic strategies, mortality rates for HCC remain high. The tumor immune microenvironment (TIME) plays a vital role in HCC progression by influencing tumor cell survival and growth. Recent studies highlight the essential role of exosomes in mediating intercellular communication within the TIME, particularly in interactions among tumor cells, immune cells, and fibroblasts. These interactions drive critical aspects of tumor development, including immune escape, angiogenesis, drug resistance, and metastasis. A detailed understanding of the molecular mechanisms by which exosomes modulate the TIME is essential for developing targeted therapies. This review systematically evaluated the roles and regulatory mechanisms of exosomes within the TIME of HCC, examining the impact of both HCC-derived and non-HCC-derived exosomes on various cellular components within the TIME. It emphasized their regulatory effects on cell phenotypes and functions, as well as their roles in HCC progression. The review also explored the potential applications of exosome-based immunotherapies, offering new insights into improving therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Yifei Ge
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lixue Jiang
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qingfu Dong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiangyu Zhong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
7
|
Nashtahosseini Z, Nejatollahi M, Fazilat A, Zarif Fakoor E, Emamvirdizadeh A, Bahadori K, Hadian NS, Valilo M. The crosstalk between exosomal miRNA and ferroptosis: A narrative review. Biol Cell 2025; 117:e2400077. [PMID: 39853758 DOI: 10.1111/boc.202400077] [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: 07/10/2024] [Revised: 12/24/2024] [Accepted: 01/06/2025] [Indexed: 01/26/2025]
Abstract
Ferroptosis is a type of cell death that multiple mechanisms and pathways contribute to the positive and negative regulation of it. For example, increased levels of reactive oxygen species (ROS) induce ferroptosis. ferroptosis unlike apoptosis, it is not dependent on caspases, but is dependent on iron. Exosomes are membrane-bound vesicles with a size of about 30 to 150 nm, contain various cellular components, including DNA, RNA, microRNAs (miRNAs), lipids, and proteins, which are genetically similar to their cells of origin. Exosomes are found in all bodily fluids, including blood, saliva, and urine. Cells often release exosomes after their fusion with the cell membrane. They play an important role in immune regulation and cell-cell communication. miRNAs, which are noncoding RNAs with a length of about 18 to 24 nucleotides, are involved in regulating gene expression after transcription. Emerging data suggests that exosomal miRNAs are implicated in various pathophysiological mechanisms of cells, including metastasis, drug resistance, and cell death. In addition, functional studies have indicated that exosomal miRNAs can play a key role in the modulation of cell death by regulating ferroptosis. Therefore, in this review, given the importance of exosomal miRNAs in ferroptosis, we decided to elucidate the relationship between exosomal miRNAs and ferroptosis in various diseases.
Collapse
Affiliation(s)
| | - Masoumeh Nejatollahi
- Research center for high school students, Education System Zanjan Province, Zanjan, Iran
| | - Ahmad Fazilat
- Department of Genetics, Motamed Cancer Institute, Breast Cancer Research Center, ACECR, Tehran, Iran
| | | | - Alireza Emamvirdizadeh
- Department of Genetics, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Kamran Bahadori
- Health center of Bahar, Hamadan University of Medical Science& Health Services, Hamadan, Iran
| | | | - Mohammad Valilo
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
8
|
Dong J, Hu Y, Liu S, Liu W, Zhu Q, Liu S, Zhang N, Liao C, Jiang G. Arsenic induces ferroptosis in HTR-8/SVneo cells and placental damage. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:176885. [PMID: 39414034 DOI: 10.1016/j.scitotenv.2024.176885] [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: 07/01/2024] [Revised: 09/24/2024] [Accepted: 10/10/2024] [Indexed: 10/18/2024]
Abstract
Placenta ferroptosis has been proven to be associated with a variety of adverse pregnancy outcomes. Arsenic, a conventional metal noxious substance, has garnered considerable attention due to traversing the placental barrier. How arsenic induces placental ferroptosis and reproductive developmental toxicities remains largely unknown. Herein, we investigated the impact of sodium arsenite (As (III)) on iron homeostasis in the placenta through both in vivo and in vitro experiments by using HTR-8/SVneo cells and ICR pregnant mice. As (III) up-regulated the expression of genes or proteins associated with iron uptake (TFRC, DMT1), iron storage (FTH, FTL), ferritin autophagy (NCOA4), and heme degradation (HO-1), and induced cell iron overload. Additionally, accumulation of the lipid hydroperoxide malondialdehyde within cells was triggered by As (III) through inhibition of the Nrf2/GPX4 signal pathway, which resulted in cellular ferroptosis. Fer-1 effectively alleviated the suppression of GPX4 induced by As (III), reduced the accumulation of intracellular lipid peroxidation product MDA, and mitigated cellular ferroptosis. As (III) affected the iron homeostasis, as evidenced by the abnormal iron accumulation in the placenta. Placental structural abnormalities and hemorrhage may be the reason for As (III) causing placental injury and subsequent poor pregnancy outcomes. This study provides new insights into understanding the mechanisms by which As (III) produces placental damage and possible fetal developmental toxicity.
Collapse
Affiliation(s)
- Jingcun Dong
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuang Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Liu
- Yale University, Yale Cancer Center, New Haven, CT 06511, United States
| | - Qingqing Zhu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Na Zhang
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, China.
| | - Chunyang Liao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, Zhejiang 310024, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, Zhejiang 310024, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
9
|
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
|
10
|
Cao M, Zou J, Shi M, Zhao D, Liu C, Liu Y, Li L, Jiang H. A promising therapeutic: Exosome-mediated mitochondrial transplantation. Int Immunopharmacol 2024; 142:113104. [PMID: 39270344 DOI: 10.1016/j.intimp.2024.113104] [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: 07/17/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
Mitochondrial dysfunction has been identified as a trigger for cellular autophagy dysfunction and programmed cell death. Emerging studies have revealed that, in pathological contexts, intercellular transfer of mitochondria takes place, facilitating the restoration of mitochondrial function, energy metabolism, and immune homeostasis. Extracellular vesicles, membranous structures released by cells, exhibit reduced immunogenicity and enhanced stability during the transfer of mitochondria. Thus, this review provides a concise overview of mitochondrial dysfunction related diseases and the mechanism of mitochondrial dysfunction in diseases progression, and the composition and functions of the extracellular vesicles, along with elucidating the principal mechanisms underlying intercellular mitochondrial transfer. In this article, we will focus on the advancements in both animal models and clinical trials concerning the therapeutic efficacy of extracellular vesicle-mediated mitochondrial transplantation across various systemic diseases in neurodegenerative diseases and cardiovascular diseases. Additionally, the review delves into the multifaceted roles of extracellular vesicle-transplanted mitochondria, encompassing anti-inflammatory actions, promotion of tissue repair, enhancement of cellular function, and modulation of metabolic and immune homeostasis within diverse pathological contexts, aiming to provide novel perspectives for extracellular vesicle transplantation of mitochondria in the treatment of various diseases.
Collapse
Affiliation(s)
- Meiling Cao
- Department of Neonatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Jiahui Zou
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Mingyue Shi
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Danyang Zhao
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Chang Liu
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yanshan Liu
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Lei Li
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Hongkun Jiang
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
11
|
Shi C, Hu S, Liu S, Jia X, Feng Y. Emerging role of exosomes during the pathogenesis of viral hepatitis, non-alcoholic steatohepatitis and alcoholic hepatitis. Hum Cell 2024; 38:26. [PMID: 39630211 DOI: 10.1007/s13577-024-01158-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/24/2024] [Indexed: 01/07/2025]
Abstract
Extracellular vesicles (EVs) refer to a diverse range of membranous vesicles that are secreted by various cell types, they can be categorized into two primary subgroups: exosomes and microvesicles. Specifically, exosomes constitute a nanosized subset of EVs characterized by their intact lipid bilayer and diameters ranging from 30 to 150 nm. These vesicles play a crucial role in intercellular communication by transporting a diverse array of biomolecules, which act as cargoes for this communication process. Exosomes have demonstrated significant implications in a wide range of biologic processes and pathologic conditions, including immunity, development, cancer, neurodegenerative diseases, and liver diseases. Liver diseases significantly contribute to the global burden of morbidity and mortality, yet their pathogenesis remains complex and effective therapies are relatively scarce. Emerging evidence suggests that exosomes play a modulatory role in the pathogenesis of liver diseases, including viral hepatitis, non-alcoholic steatohepatitis (NASH), and alcoholic hepatitis (AH). These findings bolster our confidence in the potential of exosomes as biomarkers and therapeutic tools for the diagnosis and treatment of liver diseases. In this comprehensive review, we offer a straightforward overview of exosomes and summarize the current understanding of their role in the pathogenesis of liver diseases. This provides a foundation for novel diagnostic and therapeutic approaches in the treatment of liver diseases.
Collapse
Affiliation(s)
- Congjian Shi
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, 350007, China
| | - Shuang Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
| | - Shen Liu
- Department of Pharmacy, Linquan County People's Hospital, Fuyang, 236400, Anhui, China
| | - Xiaodi Jia
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, 350007, China
| | - Yubin Feng
- Department of Pharmacy, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, 230001, Anhui, China.
| |
Collapse
|
12
|
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
|
13
|
Bai X, Li J, Guo X, Huang Y, Xu X, Tan A, Jia Y, Sun Q, Guo X, Chen J, Kang J. LncRNA MALAT1 promotes Erastin-induced ferroptosis in the HBV-infected diffuse large B-cell lymphoma. Cell Death Dis 2024; 15:819. [PMID: 39532842 PMCID: PMC11557927 DOI: 10.1038/s41419-024-07209-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
In a retrospective analysis of clinical data from 587 DLBCL (diffuse large B-cell lymphoma) patients in China, 13.8% of cases were associated with HBV (hepatitis B virus) infection, leading to distinct clinical features and poorer prognosis. Moreover, HBV infection has a more pronounced impact on the survival of the GCB (germinal center B-cell-like) type DLBCL patients compared to the ABC (activated B-cell-like) type. In this study, we found that the expression of LncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) was downregulated in the HBV-infected GCB-type DLBCL patients, and the HBV core protein (HBX) directly inhibited the MALAT1 expression in DLBCL cells. Notably, the overexpression of HBX could attenuate the Erastin-induced ferroptosis in the GCB-type DLBCLs, while MALAT1 re-expression restored sensitivity in the HBX-overexpressing DLBCLs in vitro and in vivo. Mechanistically, MALAT1 competitively hindered SFPQ (splicing factor proline and glutamine-rich) from effectively splicing the pre-mRNA of SLC7A11 (solute carrier family 7 member 11), due to a shared TTGGTCT motif, which impeded the SLC7A11 pre-mRNA maturation and hence diminished its negative regulation on ferroptosis. Together, our study identified HBX's role in inhibiting MALAT1 expression, promoting SFPQ-mediated splicing of SLC7A11 pre-mRNA, and reducing the GCB-type DLBCL sensitivity to Erastin-induced ferroptosis. Combined with the recent studies that ferroptosis may be involved in the occurrence and development of DLBCL, these findings explain our clinical data analysis that DLBCL patients with low expression of MALAT1 have poorer prognosis and shorter overall survival, and provide a valuable therapeutic target for the HBV-infected GCB-type DLBCL patients.
Collapse
MESH Headings
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/virology
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Ferroptosis/genetics
- Ferroptosis/drug effects
- Animals
- Hepatitis B virus/genetics
- Cell Line, Tumor
- Male
- Mice
- Hepatitis B/complications
- Hepatitis B/genetics
- Female
- Viral Regulatory and Accessory Proteins
- Gene Expression Regulation, Neoplastic/drug effects
- Middle Aged
- Trans-Activators/metabolism
- Trans-Activators/genetics
- Mice, Nude
Collapse
Affiliation(s)
- Xiaofei Bai
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jianguo Li
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xuecong Guo
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yinghui Huang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xu Xu
- Department of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ailing Tan
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yisha Jia
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qiaoyi Sun
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xudong Guo
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Jie Chen
- Department of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Jiuhong Kang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
14
|
Li S, Cheng F, Zhang Z, Xu R, Shi H, Yan Y. The role of hepatocyte-derived extracellular vesicles in liver and extrahepatic diseases. Biomed Pharmacother 2024; 180:117502. [PMID: 39357327 DOI: 10.1016/j.biopha.2024.117502] [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: 07/21/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
Extracellular vesicles (EVs) are vesicle-like bodies with a double membrane structure that are released from the cell membrane or secreted by cells into the extracellular environment. These include exosomes, microvesicles, and apoptotic bodies. There is growing evidence indicating that the composition of liver cell contents changes following injury. The quantity of EVs and the biologically active substances they carry vary depending on the condition of the liver cells. Hepatocytes utilize EVs to modulate the functions of different liver cells and transfer them to distant organs via the systemic circulation, thereby playing a crucial role in intercellular communication. This review provides a concise overview of the research on the effects and potential mechanisms of hepatocyte-derived EVs (Hep-EVs) on liver diseases and extrahepatic diseases under different physiological and pathological conditions. Common liver diseases discussed include non-alcoholic fatty liver disease (NAFLD), viral hepatitis, alcoholic liver disease, drug-induced liver damage, and liver cancer. Given that NAFLD is the most prevalent chronic liver disease globally, this review particularly highlights the use of hepatocyte-derived EVs in NAFLD for disease progression, diagnosis, and treatment.
Collapse
Affiliation(s)
- Shihui Li
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Fang Cheng
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Zhuan Zhang
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Ruizi Xu
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Honglei Shi
- Wujin Hospital Affiliated With Jiangsu University, Changzhou Wujin People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213004, China; Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University (Wujin Clinical College of Xuzhou Medical University), Changzhou 213017, China; Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou 213017, China.
| | - Yongmin Yan
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University (Wujin Clinical College of Xuzhou Medical University), Changzhou 213017, China; Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou 213017, China.
| |
Collapse
|
15
|
Lan W, Yang L, Tan X. Crosstalk between ferroptosis and macrophages: potential value for targeted treatment in diseases. Mol Cell Biochem 2024; 479:2523-2543. [PMID: 37880443 DOI: 10.1007/s11010-023-04871-4] [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: 06/26/2023] [Accepted: 10/05/2023] [Indexed: 10/27/2023]
Abstract
Ferroptosis is a newly identified form of programmed cell death that is connected to iron-dependent lipid peroxidization. It involves a variety of physiological processes involving iron metabolism, lipid metabolism, oxidative stress, and biosynthesis of nicotinamide adenine dinucleotide phosphate, glutathione, and coenzyme Q10. So far, it has been discovered to contribute to the pathological process of many diseases, such as myocardial infarction, acute kidney injury, atherosclerosis, and so on. Macrophages are innate immune system cells that regulate metabolism, phagocytize pathogens and dead cells, mediate inflammatory reactions, promote tissue repair, etc. Emerging evidence shows strong associations between macrophages and ferroptosis, which can provide us with a deeper comprehension of the pathological process of diseases and new targets for the treatments. In this review, we summarized the crosstalk between macrophages and ferroptosis and anatomized the application of this association in disease treatments, both non-neoplastic and neoplastic diseases. In addition, we have also addressed problems that remain to be investigated, in the hope of inspiring novel therapeutic strategies for diseases.
Collapse
Affiliation(s)
- Wanxin Lan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Lei Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Xuelian Tan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
16
|
Hou Q, Ouyang S, Xie Z, He Y, Deng Y, Guo J, Yu P, Tan X, Ma W, Li P, Yu J, Mo Q, Zhang Z, Chen D, Lin X, Liu Z, Chen X, Peng T, Li L, Xie W. Exosome is a Fancy Mobile Sower of Ferroptosis. J Cardiovasc Transl Res 2024; 17:1067-1082. [PMID: 38776048 DOI: 10.1007/s12265-024-10508-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/01/2024] [Indexed: 10/29/2024]
Abstract
Exosomes, nano-sized small extracellular vesicles, have been shown to serve as mediators between intercellular communications by transferring bioactive molecules, such as non-coding RNA, proteins, and lipids from secretory to recipient cells, modulating a variety of physiological and pathophysiological processes. Recent studies have gradually demonstrated that altered exosome charges may represent a key mechanism driving the pathological process of ferroptosis. This review summarizes the potential mechanisms and signal pathways relevant to ferroptosis and then discusses the roles of exosome in ferroptosis. As well as transporting iron, exosomes may also indirectly convey factors related to ferroptosis. Furthermore, ferroptosis may be transmitted to adjacent cells through exosomes, resulting in cascading effects. It is expected that further research on exosomes will be conducted to explore their potential in ferroptosis and will lead to the creation of new therapeutic avenues for clinical diseases.
Collapse
Affiliation(s)
- Qin Hou
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Siyu Ouyang
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhongcheng Xie
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yinling He
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yunong Deng
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jiamin Guo
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Panpan Yu
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoqian Tan
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wentao Ma
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Pin Li
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jiang Yu
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Qinger Mo
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhixia Zhang
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Dandan Chen
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoyan Lin
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhiyang Liu
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xi Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Tianhong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Liang Li
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
17
|
Ouyang A, Chen T, Feng Y, Zou J, Tu S, Jiang M, Sun H, Zhou H. The Hemagglutinin of Influenza A Virus Induces Ferroptosis to Facilitate Viral Replication. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404365. [PMID: 39159143 PMCID: PMC11497066 DOI: 10.1002/advs.202404365] [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: 04/24/2024] [Revised: 07/27/2024] [Indexed: 08/21/2024]
Abstract
Ferroptosis is a novel form of cell death caused by the accumulation of lipid peroxides in an iron-dependent manner. However, the precise mechanism underlying the exploitation of ferroptosis by influenza A viruses (IAV) remains unclear. The results demonstrate that IAV promotes its own replication through ferritinophagy by sensitizing cells to ferroptosis, with hemagglutinin identified as a key trigger in this process. Hemagglutinin interacts with autophagic receptors nuclear receptor coactivator 4 (NCOA4) and tax1-binding protein 1 (TAX1BP1), facilitating the formation of ferritin-NCOA4 condensates and inducing ferritinophagy. Further investigation shows that hemagglutinin-induced ferritinophagy causes cellular lipid peroxidation, inhibits aggregation of mitochondrial antiviral signaling protein (MAVS), and suppresses the type I interferon response, thereby contributing to viral replication. Collectively, a novel mechanism by which IAV hemagglutinin induces ferritinophagy resulting in cellular lipid peroxidation, consequently impairing MAVS-mediated antiviral immunity, is revealed.
Collapse
Affiliation(s)
- Aotian Ouyang
- National Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei430070China
| | - Tong Chen
- National Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei430070China
| | - Yi Feng
- National Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei430070China
| | - Jiahui Zou
- National Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei430070China
| | - Shaoyu Tu
- National Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei430070China
| | - Meijun Jiang
- National Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei430070China
| | - Huimin Sun
- National Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei430070China
| | - Hongbo Zhou
- National Key Laboratory of Agricultural MicrobiologyCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanHubei430070China
- Frontiers Science Center for Animal Breeding and Sustainable ProductionWuhanHubei430070China
- Hubei Hongshan LaboratoryWuhanHubei430070China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Provincethe Cooperative Innovation Center for Sustainable Pig ProductionWuhanHubei430070China
| |
Collapse
|
18
|
Song H, Zhang Q, Fang G, Luo X, Wu D, Li H, Zhou K, Zhao X, Xu F, Zhang Y, Huang A. Unraveling the Mechanisms of MicroRNA in Suppressing Hepatitis B Virus Progression: A Comprehensive Review for Designing Treatment Strategies. HEPATITIS MONTHLY 2024; 24. [DOI: 10.5812/hepatmon-144239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 06/22/2024] [Accepted: 07/13/2024] [Indexed: 01/02/2025]
Abstract
: Liver cancer and cirrhosis caused by the Hepatitis B virus (HBV) remain significant global health challenges due to the virus's high prevalence and contagious nature. Hepatitis B virus can be transmitted through various means, leading to mild or severe liver disease. Although an effective prophylactic vaccine is available, it offers limited benefits for those already chronically infected. Current treatments often fail to consistently eliminate the virus and can cause severe adverse effects. In response to these challenges, researchers have begun exploring microRNAs (miRNAs) as novel therapeutic targets. Studying miRNA-virus interactions presents a promising opportunity to identify potential therapeutic targets. By manipulating host miRNAs, researchers aim to enhance antiviral defenses, restore cellular balance, and prevent viral replication. The text concludes by highlighting the potential for personalized medicine in Hepatitis B treatment, guided by individual miRNA profiles. Numerous studies have been conducted to understand how different miRNAs inhibit HBV replication, paving the way for the development of innovative and effective therapeutic strategies.
Collapse
|
19
|
Guan J, Tang L, Wang Y, Fu M, Xia T, Zheng K, Sabi MM, Cong H, Wang J, Zhou C, Zhou H, Weiss LM, Qu H, Han B. Microsporidian EnP1 alters host cell H2B monoubiquitination and prevents ferroptosis facilitating microsporidia survival. Proc Natl Acad Sci U S A 2024; 121:e2400657121. [PMID: 39141344 PMCID: PMC11348272 DOI: 10.1073/pnas.2400657121] [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: 01/17/2024] [Accepted: 07/02/2024] [Indexed: 08/15/2024] Open
Abstract
Microsporidia are intracellular eukaryotic pathogens that pose a substantial threat to immunocompromised hosts. The way these pathogens manipulate host cells during infection remains poorly understood. Using a proximity biotinylation strategy we established that microsporidian EnP1 is a nucleus-targeted effector that modifies the host cell environment. EnP1's translocation to the host nucleus is meditated by nuclear localization signals (NLSs). In the nucleus, EnP1 interacts with host histone H2B. This interaction disrupts H2B monoubiquitination (H2Bub), subsequently impacting p53 expression. Crucially, this inhibition of p53 weakens its control over the downstream target gene SLC7A11, enhancing the host cell's resilience against ferroptosis during microsporidian infection. This favorable condition promotes the proliferation of microsporidia within the host cell. These findings shed light on the molecular mechanisms by which microsporidia modify their host cells to facilitate their survival.
Collapse
Affiliation(s)
- Jingyu Guan
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Liyuan Tang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Yongliang Wang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Ming Fu
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Tian Xia
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Kai Zheng
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Musa Makongoro Sabi
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Hua Cong
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Juncheng Wang
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Chunxue Zhou
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Huaiyu Zhou
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Louis M. Weiss
- Department of Pathology, Albert Einstein College of Medicine, New York, NY10461
| | - Hongnan Qu
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Bing Han
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| |
Collapse
|
20
|
Lu T, Zheng Y, Chen X, Lin Z, Liu C, Yuan C. The role of exosome derived miRNAs in inter-cell crosstalk among insulin-related organs in type 2 diabetes mellitus. J Physiol Biochem 2024; 80:501-510. [PMID: 38698251 DOI: 10.1007/s13105-024-01026-x] [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: 09/07/2023] [Accepted: 04/23/2024] [Indexed: 05/05/2024]
Abstract
Exosomes are small extracellular vesicles secreted by almost all cell types, and carry diverse cargo including RNA, and other substances. Recent studies have focused exosomal microRNAs (miRNAs) on various human diseases, including type 2 diabetes mellitus (T2DM) and metabolic syndrome (METS) which accompany the occurrence of insulin resistance. The regulation of insulin signaling has connected with some miRNA expression which play a significant regulatory character in insulin targeted cells or organs, such as fat, muscle, and liver. The miRNAs carried by exosomes, through the circulation in the body fluids, mediate all kinds of physiological and pathological process involved in the human body. Studies have found that exosome derived miRNAs are abnormally expressed and cross-talked with insulin targeted cells or organs to affect insulin pathways. Further investigations of the mechanisms of exosomal miRNAs in T2DM will be valuable for the diagnostic biomarkers and therapeutic targets of T2DM. This review will summarize the molecular mechanism of action of the miRNAs carried by exosomes which are secreted from insulin signaling related cells, and elucidate the pathogenesis of insulin resistance to provide a new strategy for the potential diagnostic biomarkers and therapeutic targets for the type 2 diabetes.
Collapse
Affiliation(s)
- Ting Lu
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Ying Zheng
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Xiaoling Chen
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Zhiyong Lin
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Chaoqi Liu
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China.
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
| | - Chengfu Yuan
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China.
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, China Three Gorges University, School of Medicine, Yichang, 443002, China.
| |
Collapse
|
21
|
Shi M, Jia JS, Gao GS, Hua X. Advances and challenges of exosome-derived noncoding RNAs for hepatocellular carcinoma diagnosis and treatment. Biochem Biophys Rep 2024; 38:101695. [PMID: 38560049 PMCID: PMC10979073 DOI: 10.1016/j.bbrep.2024.101695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/10/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Exosomes, also termed extracellular vesicles (EVs), are an important component of the tumor microenvironment (TME) and exert versatile effects on the molecular communications in the TME of hepatocellular carcinoma (HCC). Exosome-mediated intercellular communication is closely associated with the tumorigenesis and development of HCC. Exosomes can be extracted through ultracentrifugation and size exclusion, followed by molecular analysis through sequencing. Increasing studies have confirmed the important roles of exosome-derived ncRNAs in HCC, including tumorigenesis, progression, immune escape, and treatment resistance. Due to the protective membrane structure of exosomes, the ncRNAs carried by exosomes can evade degradation by enzymes in body fluids and maintain good expression stability. Thus, exosome-derived ncRNAs are highly suitable as biomarkers for the diagnosis and prognostic prediction of HCC, such as exosomal miR-21-5p, miR-221-3p and lncRNA-ATB. In addition, substantial studies revealed that the up-or down-regulation of exosome-derived ncRNAs had an important impact on HCC progression and response to treatment. Exosomal biomarkers, such as miR-23a, lncRNA DLX6-AS1, miR-21-5p, lncRNA TUC339, lncRNA HMMR-AS1 and hsa_circ_0004658, can reshape immune microenvironment by regulating M2-type macrophage polarization and then promote HCC development. Therefore, by controlling exosome biogenesis and modulating exosomal ncRNA levels, HCC may be inhibited or eliminated. In this current review, we summarized the recent findings on the role of exosomes in HCC progression and analyzed the relationship between exosome-derived ncRNAs and HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Min Shi
- Department of Clinical Laboratory, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| | - Jun-Su Jia
- Department of Clinical Laboratory, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| | - Guo-Sheng Gao
- Department of Clinical Laboratory, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| | - Xin Hua
- Department of Clinical Laboratory, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
22
|
Escuder-Rodríguez JJ, Liang D, Jiang X, Sinicrope FA. Ferroptosis: Biology and Role in Gastrointestinal Disease. Gastroenterology 2024; 167:231-249. [PMID: 38431204 PMCID: PMC11193643 DOI: 10.1053/j.gastro.2024.01.051] [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: 07/28/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 03/05/2024]
Abstract
Ferroptosis is a form of nonapoptotic cell death that involves iron-dependent phospholipid peroxidation induced by accumulation of reactive oxygen species, and results in plasma membrane damage and the release of damage-associated molecular patterns. Ferroptosis has been implicated in aging and immunity, as well as disease states including intestinal and liver conditions and cancer. To date, several ferroptosis-associated genes and pathways have been implicated in liver disease. Although ferroptotic cell death is associated with dysfunction of the intestinal epithelium, the underlying molecular basis is poorly understood. As the mechanisms regulating ferroptosis become further elucidated, there is clear potential to use ferroptosis to achieve therapeutic benefit.
Collapse
Affiliation(s)
- Juan-José Escuder-Rodríguez
- Department of Medicine, Gastrointestinal Research Unit, Mayo Clinic Alix School of Medicine, Rochester, Minnesota
| | - Deguang Liang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York.
| | - Frank A Sinicrope
- Department of Medicine, Gastrointestinal Research Unit, Mayo Clinic Alix School of Medicine, Rochester, Minnesota.
| |
Collapse
|
23
|
Wu X, Niu J, Shi Y. Exosomes target HBV-host interactions to remodel the hepatic immune microenvironment. J Nanobiotechnology 2024; 22:315. [PMID: 38840207 PMCID: PMC11151510 DOI: 10.1186/s12951-024-02544-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 05/09/2024] [Indexed: 06/07/2024] Open
Abstract
Chronic hepatitis B poses a significant global burden, modulating immune cells, leading to chronic inflammation and long-term damage. Due to its hepatotropism, the hepatitis B virus (HBV) cannot infect other cells. The mechanisms underlying the intercellular communication among different liver cells in HBV-infected individuals and the immune microenvironment imbalance remain elusive. Exosomes, as important intercellular communication and cargo transportation tools between HBV-infected hepatocytes and immune cells, have been shown to assist in HBV cargo transportation and regulate the immune microenvironment. However, the role of exosomes in hepatitis B has only gradually received attention in recent years. Minimal literature has systematically elaborated on the role of exosomes in reshaping the immune microenvironment of the liver. This review unfolds sequentially based on the biological processes of exosomes: exosomes' biogenesis, release, transport, uptake by recipient cells, and their impact on recipient cells. We delineate how HBV influences the biogenesis of exosomes, utilizing exosomal covert transmission, and reshapes the hepatic immune microenvironment. And based on the characteristics and functions of exosomes, potential applications of exosomes in hepatitis B are summarized and predicted.
Collapse
Affiliation(s)
- Xiaojing Wu
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, Jilin, 130021, People's Republic of China
| | - Junqi Niu
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, Jilin, 130021, People's Republic of China.
| | - Ying Shi
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, Jilin, 130021, People's Republic of China.
| |
Collapse
|
24
|
Cui H, Wang Y, Ma J, Zhou L, Li G, Li Y, Sun Y, Shen J, Ma T, Wang Q, Feng X, Dong B, Yang P, Li Y, Ma X. Advances in exosome modulation of ferroptosis for the treatment of orthopedic diseases. Pathol Res Pract 2024; 257:155312. [PMID: 38663177 DOI: 10.1016/j.prp.2024.155312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024]
Abstract
Current treatments for orthopaedic illnesses frequently result in poor prognosis, treatment failure, numerous relapses, and other unpleasant outcomes that have a significant impact on patients' quality of life. Cell-free therapy has emerged as one of the most promising options in recent decades for improving the status quo. As a result, using exosomes produced from various cells to modulate ferroptosis has been proposed as a therapeutic method for the condition. Exosomes are extracellular vesicles that secrete various bioactive chemicals that influence disease treatment and play a role in the genesis and progression of orthopaedic illnesses. Ferroptosis is a recently defined kind of controlled cell death typified by large iron ion buildup and lipid peroxidation. An increasing number of studies indicate that ferroptosis plays a significant role in orthopaedic illnesses. Exosomes, as intercellular information transfer channels, have been found to play a significant role in the regulation of ferroptosis processes. Furthermore, accumulating research suggests that exosomes can influence the course of many diseases by regulating ferroptosis in injured cells. In order to better understand the processes by which exosomes govern ferroptosis in the therapy of orthopaedic illnesses. This review discusses the biogenesis, secretion, and uptake of exosomes, as well as the mechanisms of ferroptosis and exosomes in the therapy of orthopaedic illnesses. It focuses on recent research advances and exosome mechanisms in regulating iron death for the therapy of orthopaedic illnesses. The present state of review conducted both domestically and internationally is elucidated and anticipated as a viable avenue for future therapy in the field of orthopaedics.
Collapse
Affiliation(s)
- Hongwei Cui
- Tianjin Medical University Orthopedic Clinical College, Tianjin 300050, China; Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Yan Wang
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Jianxiong Ma
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China.
| | - Liyun Zhou
- Tianjin Medical University Orthopedic Clinical College, Tianjin 300050, China; Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Guang Li
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Yiyang Li
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Yadi Sun
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Jiahui Shen
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Tiancheng Ma
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Qiyu Wang
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Xiaotian Feng
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Benchao Dong
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Peichuan Yang
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Yan Li
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| | - Xinlong Ma
- Tianjin Hospital of Tianjin University (Tianjin Hospital), Tianjin 300211, China; Tianjin Orthopedic Institute, Tianjin 300050, China; Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin 300050, China
| |
Collapse
|
25
|
Qiu X, Bi Q, Wu J, Sun Z, Wang W. Role of ferroptosis in fibrosis: From mechanism to potential therapy. Chin Med J (Engl) 2024; 137:806-817. [PMID: 37668091 PMCID: PMC10997224 DOI: 10.1097/cm9.0000000000002784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Indexed: 09/06/2023] Open
Abstract
ABSTRACT Fibrosis, which is a manifestation of the physiological response to injury characterized by excessive accumulation of extracellular matrix components, is a ubiquitous outcome of the repair process. However, in cases of repetitive or severe injury, fibrosis may become dysregulated, leading to a pathological state and organ failure. In recent years, a novel form of regulated cell death, referred to as ferroptosis, has been identified as a possible contributor to fibrosis; it is characterized by iron-mediated lipid peroxidation. It has garnered attention due to the growing body of evidence linking ferroptosis and fibrogenesis, which is believed to be driven by underlying inflammation and immune responses. Despite the increasing interest in the relationship between ferroptosis and fibrosis, a comprehensive understanding of the precise role that ferroptosis plays in the formation of fibrotic tissue remains limited. This review seeks to synthesize previous research related to the topic. We categorized the different direct and indirect mechanisms by which ferroptosis may contribute to fibrosis into three categories: (1) iron overload toxicity; (2) ferroptosis-evoked necroinflammation, with a focus on ferroptosis and macrophage interplay; and (3) ferroptosis-associated pro-fibrotic factors and pathways. Furthermore, the review considers the potential implications of these findings and highlights the utilization of ferroptosis-targeted therapies as a promising strategy for mitigating the progression of fibrosis. In conclusion, novel anti-fibrotic treatments targeting ferroptosis could be an effective treatment for fibrosis.
Collapse
Affiliation(s)
- Xuemeng Qiu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
- Department of Surgery, Third Clinical Medical College, Capital Medical University, Beijing 100020, China
| | - Qing Bi
- Urinary and Nephropathy Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Jiyue Wu
- Institute of Urology, Capital Medical University, Beijing 100020, China
| | - Zejia Sun
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Wei Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
- Urinary and Nephropathy Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
26
|
Zhou Z, Shi L, Chen B, Qian H. Regulation of regulated cell death by extracellular vesicles in acute kidney injury and chronic kidney disease. Cytokine Growth Factor Rev 2024; 76:99-111. [PMID: 38182464 DOI: 10.1016/j.cytogfr.2023.12.006] [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: 12/04/2023] [Accepted: 12/28/2023] [Indexed: 01/07/2024]
Abstract
The imbalance between proliferation and death of kidney resident cells is a crucial factor in the development of acute or chronic renal dysfunction. Acute kidney injury (AKI) is often associated with the rapid loss of tubular epithelial cells (TECs). Sustained injury leads to the loss of glomerular endothelial cells (GECs) and podocytes, which is a key mechanism in the pathogenesis of glomerular diseases. This irreversible damage resulting from progressive cell loss eventually leads to deterioration of renal function characterized by glomerular compensatory hypertrophy, tubular degeneration, and renal fibrosis. Regulated cell death (RCD), which involves a cascade of gene expression events with tight structures, plays a certain role in regulating kidney health by determining the fate of kidney resident cells. Under pathological conditions, cells in the nephron have been demonstrated to constitutively release extracellular vesicles (EVs) which act as messengers that specifically interact with recipient cells to regulate their cell death process. For therapeutic intervention, exogenous EVs have exhibited great potential for the prevention and treatment of kidney disease by modulating RCD, with enhanced effects through engineering modification. Based on the functional role of EVs, this review comprehensively explores the regulation of RCD by EVs in AKI and chronic kidney disease (CKD), with emphasis on pathogenesis and therapeutic intervention.
Collapse
Affiliation(s)
- Zixuan Zhou
- Institute of Translational Medicine of Jiangsu University, Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, Jiangsu, China; Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Linru Shi
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Binghai Chen
- Institute of Translational Medicine of Jiangsu University, Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, Jiangsu, China
| | - Hui Qian
- Institute of Translational Medicine of Jiangsu University, Department of Urology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, Jiangsu, China; Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China.
| |
Collapse
|
27
|
Huang Q, Zhong X, Li J, Hu R, Yi J, Sun J, Xu Y, Zhou X. Exosomal ncRNAs: Multifunctional contributors to the immunosuppressive tumor microenvironment of hepatocellular carcinoma. Biomed Pharmacother 2024; 173:116409. [PMID: 38460375 DOI: 10.1016/j.biopha.2024.116409] [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: 12/05/2023] [Revised: 02/23/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant liver cancer characterized by aggressive progression, unfavorable prognosis, and an increasing global health burden. Therapies that precisely target immunological checkpoints and immune cells have gained significant attention as possible therapeutics in recent years. In truth, the efficacy of immunotherapy is heavily contingent upon the tumor microenvironment (TME). Recent studies have indicated that exosomes serve as a sophisticated means of communication among biomolecules, executing an essential part in the TME of immune suppression. Exosomal non-coding RNAs (ncRNAs) can induce the activation of tumor cells and immunosuppressive immune cells that suppress the immune system, such as cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), CD+8 T cells, regulatory T cells (Tregs), and regulatory B cells (Bregs). This cell-cell crosstalk triggered by exosomal ncRNAs promotes tumor proliferation and metastasis, angiogenesis, malignant phenotype transformation, and drug resistance. Hence, it is imperative to comprehend how exosomal ncRNAs regulate tumor cells or immune cells within the TME to devise more comprehensive and productive immunotherapy programs. This study discusses the features of exosomal ncRNAs in HCC and how the activation of the exosomes redefines the tumor's immunosuppressive microenvironment, hence facilitating the advancement of HCC. Furthermore, we also explored the potential of exosomal ncRNAs as a viable biological target or natural vehicle for HCC therapy.
Collapse
Affiliation(s)
- Qi Huang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao PR China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China
| | - Xin Zhong
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China
| | - Jing Li
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao PR China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China
| | - Rui Hu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao PR China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China
| | - Jinyu Yi
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao PR China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China
| | - Jialing Sun
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China
| | - Youhua Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao PR China.
| | - Xiaozhou Zhou
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China.
| |
Collapse
|
28
|
Sun J, Chen Y, Wang T, Ali W, Ma Y, Yuan Y, Gu J, Bian J, Liu Z, Zou H. Baicalin and N-acetylcysteine regulate choline metabolism via TFAM to attenuate cadmium-induced liver fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155337. [PMID: 38241915 DOI: 10.1016/j.phymed.2024.155337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/14/2023] [Accepted: 01/05/2024] [Indexed: 01/21/2024]
Abstract
(Background): Cadmium is an environmental pollutant associated with several liver diseases. Baicalin and N-Acetylcysteine have antioxidant and hepatoprotective effects. (Purpose): However, it is unclear whether baicalin and N-Acetylcysteine can alleviate Cadmium -induced liver fibrosis by regulating metabolism, or whether they exert a synergistic effect. (Study design): We treated Cadmium-poisoned mice with baicalin, N-Acetylcysteine, or baicalin+ N-Acetylcysteine. We studied the effects of baicalin and N-Acetylcysteine on Cadmium-induced liver fibers and their specific mechanisms. (Methods): We used C57BL/6 J mice, and AML12, and HSC-6T cells to establish in vitro assays and in vivo models. (Results): Metabolomics was used to detect the effect of baicalin and N-Acetylcysteine on liver metabolism, which showed that compared with the control group, the Cadmium group had increased fatty acid and amino acid levels, with significantly reduced choline and acetylcholine contents. Baicalin and N-Acetylcysteine alleviated these Cadmium-induced metabolic changes. We further showed that choline alleviated Cadmium -induced liver inflammation and fibrosis. In addition, cadmium significantly promoted extracellular leakage of lactic acid, while choline alleviated the cadmium -induced destruction of the cell membrane structure and lactic acid leakage. Western blotting showed that cadmium significantly reduced mitochondrial transcription factor A (TFAM) and Choline Kinase α(CHKα2) levels, and baicalin and N-Acetylcysteine reversed this effect. Overexpression of Tfam in mouse liver and AML12 cells increased the expression of CHKα2 and the choline content, alleviating and cadmium-induced lactic acid leakage, liver inflammation, and fibrosis. (Conclusion): Overall, baicalin and N-Acetylcysteine alleviated cadmium-induced liver damage, inflammation, and fibrosis to a greater extent than either drug alone. TFAM represents a target for baicalin and N-Acetylcysteine, and alleviated cadmium-induced liver inflammation and fibrosis by regulating hepatic choline metabolism.
Collapse
Affiliation(s)
- Jian Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Yan Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Tao Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Waseem Ali
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Yonggang Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| |
Collapse
|
29
|
Zhang Y, Xie J. Ferroptosis-related exosomal non-coding RNAs: promising targets in pathogenesis and treatment of non-malignant diseases. Front Cell Dev Biol 2024; 12:1344060. [PMID: 38385027 PMCID: PMC10879574 DOI: 10.3389/fcell.2024.1344060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/10/2024] [Indexed: 02/23/2024] Open
Abstract
Ferroptosis, an iron-dependent form of programmed cell death, introduces a novel perspective on cellular demise. This study investigates the regulatory network of exosomal non-coding RNAs (ncRNAs), including miRNAs, circRNAs, and lncRNAs, in ferroptosis modulation. The primary goal is to examine the pathological roles of ferroptosis-related exosomal ncRNAs, particularly in ischemic reperfusion injuries. The research reveals intricate molecular interactions governing the regulatory interplay between exosomal ncRNAs and ferroptosis, elucidating their diverse roles in different non-malignant pathological contexts. Attention is given to their impact on diseases, including cardiac, cerebral, liver, and kidney ischemic injuries, as well as lung, wound, and neuronal injuries. Beyond theoretical exploration, the study provides insights into potential therapeutic applications, emphasizing the significance of mesenchymal stem cells (MSCs)-derived exosomes. Findings underscore the pivotal role of MSC-derived exosomal ncRNAs in modulating cellular responses related to ferroptosis regulation, introducing a cutting-edge dimension. This recognition emphasizes the importance of MSC-derived exosomes as crucial mediators with broad therapeutic implications. Insights unveil promising avenues for targeted interventions, capitalizing on the diverse roles of exosomal ncRNAs, providing a comprehensive foundation for future therapeutic strategies.
Collapse
Affiliation(s)
- Yiping Zhang
- School of Life Science, Fudan University, Shanghai, China
- Wanchuanhui (Shanghai) Medical Technology Co., Ltd., Shanghai, China
| | - Jun Xie
- School of Life Science, Fudan University, Shanghai, China
- Wanchuanhui (Shanghai) Medical Technology Co., Ltd., Shanghai, China
| |
Collapse
|
30
|
Lai W, Wang B, Huang R, Zhang C, Fu P, Ma L. Ferroptosis in organ fibrosis: From mechanisms to therapeutic medicines. J Transl Int Med 2024; 12:22-34. [PMID: 38525436 PMCID: PMC10956731 DOI: 10.2478/jtim-2023-0137] [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] [Indexed: 03/26/2024] Open
Abstract
Fibrosis occurs in many organs, and its sustained progress can lead to organ destruction and malfunction. Although numerous studies on organ fibrosis have been carried out, its underlying mechanism is largely unknown, and no ideal treatment is currently available. Ferroptosis is an iron-dependent process of programmed cell death that is characterized by lipid peroxidation. In the past decade, a growing body of evidence demonstrated the association between ferroptosis and fibrotic diseases, while targeting ferroptosis may serve as a potential therapeutic strategy. This review highlights recent advances in the crosstalk between ferroptosis and organ fibrosis, and discusses ferroptosis-targeted therapeutic approaches against fibrosis that are currently being explored.
Collapse
Affiliation(s)
- Weijing Lai
- Department of Nephrology, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan Province, China
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Bo Wang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Rongshuang Huang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Chuyue Zhang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Ping Fu
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Liang Ma
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| |
Collapse
|
31
|
Liu L, Ye Y, Lin R, Liu T, Wang S, Feng Z, Wang X, Cao H, Chen X, Miao J, Liu Y, Jiang K, Han Z, Li Z, Cao X. Ferroptosis: a promising candidate for exosome-mediated regulation in different diseases. Cell Commun Signal 2024; 22:6. [PMID: 38166927 PMCID: PMC11057189 DOI: 10.1186/s12964-023-01369-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/28/2023] [Indexed: 01/05/2024] Open
Abstract
Ferroptosis is a newly discovered form of cell death that is featured in a wide range of diseases. Exosome therapy is a promising therapeutic option that has attracted much attention due to its low immunogenicity, low toxicity, and ability to penetrate biological barriers. In addition, emerging evidence indicates that exosomes possess the ability to modulate the progression of diverse diseases by regulating ferroptosis in damaged cells. Hence, the mechanism by which cell-derived and noncellular-derived exosomes target ferroptosis in different diseases through the system Xc-/GSH/GPX4 axis, NAD(P)H/FSP1/CoQ10 axis, iron metabolism pathway and lipid metabolism pathway associated with ferroptosis, as well as its applications in liver disease, neurological diseases, lung injury, heart injury, cancer and other diseases, are summarized here. Additionally, the role of exosome-regulated ferroptosis as an emerging repair mechanism for damaged tissues and cells is also discussed, and this is expected to be a promising treatment direction for various diseases in the future. Video Abstract.
Collapse
Affiliation(s)
- Limin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Yulin Ye
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Rui Lin
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Sinan Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Zelin Feng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Xiaoli Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Junming Miao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Yifei Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Kui Jiang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China.
| | - Zhibo Han
- National Engineering Research Center of Cell Products, AmCellGene Engineering Co., Ltd, Tianjin, 300457, China.
- Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, Tianjin, 300457, China.
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China.
| | - Zongjin Li
- Nankai University School of Medicine, Tianjin, 300071, China.
| | - Xiaocang Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300052, China.
| |
Collapse
|
32
|
Huang R, Wu J, Ma Y, Kang K. Molecular Mechanisms of Ferroptosis and Its Role in Viral Pathogenesis. Viruses 2023; 15:2373. [PMID: 38140616 PMCID: PMC10747891 DOI: 10.3390/v15122373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Ferroptosis is a novelty form of regulated cell death, and it is mainly characterized by iron accumulation and lipid peroxidation in the cells. Its underlying mechanism is related to the amino acid, iron, and lipid metabolisms. During viral infection, pathogenic microorganisms have evolved to interfere with ferroptosis, and ferroptosis is often manipulated by viruses to regulate host cell servicing for viral reproduction. Therefore, this review provides a comprehensive overview of the mechanisms underlying ferroptosis, elucidates the intricate signaling pathways involved, and explores the pivotal role of ferroptosis in the pathogenesis of viral infections. By enhancing our understanding of ferroptosis, novel therapeutic strategies can be devised to effectively prevent and treat diseases associated with this process. Furthermore, unraveling the developmental mechanisms through which viral infections exploit ferroptosis will facilitate development of innovative antiviral agents.
Collapse
Affiliation(s)
- Riwei Huang
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (R.H.); (J.W.); (Y.M.)
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jiang Wu
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (R.H.); (J.W.); (Y.M.)
| | - Yaodan Ma
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (R.H.); (J.W.); (Y.M.)
| | - Kai Kang
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (R.H.); (J.W.); (Y.M.)
| |
Collapse
|
33
|
Gan L, Zheng L, Yao L, Lei L, Huang Y, Zeng Z, Fang N. Exosomes from adipose-derived mesenchymal stem cells improve liver fibrosis by regulating the miR-20a-5p/TGFBR2 axis to affect the p38 MAPK/NF-κB pathway. Cytokine 2023; 172:156386. [PMID: 37852157 DOI: 10.1016/j.cyto.2023.156386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/08/2023] [Accepted: 09/27/2023] [Indexed: 10/20/2023]
Abstract
OBJECTIVE Human adipose-derived mesenchymal stem cell exosomes (ADSC-Exos) are active constituents for treating liver fibrosis. This paper attempted to preliminarily explain the functional mechanism of ADSC-Exos in liver fibrosis through the p38 MAPK/NF-κB pathway. METHODS The cell models of hepatic fibrosis were established by inducing LX-2 cells with TGF-β1. Mouse models of liver fibrosis were established by treating mice with CCl4. The in vivo and in vitro models of liver fibrosis were treated with ADSC-Exos. ADSCs were identified by flow cytometry/Alizarin red/oil red O/alcian blue staining. ADSC-Exos were identified by transmission electron microscopy, nanoparticle tracking analysis, and Western blot. LX-2 cell proliferation/viability were evaluated by MTT/BrdU assays. Exosomes were tracked in vivo and body weight changes in mice were monitored. Hepatic pathological changes were observed by HE/Masson staining. α-SMA/collagen I levels in liver tissues were assessed by immunohistochemistry. HA/PIIINP concentrations were measured using the magnetic particle chemiluminescence method. Liver function was assessed using an automatic analyzer. miR-20a-5p level was measured by RT-qPCR. The mRNA levels of fibrosis markers were determined by RT-qPCR, and their protein levels and levels of MAPK/NF-κB pathway-related proteins, as well as TGFBR2 protein level were measured by Western blot. The P65 nuclear expression in mouse liver tissues was quantified by immunofluorescence. RESULTS ADSC-Exos suppressed TGF-β1-induced LX-2 cell proliferation and fibrosis and reduced mRNA and protein levels of fibrosis markers in vitro. ADSC-Exos ameliorated liver fibrosis by inhibiting the p38 MAPK/NF-κB pathway activation. ADSC-Exos inhibited activation of the p38 MAPK/NF-κB pathway via regulating the miR-20a-5p/TGFBR2 axis. The in vivo experiment asserted that ADSC-Exos were mainly distributed in the liver, and ADSC-Exos relieved liver fibrosis in mice, which was evidenced by alleviating decreased body weight, reducing collagen and enhancing liver function, and repressed the activation of the p38 MAPK/NF-κB pathway via the miR-20a-5p/TGFBR2 axis. CONCLUSION ADSC-Exos attenuated liver fibrosis by suppressing the activation of the p38 MAPK/NF-κB pathway via the miR-20a-5p/TGFBR2 axis.
Collapse
Affiliation(s)
- Lihong Gan
- Third Clinical Medical College, Nanchang University, Nanchang, China; Department of Gastroenterology, The First Hospital of Nanchang (The Third Affiliated Hospital of Nanchang University), Nanchang, China
| | - Li Zheng
- Department of Gastroenterology, The First Hospital of Nanchang (The Third Affiliated Hospital of Nanchang University), Nanchang, China
| | - Ling Yao
- Department of Gastroenterology, The First Hospital of Nanchang (The Third Affiliated Hospital of Nanchang University), Nanchang, China
| | - Ling Lei
- Department of Gastroenterology, The First Hospital of Nanchang (The Third Affiliated Hospital of Nanchang University), Nanchang, China
| | - Yaqin Huang
- Department of Gastroenterology, The First Hospital of Nanchang (The Third Affiliated Hospital of Nanchang University), Nanchang, China
| | - Zhiping Zeng
- Department of Gastroenterology, The First Hospital of Nanchang (The Third Affiliated Hospital of Nanchang University), Nanchang, China
| | - Nian Fang
- Third Clinical Medical College, Nanchang University, Nanchang, China; Department of Gastroenterology, The First Hospital of Nanchang (The Third Affiliated Hospital of Nanchang University), Nanchang, China.
| |
Collapse
|
34
|
Feng S, Tang D, Wang Y, Li X, Bao H, Tang C, Dong X, Li X, Yang Q, Yan Y, Yin Z, Shang T, Zheng K, Huang X, Wei Z, Wang K, Qi S. The mechanism of ferroptosis and its related diseases. MOLECULAR BIOMEDICINE 2023; 4:33. [PMID: 37840106 PMCID: PMC10577123 DOI: 10.1186/s43556-023-00142-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/23/2023] [Indexed: 10/17/2023] Open
Abstract
Ferroptosis, a regulated form of cellular death characterized by the iron-mediated accumulation of lipid peroxides, provides a novel avenue for delving into the intersection of cellular metabolism, oxidative stress, and disease pathology. We have witnessed a mounting fascination with ferroptosis, attributed to its pivotal roles across diverse physiological and pathological conditions including developmental processes, metabolic dynamics, oncogenic pathways, neurodegenerative cascades, and traumatic tissue injuries. By unraveling the intricate underpinnings of the molecular machinery, pivotal contributors, intricate signaling conduits, and regulatory networks governing ferroptosis, researchers aim to bridge the gap between the intricacies of this unique mode of cellular death and its multifaceted implications for health and disease. In light of the rapidly advancing landscape of ferroptosis research, we present a comprehensive review aiming at the extensive implications of ferroptosis in the origins and progress of human diseases. This review concludes with a careful analysis of potential treatment approaches carefully designed to either inhibit or promote ferroptosis. Additionally, we have succinctly summarized the potential therapeutic targets and compounds that hold promise in targeting ferroptosis within various diseases. This pivotal facet underscores the burgeoning possibilities for manipulating ferroptosis as a therapeutic strategy. In summary, this review enriched the insights of both investigators and practitioners, while fostering an elevated comprehension of ferroptosis and its latent translational utilities. By revealing the basic processes and investigating treatment possibilities, this review provides a crucial resource for scientists and medical practitioners, aiding in a deep understanding of ferroptosis and its effects in various disease situations.
Collapse
Affiliation(s)
- Shijian Feng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Dan Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yichang Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiang Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui Bao
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chengbing Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiuju Dong
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xinna Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qinxue Yang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yun Yan
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhijie Yin
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tiantian Shang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Kaixuan Zheng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaofang Huang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zuheng Wei
- Chengdu Jinjiang Jiaxiang Foreign Languages High School, Chengdu, People's Republic of China
| | - Kunjie Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Shiqian Qi
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
35
|
Yin M, Ding X, Yin S, Wang L, Zhang K, Chen Y, Liu R, Zhu C, Li W. Exosomes from hepatitis B virus-infected hepatocytes activate hepatic stellate cells and aggravate liver fibrosis through the miR-506-3p/Nur77 pathway. J Biochem Mol Toxicol 2023; 37:e23432. [PMID: 37352222 DOI: 10.1002/jbt.23432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 02/06/2023] [Accepted: 06/12/2023] [Indexed: 06/25/2023]
Abstract
Cumulative evidence indicates the important role of Nur77 in organ fibrogenesis. However, the role of Nur77 in hepatitis B virus (HBV)-related liver fibrosis (LF) remains unclear. Cells were transfected with the microRNA mimic miRNA-506-3p or inhibitor, and pcDNA3.1-Nur77 or Nur77 guide RNA. Exosomes were isolated from HBV-infected HepG2-sodium taurocholate cotransporting polypeptide cells. The levels of miR-506-3p, Nur77, and LF-related genes and proteins were detected by quantitative polymerase chain reaction (qPCR) and western blot analysis, respectively. The pathology of the liver from HBV-infected patients was examined using hematoxylin-eosin and Masson's staining. The expression of Nur77 in liver tissue was determined by immunohistochemistry, and the LF score was assessed using the METAVIR system. The relationship between miR-506-3p/Nur77 and LF score was analyzed by correlation analysis. HBV infection downregulated miR-506-3p expression and upregulated Nur77 levels in hepatocytes. Exosomes from HBV-infected hepatocytes also displayed decreased gene expression of miR-506-3p and increased expressions of Nur77- and LF-related genes in stellate cells compared with exosomes from hepatocytes with mock infection. These changes were reversed by Nur77 guide RNA. Nur77 expression in liver tissue was strongly correlated with LF, whereas serum miR-506-3p was strongly negatively correlated with LF. Exosomes from HBV-infected hepatocytes activate stellate cells and aggravate LF through the miR-506-3p/Nur77 pathway. These exosomes may be the basis of a promising therapeutic strategy.
Collapse
Affiliation(s)
- Ming Yin
- Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Critical Care Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Xiurong Ding
- Graduate School of Bengbu Medical University, Bengbu, China
- Department of Infectious Disease, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Song Yin
- Department of Infectious Disease, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
- Wannan Medical College, Wuhu, China
| | - Longmei Wang
- Department of Infectious Disease, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
- Wannan Medical College, Wuhu, China
| | - Kaiguang Zhang
- Department of Gastroenterology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Yuankun Chen
- Department of Infectious and Tropical Diseases, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Rui Liu
- Department of Infectious and Tropical Diseases, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Chuanlong Zhu
- Department of Infectious and Tropical Diseases, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
- Department of Infectious Disease, Jiangsu Provincial Hospital, Nanjing, China
| | - Wenting Li
- Department of Infectious and Tropical Diseases, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
36
|
Tang R, Luo J, Zhu X, Miao P, Tang H, Jian Y, Ruan S, Ling F, Tang M. Recent progress in the effect of ferroptosis of HSCs on the development of liver fibrosis. Front Mol Biosci 2023; 10:1258870. [PMID: 37860583 PMCID: PMC10584331 DOI: 10.3389/fmolb.2023.1258870] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/04/2023] [Indexed: 10/21/2023] Open
Abstract
Fibrosis is a common pathological process that must take place for multiple chronic liver diseases to develop into cirrhosis and liver cancer. Liver fibrosis (LF) is regulated by various cytokines and signaling pathways in its occurrence and development. Ferroptosis is an important mode of cell death caused by iron-dependent oxidative damage and is regulated by iron metabolism and lipid peroxidation signaling pathways. In recent years, numerous studies have shown that ferroptosis is closely related to LF. As the main material secreted by the extracellular matrix, hepatic stellate cells (HSCs) are a general concern in the development of LF. Therefore, targeting HSC ferroptosis against LF is crucial. This review describes the current status of treating LF by inducing HSC ferroptosis that would aid studies in better understanding the current knowledge on ferroptosis in HSCs and the future research direction in this field.
Collapse
Affiliation(s)
- Rui Tang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Luo
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaoxia Zhu
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Pengyu Miao
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Hong Tang
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yue Jian
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Sibei Ruan
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Feng Ling
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Mingxi Tang
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
37
|
Li Z, Lei Z, Cai Y, Cheng DB, Sun T. MicroRNA therapeutics and nucleic acid nano-delivery systems in bacterial infection: a review. J Mater Chem B 2023; 11:7804-7833. [PMID: 37539650 DOI: 10.1039/d3tb00694h] [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: 08/05/2023]
Abstract
Bacteria that have worked with humans for thousands of years pose a major threat to human health even today, as drug resistance has become a prominent problem. Compared to conventional drug therapy, nucleic acid-based therapies are a promising and potential therapeutic strategy for diseases in which nucleic acids are delivered through a nucleic acid delivery system to regulate gene expression in specific cells, offering the possibility of curing intractable diseases that are difficult to treat at this stage. Among the many nucleic acid therapeutic ideas, microRNA, a class of small nucleic acids with special properties, has made great strides in biology and medicine in just over two decades, showing promise in preclinical drug development. In this review, we introduce recent advances in nucleic acid delivery systems and their clinical applications, highlighting the potential of nucleic acid therapies, especially miRNAs extracted from traditional herbs, in combination with the existing set of nucleic acid therapeutic systems, to potentially open up a new line of thought in the treatment of cancer, viruses, and especially bacterial infectious diseases.
Collapse
Affiliation(s)
- Ze Li
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Yilun Cai
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| |
Collapse
|
38
|
Zhang MH, Yuan YF, Liu LJ, Wei YX, Yin WY, Zheng LZY, Tang YY, Lv Z, Zhu F. Dysregulated microRNAs as a biomarker for diagnosis and prognosis of hepatitis B virus-associated hepatocellular carcinoma. World J Gastroenterol 2023; 29:4706-4735. [PMID: 37664153 PMCID: PMC10473924 DOI: 10.3748/wjg.v29.i31.4706] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/29/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignancy with a high incidence and fatality rate worldwide. Hepatitis B virus (HBV) infection is one of the most important risk factors for its occurrence and development. Early detection of HBV-associated HCC (HBV-HCC) can improve clinical decision-making and patient outcomes. Biomarkers are extremely helpful, not only for early diagnosis, but also for the development of therapeutics. MicroRNAs (miRNAs), a subset of non-coding RNAs approximately 22 nucleotides in length, have increasingly attracted scientists' attention due to their potential utility as biomarkers for cancer detection and therapy. HBV profoundly impacts the expression of miRNAs potentially involved in the development of hepatocarcinogenesis. In this review, we summarize the current progress on the role of miRNAs in the diagnosis and treatment of HBV-HCC. From a molecular standpoint, we discuss the mechanism by which HBV regulates miRNAs and investigate the exact effect of miRNAs on the promotion of HCC. In the near future, miRNA-based diagnostic, prognostic, and therapeutic applications will make their way into the clinical routine.
Collapse
Affiliation(s)
- Ming-He Zhang
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Yu-Feng Yuan
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Li-Juan Liu
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Yu-Xin Wei
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Wan-Yue Yin
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Lan-Zhuo-Yin Zheng
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Ying-Ying Tang
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Zhao Lv
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Fan Zhu
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
- Hubei Province Key Laboratory of Allergy & Immunology, Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
39
|
Lang Z, Li Y, Lin L, Li X, Tao Q, Hu Y, Bao M, Zheng L, Yu Z, Zheng J. Hepatocyte-derived exosomal miR-146a-5p inhibits hepatic stellate cell EMT process: a crosstalk between hepatocytes and hepatic stellate cells. Cell Death Discov 2023; 9:304. [PMID: 37598186 PMCID: PMC10439924 DOI: 10.1038/s41420-023-01602-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/31/2023] [Accepted: 08/10/2023] [Indexed: 08/21/2023] Open
Abstract
Recently, Salidroside (Sal) has been demonstrated to suppress hepatic stellate cell (HSC) activation, a crucial event for liver fibrosis. Moreover, Sal has been reported to decrease hepatocyte injury. A growing number of reports have indicated that the crosstalk between hepatocytes and HSCs is very crucial for liver fibrosis development. Whether Sal-treated hepatocytes could inhibit HSC activation is unclear. Exosomes, as vital vehicles of intercellular communication, have been shown to transfer cargos between hepatocytes and HSCs. Herein, we aimed to investigate the roles of exosomal miRNAs from Sal-treated hepatocytes in HSC activation as well as liver fibrosis. Our results showed that Sal suppressed carbon tetrachloride (CCl4)-induced liver fibrosis in vivo. HSC activation as well as cell proliferation was repressed in HSCs co-cultured with Sal-treated hepatocytes. Interestingly, miR-146a-5p was up-regulated by Sal in CCl4-treated mice. Also, enhanced miR-146a-5p was found in hepatocytes isolated from Sal-treated CCl4 mice and hepatocyte-derived exosomes. Notably, hepatocyte exosomal miR-146a-5p contributed to HSC inactivation. Inhibiting miR-146a-5p in hepatocyte exosomes resulted in reduced E-cadherin (E-cad) and increased desmin in HSCs, indicating that miR-146a-5p caused HSC inactivation via epithelial-mesenchymal transition (EMT). miR-146a-5p inhibition-mediated HSC activation and EMT process were blocked down by loss of EIF5A2. Further studies revealed that EIF5A2 was a target of miR-146a-5p. Furthermore, exosomes with miR-146a-5p overexpression inhibited liver fibrosis in CCl4 mice. Collectively, exosomal miR-146a-5p from Sal-treated hepatocytes inhibits HSC activation and liver fibrosis, at least in part, by suppressing EIF5A2 and EMT process.
Collapse
Affiliation(s)
- Zhichao Lang
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yifei Li
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lifan Lin
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xinmiao Li
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Qiqi Tao
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yuhang Hu
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315000, China
| | - Menglu Bao
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lei Zheng
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhengping Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jianjian Zheng
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315000, China.
| |
Collapse
|
40
|
Chen J, Fu J, Zhao S, Zhang X, Chao Y, Pan Q, Sun H, Zhang J, Li B, Xue T, Li J, Liu C. Free Radical and Viral Infection: A Review from the Perspective of Ferroptosis. Vet Sci 2023; 10:456. [PMID: 37505861 PMCID: PMC10384322 DOI: 10.3390/vetsci10070456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/29/2023] Open
Abstract
Free radicals, including reactive oxygen species (ROS) and reactive nitrogen species (RNS), play critical roles in various physiological activities such as cell differentiation, apoptosis, and vascular tension when existing in cells at low levels. However, excessive amounts of free radicals are harmful, causing DNA damage, lipid peroxidation, protein degeneration, and abnormal cell death. Certain viral infections induce cells to produce excessive free radicals, which in multiple ways help the virus to replicate, mature, and exit. Iron is a necessary element for many intracellular enzymes, involved in both cellular activities and viral replication. Ferroptosis, a programmed cell death mode distinct from apoptosis, necrosis, and pyroptosis, is characterized by lipid peroxide accumulation and damage to the antioxidant system, affecting many cellular processes. Viral infection commonly manifests as decreased glutathione (GSH) content and down-regulated glutathione peroxidase 4 (GPX4) activity, similar to ferroptosis. Recent studies have suggested a possible relationship among free radicals, viral infections and ferroptosis. This review aims to elucidate the molecular mechanism linking free radicals and ferroptosis during viral infections and provide a new theoretical basis for studying viral pathogenesis and control.
Collapse
Affiliation(s)
- Jun Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Jinping Fu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing 210014, China
- Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Sha Zhao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing 210014, China
- Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Xiaoxi Zhang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing 210014, China
- Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Yuyang Chao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing 210014, China
- Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Qunxing Pan
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing 210014, China
- Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Huawei Sun
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing 210014, China
- Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Jingfeng Zhang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing 210014, China
- Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Bin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing 210014, China
- Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Tao Xue
- College of Medicine, Linyi University, Linyi 276000, China
| | - Jingui Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Chuanmin Liu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Nanjing 210014, China
- Key Laboratory of Veterinary Diagnosis, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- College of Medicine, Linyi University, Linyi 276000, China
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
41
|
Li T, Jiao J, Ke H, Ouyang W, Wang L, Pan J, Li X. Role of exosomes in the development of the immune microenvironment in hepatocellular carcinoma. Front Immunol 2023; 14:1200201. [PMID: 37457718 PMCID: PMC10339802 DOI: 10.3389/fimmu.2023.1200201] [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: 04/04/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Despite numerous improved treatment methods used in recent years, hepatocellular carcinoma (HCC) is still a disease with a high mortality rate. Many recent studies have shown that immunotherapy has great potential for cancer treatment. Exosomes play a significant role in negatively regulating the immune system in HCC. Understanding how these exosomes play a role in innate and adaptive immunity in HCC can significantly improve the immunotherapeutic effects on HCC. Further, engineered exosomes can deliver different drugs and RNA molecules to regulate the immune microenvironment of HCC by regulating the aforementioned immune pathway, thereby significantly improving the mortality rate of HCC. This study aimed to declare the role of exosomes in the development of the immune microenvironment in HCC and list engineered exosomes that could be used for clinical transformation therapy. These findings might be beneficial for clinical patients.
Collapse
Affiliation(s)
- Tanghua Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiapeng Jiao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Haoteng Ke
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wenshan Ouyang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Luobin Wang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jin Pan
- The Department of Electronic Engineering, The Chinese University of Hong Kong, Hongkong, Hongkong SAR, China
| | - Xin Li
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
42
|
Lv Y, Wu M, Wang Z, Wang J. Ferroptosis: From regulation of lipid peroxidation to the treatment of diseases. Cell Biol Toxicol 2023; 39:827-851. [PMID: 36459356 DOI: 10.1007/s10565-022-09778-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/11/2022] [Indexed: 12/04/2022]
Abstract
Ferroptosis is a regulated cell death mainly manifested by iron-dependent lipid peroxide accumulation. The leading cause of ferroptosis is the imbalance of intracellular oxidative systems (e.g., LOXs, POR, ROS) and antioxidant systems (e.g., GSH/GPx4, CoQ10/FSP1, BH4/GCH1), which is regulated by a complex network. In the past decade, this metabolic network has been continuously refined, and the links with various pathophysiological processes have been gradually established. Apoptosis has been regarded as the only form of regulated cell death for a long time, and the application of chemotherapeutic drugs to induce apoptosis of cancer cells is the mainstream method. However, studies have reported that cancer cells' key features are resistance to apoptosis and chemotherapeutics. For high proliferation, cancer cells often have very active lipid metabolism and iron metabolism, which pave the way for ferroptosis. Interestingly, researchers found that drug-resistant or highly aggressive cancer cells are more prone to ferroptosis. Therefore, ferroptosis may be a potential strategy to eliminate cancer cells. In addition, links between ferroptosis and other diseases, such as neurological disorders and ischemia-reperfusion injury, have also been found. Understanding these diseases from the perspective of ferroptosis may provide new insights into clinical treatment. Herein, the metabolic processes in ferroptosis are reviewed, and the potential mechanisms and targets of ferroptosis in different diseases are summarized.
Collapse
Affiliation(s)
- Yonghui Lv
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Meiying Wu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Zhe Wang
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| | - Junqing Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
43
|
Lu Y, Hu J, Chen L, Li S, Yuan M, Tian X, Cao P, Qiu Z. Ferroptosis as an emerging therapeutic target in liver diseases. Front Pharmacol 2023; 14:1196287. [PMID: 37256232 PMCID: PMC10225528 DOI: 10.3389/fphar.2023.1196287] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/02/2023] [Indexed: 06/01/2023] Open
Abstract
Ferroptosis is an iron-dependently nonapoptotic cell death characterized by excessive accumulation of lipid peroxides and cellular iron metabolism disturbances. Impaired iron homeostasis and dysregulation of metabolic pathways are contributors to ferroptosis. As a major metabolic hub, the liver synthesizes and transports plasma proteins and endogenous fatty acids. Also, it acts as the primary location of iron storage for hepcidin generation and secretion. To date, although the intricate correlation between ferroptosis and liver disorders needs to be better defined, there is no doubt that ferroptosis participates in the pathogenesis of liver diseases. Accordingly, pharmacological induction and inhibition of ferroptosis show significant potential for the treatment of hepatic disorders involved in lipid peroxidation. In this review, we outline the prominent features, molecular mechanisms, and modulatory networks of ferroptosis and its physiopathologic functions in the progression of liver diseases. Further, this review summarizes the underlying mechanisms by which ferroptosis inducers and inhibitors ameliorate liver diseases. It is noteworthy that natural active ingredients show efficacy in preclinical liver disease models by regulating ferroptosis. Finally, we analyze crucial concepts and urgent issues concerning ferroptosis as a novel therapeutic target in the diagnosis and therapy of liver diseases.
Collapse
Affiliation(s)
- Yuzhen Lu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Junjie Hu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Liang Chen
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Shan Li
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
- Department of Biochemistry, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Ming Yuan
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Xianxiang Tian
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Peng Cao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenpeng Qiu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
44
|
Chen Z, Ding W, Duan P, Lv X, Feng Y, Yin Z, Luo Z, Li Z, Zhang H, Zhou T, Tan H. HWJMSC-derived extracellular vesicles ameliorate IL-1β-induced chondrocyte injury through regulation of the BMP2/RUNX2 axis via up-regulation TFRC. Cell Signal 2023; 105:110604. [PMID: 36669606 DOI: 10.1016/j.cellsig.2023.110604] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/29/2022] [Accepted: 01/15/2023] [Indexed: 01/19/2023]
Abstract
Articular osteochondral injury is a common and frequently occurring disease in orthopedics that is caused by aging, disease, and trauma. The cytokine interleukin-1β (IL-1β) is a crucial mediator of the inflammatory response, which exacerbates damage during chronic disease and acute tissue injury. Human Wharton's jelly mesenchymal stem cell (HWJMSC) extracellular vesicles (HWJMSC-EVs) have been shown to promote cartilage regeneration. The study aimed to investigate the influence and mechanisms of HWJMSC-EVs on the viability, apoptosis, and cell cycle of IL-1β-induced chondrocytes. HWJMSC-EVs were isolated by Ribo™ Exosome Isolation Reagent kit. Nanoparticle tracking analysis was used to determine the size and concentration of HWJMSC-EVs. We characterized HWJMSC-EVs by western blot and transmission electron microscope. The differentiation, viability, and protein level of chondrocytes were measured by Alcian blue staining, Cell Counting Kit-8, and western blot, respectively. Flow cytometer was used to determine apoptosis and cell cycle of chondrocytes. The results showed that HWJMSCs relieved IL-1β-induced chondrocyte injury by inhibiting apoptosis and elevating viability and cell cycle of chondrocyte, which was reversed with exosome inhibitor (GW4869). HWJMSC-EVs were successfully extracted and proven to be uptake by chondrocytes. HWJMSC-EVs ameliorate IL-1β-induced chondrocyte injury by inhibiting cell apoptosis and elevating viability and cycle of cell, but these effects were effectively reversed by knockdown of transferrin receptor (TFRC). Notably, using bone morphogenetic protein 2 (BMP2) pathway agonist and inhibitor suggested that HWJMSC-EVs ameliorate IL-1β-induced chondrocyte injury through activating the BMP2 pathway via up-regulation TFRC. Furthermore, over-expression of runt-related transcription factor 2 (RUNX2) reversed the effects of BMP2 pathway inhibitor promotion of IL-1β-induced chondrocyte injury. These results suggested that HWJMSC-EVs ameliorate IL-1β-induced chondrocyte injury by regulating the BMP2/RUNX2 axis via up-regulation TFRC. HWJMSC-EVs may play a new insight for early medical interventions in patients with articular osteochondral injury.
Collapse
Affiliation(s)
- Zhian Chen
- Graduate School, Kunming Medical University, Kunming City, Yunnan Province, China
| | - Wei Ding
- College of Medicine Technology, Yunnan Medical Health College, Kunming City, Yunnan Province, China
| | - Peiya Duan
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming City, Yunnan Province, China
| | - Xiaoyu Lv
- Graduate School, Kunming Medical University, Kunming City, Yunnan Province, China
| | - Yujiao Feng
- Graduate School, Kunming Medical University, Kunming City, Yunnan Province, China
| | - Zhengbo Yin
- Graduate School, Kunming Medical University, Kunming City, Yunnan Province, China
| | - Zhihong Luo
- Department of Orthopaedics, People's Liberation Army Joint Logistic Support Force 920th Hospital, Kunming City, Yunnan Province, China
| | - Zhigui Li
- Department of Orthopaedics, People's Liberation Army Joint Logistic Support Force 920th Hospital, Kunming City, Yunnan Province, China
| | - Hua Zhang
- Department of Orthopaedics, People's Liberation Army Joint Logistic Support Force 920th Hospital, Kunming City, Yunnan Province, China
| | - Tianhua Zhou
- Department of Orthopaedics, People's Liberation Army Joint Logistic Support Force 920th Hospital, Kunming City, Yunnan Province, China.
| | - Hongbo Tan
- Department of Orthopaedics, People's Liberation Army Joint Logistic Support Force 920th Hospital, Kunming City, Yunnan Province, China.
| |
Collapse
|
45
|
Yin KL, Li M, Song PP, Duan YX, Ye WT, Tang W, Kokudo N, Gao Q, Liao R. Unraveling the Emerging Niche Role of Hepatic Stellate Cell-derived Exosomes in Liver Diseases. J Clin Transl Hepatol 2023; 11:441-451. [PMID: 36643031 PMCID: PMC9817040 DOI: 10.14218/jcth.2022.00326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 01/18/2023] Open
Abstract
Hepatic stellate cells (HSCs) play an essential role in various liver diseases, and exosomes are critical mediators of intercellular communication in local and distant microenvironments. Cellular crosstalk between HSCs and surrounding multiple tissue-resident cells promotes or inhibits the activation of HSCs. Substantial evidence has revealed that HSC-derived exosomes are involved in the occurrence and development of liver diseases through the regulation of retinoid metabolism, lipid metabolism, glucose metabolism, protein metabolism, and mitochondrial metabolism. HSC-derived exosomes are underpinned by vehicle molecules, such as mRNAs and microRNAs, that function in, and significantly affect, the processes of various liver diseases, such as acute liver injury, alcoholic liver disease, nonalcoholic fatty liver disease, viral hepatitis, fibrosis, and cancer. As such, numerous exosomes derived from HSCs or HSC-associated exosomes have attracted attention because of their biological roles and translational applications as potential targets for therapeutic targets. Herein, we review the pathophysiological and metabolic processes associated with HSC-derived exosomes, their roles in various liver diseases and their potential clinical application.
Collapse
Affiliation(s)
- Kun-Li Yin
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Li
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Pei-Pei Song
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Yu-Xin Duan
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wen-Tao Ye
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Tang
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Norihiro Kokudo
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Rui Liao
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
46
|
You H, Wang X, Ma L, Zhang F, Zhang H, Wang Y, Pan X, Zheng K, Kong F, Tang R. Insights into the impact of hepatitis B virus on hepatic stellate cell activation. Cell Commun Signal 2023; 21:70. [PMID: 37041599 PMCID: PMC10088164 DOI: 10.1186/s12964-023-01091-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/26/2023] [Indexed: 04/13/2023] Open
Abstract
During chronic hepatitis B virus (HBV) infection, hepatic fibrosis is a serious pathological condition caused by virus-induced liver damage. The activation of hepatic stellate cells (HSCs) is a central event in the occurrence and progression of liver fibrosis. Although accumulating evidence has shown that HBV directly stimulates HSC activation, whether the virus infects and replicates in HSCs remains controversial. Inflammation is one of the obvious characteristics of chronic HBV infection, and it has been demonstrated that persistent inflammation has a predominant role in triggering and maintaining liver fibrosis. In particular, the regulation of HSC activation by HBV-related hepatocytes via various inflammatory modulators, including TGF-β and CTGF, in a paracrine manner has been reported. In addition to these inflammation-related molecules, several inflammatory cells are essential for the progression of HBV-associated liver fibrosis. Monocytes, macrophages, Th17 cells, NK cells, as well as NKT cells, participate in the modulation of HBV-related liver fibrosis by interacting with HSCs. This review summarizes current findings on the effects of HBV and the relevant molecular mechanisms involved in HSC activation. Because HSC activation is essential for liver fibrosis, targeting HSCs is an attractive therapeutic strategy to prevent and reverse hepatic fibrosis induced by HBV infection. Video abstract.
Collapse
Affiliation(s)
- Hongjuan You
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xing Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lihong Ma
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fulong Zhang
- Imaging Department, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Huanyang Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuxin Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiucheng Pan
- Department of Infectious Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fanyun Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
47
|
Guo G, Yang W, Sun C, Wang X. Dissecting the potential role of ferroptosis in liver diseases: an updated review. Free Radic Res 2023; 57:282-293. [PMID: 37401821 DOI: 10.1080/10715762.2023.2232941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/05/2023]
Abstract
Ferroptosis is a novel form of cell death, manifested by iron-dependent, non-apoptotic manner resulting from the intracellular accumulation of large clusters of reactive oxygen species (ROS) and lipid peroxides due to abnormal iron metabolism. Since the liver is the main organ of human body for storing iron, it is essential to perform in-depth investigation on the role and mechanistic basis of ferroptosis in the context of divergent liver diseases. We previously summarized the emerging role of ferroptosis among various liver diseases, however, the past few years have been a surge in research establishing ferroptosis as the molecular basis or treatment option. This review article concentrated on the accumulating research progress of ferroptosis in a range of liver diseases such as acute liver injury/failure (ALI/ALF), immune-mediated hepatitis, alcoholic liver disease (ALD), nonalcoholic fatty liver disease and liver fibrosis. Ferroptosis may be a promising target for the prevention and treatment of various liver diseases, providing a strategy for exploring new therapeutic avenues for these entities.
Collapse
Affiliation(s)
- Gaoyue Guo
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Wanting Yang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao Sun
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
- Department of Gastroenterology, Tianjin Medical University General Hospital Airport Hospital, Tianjin, China
| | - Xiaoyu Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
48
|
Wang L, Zhou K, Wu Q, Zhu L, Hu Y, Yang X, Li D. Microanatomy of the metabolic associated fatty liver disease (MAFLD) by single-cell transcriptomics. J Drug Target 2023; 31:421-432. [PMID: 36847649 DOI: 10.1080/1061186x.2023.2185626] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
BACKGROUND Metabolic-associated fatty liver disease (MAFLD) is a major cause of liver disease worldwide and comprises non-alcoholic fatty liver (NAFL) and non-alcoholic steatohepatitis (NASH). Due to the high prevalence and poor prognosis of NASH, it is critical to identify and treat patients at risk. However, the aetiology and mechanisms remain largely unknown, warranting further analysis. METHODS We first identified differential genes in NASH by single-cell analysis of the GSE129516 dataset and conducted expression profiling data analysis of the GSE184019 dataset from the Gene Expression Omnibus (GEO) database. Then single-cell trajectory reconstruction and analysis, immune gene score, cellular communication, key gene screening, functional enrichment analysis, and immune microenvironment analysis were carried out. Finally, cell experiments were performed to verify the role of key genes in NASH. RESULTS We conducted transcriptome profiling of 30,038 single cells, including hepatocytes and non-hepatocytes from normal and steatosis adult mouse livers. Comparative analysis of hepatocytes and non-hepatocytes revealed pronounced heterogeneity as non-hepatocytes acted as major cell-communication hubs. The results showed that Hspa1b, Tfrc, Hmox1 and Map4k4 could effectively distinguish NASH tissues from normal samples. The results of scRNA-seq and qPCR indicated that the expression levels of hub genes in NASH were significantly higher than in normal cells or tissues. Further immune infiltration analysis showed significant differences in M2 macrophage distribution between healthy and metabolic-associated fatty liver samples. CONCLUSIONS Our results suggest that Hspa1b, Tfrc, Hmox1 and Map4k4 have huge prospects as diagnostic and prognostic biomarkers for NASH and may be potential therapeutic targets for NASH.
Collapse
Affiliation(s)
- Lijun Wang
- The Nanhua Affiliated Hospital, Department of Stomatology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Kebing Zhou
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Nanhua Affiliated Hospital, Department of General Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Nanhua Affiliated Hospital, Department of Gastroenterology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qing Wu
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Nanhua Affiliated Hospital, Department of General Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Lingping Zhu
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Nanhua Affiliated Hospital, Department of General Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yang Hu
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Nanhua Affiliated Hospital, Department of Gastroenterology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xuefeng Yang
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Nanhua Affiliated Hospital, Department of General Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Nanhua Affiliated Hospital, Department of Gastroenterology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Duo Li
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Nanhua Affiliated Hospital, Department of General Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
49
|
Xiang X, Gao J, Su D, Shi D. The advancements in targets for ferroptosis in liver diseases. Front Med (Lausanne) 2023; 10:1084479. [PMID: 36999078 PMCID: PMC10043409 DOI: 10.3389/fmed.2023.1084479] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
Ferroptosis is a type of regulated cell death caused by iron overload and lipid peroxidation, and its core is an imbalance of redox reactions. Recent studies showed that ferroptosis played a dual role in liver diseases, that was, as a therapeutic target and a pathogenic factor. Therefore, herein, we summarized the role of ferroptosis in liver diseases, reviewed the part of available targets, such as drugs, small molecules, and nanomaterials, that acted on ferroptosis in liver diseases, and discussed the current challenges and prospects.
Collapse
Affiliation(s)
- Xiaohong Xiang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Xiaohong Xiang
| | - Jianbo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danyang Su
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Doudou Shi
- Department of Geriatrics, The Ninth Hospital of Xi'an, Xi'an, Shaanxi, China
| |
Collapse
|
50
|
Huang X, Song Y, Wei L, Guo J, Xu W, Li M. The emerging roles of ferroptosis in organ fibrosis and its potential therapeutic effect. Int Immunopharmacol 2023; 116:109812. [PMID: 36746022 DOI: 10.1016/j.intimp.2023.109812] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/19/2022] [Accepted: 01/28/2023] [Indexed: 02/05/2023]
Abstract
Fibrosis refers to the process of excessive deposition of extracellular matrix (ECM) proteins, eventually leading to excessive scar formation. Fibrotic diseases can occur in many organs and result in high mortality. Currently, there is no effective treatment for fibrosis. As a new form of regulatory cell death (RCD), ferroptosis is mainly mediated by iron overload and lipid peroxidation. Emerging evidence shows that ferroptosis is involved in the pathogenesis of fibrotic diseases. Generally, ferroptosis of parenchymal cells exacerbates the progression of fibrosis, while ferroptosis of myofibroblasts may ameliorate it. Therefore, studying the mechanisms of ferroptosis in fibrosis and targeting ferroptosis in certain cells can provide valuable insights into the pathogenesis of fibrotic diseases. In the present review, we summarized the mechanisms and regulators of ferroptosis and then described the mechanism of fibrosis and the role of ferroptosis in fibrotic diseases, including liver fibrosis, renal fibrosis, pulmonary fibrosis, and myocardial fibrosis.
Collapse
Affiliation(s)
- Xuege Huang
- Institute of Biology and Medical Sciences, Soochow University, Building 703, 199 Ren-ai Road, Suzhou 215123, China
| | - Yahui Song
- Institute of Biology and Medical Sciences, Soochow University, Building 703, 199 Ren-ai Road, Suzhou 215123, China
| | - Lin Wei
- Institute of Biology and Medical Sciences, Soochow University, Building 703, 199 Ren-ai Road, Suzhou 215123, China
| | - Jing Guo
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Wei Xu
- Institute of Biology and Medical Sciences, Soochow University, Building 703, 199 Ren-ai Road, Suzhou 215123, China.
| | - Min Li
- Institute of Biology and Medical Sciences, Soochow University, Building 703, 199 Ren-ai Road, Suzhou 215123, China.
| |
Collapse
|