1
|
Mao Q, Luo Q, Ma SM, Teng M, Luo J. Critical role of ferroptosis in viral infection and host responses. Virology 2025; 606:110485. [PMID: 40086206 DOI: 10.1016/j.virol.2025.110485] [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: 11/19/2024] [Revised: 01/07/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Ferroptosis is an iron-dependent form of programmed cell death that plays a crucial role in regulating intracellular redox homeostasis and lipid metabolism, and in combating viral infections. Viruses have persistently evolved and adapted synergistically with their hosts over a long period and, to some extent, have been able to utilize ferroptosis to promote viral replication. Herein, we summarize the characteristics, mechanisms, and regulatory networks of ferroptosis and provide an overview of the key regulatory steps of ferroptosis involved in viral infection, together with the changes in host indicators and key regulatory signaling pathways. This study intends to deepen our understanding of the critical role of ferroptosis in viral infection, which will be meaningful for further revealing the mechanisms underlying the occurrence and progression of virus diseases, as well as for the future exploration of anti-viral strategies.
Collapse
Affiliation(s)
- Qian Mao
- Institute for Animal Health & UK-China Centre of Excellence for Research on Avian Diseases, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China; College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, People's Republic of China.
| | - Qin Luo
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, 450046, People's Republic of China
| | - Sheng-Min Ma
- Henan Joint International Research Laboratory of Veterinary Biologics Research and Application, Anyang Institute of Technology, Anyang, 455000, People's Republic of China
| | - Man Teng
- Institute for Animal Health & UK-China Centre of Excellence for Research on Avian Diseases, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China
| | - Jun Luo
- Institute for Animal Health & UK-China Centre of Excellence for Research on Avian Diseases, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China; Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, People's Republic of China.
| |
Collapse
|
2
|
Luo P, Tang Y, Chen N, Liu P, Wang J, Fan Y, Liu H, Wang K. USP21 is involved in the development of chronic hepatitis B by modulating the immune microenvironment. Eur J Med Res 2025; 30:259. [PMID: 40205504 PMCID: PMC11980114 DOI: 10.1186/s40001-025-02502-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
Hepatitis B virus (HBV) infection is a global public health challenge that alters the immune microenvironment of the liver and drives disease progression by triggering chronic inflammation that leads to hepatic cell death through multiple programmed cell death (PCD) modalities. Due to the persistence of covalently closed circular DNA in hepatocytes, there is a lack of curative drugs that can completely eradicate HBV. Therefore, revealing how HBV infection leads to changes in the hepatic immune microenvironment, as well as searching for specific molecular targets, is crucial for controlling the onset and progression of chronic hepatitis B (CHB). In this study, we used the single sample gene set enrichment analysis and CIBERSORT algorithms to assess immune cell infiltration in the livers of CHB patients. With three advanced machine learning algorithms, random forest, least absolute shrinkage and selection operator, and selected support vector machine recursive feature elimination, we identified the PCD signature genes associated with CHB from the candidate genes. We further validated that ubiquitin-specific peptidase 21 could differentiate CHB patients with different natural courses by receiver operating characteristic analysis. These findings enhance our understanding of the mechanisms of HBV infection.
Collapse
Affiliation(s)
- Pengyu Luo
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, Shandong, People's Republic of China
| | - Yuna Tang
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, Shandong, People's Republic of China
| | - Nan Chen
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, Shandong, People's Republic of China
| | - Pei Liu
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, Shandong, People's Republic of China
| | - Jing Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, Shandong, People's Republic of China
| | - Yuchen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, Shandong, People's Republic of China
- Hepatology Institute of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Huihui Liu
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, Shandong, People's Republic of China.
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012, Shandong, People's Republic of China.
- Hepatology Institute of Shandong University, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
3
|
Sun P, Liu Q, Yuan S, Wang XT, Qiu Y, Ge XY. SARS-CoV-2 Membrane Protein Induces MARCHF1/GPX4-Mediated Ferroptosis by Promoting Lipid Accumulation. J Med Virol 2025; 97:e70328. [PMID: 40186530 DOI: 10.1002/jmv.70328] [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: 10/15/2024] [Revised: 02/21/2025] [Accepted: 03/14/2025] [Indexed: 04/07/2025]
Abstract
The membrane protein (M), a key structural protein of SARS-CoV-2 that regulates virus assembly and morphogenesis, is involved in the pathological processes of multiple organ damage and metabolic disorders. This study aims to elucidate the mechanisms of M-mediated host ferroptosis and lipid accumulation during SARS-CoV-2 infection. Here, we detected that M protein enhances cellular sensitivity to ferroptosis. Additionally, we uncovered the pivotal role of perilipin-2 and sterol regulatory element-binding protein 1 in M-induced lipid accumulation. Xanthohumol, a cost-effective and orally available diacylglycerol acyltransferase inhibitor, alleviated triglyceride and total cholesterol accumulation, thereby counteracting the M-induced ferroptosis. Furthermore, we identified that the mitochondrial import inner membrane translocase subunit TIM23 and the mitochondrial import receptor subunit TOM20 homolog contribute to M-induced mitochondrial dysfunction. Notably, inhibiting lipid synthesis effectively reduced mitochondrial reactive oxygen species and transmembrane potential, indicating a cross-talk between lipid and ferro metabolic pathways. Mechanistically, glutathione peroxidase 4 (GPX4) interacts with SARS-CoV-2 M, leading to its subsequent degradation by the Membrane Associated Ring-CH-Type Finger 1 (MARCHF1) ubiquitin ligase. M-GPX4 interaction occurs at the R72 residue, which may represent a potential therapeutic target against SARS-CoV-2 infection. M modulates lipid accumulation and further impairs mitochondrial functions, ultimately resulting in ferroptosis through MARCHF1-GPX4 axis. Disrupting host-virus interactions along this pathway may provide a therapeutic strategy for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Pei Sun
- Department of Biomedical Engineering, Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Qian Liu
- Department of Biomedical Engineering, Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Shuofeng Yuan
- Department of Microbiology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xin-Tao Wang
- Department of Biomedical Engineering, Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Ye Qiu
- Department of Biomedical Engineering, Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Xing-Yi Ge
- Department of Biomedical Engineering, Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, Hunan, China
| |
Collapse
|
4
|
Li L, Wu T, Gong G, Li B, Feng J, Xu L, Zhao H, Gao X. NDRG1 alleviates Erastin-induced ferroptosis of hepatocellular carcinoma. BMC Cancer 2025; 25:522. [PMID: 40119318 PMCID: PMC11929176 DOI: 10.1186/s12885-025-13954-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: 07/17/2024] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND NDRG1, a cell differentiation-associated factor, has recently emerged as a regulator ferroptosis. Nevertheless, its role in modulating ferroptosis within hepatocellular carcinoma (HCC) remains uncharacterized. METHODS The differential expression of NDRG1 and its prognostic value were analyzed in HCC using data from TCGA and GEO. Ferroptosis in HepG2 and Huh7 cells was assessed using flow cytometry, transmission electron microscopy, and propidium iodide staining following NDRG1 knockdown using shRNA. RNA-seq was performed to characterize the mRNA expression profiles in HepG2 cells, identifying differentially expressed mRNAs (DE-mRNAs) and NDRG1-related hub genes. RESULTS NDRG1 was overexpressed in multiple malignant tumors, including HCC, and was associated with a significantly poor prognosis in HCC patients. A nomogram model integrating NDRG1 expression and clinical parameters demonstrated robust prognostic accuracy. NDRG1 knockdown potentiated erastin-induced alterations in Fe2+, total ROS, lipid ROS, and ferroptosis markers (PTGS2, ACSL4, GPX4, SLC7A11, GSH, GSSG), while exacerbating mitochondrial ultrastructural damage in HepG2 and Huh7 cells. Erastin induction elicited 1,056 DE-mRNAs, while subsequent NDRG1 knockdown revealed 1,323 DE-mRNAs in HepG2 cells. These DE-mRNAs are mainly involved in metastasis, immunity, growth, ferroptosis, and are associated with AMPK, MAPK, and PI3K/AKT pathways. Moreover, NDRG1 potentially interacted with HSPA8, CDH1, ALDOC, ANGPTL4, ANKRD37, CA9, ERBB3, FOS. qRT-PCR confirmed their expression changes consistent with RNA-seq. CONCLUSION NDRG1 exhibits strong predictive value for HCC, and accelerates tumor progression by suppressing ferroptosis.
Collapse
Affiliation(s)
- Liuzheng Li
- Hepatobiliary Surgery, The People's Hospital of Lincang, No.116 Nantang Street, Lincang, Yunnan, 677000, China
| | - Tong Wu
- Hepatobiliary Surgery, The People's Hospital of Lincang, No.116 Nantang Street, Lincang, Yunnan, 677000, China.
| | - Guocha Gong
- Hepatobiliary Surgery, The People's Hospital of Lincang, No.116 Nantang Street, Lincang, Yunnan, 677000, China
| | - Bo Li
- Hepatobiliary Surgery, The People's Hospital of Lincang, No.116 Nantang Street, Lincang, Yunnan, 677000, China
| | - Jiawei Feng
- Hepatobiliary Surgery, The People's Hospital of Lincang, No.116 Nantang Street, Lincang, Yunnan, 677000, China
| | - Leisheng Xu
- Hepatobiliary Surgery, The People's Hospital of Lincang, No.116 Nantang Street, Lincang, Yunnan, 677000, China
| | - Hairong Zhao
- Hepatobiliary Surgery, The People's Hospital of Lincang, No.116 Nantang Street, Lincang, Yunnan, 677000, China
| | - Xuechang Gao
- Hepatobiliary Surgery, The People's Hospital of Lincang, No.116 Nantang Street, Lincang, Yunnan, 677000, China
| |
Collapse
|
5
|
Sandilya V, El-Gameel D, Atashi M, Nguyen T, Fowowe M, Bhuiyan MMAA, Daramola O, Nwaiwu J, Hamdy NA, Ghanem M, El-Khordagui LK, Abdallah SM, El-Yazbi A, Mechref Y. LC-MS/MS-Profiling of Human Serum Unveils Significant Increase in Neuroinflammation and Carcinogenesis Following Chronic Organophosphate Exposure. J Proteome Res 2025; 24:1342-1355. [PMID: 39905624 DOI: 10.1021/acs.jproteome.4c00995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
The utilization of organophosphate pesticides (OPs) has escalated in response to the growing global food demand driven by a rapidly increasing population and the environmental disruptions caused by climate change. While acute exposure leads to cholinergic poisoning, chronic OP exposure has been linked to organ dysfunction, inflammation, and carcinogenesis. Serum samples from healthy individuals (n = 11), patients with acute OP exposure (n = 12), and those with chronic OP exposure (n = 31) were analyzed to discern the differentially expressed pathways after acute and chronic OP exposure. Differential expression analysis identified 132 proteins altered in chronic exposure vs control, 86 in acute exposure vs control, and 124 in chronic vs acute exposure. Pathway analysis revealed increased blood coagulation and reduced LXR/RXR activation and DCHR24 signaling in both acute and chronic exposures. Elevated levels of pro-inflammatory proteins, such as S100A8, VWF, and GPIBA, were observed, particularly in chronic exposure, highlighting significant inflammatory effects of OP exposure. These findings provide insights into the pathological mechanisms underlying chronic OP exposure and its contribution to inflammation and long-term health risks.
Collapse
Affiliation(s)
- Vishal Sandilya
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Dina El-Gameel
- Department of Pharmacy Practice, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mojgan Atashi
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Thu Nguyen
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Mojibola Fowowe
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | | | - Oluwatosin Daramola
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Judith Nwaiwu
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| | - Noha A Hamdy
- Department of Pharmacy Practice, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Maha Ghanem
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Labiba K El-Khordagui
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Salwa M Abdallah
- Center of Excellence for Toxicological Testing, Department of Mammalian and Aquatic Toxicology, Central Agricultural Pesticides Lab (CAPL), Agricultural Research Center (ARC), Giza, Egypt
| | - Ahmed El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria, Egypt
- Faculty of Pharmacy and Research & Innovation Hub, Alamein International University, Alamein, Egypt
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| |
Collapse
|
6
|
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
|
7
|
Han Z, Yu X, Wang C, Song X, Zhong X, Guo R, Yu W, Luo C. Circular RNA circHSPA8 Aggravates Metastasis by Acting as a Competitive Inhibitor of miR-195-5p to Upregulate WNT3A Expression in Breast Cancer. J Cell Mol Med 2025; 29:e70499. [PMID: 40099939 PMCID: PMC11915588 DOI: 10.1111/jcmm.70499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025] Open
Abstract
Circular RNA (circRNA) plays a vital role in the tumorigenicity and progression of cancer by regulating various biological behaviours. It acts as a microRNA sponge, disrupting transcription and the abnormal expression of oncogenes. Hsa_circ_0024715, a circRNA generated from cyclization at specific sites of the HSPA8 gene, has been found to be highly expressed in breast cancer (BC) tissue based on non-coding RNA high-throughput sequencing. However, its functions remain poorly understood. In this study, we performed qPCR to evaluate the expression of circHSPA8 in BC tissues. Survival analysis in a prospective cohort revealed that high expression of circHSPA8 is associated with poor prognosis and lymphoid node metastasis. Overexpression of circHSPA8 in MCF-7 cells significantly enhanced their proliferative and invasive abilities, whereas knockdown of circHSPA8 in MDA-MB-231 cells significantly reduced their proliferative and invasive abilities. We found that circHSPA8 can promote epithelial-mesenchymal transition (EMT) in BC cells, primarily by upregulating the expression of WNT3A. This process depends on the sponging and inhibition of miR-195-5p, which suppresses the proliferation, invasion, and metastasis of BC cells. In vivo experiments further confirmed that circHSPA8 can promote the intravasation and extravasation of BC cells as well as the formation of metastatic lesions in the lungs. In summary, these data demonstrate that circHSPA8 promotes EMT by acting as a competitive inhibitor of miR-195-5p to upregulate the expression of WNT3A in BC, suggesting that dysregulation of circRNA in BC might be a pathological factor and potential therapeutic target.
Collapse
Affiliation(s)
- Zhuoying Han
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, China
| | - Xiaojuan Yu
- Department of Clinical Oncology, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, China
| | - Chenlong Wang
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, China
| | - Xiaoyu Song
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, China
| | - Xiaomin Zhong
- Department of Clinical Oncology, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, China
| | - Renhua Guo
- Department of Clinical Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weiyong Yu
- Department of Clinical Oncology, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, China
| | - Chao Luo
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, China
- Biological Sample Bank, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, China
| |
Collapse
|
8
|
Liu R, Ye J, Wang J, Ma W, Qiu Z, Yu J, Wang W. Single-cell landscape of dynamic changes in CD8 + T cells, CD4 + T cells and exhausted T cells in hepatocellular carcinoma. Sci Rep 2025; 15:4130. [PMID: 39900964 PMCID: PMC11791069 DOI: 10.1038/s41598-025-88377-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/28/2025] [Indexed: 02/05/2025] Open
Abstract
Hepatocellular carcinoma has a high incidence and poor prognosis. In this study, we investigated the value of T-cell-related genes in prognosis by single-cell sequencing data in hepatocellular carcinoma. Twelve cases of hepatocellular carcinoma single-cell sequencing were included in the study. The high dimensional weighted gene co-expression network analysis (hdWGCNA) was used to identify gene modules associated with CD4+ T cells, CD8+ T cells and exhausted T cells. Altered signaling pathway activity in exhausted T cells was uncovered by the AUCell algorithm. xCELL, TIMER, QUANTISEQ, CIBERSORT and CIBERSORT-abs were performed to explore immune cell infiltration. Immune checkpoint inhibitor genes and TIDE methods were used to predict immunotherapy response. Finally, immunohistochemistry and real-time PCR were used to verify gene expression. The hdWGCNA algorithm identified 40 genes strongly associated with CD4+ T cells, CD8+ T cells and exhausted T cells. Seven genes were finally selected for transcriptome data modeling. The results of the three independent datasets suggested that the model had strong prognostic value. Model genes were critical factors influencing CD4+ T cell and CD8+ T cell infiltration in patients. The efficacy of PD-1 immunotherapy was higher in patients belonging to the low-risk group. Alterations in signaling pathways' activity within exhausted T cells were crucial factors contributing to the decline in immune function. Differential expression of seven genes in CD8+ T cells, CD4+ T cells and exhausted T cells were key targets for improving immunotherapy response in HCC.
Collapse
Affiliation(s)
- Rongqiang Liu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jing Ye
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jianguo Wang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wangbin Ma
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhendong Qiu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jia Yu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Weixing Wang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
9
|
Qian S, Tan G, Lei G, Zhang X, Xie Z. Programmed cell death in nasopharyngeal carcinoma: Mechanisms and therapeutic targets. Biochim Biophys Acta Rev Cancer 2025; 1880:189265. [PMID: 39809344 DOI: 10.1016/j.bbcan.2025.189265] [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: 09/19/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
Programmed cell death is a type of autonomic and orderly cell death mode controlled by genes that maintain homeostasis and growth. Tumor is a typical manifestation of an imbalance in environmental homeostasis in the human body. Currently, several tumor treatments are designed to trigger the death of tumor cells. Nasopharyngeal carcinoma is one of the most common malignant tumors in China. It displays obvious regional and ethnic differences in its incidence, being typically high in the south and low in the north of China. Nasopharyngeal carcinoma is currently considered to be a polygenic inherited disease and is often mediated by the interaction between multiple genes or between genes and the environment. Apoptosis has long been considered the key to tumor treatment, while other cell death pathways have often been overlooked. The current study provides an overview of the relationship among apoptosis, autophagy, pyroptosis, necroptosis, ferroptosis, and nasopharyngeal carcinoma, and the regulatory pathways of nasopharyngeal carcinoma based on five cell death modes were synthesized from the view of molecule. We hope this review will help explore additional, novel programmed cell death targets for the treatment of nasopharyngeal carcinoma and thus promote in-depth research.
Collapse
Affiliation(s)
- Shen'er Qian
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Otolaryngology Head and Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Guolin Tan
- Department of Otolaryngology Head and Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guang Lei
- Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School Of Medicine, Central South University, Changsha, Hunan, China
| | - Xiaowei Zhang
- Department of Otolaryngology Head and Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zuozhong Xie
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Laboratory of Otolaryngology Head and Neck Cancer, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Otology, Central South University, Changsha, Hunan, China.
| |
Collapse
|
10
|
Lin M, Mo Y, Li CM, Liu YZ, Feng XP. GRP78 as a potential therapeutic target in cancer treatment: an updated review of its role in chemoradiotherapy resistance of cancer cells. Med Oncol 2025; 42:49. [PMID: 39827214 DOI: 10.1007/s12032-024-02586-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025]
Abstract
GRP78 (Glucose-related protein 78, BiP/HSPA5) is commonly overexpressed in cancer cells. Acting as an activator of endoplasmic reticulum stress, GRP78 is involved in the resistance of cancer cells to injury. Current evidence suggests that GRP78 plays a significant role in the radiotherapy resistance and chemotherapy resistance of cancers, which is accomplished through a variety of complex pathways. These include the promotion of tumor stemness, inhibition of apoptosis, regulation of autophagy, maintenance of tumor microenvironment homeostasis, protection of dormant cells, evasion of senescence, counteraction of autoantibodies against GRP78, facilitation of DNA damage repair, suppression of ferroptosis, and modulation of metabolic reprogramming in tumor cells. Importantly, chemoradiotherapy resistance in cancers are the main reasons for treatment failure in patients, severely affecting their survival. Investigating the mechanisms of GRP78 in tumor therapeutic resistance is essential. In this article, we review the mechanisms by which GRP78 mediates cell survival and chemoradiotherapy resistance in cancers and provide an overview of clinical trials targeting GRP78 therapy.
Collapse
Affiliation(s)
- Min Lin
- Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yan Mo
- Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Cheng-Min Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Ying-Zhe Liu
- Xiangya International Medical Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Xue-Ping Feng
- Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
11
|
Xi C, Zhou J, Zheng X, Fu X, Xie M. Sodium aescinate-induced hepatotoxicity via ATF4/GSH/GPX4 axis-mediated ferroptosis. Sci Rep 2025; 15:1141. [PMID: 39774712 PMCID: PMC11706965 DOI: 10.1038/s41598-024-79723-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025] Open
Abstract
Sodium aescinate (SA), a natural plant extract with various bioactivities, is widely used to treat oedema and inflammation in clinics. However, adverse events, including liver injury, kidney injury, and phlebitis, have been reported in patients with SA in recent years. In this study, we used BALB/c mice and L02 cells to evaluate the role of ferroptosis in SA-induced liver injury. SA significantly increased AST, ALT, MDA and Fe2+, decreased GSH levels, and induced pathological changes in the liver in vivo. SA also reduced the viability of L02 cells and induced LDH release, intracellular cysteine reduction, GSH depletion, iron accumulation, ROS production, and lipid peroxidation, indicating that SA causes ferroptosis. In addition, SA inhibited transcriptional activity of activating transcription factor 4 (ATF4) and subsequently reduced the expression of the downstream genes xCT (solute carrier family 7a member 11, SLC7A11) and Cystathionine gamma-lyase (CTH) which play vital roles in GSH biosynthesis. Interestingly, the cytotoxic effects of SA were effectively attenuated by ATF4 overexpression, while they were significantly aggravated by ATF4 silencing. These results revealed that SA triggers hepatocyte ferroptosis by inhibiting the activity of ATF4, which causes an oxidative imbalance.
Collapse
Affiliation(s)
- Chen Xi
- Pharmaceutical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
| | - Jie Zhou
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China.
| | - Xin Zheng
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China
| | - Xiaoyi Fu
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China
| | - Minjuan Xie
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China
| |
Collapse
|
12
|
Fu J, Chen N, Qin T, Chen Y, Liu J, Wu H, Yan J, Xiao J, Zou J, Feng H. HSC70 functions as a negatively regulator in IFN signaling pathway via suppressing K63-linked ubiquitination of RIG-I in black carp. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105300. [PMID: 39647596 DOI: 10.1016/j.dci.2024.105300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
Heat shock cognate 70 (HSC70), a highly conserved molecular chaperone in the heat shock protein 70 (HSP70) family, plays an essential role in maintaining the homeostasis of the cellular environment. Furthermore, although previous studies have investigated potential function of HSC70 in innate antiviral immunity, further research is still required to fully elucidate its role. In this study, we cloned and characterized the HSC70 homolog gene from black carp (Mylopharyngodon piceus), which consists of 1950 nucleotides encoding 650 amino acids, migrates at approximately 71 kDa on SDS-PAGE, and is distributed in the cytoplasm. In response to different stimuli (SVCV, poly (I:C) and LPS), the transcription level of black carp HSC70 (bcHSC70) all increased to a certain extent. Luciferase reporter assay demonstrated that co-transfected bcHSC70 obviously reduced activity of interferon (IFN) promoters mediated by most factors in the RLRs pathway, and further qRT-PCR and plaque assay indicated that co-transfection of bcHSC70 with bcRIG-I decreased the bcRIG-I-mediated IFN transcription and antiviral ability resisting spring viremia of carp virus (SVCV), whereas knockdown of bcHSC70 improves the host cellular antiviral activity. Noteworthily, co-immunoprecipitation (co-IP) assay and immunofluorescence (IF) assay confirmed bcHSC70 interacts with bcRIG-I, and weaken K63-linked polyubiquitination of bcRIG-I. In summary, our study revealed that HSC70 negatively regulates IFN signaling pathway through impairing K63-linked ubiquitination of RIG-I in black carp, which provides an important basis for exploring innate immune regulatory mechanisms in teleost fish.
Collapse
Affiliation(s)
- Jiaxin Fu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Nianfeng Chen
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Tian Qin
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yixin Chen
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Ji Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Hui Wu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jun Yan
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China.
| | - Jun Xiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China.
| | - Jun Zou
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Hao Feng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| |
Collapse
|
13
|
Liao H, Liu S, Ma Q, Huang H, Goel A, Torabian P, Mohan CD, Duan C. Endoplasmic reticulum stress induced autophagy in cancer and its potential interactions with apoptosis and ferroptosis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119869. [PMID: 39490702 DOI: 10.1016/j.bbamcr.2024.119869] [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: 07/19/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
The endoplasmic reticulum (ER) is a dynamic organelle that is a site of the synthesis of proteins and lipids, contributing to the regulation of proteostasis, lipid metabolism, redox balance, and calcium storage/-dependent signaling events. The disruption of ER homeostasis due to the accumulation of misfolded proteins in the ER causes ER stress which activates the unfolded protein response (UPR) system through the activation of IRE1, PERK, and ATF6. Activation of UPR is observed in various cancers and therefore, its association with process of carcinogenesis has been of importance. Tumor cells effectively utilize the UPR system to overcome ER stress. Moreover, ER stress and autophagy are the stress response mechanisms operating together to maintain cellular homeostasis. In human cancers, ER stress-driven autophagy can function as either pro-survival or pro-death in a context-dependent manner. ER stress-mediated autophagy can have crosstalk with other types of cell death pathways including apoptosis and ferroptosis. In this connection, the present review has evaluated the role of ER stress in the regulation of autophagy-mediated tumorigenesis and its interactions with other cell death mechanisms such as apoptosis and ferroptosis. We have also comprehensively discussed the effect of ER stress-mediated autophagy on cancer progression and chemotherapeutic resistance.
Collapse
Affiliation(s)
- Haitang Liao
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China; Department of Intensive Care Unit, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, China
| | - Shuang Liu
- Department of Ultrasound, Chongqing Health Center for Women and Children/Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Qiang Ma
- Department of Oncology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - He Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Arul Goel
- University of California Santa Barbara, Santa Barbara, CA, USA
| | - Pedram Torabian
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Medical Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Chakrabhavi Dhananjaya Mohan
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Chenyang Duan
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
14
|
Gao H, Gu T, Gao X, Song Z, Liu J, Song Y, Zhang G, Sun Y. African swine fever virus enhances viral replication by increasing intracellular reduced glutathione levels, which suppresses stress granule formation. Vet Res 2024; 55:172. [PMID: 39707514 DOI: 10.1186/s13567-024-01433-4] [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: 06/15/2024] [Accepted: 09/27/2024] [Indexed: 12/23/2024] Open
Abstract
African swine fever virus (ASFV) is a DNA virus that has significantly impacted the global swine industry. Currently, there are no effective therapies or vaccines against ASFV. Stress granules (SGs), known for their antiviral properties, are not induced during ASFV infection, even though reactive oxygen species (ROS) are generated. The mechanism by which ASFV regulates SGs formation remains unclear. This study demonstrates that ASFV antagonises SGs formation and increases intracellular levels of reduced glutathione (GSH) levels. The use of the GSH inhibitor BSO and the activator NAC confirmed that the ASFV-induced increase in GSH helps to suppress SGs formation and influences viral replication. Additionally, this study revealed that ASFV enhances GSH by upregulating the antioxidant transcription factor NRF2, as well as factors involved in GSH synthesis and regeneration, such as GCLC, and those related to the ferroptosis pathway, such as SLC7A11. Furthermore, the study uncovered that ASFV manipulates intracellular GSH levels by activating the mitochondrial protein AIFM1. This regulatory mechanism helps the virus inhibit the formation of intracellular SGs, thereby creating an optimal environment for viral replication. These findings provide new insights into the molecular strategies employed by ASFV.
Collapse
Affiliation(s)
- Han Gao
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
- School of Animal Science and Technology, Foshan University, Foshan, 528225, China
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Animal Science and Technology, Foshan University, Foshan, 528225, China
| | - Taoming Gu
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
| | - Xiaopeng Gao
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
| | - Zebu Song
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
| | - Jing Liu
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
| | - Yi Song
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
| | - Guihong Zhang
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China.
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China.
| | - Yankuo Sun
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China.
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China.
| |
Collapse
|
15
|
Lyu W, Qin H, Li Q, Lu D, Shi C, Zhao K, Zhang S, Yu R, Zhang H, Zhou X, Xia S, Zhang L, Wang X, Chi X, Liu Z. Novel mechanistic insights - A brand new Era for anti-HBV drugs. Eur J Med Chem 2024; 279:116854. [PMID: 39276582 DOI: 10.1016/j.ejmech.2024.116854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024]
Abstract
Hepatitis B Virus (HBV) remains a critical global health issue, with substantial morbidity and mortality. Current therapies, including interferons and nucleoside analogs, often fail to achieve complete cure or functional eradication. This review explores recent advances in anti-HBV agents, focusing on their innovative mechanisms of action. HBV entry inhibitors target the sodium taurocholate cotransporting polypeptide (NTCP) receptor, impeding viral entry, while nucleus translocation inhibitors disrupt key viral life cycle steps, preventing replication. Capsid assembly modulators inhibit covalently closed circular DNA (cccDNA) formation, aiming to eradicate the persistent viral reservoir. Transcription inhibitors targeting cccDNA and integrated DNA offer significant potential to suppress HBV replication. Immunomodulatory agents are highlighted for their ability to enhance host immune responses, facil-itating better control and possible eradication of HBV. These novel approaches represent significant advancements in HBV therapy, providing new strategies to overcome current treatment limitations. The development of cccDNA reducers is particularly critical, as they directly target the persistent viral reservoir, offering a promising pathway towards achieving a functional cure or complete viral eradication. Continued research in this area is essential to advance the effectiveness of anti-HBV therapies.
Collapse
Affiliation(s)
- Weiping Lyu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Haoming Qin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Qi Li
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Dehua Lu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Cheng Shi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Kangchen Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Shengran Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Ruohan Yu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Huiying Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Xiaonan Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Sitian Xia
- Beijing National Day School, Beijing, 100089, PR China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Xiaoqian Wang
- Beijing Tide Pharmaceutical Co., Ltd, No.8 East Rongjing Street, Beijing Economic-Technological Development Area (BDA), Beijing, 100176, PR China.
| | - Xiaowei Chi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
16
|
Liu X, Cai Y, Zhang Y, Zhang H, Tian S, Gong Y, Song Q, Chen X, Ma X, Wen Y, Chen Y, Zeng J. Artesunate: A potential drug for the prevention and treatment from hepatitis to hepatocellular carcinoma. Pharmacol Res 2024; 210:107526. [PMID: 39617278 DOI: 10.1016/j.phrs.2024.107526] [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: 09/25/2024] [Revised: 11/14/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Liver cancer represents a multifactorial, multistage, and intricately progressive malignancy. Over the past decade, artesunate (ART), initially renowned for its anti-malarial efficacy, has been the focus of over 3000 studies uncovering its diverse pharmacological actions, including anti-inflammatory, immunoregulatory, metabolic regulatory, anti-fibrotic, and anti-cancer properties. This review highlights ART's role in the multistep progression from hepatitis to cancer and its underlying regulatory mechanisms, revealing signal transducer and activator of transcription 3 (STAT3) and ferroptosis (a novel form of programmed cell death) as promising therapeutic targets. ART demonstrates efficacy in inhibiting hepatitis virus infections, modulating inflammation, and facilitating recovery from inflammatory processes. During stages of hepatic fibrosis or cirrhosis, ART reverses fibrotic and cirrhotic changes by suppressing hepatic stellate cell activity, regulating inflammatory pathways, inhibiting hematopoietic stem cell proliferation, and inducing ferroptosis. Additionally, ART hinders hepatocellular carcinoma (HCC) cell proliferation, invasion, and metastasis, induces apoptosis and autophagy, combats drug resistance, and enhances chemosensitivity. Collectively, ART exhibits multi-step actions across multiple targets and signaling pathways, highlighting its potential as a clinical candidate for the prevention and treatment of liver cancer, from hepatitis and hepatic fibrosis to advanced HCC.
Collapse
Affiliation(s)
- Xinyue Liu
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China; Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yilin Cai
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China; Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yuanhao Zhang
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China
| | - Hetian Zhang
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China
| | - Sisi Tian
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China; Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yuxia Gong
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China; Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Qinmei Song
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China; Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Xiaotong Chen
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yu Chen
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| |
Collapse
|
17
|
Zhong C, Dong H, Ma Y, Zhuang B, Shi H, Hong L. Single-cell sequencing combined with transcriptomics and in vivo and in vitro analysis reveals the landscape of ferroptosis in myocardial ischemia-reperfusion injury. FASEB J 2024; 38:e70164. [PMID: 39520298 DOI: 10.1096/fj.202401056r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/21/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a significant risk factor for acute myocardial infarction and is closely associated with ferroptosis. This study aimed to identify key ferroptosis-related genes as potential diagnostic markers for MIRI and to explore their roles in immune infiltration and therapeutic targeting in myocardial tissue. We obtained single-cell RNA sequencing (scRNA-seq) and RNA-seq data on MIRI from the GEO database, applied Seurat and UMAP for data processing and clustering, and analyzed ligand-receptor interactions using CellPhoneDB. By scoring ferroptosis in cardiomyocytes, we identified differentially expressed genes and conducted GO and KEGG pathway analyses. A protein interaction network was then constructed using the STRING database, and seven key genes (Atp5h, Vdac2, Pkm, Cycs, Hspa8, Tpi1, Ldha) were identified through Lasso regression modeling, showing significant associations with immune responses. In vivo experiments in a mouse ischemia-reperfusion model confirmed the roles of these seven genes in MIRI via RT-qPCR. To further investigate the role of Hspa8 in ferroptosis and MIRI, siRNA knockdown experiments were performed in H9C2 rat cardiomyocytes, and its involvement in ferroptosis was validated by JC-1 staining and PCR analysis. This study reveals the importance of ferroptosis-related genes in MIRI through integrated bioinformatics and experimental approaches, offering new insights into diagnostic markers and immune-related therapeutic targets for MIRI.
Collapse
Affiliation(s)
- Chongning Zhong
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Hui Dong
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Yuting Ma
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Bingqi Zhuang
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Hongyang Shi
- Department of Food and Biological Sciences, School of Agriculture, Yanbian University, Yanji, China
| | - Lan Hong
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| |
Collapse
|
18
|
You Y, Qian Z, Jiang Y, Chen L, Wu D, Liu L, Zhang F, Ning X, Zhang Y, Xiao J. Insights into the pathogenesis of gestational and hepatic diseases: the impact of ferroptosis. Front Cell Dev Biol 2024; 12:1482838. [PMID: 39600338 PMCID: PMC11588751 DOI: 10.3389/fcell.2024.1482838] [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: 08/20/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Ferroptosis, a distinct form of non-apoptotic cell death characterized by iron dependency and lipid peroxidation, is increasingly linked to various pathological conditions in pregnancy and liver diseases. It plays a critical role throughout pregnancy, influencing processes such as embryogenesis, implantation, and the maintenance of gestation. A growing body of evidence indicates that disruptions in these processes can precipitate pregnancy-related disorders, including pre-eclampsia (PE), gestational diabetes mellitus (GDM), and intrahepatic cholestasis of pregnancy (ICP). Notably, while ICP is primarily associated with elevated maternal serum bile acid levels, its precise etiology remains elusive. Oxidative stress induced by bile acid accumulation is believed to be a significant factor in ICP pathogenesis. Similarly, the liver's susceptibility to oxidative damage underscores the importance of lipid metabolism dysregulation and impaired iron homeostasis in the progression of liver diseases such as alcoholic liver disease (ALD), non-alcoholic fatty liver disease (NAFLD), cholestatic liver injury, autoimmune hepatitis (AIH), acute liver injury, viral hepatitis, liver fibrosis, and hepatocellular carcinoma (HCC). This review discusses the shared signaling mechanisms of ferroptosis in gestational and hepatic diseases, and explores recent advances in understanding the mechanisms of ferroptosis and its potential role in the pathogenesis of gestational and hepatic disorders, with the aim of identifying viable therapeutic targets.
Collapse
Affiliation(s)
- Yilan You
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Zhiwen Qian
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Ying Jiang
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Lingyan Chen
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Danping Wu
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Lu Liu
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Feng Zhang
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Xin Ning
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Yan Zhang
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Jianping Xiao
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| |
Collapse
|
19
|
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
|
20
|
Wu M, Yan J, Qin S, Fu L, Sun S, Li W, Lv J, Chen L. Connections Between Endoplasmic Reticulum Stress and Prognosis of Hepatocarcinoma. Bioengineering (Basel) 2024; 11:1136. [PMID: 39593796 PMCID: PMC11591847 DOI: 10.3390/bioengineering11111136] [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: 09/26/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
Endoplasmic reticulum (ER) stress is a state in which misfolded or unfolded proteins accumulate in the lumen of the ER as a result of some exogenous or endogenous factors. It plays a crucial role in the pathogenesis of malignancies, affecting cell survival, proliferation, and metastasis in cancer. ER stress genes could provide new ideas for potential therapeutic targets in cancer. In our study, we aimed to construct an ER stress-related genes (ERGs) model for hepatocellular carcinoma (HCC). ERGs with differential expression and significant survival were screened to construct a prognostic model. The effectiveness of the model was successfully validated by external datasets. High and low-risk groups were classified based on risk scores. Functional analysis showed risk groups involved in the unfolded protein response, DNA repair, and other differential pathways. When compared to patients with low risk, the prognosis for HCC patients in the high-risk group might be worsened by disruptions in these pathways. Importantly, we considered genomic druggability and predicted drugs. Sorafenib-induced autophagy in HCC cells through an ES stress mechanism. Sorafenib was more sensitive for high-risk patients. In brief, our model predicted the prognosis of HCC and provided novel treatment strategies for the study of other cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Junjie Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (M.W.); (J.Y.); (S.Q.); (L.F.); (S.S.); (W.L.)
| | - Lina Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (M.W.); (J.Y.); (S.Q.); (L.F.); (S.S.); (W.L.)
| |
Collapse
|
21
|
Zeng C, Nie D, Wang X, Zhong S, Zeng X, Liu X, Qiu K, Peng X, Zhang W, Chen S, Zha X, Chen C, Chen Z, Wang W, Li Y. Combined targeting of GPX4 and BCR-ABL tyrosine kinase selectively compromises BCR-ABL+ leukemia stem cells. Mol Cancer 2024; 23:240. [PMID: 39465372 PMCID: PMC11514791 DOI: 10.1186/s12943-024-02162-0] [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: 06/20/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND In the ongoing battle against BCR-ABL+ leukemia, despite significant advances with tyrosine kinase inhibitors (TKIs), the persistent challenges of drug resistance and the enduring presence of leukemic stem cells (LSCs) remain formidable barriers to achieving a cure. METHODS In this study, we demonstrated that Disulfiram (DSF) induces ferroptosis to synergize with TKIs in inhibiting BCR-ABL+ cells, particularly targeting resistant cells and LSCs, using cell models, mouse models, and primary cells from patients. We elucidated the mechanism by which DSF promotes GPX4 degradation to induce ferroptosis through immunofluorescence, co-immunoprecipitation (CO-IP), RNA sequencing, lipid peroxidation assays, and rescue experiments. RESULTS Here, we present compelling evidence elucidating the sensitivity of DSF, an USA FDA-approved drug for alcohol dependence, towards BCR-ABL+ cells. Our findings underscore DSF's ability to selectively induce a potent cytotoxic effect on BCR-ABL+ cell lines and effectively inhibit primary BCR-ABL+ leukemia cells. Crucially, the combined treatment of DSF with TKIs selectively eradicates TKI-insensitive stem cells and resistant cells. Of particular note is DSF's capacity to disrupt GPX4 stability, elevate the labile iron pool, and intensify lipid peroxidation, ultimately leading to ferroptotic cell death. Our investigation shows that BCR-ABL expression induces alterations in cellular iron metabolism and increases GPX4 expression. Additionally, we demonstrate the indispensability of GPX4 for LSC development and the initiation/maintenance of BCR-ABL+ leukemia. Mechanical analysis further elucidates DSF's capacity to overcome resistance by reducing GPX4 levels through the disruption of its binding with HSPA8, thereby promoting STUB1-mediated GPX4 ubiquitination and subsequent proteasomal degradation. Furthermore, the combined treatment of DSF with TKIs effectively targets both BCR-ABL+ blast cells and drug-insensitive LSCs, conferring a significant survival advantage in mouse models. CONCLUSION In summary, the dual inhibition of GPX4 and BCR-ABL presents a promising therapeutic strategy to synergistically target blast cells and drug-insensitive LSCs in patients, offering potential avenues for advancing leukemia treatment.
Collapse
MESH Headings
- Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
- Phospholipid Hydroperoxide Glutathione Peroxidase/genetics
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/pathology
- Humans
- Animals
- Mice
- Protein Kinase Inhibitors/pharmacology
- Disulfiram/pharmacology
- Ferroptosis/drug effects
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Chengwu Zeng
- Guangzhou First People's Hospital, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510700, China.
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Dingrui Nie
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Department of Hematology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xianfeng Wang
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shuxin Zhong
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiangbo Zeng
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xin Liu
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Kangjie Qiu
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xueting Peng
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Wenyi Zhang
- Guangdong Key Laboratory of Bioactive Drug Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Shengting Chen
- Department of Hematology, Department of clinical laboratory, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Xianfeng Zha
- Department of Hematology, Department of clinical laboratory, First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Cunte Chen
- Guangzhou First People's Hospital, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510700, China.
| | - Zhenhua Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, 91016, USA.
| | - Weizhang Wang
- Guangdong Key Laboratory of Bioactive Drug Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Yangqiu Li
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
22
|
Murphy H, Huang Q, Jensen J, Weber N, Mendonça L, Ly H, Liang Y. Characterization of bi-segmented and tri-segmented recombinant Pichinde virus particles. J Virol 2024; 98:e0079924. [PMID: 39264155 PMCID: PMC11494906 DOI: 10.1128/jvi.00799-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024] Open
Abstract
Mammarenaviruses include several highly virulent pathogens (e.g., Lassa virus) capable of causing severe hemorrhagic fever diseases for which there are no approved vaccines and limited treatment options. Mammarenaviruses are enveloped, bi-segmented ambisense RNA viruses. There is limited knowledge about cellular proteins incorporated into progeny virion particles and their potential biological roles in viral infection. Pichinde virus (PICV) is a prototypic arenavirus used to characterize mammarenavirus replication and pathogenesis. We have developed a recombinant PICV with a tri-segmented RNA genome as a viral vector platform. Whether the tri-segmented virion differs from the wild-type bi-segmented one in viral particle morphology and protein composition has not been addressed. In this study, recombinant PICV (rPICV) virions with a bi-segmented (rP18bi) and a tri-segmented (rP18tri) genome were purified by density-gradient ultracentrifugation and analyzed by cryo-electron microscopy and mass spectrometry. Both virion types are pleomorphic with spherical morphology and have no significant difference in size despite rP18tri having denser particles. Both virion types also contain similar sets of cellular proteins. Among the highly enriched virion-associated cellular proteins are components of the endosomal sorting complex required for transport pathway and vesicle trafficking, such as ALIX, Tsg101, VPS, CHMP, and Ras-associated binding proteins, which have known functions in virus assembly and budding. Other enriched cellular proteins include peripheral and transmembrane proteins, chaperone proteins, and ribosomal proteins; their biological roles in viral infection warrant further analysis. Our study provides important insights into mammarenavirus particle formation and aids in the future development of viral vectors and antiviral discovery.IMPORTANCEMammarenaviruses, such as Lassa virus, are enveloped RNA viruses that can cause severe hemorrhagic fever diseases (Lassa fever) with no approved vaccine and limited therapeutic options. Cellular proteins incorporated into progeny virion particles and their biological roles in mammarenavirus infection have not been well characterized. Pichinde virus (PICV) is a prototypic mammarenavirus used as a surrogate model for Lassa fever. We used cryo-electron microscopy and proteomic analysis to characterize the morphology and protein contents of the purified PICV particles that package either two (bi-segmented) or three (tri-segmented) genomic RNA segments. Our results demonstrate a similar virion morphology but different particle density for the bi- and tri-segmented viral particles and reveal major virion-associated cellular proteins. This study provides important insights into the virus-host interactions that can be used for antiviral development and optimizing arenavirus-based vaccine vectors.
Collapse
Affiliation(s)
- Hannah Murphy
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Qinfeng Huang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Jacob Jensen
- Department of Biochemistry, Molecular Biology and Biophysics, College of Biological Sciences, University of Minnesota, Twin Cities, Minneapolis, Minnesota, USA
| | - Noah Weber
- Department of Biochemistry, Molecular Biology and Biophysics, College of Biological Sciences, University of Minnesota, Twin Cities, Minneapolis, Minnesota, USA
| | - Luiza Mendonça
- Department of Biochemistry, Molecular Biology and Biophysics, College of Biological Sciences, University of Minnesota, Twin Cities, Minneapolis, Minnesota, USA
| | - Hinh Ly
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Yuying Liang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| |
Collapse
|
23
|
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
|
24
|
Li Y, Wang F, Geng Z, He T, Song Y, Wu J, Wang B. HBx promotes tumorigenicity through RRM2-mediated autophagy in hepatocellular carcinoma. Cell Biosci 2024; 14:116. [PMID: 39256879 PMCID: PMC11389268 DOI: 10.1186/s13578-024-01298-2] [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: 12/01/2023] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) infection can exacerbate liver disease progression through multiple mechanisms, eventually leading to hepatocellular carcinoma (HCC). HBV-encoded oncogene X protein (HBx), a key regulatory protein of HBV infection, serves as a positive regulator of hepatocarcinogenesis. The indispensability of the M2 subunit of ribonucleotide-diphosphate reductase (RRM2) lies in its role in facilitating DNA replication and repair processes. In our previous investigation, it was postulated that the gene RRM2 exhibits elevated expression levels in several categories of malignant tumors, particularly in HBV-related HCC. Additionally, it was observed that RRM2 is present within protein complexes that are centered on HBx. In the present investigation, the objective of this work was to investigate the potential relationship between the elevated expression of RRM2 in HBV-related HCC and the influence of HBx on this expression. The study attempted to determine the specific mechanism by which RRM2 is implicated in the promotion of hepatocarcinogenesis by HBx. There have been multiple scholarly proposals suggesting that the induction of autophagy by HBx is a significant intermediary factor in the development of HCC. However, the precise carcinogenic function of HBx-induced autophagy remains a subject of debate. RESULTS This work initially investigated the impact of suppressing cellular autophagy on the malignant biological behaviors of HBx-promoted cells using an in vitro cellular model. The findings revealed that the suppression of cellular autophagy partially disrupted the oncogenic effects of HBx. In light of this, we proceeded to conduct more investigations into the regulatory association between RRM2 and HBx-induced autophagy in the upstream-downstream context. Our data indicate that HBx proteins increase the expression of RRM2. Suppression of RRM2 expression not only hinders HBx-induced autophagy, but also worsens the cellular G1/S blockage and reduces the HBx-induced malignant growth of hepatocellular carcinoma tumors, while stimulating apoptosis. CONCLUSIONS Therefore, we hypothesised that RRM2 is a potential downstream target of HBx-induced hepatocarcinogenesis, and mining the oncogenic mechanism of RRM2 is significant in exploring the preventive treatment of HBV-related HCC.
Collapse
Affiliation(s)
- Yaqun Li
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Department of Pharmacy, Department of Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Furan Wang
- Pfizer Research China, Shanghai, 200000, China
| | - Zikai Geng
- Pharmacy School, Binzhou Medical University, Yantai, Shandong Province, 264003, China
| | - Tianye He
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yun Song
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jian Wu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Bin Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
25
|
Zeng K, Huang N, Liu N, Deng X, Mu Y, Zhang X, Zhang J, Zhang C, Li Y, Li Z. LACTB suppresses liver cancer progression through regulation of ferroptosis. Redox Biol 2024; 75:103270. [PMID: 39047638 PMCID: PMC11321384 DOI: 10.1016/j.redox.2024.103270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/18/2024] [Accepted: 07/14/2024] [Indexed: 07/27/2024] Open
Abstract
Ferroptosis, driven by iron-dependent phospholipid peroxidation, is emerging as an intrinsic cancer defense mechanism. However, the regulatory networks involved in ferroptosis remain largely unknown. Here, we found that serine beta-lactamase-like protein (LACTB) inhibits liver cancer progression by regulating ferroptosis. LACTB is downregulated in liver cancer, and the ectopic expression of LACTB markedly inhibits cell viability, colony formation, and tumour growth. LACTB knockout exerts the opposite effects. Further investigation revealed that LACTB blocks HSPA8 transcription in a p53-dependent manner, resulting in the elevation of NCOA4-mediated ferritinophagy and inhibition of SLC7A11/GSH/GPX4 signalling, thereby triggering ferroptosis and suppressing liver cancer progression. Liver cancer cells with an endogenous mutation of p53 binding site in the HSPA8 promoter exhibited increased resistance to ferroptosis inducers, and the ferroptosis-promoting effect of LACTB was significantly weakened in these mutant cells. Importantly, LACTB is identified as a downstream target of lenvatinib, and adeno-associated virus-mediated overexpression and knockdown of LACTB notably enhance and attenuate the anti-tumour efficacy of lenvatinib in vivo, respectively. Taken together, our study reveals a novel action of LACTB and provides potential therapeutic strategies for enhancing the efficacy of lenvatinib in liver cancer.
Collapse
Affiliation(s)
- Kaixuan Zeng
- Department of Geriatric Hepatobiliary, Pancreatic and Spleen Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; Tumor and Immunology Center of Precision Medicine Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Na Huang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; Tumor and Immunology Center of Precision Medicine Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; Shaanxi International Cooperation Base for Inflammation and Immunity, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Nanbin Liu
- Department of Geriatric Hepatobiliary, Pancreatic and Spleen Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Xi Deng
- Department of Geriatric Hepatobiliary, Pancreatic and Spleen Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Yanhua Mu
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; Tumor and Immunology Center of Precision Medicine Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Xurui Zhang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; Tumor and Immunology Center of Precision Medicine Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jian Zhang
- Department of Geriatric Hepatobiliary, Pancreatic and Spleen Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; Tumor and Immunology Center of Precision Medicine Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Chongyu Zhang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; Tumor and Immunology Center of Precision Medicine Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; Shaanxi International Cooperation Base for Inflammation and Immunity, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Yong Li
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; Tumor and Immunology Center of Precision Medicine Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Zongfang Li
- Department of Geriatric Hepatobiliary, Pancreatic and Spleen Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; Tumor and Immunology Center of Precision Medicine Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China; Shaanxi International Cooperation Base for Inflammation and Immunity, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
26
|
Gao X, Su Q, Pan H, You Y, Ruan Z, Wu Y, Tang Z, Hu L. Arsenic-Induced Ferroptosis in Chicken Hepatocytes via the Mitochondrial ROS Pathway. Biol Trace Elem Res 2024; 202:4180-4190. [PMID: 38102534 DOI: 10.1007/s12011-023-03968-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/16/2023] [Indexed: 12/17/2023]
Abstract
Arsenic has been shown to be highly toxic and can cause liver damage. Previous studies have shown that arsenic causes severe liver damage and induces accumulation of reactive oxygen species (ROS). This study aimed to investigate the effects of ferroptosis on the liver in arsenic trioxide (ATO) and to explore the underlying mechanisms. We confirmed the hepatotoxic effects of arsenic by in vivo and in vitro experiments. After 28 days of administration of arsenic trioxide (4-mg/kg, 8-mg/kg) by gavage, chickens exhibited body weight loss and liver damage in a dose-dependent manner. In addition, in vivo and in vitro western blot and real-time fluorescence quantitative PCR analyses simultaneously indicated that ferroptosis might be the main pathway of arsenic-induced liver injury. Finally, Mito-TEMPO effectively eliminated the ROS accumulation in mitochondria, significantly attenuating the process of cellular ferroptosis. In summary, the hepatotoxic effects of arsenic are related to ferroptosis, and the hepatic ferroptosis process of arsenic is regulated by mitochondrial ROS (MtROS). Our study reveals new mechanisms of arsenic toxicity to the liver, which may deepen our understanding of arsenic toxicology.
Collapse
Affiliation(s)
- Xinglin Gao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Qian Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Hang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yanli You
- College of Life Science, Yantai University, Yantai City, 264005, Shandong Province, China
| | - Zhiyan Ruan
- School of Pharmacy, Guangdong Food & Drug Vocational College, No. 321, Longdong North Road, Tianhe District, Guangzhou, 510520, Guangdong Province, People's Republic of China
| | - Yuhan Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
27
|
Sun LL, Zhao LN, Sun J, Yuan HF, Wang YF, Hou CY, Lv P, Zhang HH, Yang G, Zhang NN, Zhang XD, Lu W. Inhibition of USP7 enhances CD8 + T cell activity in liver cancer by suppressing PRDM1-mediated FGL1 upregulation. Acta Pharmacol Sin 2024; 45:1686-1700. [PMID: 38589688 PMCID: PMC11272784 DOI: 10.1038/s41401-024-01263-2] [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: 12/04/2023] [Accepted: 03/07/2024] [Indexed: 04/10/2024]
Abstract
Lymphocyte activation gene 3 (LAG3), an immune checkpoint molecule expressed on activated T cells, functions as a negative regulator of immune responses. Persistent antigen exposure in the tumor microenvironment results in sustained LAG3 expression on T cells, contributing to T cell dysfunction. Fibrinogen-like protein 1 (FGL1) has been identified as a major ligand of LAG3, and FGL1/LAG3 interaction forms a novel immune checkpoint pathway that results in tumor immune evasion. In addition, ubiquitin-specific peptidase 7 (USP7) plays a crucial role in cancer development. In this study we investigated the role of USP7 in modulation of FGL1-mediated liver cancer immune evasion. We showed that knockdown of USP7 or treatment with USP7 inhibitor P5091 suppressed liver cancer growth by promoting CD8+ T cell activity in Hepa1-6 xenograft mice and in HepG2 or Huh7 cells co-cultured with T cells, whereas USP7 overexpression produced the opposite effect. We found that USP7 upregulated FGL1 in HepG2 and Huh7 cells by deubiquitination of transcriptional factor PR domain zinc finger protein 1 (PRDM1), which transcriptionally activated FGL1, and attenuated the CD8+ T cell activity, leading to the liver cancer growth. Interestingly, USP7 could be transcriptionally stimulated by PRDM1 as well in a positive feedback loop. P5091, an inhibitor of USP7, was able to downregulate FGL1 expression, thus enhancing CD8+ T cell activity. In an immunocompetent liver cancer mouse model, the dual blockade of USP7 and LAG3 resulted in a superior antitumor activity compared with anti-LAG3 therapy alone. We conclude that USP7 diminishes CD8+ T cell activity by a USP7/PRDM1 positive feedback loop on FGL1 production in liver cancer; USP7 might be a promising target for liver cancer immunotherapy.
Collapse
Affiliation(s)
- Lin-Lin Sun
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Li-Na Zhao
- National Key Laboratory of Drug ability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer / Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jiao Sun
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China
| | - Hong-Feng Yuan
- National Key Laboratory of Drug ability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer / Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yu-Fei Wang
- National Key Laboratory of Drug ability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer / Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Chun-Yu Hou
- National Key Laboratory of Drug ability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer / Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Pan Lv
- National Key Laboratory of Drug ability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer / Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Hui-Hui Zhang
- National Key Laboratory of Drug ability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer / Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Guang Yang
- National Key Laboratory of Drug ability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer / Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Ning-Ning Zhang
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China.
| | - Xiao-Dong Zhang
- National Key Laboratory of Drug ability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer / Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Wei Lu
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, 300060, China.
| |
Collapse
|
28
|
Zhang Y, Xu W, Peng C, Ren S, Zhang C. Intricate effects of post-translational modifications in liver cancer: mechanisms to clinical applications. J Transl Med 2024; 22:651. [PMID: 38997696 PMCID: PMC11245821 DOI: 10.1186/s12967-024-05455-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 06/29/2024] [Indexed: 07/14/2024] Open
Abstract
Liver cancer is a significant global health challenge, with hepatocellular carcinoma (HCC) being the most prevalent form, characterized by high incidence and mortality rates. Despite advances in targeted therapies and immunotherapies, the prognosis for advanced liver cancer remains poor. This underscores the urgent need for a deeper understanding of the molecular mechanisms underlying HCC to enable early detection and the development of novel therapeutic strategies. Post-translational modifications (PTMs) are crucial regulatory mechanisms in cellular biology, affecting protein functionality, interactions, and localization. These modifications, including phosphorylation, acetylation, methylation, ubiquitination, and glycosylation, occur after protein synthesis and play vital roles in various cellular processes. Recent advances in proteomics and molecular biology have highlighted the complex networks of PTMs, emphasizing their critical role in maintaining cellular homeostasis and disease pathogenesis. Dysregulation of PTMs has been associated with several malignant cellular processes in HCC, such as altered cell proliferation, migration, immune evasion, and metabolic reprogramming, contributing to tumor growth and metastasis. This review aims to provide a comprehensive understanding of the pathological mechanisms and clinical implications of various PTMs in liver cancer. By exploring the multifaceted interactions of PTMs and their impact on liver cancer progression, we highlight the potential of PTMs as biomarkers and therapeutic targets. The significance of this review lies in its potential to inform the development of novel therapeutic approaches and improve prognostic tools for early intervention in the fight against liver cancer.
Collapse
Affiliation(s)
- Yu Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Weihao Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chuanhui Peng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shenli Ren
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Cheng Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
29
|
Chen H, Hu J, Xiong X, Chen H, Liao Q, Lin B, Chen Y, Peng Y, Li Y, Cheng D, Li Z. SETD8 inhibits apoptosis and ferroptosis of Ewing's sarcoma through YBX1/RAC3 axis. Cell Death Dis 2024; 15:494. [PMID: 38987564 PMCID: PMC11237091 DOI: 10.1038/s41419-024-06882-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
Ewing's sarcoma (ES) represents a rare yet exceedingly aggressive neoplasm that poses a significant health risk to the pediatric and adolescent population. The clinical outcomes for individuals with relapsed or refractory ES are notably adverse, primarily attributed to the constrained therapeutic alternatives available. Despite significant advancements in the field, molecular pathology-driven therapeutic strategies have yet to achieve a definitive reduction in the mortality rates associated with ES. Consequently, there exists an imperative need to discover innovative therapeutic targets to effectively combat ES. To reveal the mechanism of the SETD8 (also known as lysine methyltransferase 5A) inhibitor UNC0379, cell death manners were analyzed with different inhibitors. The contributions of SETD8 to the processes of apoptosis and ferroptosis in ES cells were evaluated employing the histone methyltransferase inhibitor UNC0379 in conjunction with RNA interference techniques. The molecular regulatory mechanisms of SETD8 in ES were examined through the application of RNA sequencing (RNA-seq) and mass spectrometry-based proteomic analysis. Moreover, nude mouse xenograft models were established to explore the role of SETD8 in ES in vivo. SETD8, a sole nucleosome-specific methyltransferase that catalyzes mono-methylation of histone H4 at lysine 20 (H4K20me1), was found to be upregulated in ES, and its overexpression was associated with dismal outcomes of patients. SETD8 knockdown dramatically induced the apoptosis and ferroptosis of ES cells in vitro and suppressed tumorigenesis in vivo. Mechanistic investigations revealed that SETD8 facilitated the nuclear translocation of YBX1 through post-transcriptional regulatory mechanisms, which subsequently culminated in the transcriptional upregulation of RAC3. In summary, SETD8 inhibits the apoptosis and ferroptosis of ES cells through the YBX1/RAC3 axis, which provides new insights into the mechanism of tumorigenesis of ES. SETD8 may be a potential target for clinical intervention in ES patients.
Collapse
Affiliation(s)
- Huimou Chen
- Department of Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, No. 107 Yanjiang Road, Guangzhou, 510120, China
| | - Jing Hu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xilin Xiong
- Department of Oncology, Medical Centre of Pediatric, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107 Yanjiang Road, Guangzhou, 510120, China
| | - Hongling Chen
- Department of Clinical Laboratory, Maoming People's Hospital, Maoming, Guangdong, 525000, China
| | - Qiaofang Liao
- Department of Oncology, Huizhou First Hospital, Huizhou, Guangdong, 516000, China
| | - Biaojun Lin
- Department of Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, No. 107 Yanjiang Road, Guangzhou, 510120, China
| | - Yusong Chen
- Department of Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, No. 107 Yanjiang Road, Guangzhou, 510120, China
| | - Yanting Peng
- Department of Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, No. 107 Yanjiang Road, Guangzhou, 510120, China
| | - Yang Li
- Department of Oncology, Medical Centre of Pediatric, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107 Yanjiang Road, Guangzhou, 510120, China.
| | - Di Cheng
- Department of Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, No. 107 Yanjiang Road, Guangzhou, 510120, China.
| | - Zhihua Li
- Department of Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, No. 107 Yanjiang Road, Guangzhou, 510120, China.
| |
Collapse
|
30
|
Gong X, Wu Q, Tan Z, Lin S, Zhou J, Lin S, Wang W, Han Z, Xie T, Zhou J. Identification and validation of cuproptosis and disulfidptosis related genes in colorectal cancer. Cell Signal 2024; 119:111185. [PMID: 38643947 DOI: 10.1016/j.cellsig.2024.111185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 04/23/2024]
Abstract
Colorectal cancer, the third most prevalent malignant cancer, is associated with poor prognosis. Recent studies have investigated the mechanisms underlying cuproptosis and disulfidptosis in colorectal cancer. However, whether genes linked to these processes impact the prognosis of colorectal cancer patients through analogous mechanisms remains unclear. In this study, we developed a model of cuproptosis and disulfidptosis in colorectal cancer and concurrently explored the role of the pivotal model gene HSPA8 in colorectal cancer cell lines. Our results revealed a positive correlation between cuproptosis and disulfidptosis, both of which are emerging as protective factors for the prognosis of CRC patients. Consequently, a prognostic model encompassing HSPA8, PDCL3, CBX3, ATP6V1G1, TAF1D, RPL4, and RPL14 was constructed. Notably, the key gene in our model, HSPA8, exhibited heightened expression and was validated as a protective prognostic factor in colorectal cancer, exerting inhibitory effects on colorectal cancer cell proliferation. This study offers novel insights into the interplay between cuproptosis and disulfidptosis. The application of the prognostic model holds promise for more effectively predicting the overall survival of colorectal cancer patients.
Collapse
Affiliation(s)
- Xiaoqing Gong
- Department of Gastroenterology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China; Guangzhou Medical University, Guangzhou 511495, China
| | - Qixian Wu
- Department of Gastroenterology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China; Guangzhou Medical University, Guangzhou 511495, China
| | - Zhenlin Tan
- Peking University Shenzhen Hospital, Shenzhen 518000, China
| | - Shumao Lin
- Department of Gastroenterology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China; Guangzhou Medical University, Guangzhou 511495, China
| | - Jingdong Zhou
- Guangzhou Medical University, Guangzhou 511495, China
| | - Shihao Lin
- Guangzhou Medical University, Guangzhou 511495, China
| | - Weilin Wang
- Guangzhou Medical University, Guangzhou 511495, China
| | - Zhoujian Han
- Department of Gastroenterology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China; Guangzhou Medical University, Guangzhou 511495, China
| | - Tingting Xie
- Department of Gastroenterology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China.
| | - Jiyuan Zhou
- Department of Gastroenterology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China.
| |
Collapse
|
31
|
Li X, Liu C, Zhang R, Li Y, Ye D, Wang H, He M, Sun Y. Biosynthetic deficiency of docosahexaenoic acid causes nonalcoholic fatty liver disease and ferroptosis-mediated hepatocyte injury. J Biol Chem 2024; 300:107405. [PMID: 38788853 PMCID: PMC11231757 DOI: 10.1016/j.jbc.2024.107405] [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: 11/03/2023] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Exogenous omega-3 fatty acids, particularly docosahexaenoic acid (DHA), have shown to exert beneficial effects on nonalcoholic fatty liver disease (NAFLD), which is characterized by the excessive accumulation of lipids and chronic injury in the liver. However, the effect of endogenous DHA biosynthesis on the lipid homeostasis of liver is poorly understood. In this study, we used a DHA biosynthesis-deficient zebrafish model, elovl2 mutant, to explore the effect of endogenously biosynthesized DHA on hepatic lipid homeostasis. We found the pathways of lipogenesis and lipid uptake were strongly activated, while the pathways of lipid oxidation and lipid transport were inhibited in the liver of elovl2 mutants, leading to lipid droplet accumulation in the mutant hepatocytes and NAFLD. Furthermore, the elovl2 mutant hepatocytes exhibited disrupted mitochondrial structure and function, activated endoplasmic reticulum stress, and hepatic injury. We further unveiled that the hepatic cell death and injury was mainly mediated by ferroptosis, rather than apoptosis, in elovl2 mutants. Elevating DHA content in elovl2 mutants, either by the introduction of an omega-3 desaturase (fat1) transgene or by feeding with a DHA-rich diet, could strongly alleviate NAFLD features and ferroptosis-mediated hepatic injury. Together, our study elucidates the essential role of endogenous DHA biosynthesis in maintaining hepatic lipid homeostasis and liver health, highlighting that DHA deficiency can lead to NAFLD and ferroptosis-mediated hepatic injury.
Collapse
Affiliation(s)
- Xuehui Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Chengjie Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ru Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yi Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ding Ye
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Houpeng Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Mudan He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yonghua Sun
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.
| |
Collapse
|
32
|
Jin X, Jin W, Tong L, Zhao J, Zhang L, Lin N. Therapeutic strategies of targeting non-apoptotic regulated cell death (RCD) with small-molecule compounds in cancer. Acta Pharm Sin B 2024; 14:2815-2853. [PMID: 39027232 PMCID: PMC11252466 DOI: 10.1016/j.apsb.2024.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/29/2024] [Accepted: 03/18/2024] [Indexed: 07/20/2024] Open
Abstract
Regulated cell death (RCD) is a controlled form of cell death orchestrated by one or more cascading signaling pathways, making it amenable to pharmacological intervention. RCD subroutines can be categorized as apoptotic or non-apoptotic and play essential roles in maintaining homeostasis, facilitating development, and modulating immunity. Accumulating evidence has recently revealed that RCD evasion is frequently the primary cause of tumor survival. Several non-apoptotic RCD subroutines have garnered attention as promising cancer therapies due to their ability to induce tumor regression and prevent relapse, comparable to apoptosis. Moreover, they offer potential solutions for overcoming the acquired resistance of tumors toward apoptotic drugs. With an increasing understanding of the underlying mechanisms governing these non-apoptotic RCD subroutines, a growing number of small-molecule compounds targeting single or multiple pathways have been discovered, providing novel strategies for current cancer therapy. In this review, we comprehensively summarized the current regulatory mechanisms of the emerging non-apoptotic RCD subroutines, mainly including autophagy-dependent cell death, ferroptosis, cuproptosis, disulfidptosis, necroptosis, pyroptosis, alkaliptosis, oxeiptosis, parthanatos, mitochondrial permeability transition (MPT)-driven necrosis, entotic cell death, NETotic cell death, lysosome-dependent cell death, and immunogenic cell death (ICD). Furthermore, we focused on discussing the pharmacological regulatory mechanisms of related small-molecule compounds. In brief, these insightful findings may provide valuable guidance for investigating individual or collaborative targeting approaches towards different RCD subroutines, ultimately driving the discovery of novel small-molecule compounds that target RCD and significantly enhance future cancer therapeutics.
Collapse
Affiliation(s)
- Xin Jin
- Department of Ultrasound, Department of Medical Oncology and Department of Hematology, the First Hospital of China Medical University, China Medical University, Shenyang 110001, China
| | - Wenke Jin
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Linlin Tong
- Department of Ultrasound, Department of Medical Oncology and Department of Hematology, the First Hospital of China Medical University, China Medical University, Shenyang 110001, China
| | - Jia Zhao
- Department of Ultrasound, Department of Medical Oncology and Department of Hematology, the First Hospital of China Medical University, China Medical University, Shenyang 110001, China
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Na Lin
- Department of Ultrasound, Department of Medical Oncology and Department of Hematology, the First Hospital of China Medical University, China Medical University, Shenyang 110001, China
| |
Collapse
|
33
|
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
|
34
|
Shu YJ, Lao B, Qiu YY. Research progress of ferroptosis regulating lipid peroxidation and metabolism in occurrence and development of primary liver cancer. World J Gastrointest Oncol 2024; 16:2335-2349. [PMID: 38994128 PMCID: PMC11236230 DOI: 10.4251/wjgo.v16.i6.2335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/06/2024] [Accepted: 04/11/2024] [Indexed: 06/13/2024] Open
Abstract
As a highly aggressive tumor, the pathophysiological mechanism of primary liver cancer has attracted much attention. In recent years, factors such as ferroptosis regulation, lipid peroxidation and metabolic abnormalities have emerged in the study of liver cancer, providing a new perspective for understanding the development of liver cancer. Ferroptosis regulation, lipid peroxidation and metabolic abnormalities play important roles in the occurrence and development of liver cancer. The regulation of ferroptosis is involved in apoptosis and necrosis, affecting cell survival and death. Lipid peroxidation promotes oxidative damage and promotes the invasion of liver cancer cells. Metabolic abnormalities, especially the disorders of glucose and lipid metabolism, directly affect the proliferation and growth of liver cancer cells. Studies of ferroptosis regulation and lipid peroxidation may help to discover new therapeutic targets and improve therapeutic outcomes. The understanding of metabolic abnormalities can provide new ideas for the prevention of liver cancer, and reduce the risk of disease by adjusting the metabolic process. This review focuses on the key roles of ferroptosis regulation, lipid peroxidation and metabolic abnormalities in this process.
Collapse
Affiliation(s)
- Yu-Jie Shu
- Department of Gastroenterology, Yinzhou District Second Hospital, Ningbo 315199, Zhejiang Province, China
| | - Bo Lao
- Department of Gastroenterology, Yinzhou District Second Hospital, Ningbo 315199, Zhejiang Province, China
| | - Ying-Yang Qiu
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| |
Collapse
|
35
|
Xiao H, Chen H, Zhang L, Duolikun M, Zhen B, Kuerban S, Li X, Wang Y, Chen L, Lin J. Cytoskeletal gene alterations linked to sorafenib resistance in hepatocellular carcinoma. World J Surg Oncol 2024; 22:152. [PMID: 38849867 PMCID: PMC11157844 DOI: 10.1186/s12957-024-03417-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/20/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Although sorafenib has been consistently used as a first-line treatment for advanced hepatocellular carcinoma (HCC), most patients will develop resistance, and the mechanism of resistance to sorafenib needs further study. METHODS Using KAS-seq technology, we obtained the ssDNA profiles within the whole genome range of SMMC-7721 cells treated with sorafenib for differential analysis. We then intersected the differential genes obtained from the analysis of hepatocellular carcinoma patients in GSE109211 who were ineffective and effective with sorafenib treatment, constructed a PPI network, and obtained hub genes. We then analyzed the relationship between the expression of these genes and the prognosis of hepatocellular carcinoma patients. RESULTS In this study, we identified 7 hub ERGs (ACTB, CFL1, ACTG1, ACTN1, WDR1, TAGLN2, HSPA8) related to drug resistance, and these genes are associated with the cytoskeleton. CONCLUSIONS The cytoskeleton is associated with sorafenib resistance in hepatocellular carcinoma. Using KAS-seq to analyze the early changes in tumor cells treated with drugs is feasible for studying the drug resistance of tumors, which provides reference significance for future research.
Collapse
Affiliation(s)
- Hong Xiao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Hainan, China
| | - Hangyu Chen
- Department of Pharmacy, Peking University Third Hospital, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China
| | - Lei Zhang
- Department of Pharmacy, Peking University Third Hospital, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China
| | - Maimaitiyasen Duolikun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Hainan, China
| | - Baixin Zhen
- Department of Pharmacology, Xinjiang Medical University, Urumqi, China
| | - Subinuer Kuerban
- Department of Pharmacology, Xinjiang Medical University, Urumqi, China
| | - Xuehui Li
- Department of Pharmacology, Xinjiang Medical University, Urumqi, China
| | - Yuxi Wang
- Department of Pharmacy, Peking University Third Hospital, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China
| | - Long Chen
- Department of Pharmacy, Peking University Third Hospital, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China.
- Peking University, Third Hospital Cancer Center, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China.
| | - Jian Lin
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Hainan, China.
- Department of Pharmacy, Peking University Third Hospital, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China.
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Peking University, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China.
- Peking University, Third Hospital Cancer Center, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China.
| |
Collapse
|
36
|
Chen F, Kang R, Tang D, Liu J. Ferroptosis: principles and significance in health and disease. J Hematol Oncol 2024; 17:41. [PMID: 38844964 PMCID: PMC11157757 DOI: 10.1186/s13045-024-01564-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/02/2024] [Indexed: 06/09/2024] Open
Abstract
Ferroptosis, an iron-dependent form of cell death characterized by uncontrolled lipid peroxidation, is governed by molecular networks involving diverse molecules and organelles. Since its recognition as a non-apoptotic cell death pathway in 2012, ferroptosis has emerged as a crucial mechanism in numerous physiological and pathological contexts, leading to significant therapeutic advancements across a wide range of diseases. This review summarizes the fundamental molecular mechanisms and regulatory pathways underlying ferroptosis, including both GPX4-dependent and -independent antioxidant mechanisms. Additionally, we examine the involvement of ferroptosis in various pathological conditions, including cancer, neurodegenerative diseases, sepsis, ischemia-reperfusion injury, autoimmune disorders, and metabolic disorders. Specifically, we explore the role of ferroptosis in response to chemotherapy, radiotherapy, immunotherapy, nanotherapy, and targeted therapy. Furthermore, we discuss pharmacological strategies for modulating ferroptosis and potential biomarkers for monitoring this process. Lastly, we elucidate the interplay between ferroptosis and other forms of regulated cell death. Such insights hold promise for advancing our understanding of ferroptosis in the context of human health and disease.
Collapse
Affiliation(s)
- Fangquan Chen
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, 75390, USA.
| | - Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China.
| |
Collapse
|
37
|
Guo J, Yan W, Duan H, Wang D, Zhou Y, Feng D, Zheng Y, Zhou S, Liu G, Qin X. Therapeutic Effects of Natural Products on Liver Cancer and Their Potential Mechanisms. Nutrients 2024; 16:1642. [PMID: 38892575 PMCID: PMC11174683 DOI: 10.3390/nu16111642] [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: 04/23/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Liver cancer ranks third globally among causes of cancer-related deaths, posing a significant public health challenge. However, current treatments are inadequate, prompting a growing demand for novel, safe, and effective therapies. Natural products (NPs) have emerged as promising candidates in drug development due to their diverse biological activities, low toxicity, and minimal side effects. This paper begins by reviewing existing treatment methods and drugs for liver cancer. It then summarizes the therapeutic effects of NPs sourced from various origins on liver cancer. Finally, we analyze the potential mechanisms of NPs in treating liver cancer, including inhibition of angiogenesis, migration, and invasion; regulation of the cell cycle; induction of apoptosis, autophagy, pyroptosis, and ferroptosis; influence on tumor metabolism; immune regulation; regulation of intestinal function; and regulation of key signaling pathways. This systematic review aims to provide a comprehensive overview of NPs research in liver cancer treatment, offering a foundation for further development and application in pharmaceuticals and functional foods.
Collapse
Affiliation(s)
- Jinhong Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Wenjie Yan
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Hao Duan
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Diandian Wang
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Yaxi Zhou
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Duo Feng
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing 100081, China;
| | - Yue Zheng
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China;
| | - Shiqi Zhou
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Gaigai Liu
- Beijing Key Laboratory of Bioactive Substances and Functional Food, Beijing Union University, Beijing 100023, China; (J.G.); (W.Y.); (H.D.); (D.W.); (Y.Z.); (S.Z.); (G.L.)
| | - Xia Qin
- Graduate Department, Beijing Union University, Beijing 100101, China
| |
Collapse
|
38
|
Liu J, Song K, Lin B, Chen Z, Liu Y, Qiu X, He Q, Zuo Z, Yao X, Huang X, Liu Z, Liu Z, Huang Q, Guo X. The suppression of HSPA8 attenuates NLRP3 ubiquitination through SKP2 to promote pyroptosis in sepsis-induced lung injury. Cell Biosci 2024; 14:56. [PMID: 38698431 PMCID: PMC11064404 DOI: 10.1186/s13578-024-01239-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/25/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Acute lung injury (ALI) is strongly associated with hospitalization and mortality in patients with sepsis. Recent evidence suggests that pyroptosis mediated by NLRP3(NOD-, LRR- and pyrin domain-containing 3) inflammasome activation plays a key role in sepsis. However, the mechanism of NLRP3 inflammasome activation in sepsis-induced lung injury remains unclear. RESULTS in this study, we demonstrated that NLRP3 inflammasome was activated by the down-regulation of heat shock protein family A member 8 (HSPA8) in Lipopolysaccharide (LPS) and adenosine triphosphate (ATP)-treated mouse alveolar epithelial cells (AECs). Geranylgeranylacetone (GGA)-induced HSPA8 overexpression in cecum ligation and puncture (CLP) mice could significantly reduce systemic inflammatory response and mortality, effectively protect lung function, whilst HSPA8 inhibitor VER155008 aggravated this effect. The inhibition of HSPA8 was involved in sepsis induced acute lung injury by promoting pyroptosis of AECs. The down-regulation of HSPA8 activated NLRP3 inflammasome to mediate pyroptosis by promoting the degradation of E3 ubiquitin ligase S-phase kinase-associated protein 2 (SKP2). In addition, when stimulated by LPS and ATP, down-regulated SKP2 promoted pyroptosis of AECs by further attenuating ubiquitination of NLRP3. Adeno-associated virus 9-SKP2(AAV9-SKP2) could promote NLRP3 ubiquitination and degradation, alleviate lung injury and inhibit systemic inflammatory response in vivo. CONCLUSION in summary, our study shows there is strong statistical evidence that the suppression of HSPA8 mediates alveolar epithelial pyroptosis by promoting the degradation of E3 ubiquitin ligase SKP2 and subsequently attenuating the ubiquitination of NLRP3 to activate the NLRP3 inflammasome, which provides a new perspective and therapeutic target for the treatment of sepsis-induced lung injury.
Collapse
Affiliation(s)
- Jinlian Liu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ke Song
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Bingqi Lin
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhenfeng Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yan Liu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xianshuai Qiu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qi He
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zirui Zuo
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiaodan Yao
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoxia Huang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhuanhua Liu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhifeng Liu
- Department of Medicine intensive care unit , National Clinical Research Center for Geriatric Diseases (Chinese PLA General Hospital), General Hospital of Southern Theatre Command of PLA, Guangdong Branch Center, Guangzhou, Guangdong, China.
| | - Qiaobing Huang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- School of Basic Medical Sciences, Southern Medical University, 1023 Shatai Road, Tonghe, Guangzhou, 510515, China.
| | - Xiaohua Guo
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, National Experimental Education Demonstration Center for Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- School of Basic Medical Sciences, Southern Medical University, 1023 Shatai Road, Tonghe, Guangzhou, 510515, China.
| |
Collapse
|
39
|
Liu F, Zhang Y, Xia X, Han J, Cao L. Honokiol induces ferroptosis in ovarian cancer cells through the regulation of YAP by OTUB2. J Obstet Gynaecol Res 2024; 50:864-872. [PMID: 38480480 DOI: 10.1111/jog.15922] [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: 10/27/2023] [Accepted: 03/03/2024] [Indexed: 04/30/2024]
Abstract
BACKGROUND Ovarian cancer (OVCA) is prevalent in female reproductive organs. Despite recent advances, clinical outcomes remain poor, warranting fresh treatment avenues. Honokiol has an inhibitory effect on proliferation, invasion, and survival of cancer cells in vitro and in vivo. Therefore, this study intended to explore specific molecular mechanism by which honokiol affected OVCA progression. METHODS Bioinformatics analyzed the drug honokiol that bound to OTU deubiquitinase, ubiquitin aldehyde binding 2 (OTUB2). Cellular thermal shift assay (CETSA) verified the binding relationship between honokiol and OTUB2. Cell counting kit 8 (CCK-8) tested the IC50 value and cell viability of OVCA cells after honokiol treatment. Corresponding assay kits determined malonic dialdehyde (MDA) and Fe2+ levels in OVCA cells. Flow cytometry measured reactive oxygen species levels. Western blot detected OTUB2, SLC7A11, and transcriptional co-activators Yes-associated protein (YAP) expression, and quantitative polymerase chain reaction (qPCR) detected OTUB2 expression. Immunohistochemistry (IHC) detected the expression level of Ki67 protein in tumor tissues. RESULTS Honokiol was capable of inducing ferroptosis in OVCA cells. CETSA confirmed that honokiol could bind to OTUB2. Further cell functional and molecular experiments revealed that honokiol induced ferroptosis in OVCA cells via repression of YAP signaling pathway through binding to OTUB2. In addition, in vivo experiments have confirmed that honokiol could inhibit the growth of OVCA. CONCLUSION Honokiol induced ferroptosis in OVCA cells via repression of YAP signaling pathway through binding to OTUB2, implicating that OTUB2 may be an effective target for OVCA treatment, and our study results may provide new directions for development of more effective OVCA treatment strategies.
Collapse
Affiliation(s)
- Fang Liu
- Department of Gynecology, The Second Affiliated Hospital of Jiaxing University, Zhejiang, China
| | - Yufang Zhang
- Department of Gynecology, The Second Affiliated Hospital of Jiaxing University, Zhejiang, China
| | - Xinyi Xia
- Department of Gynecology, The Second Affiliated Hospital of Jiaxing University, Zhejiang, China
| | - Jing Han
- Department of Gynecology, The Second Affiliated Hospital of Jiaxing University, Zhejiang, China
| | - Linyan Cao
- Department of Gynecology, The Second Affiliated Hospital of Jiaxing University, Zhejiang, China
| |
Collapse
|
40
|
Niu C, Zhang J, Okolo PI. Liver cancer wars: plant-derived polyphenols strike back. Med Oncol 2024; 41:116. [PMID: 38625672 DOI: 10.1007/s12032-024-02353-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/06/2024] [Indexed: 04/17/2024]
Abstract
Liver cancer currently represents the leading cause of cancer-related death worldwide. The majority of liver cancer arises in the context of chronic inflammation and cirrhosis. Surgery, radiation therapy, and chemotherapy have been the guideline-recommended treatment options for decades. Despite enormous advances in the field of liver cancer therapy, an effective cure is yet to be found. Plant-derived polyphenols constitute a large family of phytochemicals, with pleiotropic effects and little toxicity. They can drive cellular events and modify multiple signaling pathways which involves initiation, progression and metastasis of liver cancer and play an important role in contributing to anti-liver cancer drug development. The potential of plant-derived polyphenols for treating liver cancer has gained attention from research clinicians and pharmaceutical scientists worldwide in the last decades. This review overviews hepatic carcinogenesis and briefly discusses anti-liver cancer mechanisms associated with plant-derived polyphenols, specifically involving cell proliferation, apoptosis, autophagy, angiogenesis, oxidative stress, inflammation, and metastasis. We focus on plant-derived polyphenols with experiment-based chemopreventive and chemotherapeutic properties against liver cancer and generalize their basic molecular mechanisms of action. We also discuss potential opportunities and challenges in translating plant-derived polyphenols from preclinical success into clinical applications.
Collapse
Affiliation(s)
- Chengu Niu
- Internal Medicine Residency Program, Rochester General Hospital, 1425 Portland Avenue, Rochester, NY, 14621, USA.
| | - Jing Zhang
- Rainier Springs Behavioral Health Hospital, 2805 NE 129th St, Vancouver, WA, 98686, USA
| | - Patrick I Okolo
- Division of Gastroenterology, Rochester General Hospital, Rochester, NY, 14621, USA
| |
Collapse
|
41
|
Yang P, Li H, Sun M, Guo X, Liao Y, Hu M, Ye P, Liu R. Zinc deficiency drives ferroptosis resistance by lactate production in esophageal squamous cell carcinoma. Free Radic Biol Med 2024; 213:512-522. [PMID: 38301975 DOI: 10.1016/j.freeradbiomed.2024.01.041] [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: 12/27/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/03/2024]
Abstract
Trace metal zinc is involved in key processes of solid tumors by its antioxidant properties, while the role of zinc at the onset of esophageal squamous cell carcinoma (ESCC) remains controversial. This study aimed to determine whether zinc is associated with the ESCC and underlying molecular events involving malignant progression. Based on a case-control study, we found serum and urine zinc were decreased and correlated with ESCC progression. Thus, an in vitro model for zinc deficiency (ZD) was established, and we found that ZD contributed to the proliferation, migration, and invasion of EC109 cells. Untargeted metabolomics identified 59 upregulated metabolites and 6 downregulated metabolites, among which glycolysis and ferroptosis-related oxidation of chain fatty acids might play crucial steps in ZD-treated molecular events. Interestingly, ZD disrupted redox homeostasis and enhanced cytosolic Fe2+ of EC109 cells, while lipid peroxidation, the key marker of ferroptosis occurrence, was decreased after ZD treatment. The mechanism underlying these changes may involve ZD-enhanced ESCC glycolysis and lactate production, which confer ferroptosis resistance by inhibiting of p-AMPK and leading to the upregulation of SREBP1 and SCD1 to enhance the production of anti-ferroptosis monounsaturated fatty acids.
Collapse
Affiliation(s)
- Peiyan Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Hui Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Mingjun Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Xinxin Guo
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yinghao Liao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Mohan Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Ping Ye
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
42
|
Liu X, Sun K, Yang H, Zou D, Xia L, Lu K, Meng X, Li Y. Molecular subtype identification and prognosis stratification based on lysosome-related genes in breast cancer. Heliyon 2024; 10:e25643. [PMID: 38420434 PMCID: PMC10900431 DOI: 10.1016/j.heliyon.2024.e25643] [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: 05/22/2023] [Revised: 01/13/2024] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Background Lysosomes are known to have a significant impact on the development and recurrence of breast cancer. However, the association between lysosome-related genes (LRGs) and breast cancer remains unclear. This study aims to explore the potential role of LRGs in predicting the prognosis and treatment response of breast cancer. Methods Breast cancer gene expression profile data and clinical information were downloaded from TCGA and GEO databases, and prognosis-related LRGs were screened for consensus clustering analysis. Lasso Cox regression analysis was used to construct risk features derived from LRGs, and immune cell infiltration, immune therapy response, drug sensitivity, and clinical pathological feature differences were evaluated for different molecular subtypes and risk groups. A nomogram based on risk features derived from LRGs was constructed and evaluated. Results Our study identified 176 differentially expressed LRGs that are associated with breast cancer prognosis. Based on these genes, we divided breast cancer into two molecular subtypes with significant prognostic differences. We also found significant differences in immune cell infiltration between these subtypes. Furthermore, we constructed a prognostic risk model consisting of 7 LRGs, which effectively divides breast cancer patients into high-risk and low-risk groups. Patients in the low-risk group have better prognostic characteristics, respond better to immunotherapy, and have lower sensitivity to chemotherapy drugs, indicating that the low-risk group is more likely to benefit from immunotherapy and chemotherapy. Additionally, the risk score based on LRGs is significantly correlated with immune cell infiltration, including CD8 T cells and macrophages. This risk score model, along with age, chemotherapy, clinical stage, and N stage, is an independent prognostic factor for breast cancer. Finally, the nomogram composed of these factors has excellent performance in predicting overall survival of breast cancer. Conclusions In conclusion, this study has constructed a novel LRG-derived breast cancer risk feature, which performs well in prognostic prediction when combined with clinical pathological features.
Collapse
Affiliation(s)
- Xiaozhen Liu
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Kewang Sun
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Hongjian Yang
- Department of Breast Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
| | - Dehomg Zou
- Department of Breast Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
| | - Lingli Xia
- Department of Breast Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
| | - Kefeng Lu
- Department of Outpatient Service, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Xuli Meng
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Yongfeng Li
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
43
|
Yang B, Wang S, Yang Y, Li X, Yu F, Wang T. Endoplasmic reticulum stress in breast cancer: a predictive model for prognosis and therapy selection. Front Immunol 2024; 15:1332942. [PMID: 38440732 PMCID: PMC10910050 DOI: 10.3389/fimmu.2024.1332942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Background Breast cancer (BC) is a leading cause of mortality among women, underscoring the urgent need for improved therapeutic predictio. Developing a precise prognostic model is crucial. The role of Endoplasmic Reticulum Stress (ERS) in cancer suggests its potential as a critical factor in BC development and progression, highlighting the importance of precise prognostic models for tailored treatment strategies. Methods Through comprehensive analysis of ERS-related gene expression in BC, utilizing both single-cell and bulk sequencing data from varied BC subtypes, we identified eight key ERS-related genes. LASSO regression and machine learning techniques were employed to construct a prognostic model, validated across multiple datasets and compared with existing models for its predictive accuracy. Results The developed ERS-model categorizes BC patients into distinct risk groups with significant differences in clinical prognosis, confirmed by robust ROC, DCA, and KM analyses. The model forecasts survival rates with high precision, revealing distinct immune infiltration patterns and treatment responsiveness between risk groups. Notably, we discovered six druggable targets and validated Methotrexate and Gemcitabine as effective agents for high-risk BC treatment, based on their sensitivity profiles and potential for addressing the lack of active targets in BC. Conclusion Our study advances BC research by establishing a significant link between ERS and BC prognosis at both the molecular and cellular levels. By stratifying patients into risk-defined groups, we unveil disparities in immune cell infiltration and drug response, guiding personalized treatment. The identification of potential drug targets and therapeutic agents opens new avenues for targeted interventions, promising to enhance outcomes for high-risk BC patients and paving the way for personalized cancer therapy.
Collapse
Affiliation(s)
- Bin Yang
- Research Laboratory Center, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
- NHC Key Laboratory of Pulmonary Immune-Related Diseases, Guizhou Provincial People's Hospital, Guizhou University, Guiyang, Guizhou, China
| | - Shu Wang
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yanfang Yang
- Research Laboratory Center, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
- NHC Key Laboratory of Pulmonary Immune-Related Diseases, Guizhou Provincial People's Hospital, Guizhou University, Guiyang, Guizhou, China
| | - Xukui Li
- Research Laboratory Center, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
- NHC Key Laboratory of Pulmonary Immune-Related Diseases, Guizhou Provincial People's Hospital, Guizhou University, Guiyang, Guizhou, China
| | - Fuxun Yu
- Research Laboratory Center, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
- NHC Key Laboratory of Pulmonary Immune-Related Diseases, Guizhou Provincial People's Hospital, Guizhou University, Guiyang, Guizhou, China
| | - Tao Wang
- Research Laboratory Center, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
- NHC Key Laboratory of Pulmonary Immune-Related Diseases, Guizhou Provincial People's Hospital, Guizhou University, Guiyang, Guizhou, China
| |
Collapse
|
44
|
Li J, Li PT, Wu W, Ding BN, Wen YG, Cai HL, Liu SX, Hong T, Zhang JF, Zhou JD, Qian LY, Du J. POU2F2-mediated upregulation of lncRNA PTPRG-AS1 inhibits ferroptosis in breast cancer via miR-376c-3p/SLC7A11 axis. Epigenomics 2024; 16:215-231. [PMID: 38318853 DOI: 10.2217/epi-2023-0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024] Open
Abstract
Background: Triple-negative breast cancer (TNBC) is a subtype of BC with high rates of mortality. The mechanism of PTPRG-AS1 in ferroptosis of TNBC was investigated. Methods: Chromatin immunoprecipitation and dual-luciferase reporter assays were used to measure intermolecular relationships. MTT and colony formation assays detected cell viability and proliferation. Kits detected Fe2+ and reactive oxygen species levels. The role of PTPRG-AS1 in tumor growth was analyzed in vivo. Results: PTPRG-AS1 was increased in TNBC tissues and cells. PTPRG-AS1 silencing increased the reduction of glutathione and GPX4, increased Fe2+ and reactive oxygen species in erastin-treated cells and inhibited proliferation. POU2F2 transcriptionally upregulated PTPRG-AS1. PTPRG-AS1 targeted miR-376c-3p to upregulate SLC7A11. PTPRG-AS1 knockdown suppressed tumor growth in vivo. Conclusion: POU2F2 transcriptionally activates PTPRG-AS1 to modulate ferroptosis and proliferation by miR-376c-3p/SLC7A11, promoting TNBC.
Collapse
Affiliation(s)
- Jun Li
- Department of Breast & Thyroid Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Pei-Ting Li
- Department of Breast & Thyroid Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Wei Wu
- Department of Breast & Thyroid Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Bo-Ni Ding
- Department of Breast & Thyroid Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Yan-Guang Wen
- Department of Breast & Thyroid Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Hai-Lin Cai
- Department of Breast & Thyroid Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Shuang-Xi Liu
- Department of Breast & Thyroid Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Tao Hong
- Department of Breast & Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, Jiangxi Province, China
| | - Jian-Fei Zhang
- Department of Plastic Surgery, The Second Affiliated Hospital of the University of South China, Hengyang, 421000, Hunan Province, China
| | - Jian-Da Zhou
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Li-Yuan Qian
- Department of Breast & Thyroid Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Juan Du
- Department of Breast & Thyroid Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| |
Collapse
|
45
|
Shen X, Dong P, Kong J, Sun N, Wang F, Sang L, Xu Y, Zhang M, Chen X, Guo R, Wang S, Lin Q, Jiang Z, Xu S, Zhang C, Bian Z, Wang W, Guo R. Targeted single-cell RNA sequencing analysis reveals metabolic reprogramming and the ferroptosis-resistant state in hematologic malignancies. Cell Biochem Funct 2023; 41:1343-1356. [PMID: 37823726 DOI: 10.1002/cbf.3869] [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: 07/11/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023]
Abstract
Hematologic malignancies are the most common hematopoietic diseases and a major public health concern. However, the mechanisms underlying myeloid tumors remain unknown owing to the intricate interplay between mutations and diverse clonal evolution patterns, as evidenced by the analysis of bulk cell-derived omics data. Several single-cell omics techniques have been used to characterize the hierarchies and altered immune microenvironments of hematologic malignancies. The comprehensive single-cell atlas of hematologic malignancies provides novel opportunities for personalized combinatorial targeted treatments, avoiding unwanted chemo-toxicity. In the present study, we performed transcriptome sequencing by combining single-cell RNA sequencing (scRNA-seq) with a targeted oncogenic gene panel for acute myeloid leukemia, overcoming the limitations of scRNA-seq in detecting oncogenic mutations. The distribution of oncogenic IDH1, IDH2, and KRAS mutations in each cell type was identified in the bone marrow (BM) samples of each patient. Our findings suggest that ferroptosis and metabolic reprogramming are involved in the tumorigenesis and chemotherapy resistance of oncogenic mutation-carrying cells. Biological progression via IDH1, IDH2, and KRAS mutations arrests hematopoietic maturation. Our study findings provide a rationale for using primary BM cells for personalized treatment in clinical settings.
Collapse
Affiliation(s)
- Xiaohui Shen
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peiyuan Dong
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingjing Kong
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Nannan Sun
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fang Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lina Sang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Xu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengmeng Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoli Chen
- Jiangxi Health Commission Key Laboratory of Leukemia, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi, China
| | - Rong Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuya Wang
- Department of Blood Transfusion, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Quande Lin
- Department of Hematology, The Afliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shan Xu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Congli Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhilei Bian
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Weimin Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
46
|
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
|
47
|
Liu Y, Pang Z, Wang J, Wang J, He J, Ji B, Zhang L, Ren M. Heat shock protein family A member 8 is a prognostic marker for bladder cancer: Evidences based on experiments and machine learning. J Cell Mol Med 2023; 27:3995-4008. [PMID: 37771276 PMCID: PMC10746959 DOI: 10.1111/jcmm.17977] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/03/2023] [Accepted: 09/16/2023] [Indexed: 09/30/2023] Open
Abstract
Heat shock protein member 8 (HSPA8) is one of the most abundant chaperones in eukaryotic cells, but its biological roles in bladder cancer (BC) are largely unclear. First, we observed that HSPA8 was abundant in both cell lines and tissues of BC, and the HSPA8-high group had poorer T stages and overall survival (OS) than the HSPA8-low group in the TCGA patients. Next, when we knocked down HSPA8 in BC cells, the growth and migration abilities were significantly decreased, the apoptosis rates were significantly increased, and the Ki67 fluorescence intensity was decreased in BC cells. Moreover, caspase 3 was significantly decreased with overexpression of HSPA8 in BC cells. After that, a machine learning prognostic model was created based on the expression of HSPA8 by applying LASSO Cox regression in TCGA and GEO patients. The model indicated that the low-risk (LR) group with BC had better tumour stages, lymphovascular invasion, and OS than the high-risk (HR) group. Additionally, the risk score was demonstrated to be an independent risk factor for the prognosis of BC by univariate and multivariate Cox analyses. Moreover, the HR group showed a greater rate of TP53 mutations and was mostly enriched in the ECM-receptor interaction pathway than the LR group. Importantly, lower CD8+ T-cell and NK cell infiltration, higher immune exclusion scores, higher expression of PD-L1 and CTLA4 and poorer immune checkpoint therapy effects were found in the HR group. These findings demonstrated how crucial HSPA8 plays a role in determining the prognosis of bladder cancer.
Collapse
Affiliation(s)
- Yang Liu
- Department of Urinary SurgeryFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Zhong‐qi Pang
- Department of Urinary SurgeryFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Jian‐she Wang
- Department of Urinary SurgeryFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Jin‐feng Wang
- Department of Urinary SurgeryFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Jia‐xin He
- Department of Urinary SurgeryFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Bo Ji
- Department of Urinary SurgeryFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Lu Zhang
- Department of Urinary SurgeryFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Ming‐hua Ren
- Department of Urinary SurgeryFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| |
Collapse
|
48
|
Yang D, Zhang M, Chen W, Lu Q, Wan S, Du X, Li Y, Li B, Wu W, Wang C, Li N, Peng S, Tang H, Hua J. UCHL1 maintains microenvironmental homeostasis in goat germline stem cells. FASEB J 2023; 37:e23306. [PMID: 37934018 DOI: 10.1096/fj.202301674rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/08/2023]
Abstract
Spermatogonial stem cells (SSCs) play a crucial role in mammalian spermatogenesis and maintain the stable inheritance of the germline in livestock. However, stress and bacterial or viral infections can disrupt immune homeostasis of the testes, thereby leading to spermatogenesis destruction and infertility, which severely affects the health and productivity of mammals. This study aimed to explore the effect of ubiquitin C-terminal hydrolase L1 (UCHL1) knockdown (KD) in goat SSCs and mouse testes and investigate the potential anti-inflammatory function of UCHL1 in a poly(I:C)-induced inflammation model to maintain microenvironmental homeostasis. In vitro, the downregulation of UCHL1 (UCHL1 KD) in goat SSCs increased the expression levels of apoptosis and inflammatory factors and inhibited the self-renewal and proliferation of SSCs. In vivo, the structure of seminiferous tubules and spermatogenic cells was disrupted after UCHL1 KD, and the expression levels of apoptosis- and inflammation-related proteins were significantly upregulated. Furthermore, UCHL1 inhibited the TLR3/TBK1/IRF3 pathway to resist poly(I:C)-induced inflammation in SSCs by antagonizing HSPA8 and thus maintaining SSC autoimmune homeostasis. Most importantly, the results of this study showed that UCHL1 maintained immune homeostasis of SSCs and spermatogenesis. UCHL1 KD not only inhibited the self-renewal and proliferation of goat SSCs and spermatogenesis but was also involved in the inflammatory response of goat SSCs. Additionally, UCHL1 has an antiviral function in SSCs by antagonizing HSPA8, which provides an important basis for exploring the specific mechanisms of UCHL1 in goat spermatogenesis.
Collapse
Affiliation(s)
- Donghui Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Mengfei Zhang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Wenbo Chen
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Qizhong Lu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shicheng Wan
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Xiaomin Du
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
- Shaanxi Provincial Engineering and Technology Research Center of Cashmere Goats, College of Life Sciences, Yulin University, Yulin, China
| | - Yunxiang Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Balun Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Wenping Wu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Congliang Wang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Na Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Sha Peng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
49
|
Zhang Z, Zhao Y, Wang Y, Zhao Y, Guo J. Autophagy/ferroptosis in colorectal cancer: Carcinogenic view and nanoparticle-mediated cell death regulation. ENVIRONMENTAL RESEARCH 2023; 238:117006. [PMID: 37669735 DOI: 10.1016/j.envres.2023.117006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/19/2023] [Accepted: 08/26/2023] [Indexed: 09/07/2023]
Abstract
The cell death mechanisms have a long history of being evaluated in diseases and pathological events. The ability of triggering cell death is considered to be a promising strategy in cancer therapy, but some mechanisms have dual functions in cancer, requiring more elucidation of underlying factors. Colorectal cancer (CRC) is a disease and malignant condition of colon and rectal that causes high mortality and morbidity. The autophagy targeting in CRC is therapeutic importance and this cell death mechanism can interact with apoptosis in inhibiting or increasing apoptosis. Autophagy has interaction with ferroptosis as another cell death pathway in CRC and can accelerate ferroptosis in suppressing growth and invasion. The dysregulation of autophagy affects the drug resistance in CRC and pro-survival autophagy can induce drug resistance. Therefore, inhibition of protective autophagy enhances chemosensitivity in CRC cells. Moreover, autophagy displays interaction with metastasis and EMT as a potent regulator of invasion in CRC cells. The same is true for ferroptosis, but the difference is that function of ferroptosis is determined and it can reduce viability. The lack of ferroptosis can cause development of chemoresistance in CRC cells and this cell death mechanism is regulated by various pathways and mechanisms that autophagy is among them. Therefore, current review paper provides a state-of-art analysis of autophagy, ferroptosis and their crosstalk in CRC. The nanoparticle-mediated regulation of cell death mechanisms in CRC causes changes in progression. The stimulation of ferroptosis and control of autophagy (induction or inhibition) by nanoparticles can impair CRC progression. The engineering part of nanoparticle synthesis to control autophagy and ferroptosis in CRC still requires more attention.
Collapse
Affiliation(s)
- Zhibin Zhang
- Chengde Medical College, College of Traditional Chinese Medicine, Chengde, Hebei, 067000, China.
| | - Yintao Zhao
- Chengde Medical College, Chengde, Hebei, 067000, China
| | - Yuman Wang
- Chengde Medical College, Chengde, Hebei, 067000, China
| | - Yutang Zhao
- Chengde Medical College, Chengde, Hebei, 067000, China
| | - Jianen Guo
- Chengde Medical College, Chengde, Hebei, 067000, China
| |
Collapse
|
50
|
Wang S, Wang YF, Yang G, Zhang HH, Yuan HF, Hou CY, Zhao LN, Suo YH, Sun J, Sun LL, Lv P, Sun Y, Zhang NN, Zhang XD, Lu W. Heat shock protein family A member 8 serving as a co-activator of transcriptional factor ETV4 up-regulates PHLDA2 to promote the growth of liver cancer. Acta Pharmacol Sin 2023; 44:2525-2536. [PMID: 37474643 PMCID: PMC10692233 DOI: 10.1038/s41401-023-01133-3] [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: 01/09/2023] [Accepted: 07/05/2023] [Indexed: 07/22/2023]
Abstract
Heat shock protein family A member 8 (HSPA8) participates in the folding or degradation of misfolded proteins under stress and plays critical roles in cancer. In this study, we investigated the function of HSPA8 in the development of liver cancer. By analyzing the TCGA transcriptome dataset, we found that HSPA8 was upregulated in 134 clinical liver cancer tissue samples, and positively correlated with poor prognosis. IHC staining showed the nuclear and cytoplasmic localization of HSPA8 in liver cancer cells. Knockdown of HSPA8 resulted in a decrease in the proliferation of HepG2 and Huh-7 cells. ChIP-seq and RNA-seq analysis revealed that HSPA8 bound to the promoter of pleckstrin homology-like domain family A member 2 (PHLDA2) and regulated its expression. The transcription factor ETV4 in HepG2 cells activated PHLDA2 transcription. HSPA8 and ETV4 could interact with each other in the cells and colocalize in the nucleus. From a functional perspective, we demonstrated that HSPA8 upregulated PHDLA2 through the coactivating transcription factor ETV4 to enhance the growth of liver cancer in vitro and in vivo. From a therapeutic perspective, we identified both HSPA8 and PHDLA2 as novel targets in the treatment of HCC. In conclusion, this study demonstrates that HSPA8 serves as a coactivator of ETV4 and upregulates PHLDA2, leading to the growth of HCC, and is a potential therapeutic target in HCC treatment.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, China
| | - Yu-Fei Wang
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Guang Yang
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Hui-Hui Zhang
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Hong-Feng Yuan
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Chun-Yu Hou
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Li-Na Zhao
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yu-Hong Suo
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, China
| | - Jiao Sun
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, China
| | - Lin-Lin Sun
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, China
| | - Pan Lv
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yan Sun
- Department of Pathology, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China.
| | - Ning-Ning Zhang
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, China.
| | - Xiao-Dong Zhang
- Department of Gastrointestinal Cancer Biology, Tianjin Cancer Institute, Tianjin Medical University Cancer Institute, and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Wei Lu
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, China.
| |
Collapse
|