1
|
Taha SR, Karimi M, Mahdavi B, Yousefi Tehrani M, Bemani A, Kabirian S, Mohammadi J, Jabbari S, Hushmand M, Mokhtar A, Pourhanifeh MH. Crosstalk between non-coding RNAs and programmed cell death in colorectal cancer: implications for targeted therapy. Epigenetics Chromatin 2025; 18:3. [PMID: 39810224 PMCID: PMC11734566 DOI: 10.1186/s13072-024-00560-8] [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: 09/17/2024] [Accepted: 11/13/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) remains one of the most common causes of cancer-related mortality worldwide. Its progression is influenced by complex interactions involving genetic, epigenetic, and environmental factors. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), have been identified as key regulators of gene expression, affecting diverse biological processes, notably programmed cell death (PCD). OBJECTIVE This review aims to explore the relationship between ncRNAs and PCD in CRC, focusing on how ncRNAs influence cancer cell survival, proliferation, and treatment resistance. METHODS A comprehensive literature analysis was conducted to examine recent findings on the role of ncRNAs in modulating various PCD mechanisms, including apoptosis, autophagy, necroptosis, and pyroptosis, and their impact on CRC development and therapeutic response. RESULTS ncRNAs were found to significantly regulate PCD pathways, impacting tumor growth, metastasis, and treatment sensitivity in CRC. Their influence on these pathways highlights the potential of ncRNAs as biomarkers for early CRC detection and as targets for innovative therapeutic interventions. CONCLUSION Understanding the involvement of ncRNAs in PCD regulation offers new insights into CRC biology. The targeted modulation of ncRNA-PCD interactions presents promising avenues for personalized cancer treatment, which may improve patient outcomes by enhancing therapeutic effectiveness and reducing resistance.
Collapse
Affiliation(s)
- Seyed Reza Taha
- Department of Pathology and Immunology, Washington University School of Medicine, St. LouisWashington, MO, USA
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Karimi
- Faculty of Medicine, Bogomolets National Medical University (NMU), Kiev, Ukraine.
| | - Bahar Mahdavi
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | | | - Ali Bemani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Shahriar Kabirian
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Javad Mohammadi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sina Jabbari
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Meysam Hushmand
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mokhtar
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
- PAKAN Institute, Tehran, Iran.
| |
Collapse
|
2
|
Ma L, Liu Y, Ren Y, Mi N, Fang J, Bao R, Xu X, Zhang H, Tang Y. Integrating bioinformatics and machine learning to uncover lncRNA LINC00269 as a key regulator in Parkinson's disease via pyroptosis pathways. Eur J Med Res 2024; 29:582. [PMID: 39696629 DOI: 10.1186/s40001-024-02201-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: 08/02/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Pyroptosis, a specific type of programmed cell death, which has become a significant factor to Parkinson's disease (PD). Concurrently, long non-coding RNAs (lncRNAs) have garnered attention for their regulatory roles in neurodegenerative disorders. This study was designed to ascertain the key lncRNAs in pyroptosis pathways of PD and elucidate their regulatory mechanisms. METHODS Employing a combination of bioinformatics and machine learning, we analyzed PD data sets GSE133347 and GSE110716. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) recognized different lncRNAs. Through various algorithms such as Least Absolute Shrinkage and Selection Operator (LASSO), Random Forest (RF), and Weighted Gene Co-expression Network Analysis (WGCNA), we recognized LINC01606 and LINC00269, which are key factors during the emergence and development of PD. Furthermore, experimental validation was conducted in PD mouse models to confirm these bioinformatics findings. RESULTS The analysis showed that there were a large number of apoptosis-related gene expression changes in Parkinson's syndrome, for example, CASP1 and GSDME were up-regulated, and CASP9 and AIM2 were down-regulated. Among the lncRNAs, LINC01606 and LINC00269 were identified as potential modulators of pyroptosis. Notably, LINC00269 was observed to be significantly downregulated in the brain tissues of a PD mouse model, supporting its involvement in PD. The study also highlighted potential interactions of these lncRNAs with genes like ONECUT2, PRLR, CTNNA3, and LRP2. CONCLUSIONS This study identifies LINC00269 as a potential contributor to pyroptosis pathways in PD. While further investigation is required to fully elucidate its role, these findings provide new insights into PD pathogenesis and suggest potential avenues for future research on diagnostic and therapeutic targets. The study underscores the importance of integrating bioinformatics with experimental validation in neurodegenerative disease research.
Collapse
Affiliation(s)
- LiLi Ma
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Number 23, You Zheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China
- Department of Neurology, Jilin City Hospital of Chemical Industry, Jilin City, Jilin, China
| | - Yue Liu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang Province, China
| | - Yajing Ren
- School of Medical and Life Sciences, Chengdu University of TCM, Cheng du City, 611137, Sichuan Province, China
| | - Na Mi
- Department of Neurology, Chi Feng Municipal Hospital, Chi Feng City, 024000, Inner Mongolia Autonomous Region, China
| | - Jing Fang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150081, People's Republic of China
| | - Rui Bao
- Department of Rehabilitation, The Third Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Xiuzhi Xu
- General Medical Department, Heilongjiang Provincial Hospital, Harbin, 150036, Heilongjiang Province, China
| | - Hongjia Zhang
- Department of Neurology, Jilin City Hospital of Chemical Industry, Jilin City, Jilin, China.
| | - Ying Tang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Number 23, You Zheng Street, Nan Gang District, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
3
|
Chen Y, Jiang Z, Li X. New insights into crosstalk between Nrf2 pathway and ferroptosis in lung disease. Cell Death Dis 2024; 15:841. [PMID: 39557840 PMCID: PMC11574213 DOI: 10.1038/s41419-024-07224-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024]
Abstract
Ferroptosis is a distinctive process of cellular demise that is linked to amino acid metabolism, lipid oxidation, and iron oxidation. The ferroptosis cascade genes, which are closely associated with the onset of lung diseases, are among the regulatory targets of nuclear factor erythroid 2-related factor 2 (Nrf2). Although the regulation of ferroptosis is mostly mediated by Nrf2, the precise roles and underlying regulatory mechanisms of ferroptosis and Nrf2 in lung illness remain unclear. This review provides new insights from recent discoveries involving the modulation of Nrf2 and ferroptosis in a range of lung diseases. It also systematically describes regulatory mechanisms involving lipid peroxidation, intracellular antioxidant levels, ubiquitination of Nrf2, and expression of FSP1 and GPX4. Finally, it summarises active ingredients and drugs with potential for the treatment of lung diseases. With the overarching aim of expediting improvements in treatment, this review provides a reference for novel therapeutic mechanisms and offers suggestions for the development of new medications for a variety of lung disorders.
Collapse
Affiliation(s)
- Yonghu Chen
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, 133002, P. R. China
| | - Zhe Jiang
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, 133002, P. R. China.
| | - Xuezheng Li
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, 133002, P. R. China.
| |
Collapse
|
4
|
Abida, Altamimi ASA, Ghaboura N, Balaraman AK, Rajput P, Bansal P, Rawat S, Alanazi FJ, Alruwaili AN, Aldhafeeri NA, Ali H, Deb PK. Therapeutic Potential of lncRNAs in Regulating Disulfidptosis for Cancer Treatment. Pathol Res Pract 2024; 263:155657. [PMID: 39437641 DOI: 10.1016/j.prp.2024.155657] [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: 09/02/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
Non-coding RNAs (lncRNAs) play critical roles in various cellular processes, including a novel form of regulated cell death known as disulfidptosis, characterized by accumulating protein disulfide bonds and severe endoplasmic reticulum stress. This review highlights the therapeutic potential of lncRNAs in regulating disulfidptosis for cancer treatment, emphasizing their influence on key pathway components such as GPX4, SLC7A11, and PDIA family members. Recent studies have demonstrated that targeting specific lncRNAs can sensitize cancer cells to disulfidptosis, offering a promising approach to cancer therapy. The regulation of disulfidptosis by lncRNAs involves various signaling pathways, including oxidative stress, ER stress, and calcium signaling. This review also discusses the molecular mechanisms underlying lncRNA regulation of disulfidptosis, the challenges of developing lncRNA-based therapies, and the future potential of this rapidly advancing field in cancer research.
Collapse
Affiliation(s)
- Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Nehmat Ghaboura
- Department of Pharmacy Practice, Pharmacy Program, Batterjee Medical College, PO Box 6231, Jeddah 21442, Saudi Arabia
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Pranchal Rajput
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India.
| | - Pooja Bansal
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Sushama Rawat
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Fadiyah Jadid Alanazi
- Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Abeer Nuwayfi Alruwaili
- Department of Nursing Administration and Education, College of Nursing, Jouf University, Al Jouf 72388, Saudi Arabia
| | - Nouf Afit Aldhafeeri
- College of Nursing, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haider Ali
- Center for Global health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Pran Kishore Deb
- Department of Pharmaceutical Sciences and Technology, Birla Institue of Technology (BIT), Mesra, Ranchi, Jharkhand 835215, India
| |
Collapse
|
5
|
Ouyang S, Zeng Z, He J, Luo L. Epigenetic regulation of targeted ferroptosis: A new strategy for drug development. J Pharm Anal 2024; 14:101012. [PMID: 39850234 PMCID: PMC11755343 DOI: 10.1016/j.jpha.2024.101012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 01/25/2025] Open
Abstract
Ferroptosis is a newly discovered form of cell death that is influenced by iron levels and is triggered by cellular metabolism and excessive lipid peroxidation. Epigenetic regulation plays a crucial role in the development and progression of diseases, making it essential to understand these mechanisms in order to identify potential targets for drug development and clinical treatment. The intersection of ferroptosis and epigenetics has opened up new avenues for research in drug development, offering innovative strategies for combating diseases. Recent studies have shown that epigenetic modifications can impact pathways related to ferroptosis, potentially leading to organ dysfunction. Despite the increasing focus on this relationship, the role of epigenetic regulation in drug development remains largely unexplored. This article explores current research on the interplay between epigenetic regulation and ferroptosis, delving into their regulatory mechanisms and discussing the effects of existing epigenetic modification regulators on diseases. Additionally, we highlight ongoing research on epigenetic factors involved in targeting ferroptosis in cancer, providing new insights for the development of cancer treatments.
Collapse
Affiliation(s)
- Shengli Ouyang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Zeyao Zeng
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Jieyi He
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| |
Collapse
|
6
|
Lin Y, Zhao W, Pu R, Lv Z, Xie H, Li Y, Zhang Z. Long non‑coding RNAs as diagnostic and prognostic biomarkers for colorectal cancer (Review). Oncol Lett 2024; 28:486. [PMID: 39185489 PMCID: PMC11342420 DOI: 10.3892/ol.2024.14619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
Colorectal cancer (CRC) ranks as the 3rd most common cancer globally and is the 2nd leading cause of cancer-related death. Owing to the lack of specific early symptoms and the limitations of existing early diagnostic methods, most patients with CRC are diagnosed at advanced stages. To overcome these challenges, researchers have increasingly focused on molecular biomarkers, with particular interest in long non-coding RNAs (lncRNAs). These non-protein-coding RNAs, which exceed 200 nucleotides in length, play critical roles in the development and progression of CRC. The stability and detectability of lncRNAs in the circulatory system make them promising candidate biomarkers. The analysis of circulating lncRNAs in peripheral blood represents a potential option for minimally invasive diagnostic tests based on liquid biopsy samples. The present review aimed to evaluate the efficacy of lncRNAs with altered expression levels in peripheral blood as diagnostic markers for CRC. Additionally, the clinical significance of lncRNAs as prognostic markers for this disease were summarized.
Collapse
Affiliation(s)
- Yuning Lin
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, Fujian 361009, P.R. China
| | - Wenzhen Zhao
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, Fujian 361009, P.R. China
| | - Ruonan Pu
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, Fujian 361009, P.R. China
| | - Zhenyi Lv
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, Fujian 361009, P.R. China
| | - Hongyan Xie
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, Fujian 361009, P.R. China
| | - Ying Li
- Department of Ultrasonography, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Zhongying Zhang
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, Fujian 361009, P.R. China
| |
Collapse
|
7
|
Wang J, Ying L, Xiong H, Zhou DR, Wang YX, Che HL, Zhong ZF, Wu GS, Ge YJ. Comprehensive analysis of stearoyl-coenzyme A desaturase in prostate adenocarcinoma: insights into gene expression, immune microenvironment and tumor progression. Front Immunol 2024; 15:1460915. [PMID: 39351232 PMCID: PMC11439642 DOI: 10.3389/fimmu.2024.1460915] [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: 07/07/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
Prostate adenocarcinoma (PRAD) is a prevalent global malignancy which depends more on lipid metabolism for tumor progression compared to other cancer types. Although Stearoyl-coenzyme A desaturase (SCD) is documented to regulate lipid metabolism in multiple cancers, landscape analysis of its implications in PRAD are still missing at present. Here, we conducted an analysis of diverse cancer datasets revealing elevated SCD expression in the PRAD cohort at both mRNA and protein levels. Interestingly, the elevated expression was associated with SCD promoter hypermethylation and genetic alterations, notably the L134V mutation. Integration of comprehensive tumor immunological and genomic data revealed a robust positive correlation between SCD expression levels and the abundance of CD8+ T cells and macrophages. Further analyses identified significant associations between SCD expression and various immune markers in tumor microenvironment. Single-cell transcriptomic profiling unveiled differential SCD expression patterns across distinct cell types within the prostate tumor microenvironment. The Gene Ontology and Kyoto Encyclopedia of Genes and Genome analyses showed that SCD enriched pathways were primarily related to lipid biosynthesis, cholesterol biosynthesis, endoplasmic reticulum membrane functions, and various metabolic pathways. Gene Set Enrichment Analysis highlighted the involvement of elevated SCD expression in crucial cellular processes, including the cell cycle and biosynthesis of cofactors pathways. In functional studies, SCD overexpression promoted the proliferation, metastasis and invasion of prostate cancer cells, whereas downregulation inhibits these processes. This study provides comprehensive insights into the multifaceted roles of SCD in PRAD pathogenesis, underscoring its potential as both a therapeutic target and prognostic biomarker.
Collapse
Affiliation(s)
- Jie Wang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Liang Ying
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - He Xiong
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Duan-Rui Zhou
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yi-Xuan Wang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Hui-Lian Che
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zhang-Feng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
| | - Guo-Sheng Wu
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yun-Jun Ge
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
8
|
Yu X, Xu J, Song B, Zhu R, Liu J, Liu YF, Ma YJ. The role of epigenetics in women's reproductive health: the impact of environmental factors. Front Endocrinol (Lausanne) 2024; 15:1399757. [PMID: 39345884 PMCID: PMC11427273 DOI: 10.3389/fendo.2024.1399757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 08/28/2024] [Indexed: 10/01/2024] Open
Abstract
This paper explores the significant role of epigenetics in women's reproductive health, focusing on the impact of environmental factors. It highlights the crucial link between epigenetic modifications-such as DNA methylation and histones post-translational modifications-and reproductive health issues, including infertility and pregnancy complications. The paper reviews the influence of pollutants like PM2.5, heavy metals, and endocrine disruptors on gene expression through epigenetic mechanisms, emphasizing the need for understanding how dietary, lifestyle choices, and exposure to chemicals affect gene expression and reproductive health. Future research directions include deeper investigation into epigenetics in female reproductive health and leveraging gene editing to mitigate epigenetic changes for improving IVF success rates and managing reproductive disorders.
Collapse
Affiliation(s)
- Xinru Yu
- College Of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jiawei Xu
- College Of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine School, Jinan, Shandong, China
| | - Bihan Song
- College Of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine School, Jinan, Shandong, China
| | - Runhe Zhu
- College Of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine School, Jinan, Shandong, China
| | - Jiaxin Liu
- College Of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yi Fan Liu
- Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ying Jie Ma
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
9
|
Trujano-Camacho S, Cantú-de León D, Pérez-Yepez E, Contreras-Romero C, Coronel-Hernandez J, Millan-Catalan O, Rodríguez-Dorantes M, López-Camarillo C, Gutiérrez-Ruiz C, Jacobo-Herrera N, Pérez-Plasencia C. HOTAIR Promotes the Hyperactivation of PI3K/Akt and Wnt/β-Catenin Signaling Pathways via PTEN Hypermethylation in Cervical Cancer. Cells 2024; 13:1484. [PMID: 39273054 PMCID: PMC11394386 DOI: 10.3390/cells13171484] [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: 08/01/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
The mechanisms underlying the sustained activation of the PI3K/AKT and Wnt/β-catenin pathways mediated by HOTAIR in cervical cancer (CC) have not been extensively described. To address this knowledge gap in the literature, we explored the interactions between these pathways by driving HOTAIR expression levels in HeLa cells. Our findings reveal that HOTAIR is a key regulator in sustaining the activation of both signaling pathways. Specifically, altering HOTAIR expression-either by knockdown or overexpression-significantly influenced the transcriptional activity of the PI3K/AKT and Wnt/β-catenin pathways. Additionally, we discovered that HIF1α directly induces HOTAIR transcription, which in turn leads to the epigenetic silencing of the PTEN promoter via DNMT1. This process leads to the sustained activation of both pathways, highlighting a novel regulatory axis involving HOTAIR and HIF1α in cervical cancer. Our results suggest a new model in which HOTAIR sustains reciprocal activation of the PI3K/AKT and Wnt/β-catenin pathways through the HOTAIR/HIF1α axis, thereby contributing to the oncogenic phenotype of cervical cancer.
Collapse
Affiliation(s)
- Samuel Trujano-Camacho
- Experimental Biology PhD Program, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico;
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (D.C.-d.L.); (E.P.-Y.); (C.C.-R.); (J.C.-H.); (O.M.-C.)
| | - David Cantú-de León
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (D.C.-d.L.); (E.P.-Y.); (C.C.-R.); (J.C.-H.); (O.M.-C.)
| | - Eloy Pérez-Yepez
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (D.C.-d.L.); (E.P.-Y.); (C.C.-R.); (J.C.-H.); (O.M.-C.)
| | - Carlos Contreras-Romero
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (D.C.-d.L.); (E.P.-Y.); (C.C.-R.); (J.C.-H.); (O.M.-C.)
| | - Jossimar Coronel-Hernandez
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (D.C.-d.L.); (E.P.-Y.); (C.C.-R.); (J.C.-H.); (O.M.-C.)
| | - Oliver Millan-Catalan
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (D.C.-d.L.); (E.P.-Y.); (C.C.-R.); (J.C.-H.); (O.M.-C.)
| | | | - Cesar López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Ciudad de México 03100, Mexico;
| | - Concepción Gutiérrez-Ruiz
- Laboratory of Experimental Medicine, Translational Medicine Unit, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cardiología Ignacio Chávez, Tlalpan, Mexico City 14080, Mexico;
- Department of Health Sciences, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico
| | - Nadia Jacobo-Herrera
- Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Av. Vasco de Quiroga 15, Col. Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico;
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (D.C.-d.L.); (E.P.-Y.); (C.C.-R.); (J.C.-H.); (O.M.-C.)
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla 54090, Mexico
| |
Collapse
|
10
|
Li P, Ma X, Huang D. Role of the lncRNA/Wnt signaling pathway in digestive system cancer: a literature review. Eur J Med Res 2024; 29:447. [PMID: 39218950 PMCID: PMC11367813 DOI: 10.1186/s40001-024-02033-w] [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: 03/24/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
The long noncoding RNA (lncRNA)/Wingless (Wnt) axis is often dysregulated in digestive system tumors impacting critical cellular processes. Abnormal expression of specific Wnt-related lncRNAs such as LINC01606 (promotes motility), SLCO4A1-AS1 (promotes motility), and SH3BP5-AS1 (induces chemoresistance), plays a crucial role in these malignancies. These lncRNAs are promising targets for cancer diagnosis and therapy, offering new treatment perspectives. The lncRNAs, NEF and GASL1, differentially expressed in plasma show diagnostic potential for esophageal squamous cell carcinoma and gastric cancer, respectively. Additionally, Wnt pathway inhibitors like XAV-939 have demonstrated preclinical efficacy, underscoring their therapeutic potential. This review comprehensively analyzes the lncRNA/Wnt axis, highlighting its impact on cell proliferation, motility, and chemoresistance. By elucidating the complex molecular mechanisms of the lncRNA/Wnt axis, we aim to identify potential therapeutic targets for digestive system tumors to pave the way for the development of targeted treatment strategies.
Collapse
Affiliation(s)
- Penghui Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China.
| | - Xiao Ma
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Di Huang
- Department of Child Health Care, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| |
Collapse
|
11
|
Wu L, Cheng Y, Wang R, Sun S, Ma B, Zhang Z. NDRG2 regulates glucose metabolism and ferroptosis of OGD/R-treated astrocytes by the Wnt/β-catenin signaling. J Biochem Mol Toxicol 2024; 38:e23827. [PMID: 39193856 DOI: 10.1002/jbt.23827] [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: 10/14/2023] [Revised: 07/10/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Ischemic stroke is one main type of cerebrovascular disorders with leading cause of death and disability worldwide. Astrocytes are the only nerve cell type storing glycogen in the brain, which regulate the glucose metabolism and handle the energy supply and survive of neurons. Astrocyte ferroptosis contributes to neuron injury in brain disorders. N-myc downstream-regulated gene 2 (NDRG2) has been implicated in the progression of brain diseases, including ischemic stroke. However, whether NDRG2 could affect the glucose metabolism and ferroptosis of astrocytes during ischemic stroke remains largely unknown. Mouse astrocytes were treated with oxygen-glucose deprivation/reoxygenation (OGD/R) to establish the in vitro model. Glial fibrillary acidic protein, NDRG2, Wnt3a and β-catenin expression levels were detected by immunofluorescence staining and western blot analyses. Glucose metabolism was investigated by glucose uptake, lactate production, nicotinamide adenine dinucleotide phosphate hydrogen/nicotinamide adenine dinucleotide phosphate (NADPH/NADP+), ATP and glycolysis enzymes (HK2, PKM2 and lactate dehydrogenase A [LDHA]) levels. Ferroptosis was assessed via reactive oxygen species (ROS), glutathione (GSH), iron and ferroptosis-related markers (GPX4 and PTGS2) contents. Glycolysis enzymes and ferroptosis-related markers levels were measured via western blot. NDRG2 expression was elevated in OGD/R-induced astrocytes. NDRG2 overexpression aggravated OGD/R-induced loss of glucose metabolism through reducing glucose uptake, lactate production, NADPH/NADP+ and ATP levels. NDRG2 upregulation exacerbated OGD/R-caused reduction of glycolysis enzymes (HK2, PKM2 and LDHA) levels. NDRG2 promoted OGD/R-induced ferroptosis of astrocytes by increasing ROS, iron and PTGS2 levels and decreasing GSH and GPX4 levels. NDRG2 overexpression enhanced OGD/R-induced decrease of Wnt/β-catenin signaling activation by reducing Wnt3a and β-catenin expression. NDRG2 silencing played an opposite effect. Inhibition of Wnt/β-catenin signaling activation by IWR-1 attenuated the influences of NDRG2 knockdown on glucose metabolism, glycolysis enzymes levels and ferroptosis. These findings demonstrated that NDRG2 contributes to OGD/R-induced inhibition of glucose metabolism and promotion of ferroptosis in astrocytes through inhibiting Wnt/β-catenin signaling activation, which might be associated with ischemic stroke progression.
Collapse
Affiliation(s)
- Lin Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Yingying Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Runfeng Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Shukai Sun
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Bo Ma
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhiguo Zhang
- Department of Neurosurgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
12
|
Huang K, Yu L, Lu D, Zhu Z, Shu M, Ma Z. Long non-coding RNAs in ferroptosis, pyroptosis and necroptosis: from functions to clinical implications in cancer therapy. Front Oncol 2024; 14:1437698. [PMID: 39267831 PMCID: PMC11390357 DOI: 10.3389/fonc.2024.1437698] [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: 05/24/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
As global population ageing accelerates, cancer emerges as a predominant cause of mortality. Long non-coding RNAs (lncRNAs) play crucial roles in cancer cell growth and death, given their involvement in regulating downstream gene expression levels and numerous cellular processes. Cell death, especially non-apoptotic regulated cell death (RCD), such as ferroptosis, pyroptosis and necroptosis, significantly impacts cancer proliferation, invasion and metastasis. Understanding the interplay between lncRNAs and the diverse forms of cell death in cancer is imperative. Modulating lncRNA expression can regulate cancer onset and progression, offering promising therapeutic avenues. This review discusses the mechanisms by which lncRNAs modulate non-apoptotic RCDs in cancer, highlighting their potential as biomarkers for various cancer types. Elucidating the role of lncRNAs in cell death pathways provides valuable insights for personalised cancer interventions.
Collapse
Affiliation(s)
- Ke Huang
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Li Yu
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Dingci Lu
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Ziyi Zhu
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Min Shu
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Zhaowu Ma
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
13
|
Liu C, Zou Z, Lu S, Jin K, Shen Y, Huang T, Li W, Zhou G. CircPKN2 promotes ferroptosis in bladder cancer by promoting the ubiquitination of Stearoyl-CoA Desaturase 1. Cancer Gene Ther 2024; 31:1251-1265. [PMID: 38802550 DOI: 10.1038/s41417-024-00784-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 05/05/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024]
Abstract
Bladder cancer (BC) is one of the most common malignancies in the male urinary system and currently lacks an optimal treatment strategy. To elucidate the pathogenic mechanisms of BC from the perspective of circular RNAs, we conducted this study. Building upon our previous research, a novel circRNA, circPKN2, captured our interest due to its significant downregulation in BC, and its close association with the prognosis of BC patients. Our research findings indicate that circPKN2 can inhibit the proliferation and migration of BC cells in vitro. Furthermore, we discovered that circPKN2 exerts its anti-cancer effects in BC by promoting ferroptosis. Mechanistic studies revealed that circPKN2 recruits STUB1 to facilitate the ubiquitination of SCD1, thereby suppressing the WNT pathway and promoting ferroptosis in BC. Additionally, our research unveiled the regulatory role of the splicing factor QKI in the biogenesis of circPKN2. Animal studies demonstrated that circPKN2 enhances ferroptosis in BC cells in vivo, inhibiting tumor growth and metastasis. The discovery of the anti-cancer factor circPKN2 holds promise for providing new therapeutic targets in the prevention and treatment of BC.
Collapse
Affiliation(s)
- Changkun Liu
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
| | - Zhuo Zou
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Shengming Lu
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Kun Jin
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Ye Shen
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Tianbao Huang
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Weijian Li
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Guangchen Zhou
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| |
Collapse
|
14
|
Xu D, Han S, Yue X, Xu X, Huang T. METTL14 Suppresses Tumor Stemness and Metastasis of Colon Cancer Cells by Modulating m6A-Modified SCD1. Mol Biotechnol 2024; 66:2095-2105. [PMID: 37592151 DOI: 10.1007/s12033-023-00843-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 07/27/2023] [Indexed: 08/19/2023]
Abstract
Colon cancer (CC) is a malignant disease of the digestive tract, and its rising prevalence poses a grave threat to people's health. N6-methyladenosine (m6A) modification is essential for various crucial life processes through modulating gene expression. Methyltransferase-like 14 (METTL14), the m6A methylation transferase core protein, and its aberrant expression is intimately correlated to tumor development. This study was conducted to probe the impacts and specific mechanisms of METTL14 on the biological process of CC. Bioinformatics data disclosed that METTL14 was significantly attenuated in CC. Functional assays were executed to ascertain how METTL14 affected CC tumorigenicity, and METTL14 overexpression caused a notable decline in viability, migration, invasion, and stemness phenotype of CC cells. Then, in-depth mechanistic studies displayed that stearoyl-CoA desaturase 1 (SCD1) was a downstream target gene of METTL14-mediated m6A modification. METTL14 overexpression substantially augmented the m6A modification of SCD1 mRNA and diminished the SCD1 mRNA level. In addition, we revealed that YTHDF2 was the m6A reader to recognize METTL14 m6A-modified SCD1 mRNA and abolish its stability. Finally, we also validated that METTL14 might impede the tumorigenic process of CC through SCD1 mediated Wnt/β-catenin signaling. Taken together, this study presented that METTL14 performed as a potential therapeutic target in CC with important implications for the prognosis amelioration of CC patients.
Collapse
Affiliation(s)
- Dehua Xu
- Gastrointestinal Surgery, Suzhou Xiangcheng Peoples' Hospital, No. 1060, Huayuan Road, Xiangcheng District, Suzhou, 215131, Jiangsu, China
| | - Shuguang Han
- Gastrointestinal Surgery, Suzhou Xiangcheng Peoples' Hospital, No. 1060, Huayuan Road, Xiangcheng District, Suzhou, 215131, Jiangsu, China
| | - Xiaoguang Yue
- Gastrointestinal Surgery, Suzhou Xiangcheng Peoples' Hospital, No. 1060, Huayuan Road, Xiangcheng District, Suzhou, 215131, Jiangsu, China
| | - Xiangyu Xu
- Gastrointestinal Surgery, Suzhou Xiangcheng Peoples' Hospital, No. 1060, Huayuan Road, Xiangcheng District, Suzhou, 215131, Jiangsu, China
| | - Tieao Huang
- Gastrointestinal Surgery, Suzhou Xiangcheng Peoples' Hospital, No. 1060, Huayuan Road, Xiangcheng District, Suzhou, 215131, Jiangsu, China.
| |
Collapse
|
15
|
Mao X, Wang L, Chen Z, Huang H, Chen J, Su J, Li Z, Shen G, Ren Y, Li Z, Wang W, Ou J, Guo W, Hu Y. SCD1 promotes the stemness of gastric cancer stem cells by inhibiting ferroptosis through the SQLE/cholesterol/mTOR signalling pathway. Int J Biol Macromol 2024; 275:133698. [PMID: 38972654 DOI: 10.1016/j.ijbiomac.2024.133698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Cancer stem cells (CSCs) play a substantial role in cancer onset and recurrence. Anomalous iron and lipid metabolism have been documented in CSCs, suggesting that ferroptosis, a recently discovered form of regulated cell death characterised by lipid peroxidation, could potentially exert a significant influence on CSCs. However, the precise role of ferroptosis in gastric cancer stem cells (GCSCs) remains unknown. To address this gap, we screened ferroptosis-related genes in GCSCs using The Cancer Genome Atlas and corroborated our findings through quantitative polymerase chain reaction and western blotting. These results indicate that stearoyl-CoA desaturase (SCD1) is a key player in the regulation of ferroptosis in GCSCs. This study provides evidence that SCD1 positively regulates the transcription of squalene epoxidase (SQLE) by eliminating transcriptional inhibition of P53. This mechanism increases the cholesterol content and the elevated cholesterol regulated by SCD1 inhibits ferroptosis via the mTOR signalling pathway. Furthermore, our in vivo studies showed that SCD1 knockdown or regulation of cholesterol intake affects the stemness of GCSCs and their sensitivity to ferroptosis inducers. Thus, targeting the SCD1/squalene epoxidase/cholesterol signalling axis in conjunction with ferroptosis inducers may represent a promising therapeutic approach for the treatment of gastric cancer based on GCSCs.
Collapse
Affiliation(s)
- Xinyuan Mao
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Lingzhi Wang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Zhian Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Huilin Huang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Jialin Chen
- Hepatobiliary and Pancreatic Center, The First Affiliated Hospital, Sun Yat-sen University, 510515, PR China
| | - Jin Su
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China; Department of General Surgery, Zhuzhou Hospital affiliated to Xiangya School of Medicine, Central South University, Zhuzhou 412000, PR China
| | - Zhenhao Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Guodong Shen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Yingxin Ren
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Zhenyuan Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Weisheng Wang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Jinzhou Ou
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Weihong Guo
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China.
| | - Yanfeng Hu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
16
|
Xu L, Wen B, Wu Q, Lu S, Liao J, Mo L, Li Q, Tong X, Yan H. Long non-coding RNA KB-1460A1.5 promotes ferroptosis by inhibiting mTOR/SREBP-1/SCD1-mediated polyunsaturated fatty acid desaturation in glioma. Carcinogenesis 2024; 45:487-499. [PMID: 38422369 DOI: 10.1093/carcin/bgae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/03/2024] [Accepted: 02/28/2024] [Indexed: 03/02/2024] Open
Abstract
Ferroptosis is a new form of regulated cell death caused by the iron-dependent peroxidation of phospholipids and is related to cell metabolism, redox homeostasis and various signalling pathways related to cancer. The long non-coding RNA (lncRNA) KB-1460A1.5 acts as a tumour suppressor gene to regulate tumour growth in gliomas, but its molecular network regulatory mechanism is still unclear. In this study, we found that KB-1460A1.5 can induce ferroptosis in glioma and enhance sensitivity to RSL3, a ferroptosis inducer. Tandem mass tag proteomics and nontargeted metabolomics suggest that KB-1460A1.5 affects polyunsaturated fatty acid metabolic processes. Gas chromatography-mass spectrometry-based medium- and long-chain fatty acid-targeted metabolomics confirmed that upregulation of KB-1460A1.5 decreased the levels of monounsaturated fatty acids, oleic acid (OA) and palmitoleic acid (PO) in glioma cells. The addition of OA and PO restored KB-1460A1.5-induced cellular ferroptosis. Molecularly, KB-1460A1.5 inhibited the mammalian target of rapamycin signalling pathway to suppress the expression of downstream sterol regulatory element-binding protein 1 (SREBP-1), thereby attenuating the stearoyl-CoA desaturase-1 (SCD1)-mediated desaturation of polyunsaturated fatty acids. Finally, an animal model of subcutaneous glioma confirmed that KB-1460A1.5 could inhibit tumour progression, SREBP-1/SCD1 expression and ferroptosis. In conclusion, increasing the expression level of KB-1460A1.5 in glioma can promote the induction of oxidative stress and ferroptosis in cancer cells through SREBP-1/SCD1-mediated adipogenesis, demonstrating therapeutic potential in preclinical models.
Collapse
Affiliation(s)
- Lixia Xu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Key Laboratory of Cerebral Blood Flow Reconstruction and Head and Neck Tumour New Technology Translation, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300070, China
| | - Binli Wen
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300070, China
| | - Qiaoli Wu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Key Laboratory of Cerebral Blood Flow Reconstruction and Head and Neck Tumour New Technology Translation, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300070, China
| | - Shan Lu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Key Laboratory of Cerebral Blood Flow Reconstruction and Head and Neck Tumour New Technology Translation, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300070, China
| | - Jianwen Liao
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Key Laboratory of Cerebral Blood Flow Reconstruction and Head and Neck Tumour New Technology Translation, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300070, China
| | - Lidong Mo
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Key Laboratory of Cerebral Blood Flow Reconstruction and Head and Neck Tumour New Technology Translation, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300070, China
| | - Qingguo Li
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Key Laboratory of Cerebral Blood Flow Reconstruction and Head and Neck Tumour New Technology Translation, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300070, China
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Xiaoguang Tong
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Key Laboratory of Cerebral Blood Flow Reconstruction and Head and Neck Tumour New Technology Translation, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300070, China
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Hua Yan
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Key Laboratory of Cerebral Blood Flow Reconstruction and Head and Neck Tumour New Technology Translation, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300070, China
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350, China
| |
Collapse
|
17
|
Xiao F, Zhu H, Xiong Y, Guo Y, Zhang Z, Zeng J, Xiao Y, Liao B, Shang X, Zhao S, Hu G, Huang K, Guo H. Positive feedback loop of c-myc/XTP6/NDH2/NF-κB to promote malignant progression in glioblastoma. J Exp Clin Cancer Res 2024; 43:187. [PMID: 38965580 PMCID: PMC11225266 DOI: 10.1186/s13046-024-03109-5] [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: 04/18/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Recent studies have highlighted the significant role of the NF-κB signaling pathway in the initiation and progression of cancer. Furthermore, long noncoding RNAs (lncRNAs) have been identified as pivotal regulators in sustaining the NF-κB signaling pathway's functionality. Despite these findings, the underlying molecular mechanisms through which lncRNAs influence the NF-κB pathway remain largely unexplored. METHODS Bioinformatic analyses were utilized to investigate the differential expression and prognostic significance of XTP6. The functional roles of XTP6 were further elucidated through both in vitro and in vivo experimental approaches. To estimate the interaction between XTP6 and NDH2, RNA pulldown and RNA Immunoprecipitation (RIP) assays were conducted. The connection between XTP6 and the IκBα promoter was examined using Chromatin Isolation by RNA Purification (ChIRP) assays. Additionally, Chromatin Immunoprecipitation (ChIP) assays were implemented to analyze the binding affinity of c-myc to the XTP6 promoter, providing insights into the regulatory mechanisms at play. RESULTS XTP6 was remarkedly upregulated in glioblastoma multiforme (GBM) tissues and was connected with adverse prognosis in GBM patients. Our investigations revealed that XTP6 can facilitate the malignant progression of GBM both in vitro and in vivo. Additionally, XTP6 downregulated IκBα expression by recruiting NDH2 to the IκBα promoter, which resulted in elevated levels of H3K27me3, thereby reducing the transcriptional activity of IκBα. Moreover, the progression of GBM was further driven by the c-myc-mediated upregulation of XTP6, establishing a positive feedback loop with IκBα that perpetuated the activation of the NF-κB signaling pathway. Notably, the application of an inhibitor targeting the NF-κB signaling pathway effectively inhibited the continuous activation induced by XTP6, leading to a significant reduction in tumor formation in vivo. CONCLUSION The results reveal that XTP6 unveils an innovative epigenetic mechanism instrumental in the sustained activation of the NF-κB signaling pathway, suggesting a promising therapeutic target for the treatment of GBM.
Collapse
Affiliation(s)
- Feng Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Hong Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yaping Xiong
- Departments of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yun Guo
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zhe Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Jie Zeng
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yao Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Bin Liao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Xuesong Shang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Siyi Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Guowen Hu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Kai Huang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Hua Guo
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China.
- JXHC key Laboratory of Neurological medicine, Nanchang University, Nanchang, Jiangxi, 330006, China.
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi, 330006, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
18
|
Lin Y, Zhao W, Lv Z, Xie H, Li Y, Zhang Z. The functions and mechanisms of long non-coding RNA in colorectal cancer. Front Oncol 2024; 14:1419972. [PMID: 39026978 PMCID: PMC11254705 DOI: 10.3389/fonc.2024.1419972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
CRC poses a significant challenge in the global health domain, with a high number of deaths attributed to this disease annually. If CRC is detected only in its advanced stages, the difficulty of treatment increases significantly. Therefore, biomarkers for the early detection of CRC play a crucial role in improving patient outcomes and increasing survival rates. The development of a reliable biomarker for early detection of CRC is particularly important for timely diagnosis and treatment. However, current methods for CRC detection, such as endoscopic examination, blood, and stool tests, have certain limitations and often only detect cases in the late stages. To overcome these constraints, researchers have turned their attention to molecular biomarkers, which are considered a promising approach to improving CRC detection. Non-invasive methods using biomarkers such as mRNA, circulating cell-free DNA, microRNA, LncRNA, and proteins can provide more reliable diagnostic information. These biomarkers can be found in blood, tissue, stool, and volatile organic compounds. Identifying molecular biomarkers with high sensitivity and specificity for the early and safe, economic, and easily measurable detection of CRC remains a significant challenge for researchers.
Collapse
Affiliation(s)
- Yuning Lin
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, China
| | - Wenzhen Zhao
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, China
| | - Zhenyi Lv
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, China
| | - Hongyan Xie
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, China
| | - Ying Li
- Ultrasonography Department, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Zhongying Zhang
- Medical Laboratory, Xiamen Humanity Hospital, Fujian Medical University, Xiamen, China
| |
Collapse
|
19
|
Wang J, Zhang Q, Zhou D, Wang Y, Che H, Ge Y, Zhong Z, Wu G. Systematic analysis of fatty acid desaturases in breast invasive carcinoma: The prognosis, gene mutation, and tumor immune microenvironment. Medicine (Baltimore) 2024; 103:e38597. [PMID: 38905386 PMCID: PMC11191958 DOI: 10.1097/md.0000000000038597] [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: 04/29/2024] [Accepted: 05/24/2024] [Indexed: 06/23/2024] Open
Abstract
Breast invasive carcinoma (BRCA) is one of the most common cancers in women, with its malignant progression significantly influenced by intracellular fatty acid (FA) desaturation. Stearoyl-coenzyme A desaturase (SCD) and fatty acid desaturase 2 (FADS2) are two key rate-limiting enzymes that catalyze the FA desaturation process and cooperate to accelerate lipid metabolic activities. In this study, we investigated the potential functions of SCD and FADS2 in BRCA using bioinformatic analysis and experimental validation. The gene expression profiling interactive analysis database showed that the expression of SCD or FADS2 genes was positively linked to worse overall survival and disease-free survival in the Cancer Genome Atlas database-BRCA. The University of Alabama at Birmingham cancer data analysis portal database indicates that the expression and methylation levels of SCD or FADS2 are associated with various clinicopathological factors in patients with BRCA. Moreover, the tumor immune estimation resource and TISCH databases showed a significant positive correlation between the expression of SCD and the abundance of CD8+ T cells and macrophage cell infiltration, while the expression of FADS2 was positively correlated with the abundance of B cells. Meanwhile, SCD or FADS2 had a higher expression in monocytes/macrophages analyzed the BRCA_GSE143423 and BRCA_GSE114727_inDrop datasets. Mechanistically, the Search Tool for the Retrieval of Distant Genes and CancerSEA databases showed that SCD and FADS2 were upregulated in several cell biology signaling pathways, particularly in inflammation, apoptosis, and DNA repair. Finally, SCD or FADS2 knockdown inhibited the proliferation of MCF-7 and MDA-MB-231 cells. In summary, SCD and FADS2 play significant roles in BRCA development, suggesting that they may serve as potential therapeutic targets for BRCA treatment.
Collapse
Affiliation(s)
- Jie Wang
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Qian Zhang
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Duanrui Zhou
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yixuan Wang
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Huilian Che
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yunjun Ge
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Guosheng Wu
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Jiangnan University Medical Center, Wuxi, China
| |
Collapse
|
20
|
Song P, Gao Z, Bao Y, Chen L, Huang Y, Liu Y, Dong Q, Wei X. Wnt/β-catenin signaling pathway in carcinogenesis and cancer therapy. J Hematol Oncol 2024; 17:46. [PMID: 38886806 PMCID: PMC11184729 DOI: 10.1186/s13045-024-01563-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
The Wnt/β-catenin signaling pathway plays a crucial role in various physiological processes, encompassing development, tissue homeostasis, and cell proliferation. Under normal physiological conditions, the Wnt/β-catenin signaling pathway is meticulously regulated. However, aberrant activation of this pathway and downstream target genes can occur due to mutations in key components of the Wnt/β-catenin pathway, epigenetic modifications, and crosstalk with other signaling pathways. Consequently, these dysregulations contribute significantly to tumor initiation and progression. Therapies targeting the Wnt/β-catenin signaling transduction have exhibited promising prospects and potential for tumor treatment. An increasing number of medications targeting this pathway are continuously being developed and validated. This comprehensive review aims to summarize the latest advances in our understanding of the role played by the Wnt/β-catenin signaling pathway in carcinogenesis and targeted therapy, providing valuable insights into acknowledging current opportunities and challenges associated with targeting this signaling pathway in cancer research and treatment.
Collapse
Affiliation(s)
- Pan Song
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Zirui Gao
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yige Bao
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Li Chen
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yuhe Huang
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Yanyan Liu
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China
| | - Qiang Dong
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Agent Target, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, P.R. China.
| |
Collapse
|
21
|
Mai Z, Li Y, Zhang L, Zhang H. Citraconate promotes the malignant progression of colorectal cancer by inhibiting ferroptosis. Am J Cancer Res 2024; 14:2790-2804. [PMID: 39005662 PMCID: PMC11236773 DOI: 10.62347/lwrs3363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/24/2024] [Indexed: 07/16/2024] Open
Abstract
Metastasis is a principal factor in the poor prognosis of colorectal cancer. Recent studies have found microbial metabolites regulate colorectal cancer metastasis. By analyzing metabolomics data, we identified an essential fecal metabolite citraconate that potentially promotes colorectal cancer metastasis. Next, we tried to reveal its effect on colorectal cancer and the underlying mechanism. Firstly, the response of colorectal cancer cells (HCT116 and MC38 cells) to citraconate was assessed by Cell Counting Kit-8 assay, clonogenic assay, transwell migration and invasion assay. Moreover, we utilized an intra-splenic injection model to evaluate the effect of citraconate on colorectal cancer liver metastasis in vivo. Then molecular approaches were employed, including RNA sequencing, mass spectrometry-based metabolomics, western blot, quantitative real-time PCR, cell ferrous iron colorimetric assay and intracellular malondialdehyde measurement. In vitro, citraconate promotes the growth of colorectal cancer cells. In vivo, citraconate aggravated liver metastasis of colorectal cancer. Mechanistically, downstream genes of NRF2, NQO1, GCLC, and GCLM high expression induced by citraconate resulted in resistance to ferroptosis of colorectal cancer cells. In summary, citraconate promotes the malignant progression of colorectal cancer through NRF2-mediated ferroptosis resistance in colorectal cancer cells. Furthermore, our study indicates that fecal metabolite may be crucial in colorectal cancer development.
Collapse
Affiliation(s)
- Zongjiong Mai
- The Fifth Affiliated Hospital of Sun Yat-sen University Zhuhai, Guangdong, China
| | - Yanyu Li
- The Fifth Affiliated Hospital of Sun Yat-sen University Zhuhai, Guangdong, China
| | - Lei Zhang
- The Fifth Affiliated Hospital of Sun Yat-sen University Zhuhai, Guangdong, China
| | - Hongyu Zhang
- The Fifth Affiliated Hospital of Sun Yat-sen University Zhuhai, Guangdong, China
| |
Collapse
|
22
|
Hang X, Du Z, Song J. Exploring the prognostic significance of iron death‑related lncRNAs in colorectal cancer: A systematic review and meta‑analysis. Oncol Lett 2024; 27:284. [PMID: 38736739 PMCID: PMC11083912 DOI: 10.3892/ol.2024.14417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 04/05/2024] [Indexed: 05/14/2024] Open
Abstract
Colorectal cancer is a significant health challenge globally, with its prognosis associated with the profile of molecular biomarkers. Recently, the role of iron death-associated long non-coding (lnc)RNAs in cancer development has garnered attention; however, their expression patterns and prognostic value in colorectal cancer remain poorly elucidated. The present study aimed to assess the expression levels of iron death-related lncRNAs in colorectal cancer tissues and evaluate their relationship with patient outcomes through a comprehensive meta-analysis. Systematic searches were performed across multiple databases, including PubMed, Embase, Web of Science, Cochrane Library, CNKI, Wanfang and VIP databases, to identify studies relevant to the objective of the present study. Selected studies met predetermined inclusion criteria, from which data were extracted. R software version 4.3.1 was used for the meta-analysis, evaluating the association between iron death-related lncRNAs expression and colorectal cancer prognosis. Publication bias was assessed using funnel plot analysis, and Begg's and Egger's test. A total of 11 studies, including 3,984 patients with colorectal cancer, were included in the present meta-analysis. The results demonstrated a significant association between iron death-related lncRNAs and tumor stage classification [odds ratio (OR)=2.00; 95% confidence interval (CI), 1.77-2.24]. Notably, a significant association was also revealed between iron death-related lncRNAs and T stage classification in colorectal cancer (OR=1.82; 95% CI, 1.50-2.20). Furthermore, a statistically significant association was demonstrated between iron death-related lncRNAs and lymph node metastasis in colorectal cancer (OR=1.31; 95% CI, 1.03-1.66). The findings also highlighted a significant association between iron death-associated lncRNA and distant metastasis in colon cancer (OR=2.04; 95% CI, 1.62-2.56). Moreover, a significant association between iron death-related lncRNAs and risk score in colorectal cancer was revealed (OR=1.75; 95% CI, 1.25-2.46). In conclusion, the findings of the present meta-analysis underscore the potential of high ferroptosis-associated lncRNA expression as an indicator of adverse outcomes in colorectal cancer, suggesting their viability as biomarkers for cancer progression and prognosis. This insight opens potential new avenues for clinical application and further research into colorectal cancer management.
Collapse
Affiliation(s)
- Xiaoli Hang
- Clinical Laboratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang 212300, P.R. China
| | - Zhongxiang Du
- Clinical Laboratory, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang 212300, P.R. China
| | - Jie Song
- Clinical Laboratory, Nantong Fourth People's Hospital, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
23
|
Yao H, Liu P, Yao L, Li X. Establishment of disulfidptosis-related LncRNA signature as biomarkers in colon adenocarcinoma. Cancer Cell Int 2024; 24:183. [PMID: 38802854 PMCID: PMC11131243 DOI: 10.1186/s12935-024-03374-6] [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: 12/12/2023] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
PURPOSE Metabolic reprogramming is a hallmark of cancer and plays a key role in precision oncology treatment. Long non-coding RNAs (lncRNAs) regulate cancer cell behavior, including metabolism. Disulfidptosis, a newly identified form of regulated cell death triggered by glucose starvation, has yet to be fully understood in colon adenocarcinoma (COAD). This study aimed to confirm the existence and role of disulfidptosis in COAD and identify disulfidptosis-related lncRNAs that may be targeted to induce disulfidptosis in COAD. METHODS PI and F-actin staining were used to observe disulfidptosis in COAD cell lines. Disulfidptosis-related lncRNAs were identified based on the expression of disulfidptosis-associated genes in the TCGA-COAD database. A four-lncRNA signature for disulfidptosis was established. Subsequently, loss-of-function assays explored the roles of AC013652.1 and MCM3AP-AS1 in disulfidptosis. RESULTS Disulfidptosis was observed in COAD cells under glucose starvation and could be reversed by agents that prevent disulfide stress, such as dithiothreitol (DTT) and tris-(2-carboxyethyl)-phosphine (TCEP). The prognostic value of disulfidptosis-associated genes in COAD patients was confirmed, with higher expression indicating longer survival. A disulfidptosis-related lncRNA signature comprising four lncRNAs was established based on the expression of these genes. Among these, AC013652.1 and MCM3AP-AS1 predicted worse prognoses. Furthermore, inhibiting AC013652.1 or MCM3AP-AS1 increased disulfidptosis-associated gene expression and cellular death, which could be reversed by DTT and TCEP. CONCLUSIONS This study provides hitherto undocumented evidence of the existence of disulfidptosis and the prognostic value of disulfidptosis-associated genes in COAD. Importantly, we identified lncRNAs AC013652.1 and MCM3AP-AS1, which suppress disulfidptosis and may serve as potential therapeutic targets for COAD.
Collapse
Affiliation(s)
- Hongfei Yao
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People's Republic of China
| | - Peng Liu
- Department of Radiotherapy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Linli Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China.
| | - Xiao Li
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People's Republic of China.
| |
Collapse
|
24
|
Chen H, Han Z, Su J, Song X, Ma Q, Lin Y, Ran Z, Li X, Mou R, Wang Y, Li D. Ferroptosis and hepatocellular carcinoma: the emerging role of lncRNAs. Front Immunol 2024; 15:1424954. [PMID: 38846953 PMCID: PMC11153672 DOI: 10.3389/fimmu.2024.1424954] [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: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 06/09/2024] Open
Abstract
Hepatocellular carcinoma is the most common form of primary liver cancer and poses a significant challenge to the medical community because of its high mortality rate. In recent years, ferroptosis, a unique form of cell death, has garnered widespread attention. Ferroptosis, which is characterized by iron-dependent lipid peroxidation and mitochondrial alterations, is closely associated with the pathological processes of various diseases, including hepatocellular carcinoma. Long non-coding RNAs (lncRNAs), are a type of functional RNA, and play crucial regulatory roles in a variety of biological processes. In this manuscript, we review the regulatory roles of lncRNAs in the key aspects of ferroptosis, and summarize the research progress on ferroptosis-related lncRNAs in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Haoran Chen
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Zhongyu Han
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Junyan Su
- The First People’s Hospital of Longquanyi District, Chengdu, China
| | - Xuanliang Song
- The First People’s Hospital of Longquanyi District, Chengdu, China
| | - Qingquan Ma
- The First People’s Hospital of Longquanyi District, Chengdu, China
| | - Yumeng Lin
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Zijin Ran
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Xueping Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rongkun Mou
- Department of General Surgery, The Third Hospital of Mianyang, Mianyang, China
| | - Yi Wang
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Dongxuan Li
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| |
Collapse
|
25
|
Zhang B, Ye Q. Linc00662 sponges miR-15b-5p to promote hypopharyngeal squamous cell carcinoma progression by facilitating cancer stem cell-like phenotypes. J Cancer 2024; 15:3781-3793. [PMID: 38911389 PMCID: PMC11190759 DOI: 10.7150/jca.95852] [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: 03/01/2024] [Accepted: 04/28/2024] [Indexed: 06/25/2024] Open
Abstract
Background: Long non-coding RNAs (lncRNAs) are associated with multiple head and neck tumors and play important roles in cancer. This study explored the molecular mechanism of Linc00662 in hypopharyngeal squamous cell carcinoma (HSCC). Methods: Real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to detect gene expression in HSCC tissues. The viability and proliferation of tumor cells were measured using CCK-8 assays. HSCC cell apoptosis was measured using flow cytometry and western blotting. Cell stemness was examined using the sphere formation assay. A xenograft tumor model was established to investigate the role of Linc00662 in vivo. Results: The expression level of Linc00662 in HSCC tissues was significantly higher than that in adjacent normal tissues. The expression of Linc00662 had no significant relationship with the tumor stage. Patients with high Linc00662 expression were found to have shorter overall survival than those with low Linc00662 expression. Linc00662 over-expression promoted cell viability and inhibited apoptosis. Using online databases and a dual luciferase reporter, miR-15b-5p was confirmed as a potential downstream sponge of Linc00662. Moreover, Linc00662 was negatively associated with miR-15b-5p in HSCC cells. Depletion of miR-15b-5p can reverse the function of Linc00662 in vivo and in vitro. Furthermore, Linc00662 promotes tumor growth, which was abolished by miR-15b-5p mimics. Importantly, the stemness of cancer stem cells was mediated by the Linc00662/miR-15b-5p axis. Conclusion: Patients with HSCC with high Linc00662 showed poor prognosis and high Linc00662 induced stemness of tumor cells by targeting miR-15b-5p. Linc00662 may serve as a novel diagnostic and target marker for head and neck squamous cell carcinoma.
Collapse
Affiliation(s)
| | - Qing Ye
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| |
Collapse
|
26
|
Meng X, Peng F, Yu S, Chi X, Wang W, Shao S. Knockdown of NADK promotes LUAD ferroptosis via NADPH/FSP1 axis. J Cancer Res Clin Oncol 2024; 150:228. [PMID: 38700533 PMCID: PMC11068837 DOI: 10.1007/s00432-024-05752-z] [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: 03/12/2024] [Accepted: 04/18/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Lung cancer is a serious threat to human health and is the first leading cause of cancer death. Ferroptosis, a newly discovered form of programmed cell death associated with redox homeostasis, is of particular interest in the lung cancer, given the high oxygen environment of lung cancer. NADPH has reducing properties and therefore holds the potential to resist ferroptosis. Resistance to ferroptosis exists in lung cancer, but the role of NADK in regulating ferroptosis in lung cancer has not been reported yet. METHODS Immunohistochemistry (IHC) was used to analyse the expression of NADK in 86 cases of lung adenocarcinoma(LUAD) and adjacent tissues, and a IHC score was assigned to each sample. Chi-square and kaplan-meier curve was performed to analyse the differences in metastasis and five-year survival between the two groups with NADK high or low scores. Proliferation of NADK-knockdown LUAD cell lines was detected in vivo and vitro. Furthermore, leves of ROS, MDA and Fe2+ were measured to validate the effect and mechanism of NADK on ferroptosis in LUAD. RESULTS The expression of NADK was significantly evaluated in LUAD tissues as compared to adjacent non-cancerous tissues. The proliferation of NADK-knockdown cells was inhibited both in vivo and vitro, and increasing levels of intracellular ROS, Fe2+ and lipid peroxide products (MDA) were observed. Furthermore, NADK-knockdown promoted the ferroptosis of LUAD cells induced by Erastin/RSL3 by regulating the level of NADPH and the expression of FSP1. Knockdown of NADK enhanced the sensitivities of LUAD cells to Erastin/RSL3-induced ferroptosis by regulating NADPH level and FSP1 expression. CONCLUSIONS NADK is over-expressed in LUAD patients. Knockdown of NADK inhibited the proliferation of LUAD cells both in vitro and in vivo and promotes the Erastin/RSL3-induced ferroptosis of LUAD cells by down-regulating the NADPH/FSP1 axis.
Collapse
Affiliation(s)
- Xiangpeng Meng
- Liaoning Key Laboratory of Proteomics, Dalian Medical University, Dalian, 116044, China
| | - Fang Peng
- Department of Pathologic, The Second Hospital of Dalian Medical University, Dalian, 116011, China
| | - Shijie Yu
- Liaoning Key Laboratory of Proteomics, Dalian Medical University, Dalian, 116044, China
| | - Xinming Chi
- Liaoning Key Laboratory of Proteomics, Dalian Medical University, Dalian, 116044, China
| | - Wenchi Wang
- Liaoning Key Laboratory of Proteomics, Dalian Medical University, Dalian, 116044, China
| | - Shujuan Shao
- Liaoning Key Laboratory of Proteomics, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
27
|
Li SQ, Lv F, Xu WT, Yin YX, Wei HT, Li KZ, Hu BL. lncRNA SNHG4 inhibits ferroptosis by orchestrating miR-150-5p/c-Myb axis in colorectal cancer. Int J Biol Macromol 2024; 268:131961. [PMID: 38692535 DOI: 10.1016/j.ijbiomac.2024.131961] [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: 01/15/2024] [Revised: 04/07/2024] [Accepted: 04/27/2024] [Indexed: 05/03/2024]
Abstract
LncRNAs have shown to regulate ferroptosis in colorectal cancer (CRC), but the mechanism remains largely unknown. This study unveiled the mechanism of SNHG4 underlying ferroptosis in CRC. RNA-seq and RT-PCR assay confirmed SNHG4 was decreased after Erastin treatment in CRC cells. Overexpression of SNHG4 inhibited and silence promoted CRC cells ferroptosis. SNHG4 was positively correlated to c-Myb in CRC tissues and both located in cytoplasm of CRC cells. RIP and RNA pull-down assays verified the interaction between SNHG4 and c-Myb. Silence of c-Myb alleviated the suppressing effect on ferroptosis by SNHG4 in CRC cells. Dual-luciferase reporter assay revealed that SNHG4 sponging miR-150-5p in CRC cells. Overexpression of SNHG4 decreased the miR-150-5p and increased c-Myb expression. c-Myb was a direct target gene of miR-150-5p in CRC cells. Moreover, effect of CDO1 on ferroptosis was regulated transcriptionally by c-Myb, overexpression of c-Myb reduce CDO1 expression and enhance the GPX4 levels. The animal models confirmed that regulatory effect of SNHG4 on miR-150-5p and c-Myb after inducing ferroptosis. We concluded that SNHG4 inhibited Erastin-induce ferroptosis in CRC, this effect is via sponging miR-150-5p to regulate c-Myb expression, and activated CDO1/GPX4 axis. These findings provide insights into the regulatory mechanism of SNHG4 on ferroptosis.
Collapse
Affiliation(s)
- Si-Qi Li
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi, China
| | - Feng Lv
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi, China
| | - Wen-Ting Xu
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi, China
| | - Yi-Xin Yin
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi, China
| | - Hao-Tang Wei
- Department of Gastrointestinal Surgery, Third Affiliated Hospital of Guangxi Medical University, Nanning 530031, China
| | - Ke-Zhi Li
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi, China
| | - Bang-Li Hu
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi, China.
| |
Collapse
|
28
|
Ciftci YC, Vatansever İE, Akgül B. Unraveling the intriguing interplay: Exploring the role of lncRNAs in caspase-independent cell death. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1862. [PMID: 38837618 DOI: 10.1002/wrna.1862] [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: 11/05/2023] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024]
Abstract
Cell death plays a crucial role in various physiological and pathological processes. Until recently, programmed cell death was mainly attributed to caspase-dependent apoptosis. However, emerging evidence suggests that caspase-independent cell death (CICD) mechanisms also contribute significantly to cellular demise. We and others have reported and functionally characterized numerous long noncoding RNAs (lncRNAs) that modulate caspase-dependent apoptotic pathways potentially in a pathway-dependent manner. However, the interplay between lncRNAs and CICD pathways has not been comprehensively documented. One major reason for this is that most CICD pathways have been recently discovered with some being partially characterized at the molecular level. In this review, we discuss the emerging evidence that implicates specific lncRNAs in the regulation and execution of CICD. We summarize the diverse mechanisms through which lncRNAs modulate different forms of CICD, including ferroptosis, necroptosis, cuproptosis, and others. Furthermore, we highlight the intricate regulatory networks involving lncRNAs, protein-coding genes, and signaling pathways that orchestrate CICD in health and disease. Understanding the molecular mechanisms and functional implications of lncRNAs in CICD may unravel novel therapeutic targets and diagnostic tools for various diseases, paving the way for innovative strategies in disease management and personalized medicine. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Yusuf Cem Ciftci
- Noncoding RNA Laboratory, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Gülbahçeköyü, Urla, Turkey
| | - İpek Erdoğan Vatansever
- Noncoding RNA Laboratory, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Gülbahçeköyü, Urla, Turkey
| | - Bünyamin Akgül
- Noncoding RNA Laboratory, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Gülbahçeköyü, Urla, Turkey
| |
Collapse
|
29
|
Pang Q, Tang Z, Luo L. The crosstalk between oncogenic signaling and ferroptosis in cancer. Crit Rev Oncol Hematol 2024; 197:104349. [PMID: 38626848 DOI: 10.1016/j.critrevonc.2024.104349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 03/13/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
Ferroptosis, a novel form of cell death regulation, was identified in 2012. It is characterized by unique features that differentiate it from other types of cell death, including necrosis, apoptosis, autophagy, and pyroptosis. Ferroptosis is defined by an abundance of iron ions and lipid peroxidation, resulting in alterations in subcellular structures, an elevation in reactive oxygen species (ROS), a reduction in glutathione (GSH) levels, and an augmentation in Fe (II) cytokines. Ferroptosis, a regulated process, is controlled by an intricate network of signaling pathways, where multiple stimuli can either enhance or hinder the process. This review primarily examines the defensive mechanisms of ferroptosis and its interaction with the tumor microenvironment. The analysis focuses on the pathways that involve AMPK, p53, NF2, mTOR, System Xc-, Wnt, Hippo, Nrf2, and cGAS-STING. The text discusses the possibilities of employing a combination therapy that targets several pathways for the treatment of cancer. It emphasizes the necessity for additional study in this field.
Collapse
Affiliation(s)
- Qianghu Pang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Zhirou Tang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang,School of Ocean and Tropical Medicine. Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
30
|
Yu S, Cui X, Zhou S, Li Y, Feng W, Zhang X, Zhong Y, Zhang P. THOC7-AS1/OCT1/FSTL1 axis promotes EMT and serves as a therapeutic target in cutaneous squamous cell carcinoma. J Transl Med 2024; 22:347. [PMID: 38605354 PMCID: PMC11010364 DOI: 10.1186/s12967-024-05116-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/20/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND THOC7-AS1 and FSTL1 expression are frequently upregulated in cutaneous squamous cell carcinoma (cSCC). However, their molecular biological mechanisms remain elusive and their potential as therapeutic targets needs urgent exploration. METHODS Human tissue samples were used to evaluate clinical parameters. In vitro and in vivo experiments assessed biological functions. Quantitative PCR, western blot, immunohistochemistry, immunocytochemistry, immunoprecipitation, RNA fluorescence in situ hybridization, RNA pull-down, RNA immunoprecipitation, silver staining, chromatin immunoprecipitation, dual luciferase reporter assays etc. were utilized to explore the molecular biological mechanisms. RESULTS We found FSTL1 is an oncogene in cSCC, with high expression in tumor tissues and cells. Its elevated expression closely associates with tumor size and local tissue infiltration. In vitro and in vivo, high FSTL1 expression promotes cSCC proliferation, migration and invasion, facilitating malignant behaviors. Mechanistically, FSTL1 interacts with ZEB1 to promote epithelial-to-mesenchymal transition (EMT) in cSCC cells. Exploring upstream regulation, we found THOC7-AS1 can interact with OCT1, which binds the FSTL1 promoter region and promotes FSTL1 expression, facilitating cSCC progression. Finally, treating tumors with THOC7-AS1 antisense oligonucleotides inhibited cSCC proliferative and migratory abilities, delaying tumor progression. CONCLUSIONS The THOC7-AS1/OCT1/FSTL1 axis regulates EMT and promotes tumor progression in cSCC. This study provides clues and ideas for cSCC targeted therapy.
Collapse
Affiliation(s)
- Site Yu
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, P.R. China
| | - Xu Cui
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, P.R. China
| | - Situo Zhou
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, P.R. China
| | - Yun Li
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, P.R. China
| | - Wenjie Feng
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, P.R. China
| | - Xiangjun Zhang
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, P.R. China
| | - Yuhui Zhong
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, P.R. China
| | - Pihong Zhang
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, P.R. China.
| |
Collapse
|
31
|
Dai Y, Wei X, Jiang T, Wang Q, Li Y, Ruan N, Luo P, Huang J, Yang Y, Yan Q, Zhang C, Liu Y. Ferroptosis in age-related vascular diseases: Molecular mechanisms and innovative therapeutic strategies. Biomed Pharmacother 2024; 173:116356. [PMID: 38428313 DOI: 10.1016/j.biopha.2024.116356] [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: 01/08/2024] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024] Open
Abstract
Aging, an inevitable aspect of human existence, serves as one of the predominant risk factors for vascular diseases. Delving into the mystery of vascular disease's pathophysiology, the profound involvement of programmed cell death (PCD) has been extensively demonstrated. PCD is a fundamental biological process that plays a crucial role in both normal physiology and pathology, including a recently discovered form, ferroptosis. Ferroptosis is characterized by its reliance on iron and lipid peroxidation, and its significant involvement in vascular disease pathophysiology has been increasingly acknowledged. This phenomenon not only offers a promising therapeutic target but also deepens our understanding of the complex relationship between ferroptosis and age-related vascular diseases. Consequently, this article aims to thoroughly review the mechanisms that enable the effective control and inhibition of ferroptosis. It focuses on genetic and pharmacological interventions, with the goal of developing innovative therapeutic strategies to combat age-related vascular diseases.
Collapse
Affiliation(s)
- Yue Dai
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiuxian Wei
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Jiang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Wang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yi Li
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Nan Ruan
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pengcheng Luo
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jingwen Huang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Nursing, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Yang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Yan
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu Liu
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
32
|
Chen L, Lin W, Zhang H, Geng S, Le Z, Wan F, Huang Q, Chen H, Liu X, Lu JJ, Kong L. TRIB3 promotes malignancy of head and neck squamous cell carcinoma via inhibiting ferroptosis. Cell Death Dis 2024; 15:178. [PMID: 38429254 PMCID: PMC10907716 DOI: 10.1038/s41419-024-06472-5] [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: 08/09/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 03/03/2024]
Abstract
Tribbles pseudokinase 3 (TRIB3) has been identified recently as a novel oncogene in several cancers. Still, further extensive research is imperative to elucidate its function and the molecular mechanisms underlying its involvement in the progression of head and neck squamous cell carcinoma (HNSCC). In our study, we found that TRIB3 silencing significantly promoted cell death by inducing ferroptosis. The interaction of TRIB3 with Transcription Factor 4 (TCF4) and β-catenin created a heterotrimeric complex, which directly interacts with the ALOXE3 promoter, detrimentally impacting its activation. The consequential partial neutralization of ferroptosis induced by TRIB3 deficiency is observed through the implementation of ALOXE3 knockdown. Furthermore, the study demonstrated that the molecular inhibitor hesperidin, targeting TRIB3, not only reduced cell malignancy but also induced ferroptosis, thereby suppressing tumor growth. Overall, our findings unequivocally validate the proposition that TRIB3 deficiency precipitates the iron death mechanism, thereby indicating that the strategic targeting of TRIB3 could emerge as an innovative therapeutic strategy for HNSCC.
Collapse
Affiliation(s)
- Li Chen
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Wanzun Lin
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Haojiong Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Shikai Geng
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Ziyu Le
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Fangzhu Wan
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Qingting Huang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Huaiyuan Chen
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Xingyu Liu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Jiade J Lu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China
| | - Lin Kong
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China.
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, 201321, China.
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, 201321, China.
| |
Collapse
|
33
|
Han Z, Luo W, Shen J, Xie F, Luo J, Yang X, Pang T, Lv Y, Li Y, Tang X, He J. Non-coding RNAs are involved in tumor cell death and affect tumorigenesis, progression, and treatment: a systematic review. Front Cell Dev Biol 2024; 12:1284934. [PMID: 38481525 PMCID: PMC10936223 DOI: 10.3389/fcell.2024.1284934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/08/2024] [Indexed: 11/02/2024] Open
Abstract
Cell death is ubiquitous during development and throughout life and is a genetically determined active and ordered process that plays a crucial role in regulating homeostasis. Cell death includes regulated cell death and non-programmed cell death, and the common types of regulatory cell death are necrosis, apoptosis, necroptosis, autophagy, ferroptosis, and pyroptosis. Apoptosis, Necrosis and necroptosis are more common than autophagy, ferroptosis and pyroptosis among cell death. Non-coding RNAs are regulatory RNA molecules that do not encode proteins and include mainly microRNAs, long non-coding RNAs, and circular RNAs. Non-coding RNAs can act as oncogenes and tumor suppressor genes, with significant effects on tumor occurrence and development, and they can also regulate tumor cell autophagy, ferroptosis, and pyroptosis at the transcriptional or post-transcriptional level. This paper reviews the recent research progress on the effects of the non-coding RNAs involved in autophagy, ferroptosis, and pyroptosis on tumorigenesis, tumor development, and treatment, and looks forward to the future direction of this field, which will help to elucidate the molecular mechanisms of tumorigenesis and tumor development, as well as provide a new vision for the treatment of tumors.
Collapse
Affiliation(s)
- Zeping Han
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
- Rehabilitation Medicine Institute of Panyu District, Guangzhou, China
| | - Wenfeng Luo
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Jian Shen
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Fangmei Xie
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Jinggen Luo
- Department of General Surgery, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Xiang Yang
- Department of Gynaecology and Obstetrics, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Ting Pang
- Clinical Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Yubing Lv
- Clinical Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Yuguang Li
- He Xian Memorial Hospital, Southern Medical University, Guangzhou, China
| | - Xingkui Tang
- Department of General Surgery, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Jinhua He
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
- Rehabilitation Medicine Institute of Panyu District, Guangzhou, China
| |
Collapse
|
34
|
Naik U, Amin SE, Elsayad M, Saluja K. Pleomorphic Adenoma with a Novel Gene Rearrangement-LINC01606::PLAG1. Head Neck Pathol 2024; 18:10. [PMID: 38393488 PMCID: PMC10891012 DOI: 10.1007/s12105-024-01612-x] [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: 11/09/2023] [Accepted: 01/09/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Pleomorphic adenoma is a well-known benign salivary gland neoplasm characterized by the presence of varying proportions of three different components, including bi-layered ducts, myoepithelial cells, and admixed within a chondromyxoid/fibrous stroma. METHOD We report an interesting case of an adult male who presented with bleeding from an extensively degenerated parotid gland mass, concerning for a vascular neoplasm versus primary malignant tumor. Microscopically, majority of the viable tumor exhibited diffuse proliferation of spindle to epithelioid cells, with focal areas depicting cribriform glands, ducts, and scant chondromyxoid stroma. RESULT Next-generation sequencing (NGS) RNA-based fusion panel analysis identified a gene rearrangement involving the pleomorphic adenoma gene 1 (PLAG1), with a novel, cryptogenic fusion partner known as LINC01606; [LINC01606::PLAG1; inv(8;8)(8q12.1;8q12.1)]. CONCLUSION To the best of our knowledge, this is the first documented case of a long non-coding RNA (lnc-RNA) serving as a rearrangement partner with the PLAG1 gene. We reviewed the molecular characteristics of this entity and explored the potential role of LINC01606::PLAG1 in the tumorigenesis of pleomorphic adenoma.
Collapse
Affiliation(s)
- Udit Naik
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Sara E Amin
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Mahmoud Elsayad
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Karan Saluja
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA.
| |
Collapse
|
35
|
Xue W, Yang L, Chen C, Ashrafizadeh M, Tian Y, Sun R. Wnt/β-catenin-driven EMT regulation in human cancers. Cell Mol Life Sci 2024; 81:79. [PMID: 38334836 PMCID: PMC10857981 DOI: 10.1007/s00018-023-05099-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/09/2023] [Accepted: 12/20/2023] [Indexed: 02/10/2024]
Abstract
Metastasis accounts for 90% of cancer-related deaths among the patients. The transformation of epithelial cells into mesenchymal cells with molecular alterations can occur during epithelial-mesenchymal transition (EMT). The EMT mechanism accelerates the cancer metastasis and drug resistance ability in human cancers. Among the different regulators of EMT, Wnt/β-catenin axis has been emerged as a versatile modulator. Wnt is in active form in physiological condition due to the function of GSK-3β that destructs β-catenin, while ligand-receptor interaction impairs GSK-3β function to increase β-catenin stability and promote its nuclear transfer. Regarding the oncogenic function of Wnt/β-catenin, its upregulation occurs in human cancers and it can accelerate EMT-mediated metastasis and drug resistance. The stimulation of Wnt by binding Wnt ligands into Frizzled receptors can enhance β-catenin accumulation in cytoplasm that stimulates EMT and related genes upon nuclear translocation. Wnt/β-catenin/EMT axis has been implicated in augmenting metastasis of both solid and hematological tumors. The Wnt/EMT-mediated cancer metastasis promotes the malignant behavior of tumor cells, causing therapy resistance. The Wnt/β-catenin/EMT axis can be modulated by upstream mediators in which non-coding RNAs are main regulators. Moreover, pharmacological intervention, mainly using phytochemicals, suppresses Wnt/EMT axis in metastasis suppression.
Collapse
Affiliation(s)
- Wenhua Xue
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Lin Yang
- Department of Hepatobiliary Surgery, Xianyang Central Hospital, Xianyang, 712000, Shaanxi, China
| | - Chengxin Chen
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Milad Ashrafizadeh
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, USA.
| | - Ranran Sun
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
36
|
Liu S, Yue M, Lu Y, Wang Y, Luo S, Liu X, Jiang J. Advancing the frontiers of colorectal cancer treatment: harnessing ferroptosis regulation. Apoptosis 2024; 29:86-102. [PMID: 37752371 DOI: 10.1007/s10495-023-01891-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 09/28/2023]
Abstract
In recent years, colorectal cancer incidence and mortality have increased significantly due to poor lifestyle choices. Despite the development of various treatments, their effectiveness against advanced/metastatic colorectal cancer remains unsatisfactory due to drug resistance. However, ferroptosis, a novel iron-dependent cell death process induced by lipid peroxidation and elevated reactive oxygen species (ROS) levels along with reduced activity of the glutathione peroxidase 4 (GPX4) antioxidant enzyme system, shows promise as a therapeutic target for colorectal cancer. This review aims to delve into the regulatory mechanisms of ferroptosis in colorectal cancer, providing valuable insights into potential therapeutic approaches. By targeting ferroptosis, new avenues can be explored for innovative therapies to combat colorectal cancer more effectively. In addition, understanding the molecular pathways involved in ferroptosis may help identify biomarkers for prognosis and treatment response, paving the way for personalized medicine approaches. Furthermore, exploring the interplay between ferroptosis and other cellular processes can uncover combination therapies that enhance treatment efficacy. Investigating the tumor microenvironment's role in regulating ferroptosis may offer strategies to sensitize cancer cells to cell death induction, leading to improved outcomes. Overall, ferroptosis presents a promising avenue for advancing the treatment of colorectal cancer and improving patient outcomes.
Collapse
Affiliation(s)
- Siyue Liu
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Ming Yue
- Department of Pharmacy, Tongji Medical College, the Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Yukang Lu
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Ying Wang
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Shiwen Luo
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Xiaoliu Liu
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China.
| | - Jue Jiang
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, China.
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, 430065, China.
| |
Collapse
|
37
|
Li S, Peng M, Tan S, Oyang L, Lin J, Xia L, Wang J, Wu N, Jiang X, Peng Q, Zhou Y, Liao Q. The roles and molecular mechanisms of non-coding RNA in cancer metabolic reprogramming. Cancer Cell Int 2024; 24:37. [PMID: 38238756 PMCID: PMC10795359 DOI: 10.1186/s12935-023-03186-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/20/2023] [Indexed: 01/22/2024] Open
Abstract
One of the key features of cancer is energy metabolic reprogramming which is tightly related to cancer proliferation, invasion, metastasis, and chemotherapy resistance. NcRNAs are a class of RNAs having no protein-coding potential and mainly include microRNAs, lncRNAs and circRNAs. Accumulated evidence has suggested that ncRNAs play an essential role in regulating cancer metabolic reprogramming, and the altered metabolic networks mediated by ncRNAs primarily drive carcinogenesis by regulating the expression of metabolic enzymes and transporter proteins. Importantly, accumulated research has revealed that dysregulated ncRNAs mediate metabolic reprogramming contributing to the generation of therapeutic tolerance. Elucidating the molecular mechanism of ncRNAs in cancer metabolic reprogramming can provide promising metabolism-related therapeutic targets for treatment as well as overcome therapeutic tolerance. In conclusion, this review updates the latest molecular mechanisms of ncRNAs related to cancer metabolic reprogramming.
Collapse
Affiliation(s)
- Shizhen Li
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha, 410013, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Mingjing Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Shiming Tan
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Linda Oyang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Jinguan Lin
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Longzheng Xia
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Jiewen Wang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Nayiyuan Wu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Qiu Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
38
|
Zhang X, Li X, Xia R, Zhang HS. Ferroptosis resistance in cancer: recent advances and future perspectives. Biochem Pharmacol 2024; 219:115933. [PMID: 37995980 DOI: 10.1016/j.bcp.2023.115933] [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: 08/01/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
Ferroptosis is an iron-dependent, non-apoptotic form of regulated cell death and has been implicated in the occurrence and development of various diseases, including heart disease, nervous system diseases and cancer. Ferroptosis induction recently emerged as an attractive strategy for cancer therapy. Ferroptosis has become a potential target for intervention in these diseases or injuries in relevant preclinical models. This review summarizes recent progress on the mechanisms of ferroptosis resistance in cancer, highlights redox status and metabolism's role in it. Combination therapy for ferroptosis has great potential in cancer treatment, especially malignant tumors that are resistant to conventional therapies. This review will lead us to have a comprehensive understanding of the future exploration of ferroptosis and cancer therapy. A deeper understanding of the relationship between ferroptosis resistance and metabolism reprogramming may provide new strategies for tumor treatment and drug development based on ferroptosis.
Collapse
Affiliation(s)
- Xing Zhang
- Faculty of Environment and Life, Beijing University of Technology, Pingleyuan 100(#), District of Chaoyang, Beijing 100124, China
| | - Xiang Li
- Faculty of Environment and Life, Beijing University of Technology, Pingleyuan 100(#), District of Chaoyang, Beijing 100124, China
| | - Ran Xia
- Faculty of Environment and Life, Beijing University of Technology, Pingleyuan 100(#), District of Chaoyang, Beijing 100124, China
| | - Hong-Sheng Zhang
- Faculty of Environment and Life, Beijing University of Technology, Pingleyuan 100(#), District of Chaoyang, Beijing 100124, China.
| |
Collapse
|
39
|
Gong H, Li Z, Wu Z, Lian G, Su Z. Modulation of ferroptosis by non‑coding RNAs in cancers: Potential biomarkers for cancer diagnose and therapy. Pathol Res Pract 2024; 253:155042. [PMID: 38184963 DOI: 10.1016/j.prp.2023.155042] [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: 11/06/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 01/09/2024]
Abstract
Ferroptosis is a recently discovered cell programmed death. Extensive researches have indicated that ferroptosis plays an essential role in tumorigenesis, development, migration and chemotherapy drugs resistance, which makes it become a new target for tumor therapy. Non-coding RNAs (ncRNAs) are considered to control a wide range of cellular processes by modulating gene expression. Recent studies have indicated that ncRNAs regulate the process of ferroptosis via various pathway to affect the development of cancer. However, the regulation network remains ambiguous. In this review, we outlined the major metabolic processes of ferroptosis and concluded the relationship between ferroptosis-related ncRNAs and cancer progression. In addition, the prospect of ncRNAs being new therapeutic targets and early diagnosis biomarkers for cancer by regulating ferroptosis were presented, and the possible obstacles were also predicted. This could help in discovering novel cancer early diagnostic methods and therapeutic approaches.
Collapse
Affiliation(s)
- Huifang Gong
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zheng Li
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhimin Wu
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Gaojian Lian
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Zehong Su
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
40
|
Cao MY, Zhang ZD, Hou XR, Wang XP. The Potential Role of Non-coding RNAs in Regulating Ferroptosis in Cancer: Mechanisms and Application Prospects. Anticancer Agents Med Chem 2024; 24:1182-1196. [PMID: 39021186 DOI: 10.2174/0118715206322163240710112404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/03/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024]
Abstract
Cancer is the second leading cause of death globally. Despite some successes, conventional cancer treatments are insufficient to address the growing problem of drug resistance in tumors and to achieve efficient treatment outcomes. Therefore, there is an urgent need to explore new therapeutic options. Ferroptosis, a type of iron- and reactive oxygen species-dependent regulated cell death, has been closely associated with cancer development and progression. Non-coding RNAs (ncRNAs) are a class of RNAs that do not code for proteins, and studies have demonstrated their involvement in the regulation of ferroptosis in cancer. This review aims to explore the molecular regulatory mechanisms of ncRNAs involved in ferroptosis in cancer and to emphasize the feasibility of ferroptosis and ncRNAs as novel therapeutic strategies for cancer. We conducted a systematic and extensive literature review using PubMed, Google Scholar, Web of Science, and various other sources to identify relevant studies on ferroptosis, ncRNAs, and cancer. A deeper understanding of ferroptosis and ncRNAs could facilitate the development of new cancer treatment strategies.
Collapse
Affiliation(s)
- Ming-Yuan Cao
- School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, 712082, P.R. China
| | - Zhen-Dong Zhang
- School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, 712082, P.R. China
| | - Xin-Rui Hou
- School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, 712082, P.R. China
| | - Xiao-Ping Wang
- School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, 712082, P.R. China
| |
Collapse
|
41
|
Sun B, Yue SG. Expression of long noncoding RNA MEG3 and microRNA-302b-3p in colon cancer: Correlation with clinical stage and value in predicing prognosis after surgical treatment. Shijie Huaren Xiaohua Zazhi 2023; 31:981-988. [DOI: 10.11569/wcjd.v31.i23.981] [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: 09/15/2023] [Revised: 11/08/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND More and more long non-coding RNAs and microRNAs have been found to have significant changes in expression levels during the occurrence and development of tumors, which can affect the expression of tumor suppressor genes or oncogenes and play an important role in the proliferation and metastasis of cancer cells.
AIM To investigate the correlation between the expression of long non-coding RNA maternal imprinted gene 3 (LncRNA MEG3) and microRNA (miR)-302b-3p in colon cancer and clinical stage and analyze their value in predicting the prognosis after surgical treatment.
METHODS A total of 97 patients with colon cancer treated at Jinhua Hospital of TCM from January 2017 to March 2022 were selected to compare the expression of LncRNA MEG3 and miR-302b-3p in different tissues, analyze the correlation between the expression of LncRNA MEG3 and miR-302b-3p and clinical pathological characteristics, compare the recurrence in patients with different LncRNA MEG3 and miR-302b-3p expression, analyze the factors affecting the recurrence of colon cancer after surgery, and analyze the impact of the interaction between LncRNA MEG3 and miR-302b-3p on the recurrence of colon cancer. The predictive value of LncRNA MEG3 and miR-302b-3p expression for the recurrence of colon cancer after surgery was evaluated.
RESULTS The expression of LncRNA MEG3 and miR-302b-3p in colon cancer tissues was lower than that in tumor-adjacent tissues (P < 0.05). The expression of LncRNA MEG3 and miR-302b-3p in colon cancer tissues was not correlated with sex, age, or tumor size (P > 0.05), but was correlated with tumor differentiation, clinical stage, and lymph node metastasis (P < 0.05). In colon cancer tissues, the recurrence rate in patients with high LncRNA MEG3 and miR-302b-3p expression was lower than that of patients with lower LncRNA MEG3 and miR-302b-3p expression (P < 0.05). Tumor differentiation degree, clinical stage, and lymph node metastasis were all identified to be risk factors for colon cancer recurrence, and LncRNA MEG3 and miR-302b-3p expression were protective factors for colon cancer recurrence (P < 0.05). The interaction analysis showed that the synergistic effect of simultaneous exposure to LncRNA MEG3 and miR-302b-3p was 15.888 times greater than the effect of exposure to either LncRNA MEG3 or miR-302b-3p alone, and when simultaneously exposing to both, 56.98% of the risk of colon cancer recurrence was attributed to their synergistic effect. The area under the curve (AUC) (95% confidence interval [CI]) of LncRNA MEG3 and miR-302b-3p in predicting the prognosis of colon cancer patients was 0.720 (0.620-0.807) and 0.767 (0.670-0.847), respectively, and that of the combined prediction was 0.892 (0.813-0.946), with a sensitivity and specificity of 92.31% and 83.33%, respectively, which were significantly higher than those of either LncRNA MEG3 or miR-302b-3p alone.
CONCLUSION The down-regulated expression of LncRNA MEG3 and miR-302b-3p in colon cancer is related to clinical stage. Clinical detection of their expression can be used to determine the malignant degree of tumor and predict the prognosis of surgical treatment, thus providing reference for adjustment of clinical treatment plan.
Collapse
Affiliation(s)
- Bing Sun
- Second Department of Surgery, Jinhua Hospital of Traditional Chinese Medicine, Jinhua 321017, Zhejiang Province, China
| | - Shi-Guo Yue
- Second Department of Surgery, Jinhua Hospital of Traditional Chinese Medicine, Jinhua 321017, Zhejiang Province, China
| |
Collapse
|
42
|
Guo Z, Huo X, Li X, Jiang C, Xue L. Advances in regulation and function of stearoyl-CoA desaturase 1 in cancer, from bench to bed. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2773-2785. [PMID: 37450239 DOI: 10.1007/s11427-023-2352-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/23/2023] [Indexed: 07/18/2023]
Abstract
Stearoyl-CoA desaturase 1 (SCD1) converts saturated fatty acids to monounsaturated fatty acids. The expression of SCD1 is increased in many cancers, and the altered expression contributes to the proliferation, invasion, sternness and chemoresistance of cancer cells. Recently, more evidence has been reported to further support the important role of SCD1 in cancer, and the regulation mechanism of SCD1 has also been focused. Multiple factors are involved in the regulation of SCD1, including metabolism, diet, tumor microenvironment, transcription factors, non-coding RNAs, and epigenetics modification. Moreover, SCD1 is found to be involved in regulating ferroptosis resistance. Based on these findings, SCD1 has been considered as a potential target for cancer treatment. However, the resistance of SCD1 inhibition may occur in certain tumors due to tumor heterogeneity and metabolic plasticity. This review summarizes recent advances in the regulation and function of SCD1 in tumors and discusses the potential clinical application of targeting SCD1 for cancer treatment.
Collapse
Affiliation(s)
- Zhengyang Guo
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
| | - Xiao Huo
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
| | - Xianlong Li
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
| | - Changtao Jiang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China.
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University and the Key Laboratory of Molecular Cardiovascular Science (Peking University), Ministry of Education, Beijing, 100191, China.
| | - Lixiang Xue
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China.
- Peking University Third Hospital Cancer Center, Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
43
|
Lee J, Roh JL. Epigenetic modulation of ferroptosis in cancer: Identifying epigenetic targets for novel anticancer therapy. Cell Oncol (Dordr) 2023; 46:1605-1623. [PMID: 37438601 DOI: 10.1007/s13402-023-00840-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2023] [Indexed: 07/14/2023] Open
Abstract
Ferroptosis is a newly recognized form of oxidative-regulated cell death resulting from iron-mediated lipid peroxidation accumulation. Radical-trapping antioxidant systems can eliminate these oxidized lipids and prevent disrupting the integrity of cell membranes. Epigenetic modifications can regulate ferroptosis by altering gene expression or cell phenotype without permanent sequence changes. These mechanisms include DNA methylation, histone modifications, RNA modifications, and noncoding RNAs. Epigenetic alterations in cancer can control the expression of ferroptosis regulators or related pathways, leading to changes in cell sensitivity to ferroptosis inducers or cancer progression. Epigenetic alterations in cancer are influenced by a wide range of cancer hallmarks, contributing to therapeutic resistance. Targeting epigenetic alterations is a promising approach to overcoming cancer resilience. However, the exact mechanisms involved in different types of cancer remain unresolved. Discovering more ferroptosis-associated epigenetic targets and interventions can help overcome current barriers in anticancer therapy. Many papers on epigenetic modifications of ferroptosis have been continuously published, making it essential to summarize the current state-of-the-art in the epigenetic regulation of ferroptosis in human cancer.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-do, 13496, Republic of Korea
- Department of Biomedical Science, General Graduate School, CHA University, Seongnam, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-do, 13496, Republic of Korea.
- Department of Biomedical Science, General Graduate School, CHA University, Seongnam, Republic of Korea.
| |
Collapse
|
44
|
Lu Q, Liang Y, Meng X, Zhao Y, Fan H, Hou S. The Role of Long Noncoding RNAs in Intestinal Health and Diseases: A Focus on the Intestinal Barrier. Biomolecules 2023; 13:1674. [PMID: 38002356 PMCID: PMC10669616 DOI: 10.3390/biom13111674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/04/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
The gut is the body's largest immune organ, and the intestinal barrier prevents harmful substances such as bacteria and toxins from passing through the gastrointestinal mucosa. Intestinal barrier dysfunction is closely associated with various diseases. However, there are currently no FDA-approved therapies targeting the intestinal epithelial barriers. Long noncoding RNAs (lncRNAs), a class of RNA transcripts with a length of more than 200 nucleotides and no coding capacity, are essential for the development and regulation of a variety of biological processes and diseases. lncRNAs are involved in the intestinal barrier function and homeostasis maintenance. This article reviews the emerging role of lncRNAs in the intestinal barrier and highlights the potential applications of lncRNAs in the treatment of various intestinal diseases by reviewing the literature on cells, animal models, and clinical patients. The aim is to explore potential lncRNAs involved in the intestinal barrier and provide new ideas for the diagnosis and treatment of intestinal barrier damage-associated diseases in the clinical setting.
Collapse
Affiliation(s)
- Qianying Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (X.M.); (S.H.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yangfan Liang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (X.M.); (S.H.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Xiangyan Meng
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (X.M.); (S.H.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yanmei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (X.M.); (S.H.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (X.M.); (S.H.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Shike Hou
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (X.M.); (S.H.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| |
Collapse
|
45
|
Zheng F, Bi JC, Wei YY, Wang Y, Zhang Q, Liang CL, Wu J, Dai Z. MiR-124-3p mediates gastric cancer cell ferroptosis induced by an anti-cancer drug polyphyllin I. Front Pharmacol 2023; 14:1285799. [PMID: 38027010 PMCID: PMC10664244 DOI: 10.3389/fphar.2023.1285799] [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: 08/30/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Background: Ferroptosis is an emerging type of regulated cell death and associated with antitumoral therapy, while some microRNAs have been shown to regulate the tumorigenesis and cancer progression. Meanwhile, polyphyllin I (PPI) has exhibited antitumoral effects by promoting cancer cell apoptosis and ferroptosis. However, it is unclear whether PPI induces cancer cell ferroptosis by regulating microRNAs. Methods: We used two gastric cancer cell lines (AGS and MKN-45) to set up a tumor model of the nude mice, which were then treated daily with PPI to measure the cancer growth in vitro and in vivo. Ferroptosis was measured using immunofluorescence staining and flow cytometric analysis according to levels of intracellular ROS, lipid ROS and ferrous ions. Moreover, NRF2 expression was measured by Western blotting. In some experiments, the mimics or inhibitors of miR-124-3p were used to further confirm its involvement in PPI-induced cancer cell ferroptosis. Results: Here we found that miR-124-3p mediated cancer ferroptosis and tumor repression induced by PPI since PPI increased miR-124-3p expression in gastric cancer cells and promoted their ferroptosis, whereas inhibition of miR-124-3p mostly abolished the effects of PPI on tumor growth, ferroptosis and NRF2 expression. Moreover, miR-124-3p mimics promoted cancer cell ferroptosis by downregulating NRF2 through directly targeting 3'-UTR region of NRF2, confirming a role for miR-124-3p in regulating PPI-induced ferroptosis. Conclusion: PPI exerts its antitumoral effects on the gastric cancer by promoting cell ferroptosis via regulating miR-124-3p. Our findings have clinical implications for cancer chemotherapy.
Collapse
Affiliation(s)
- Fang Zheng
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jian-Can Bi
- Department of Internal Medicine, The First People’s Hospital of Jingdezhen, Jingdezhen, Jiangxi, China
| | - Yu-Yan Wei
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yeshu Wang
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Qunfang Zhang
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chun-Ling Liang
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianwei Wu
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhenhua Dai
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
46
|
Li H, Liu J, Qin X, Sun J, Liu Y, Jin F. Function of Long Noncoding RNAs in Glioma Progression and Treatment Based on the Wnt/β-Catenin and PI3K/AKT Signaling Pathways. Cell Mol Neurobiol 2023; 43:3929-3942. [PMID: 37747595 DOI: 10.1007/s10571-023-01414-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/09/2023] [Indexed: 09/26/2023]
Abstract
Gliomas are a deadly primary malignant tumor of the central nervous system, with glioblastoma (GBM) representing the most aggressive type. The clinical prognosis of GBM patients remains bleak despite the availability of multiple options for therapy, which has needed us to explore new therapeutic methods to face the rapid progression, short survival, and therapy resistance of glioblastomas. As the Human Genome Project advances, long noncoding RNAs (lncRNAs) have attracted the attention of researchers and clinicians in cancer research. Numerous studies have found aberrant expression of signaling pathways in glioma cells. For example, lncRNAs not only play an integral role in the drug resistance process by regulating the Wnt/β-catenin or PI3K/Akt signaling but are also involved in a variety of malignant biological behaviors such as glioma proliferation, migration, invasion, and tumor apoptosis. Therefore, the present review systematically assesses the existing research evidence on the malignant progression and drug resistance of glioma, focusing on the critical role and potential function of lncRNAs in the Wnt/β-catenin and PI3K/Akt classical pathways to promote and encourage further research in this field.
Collapse
Affiliation(s)
- Hanyun Li
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Jilan Liu
- Department of Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, China
| | - Xianyun Qin
- Department of Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, China
| | - Jikui Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan, 250014, China.
| | - Yan Liu
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- School of Mental Health, Jining Medical University, Jining, 272013, China.
| | - Feng Jin
- The Affiliated Qingdao Central Hospital of Qingdao University, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao, 266042, China.
| |
Collapse
|
47
|
Shan C, Liang Y, Wang K, Li P. Noncoding RNAs in cancer ferroptosis: From biology to clinical opportunity. Biomed Pharmacother 2023; 165:115053. [PMID: 37379641 DOI: 10.1016/j.biopha.2023.115053] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
Ferroptosis is a recently discovered pattern of programmed cell death that is nonapoptotic and irondependent. It is involved in lipid peroxidation dependent on reactive oxygen species. Ferroptosis has been verified to play a crucial regulatory role in a variety of pathological courses of disease, in particularly cancer. Emerging research has highlighted the potential of ferroptosis in tumorigenesis, cancer development and resistance to chemotherapy. However, the regulatory mechanism of ferroptosis remains unclear, which limits the application of ferroptosis in cancer treatment. Noncoding RNAs (ncRNAs) are noncoding transcripts that regulate gene expression in various ways to affect the malignant phenotypes of cancer cells. At present, the biological function and underlying regulatory mechanism of ncRNAs in cancer ferroptosis have been partially elucidated. Herein, we summarize the current knowledge of the central regulatory network of ferroptosis, with a focus on the regulatory functions of ncRNAs in cancer ferroptosis. The clinical application and prospects of ferroptosis-related ncRNAs in cancer diagnosis, prognosis and anticancer therapies are also discussed. Elucidating the function and mechanism of ncRNAs in ferroptosis, along with assessing the clinical significance of ferroptosis-related ncRNAs, provides new perspectives for understanding cancer biology and treatment approaches, which may benefit numerous cancer patients in the future.
Collapse
Affiliation(s)
- Chan Shan
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Yan Liang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Kun Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
48
|
Zhang X, Ma L, Wang J. Cross-Regulation Between Redox and Epigenetic Systems in Tumorigenesis: Molecular Mechanisms and Clinical Applications. Antioxid Redox Signal 2023; 39:445-471. [PMID: 37265163 DOI: 10.1089/ars.2023.0253] [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] [Indexed: 06/03/2023]
Abstract
Significance: Redox and epigenetics are two important regulatory processes of cell physiological functions. The cross-regulation between these processes has critical effects on the occurrence and development of various types of tumors. Recent Advances: The core factor that influences redox balance is reactive oxygen species (ROS) generation. The ROS functions as a double-edged sword in tumors: Low levels of ROS promote tumors, whereas excessive ROS induces various forms of tumor cell death, including apoptosis and ferroptosis as well as necroptosis and pyroptosis. Many studies have shown that the redox balance is influenced by epigenetic mechanisms such as DNA methylation, histone modification, chromatin remodeling, non-coding RNAs (microRNA, long non-coding RNA, and circular RNA), and RNA N6-methyladenosine modification. Several oxidizing or reducing substances also affect the epigenetic state. Critical Issues: In this review, we summarize research on the cross-regulation between redox and epigenetics in cancer and discuss the relevant molecular mechanisms. We also discuss the current research on the clinical applications. Future Directions: Future research can use high-throughput methods to analyze the molecular mechanisms of the cross-regulation between redox and epigenetics using both in vitro and in vivo models in more detail, elucidate regulatory mechanisms, and provide guidance for clinical treatment. Antioxid. Redox Signal. 39, 445-471.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Lifang Ma
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Jiayi Wang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
49
|
Zheng X, Zhang C. The Regulation of Ferroptosis by Noncoding RNAs. Int J Mol Sci 2023; 24:13336. [PMID: 37686142 PMCID: PMC10488123 DOI: 10.3390/ijms241713336] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
As a novel form of regulated cell death, ferroptosis is characterized by intracellular iron and lipid peroxide accumulation, which is different from other regulated cell death forms morphologically, biochemically, and immunologically. Ferroptosis is regulated by iron metabolism, lipid metabolism, and antioxidant defense systems as well as various transcription factors and related signal pathways. Emerging evidence has highlighted that ferroptosis is associated with many physiological and pathological processes, including cancer, neurodegeneration diseases, cardiovascular diseases, and ischemia/reperfusion injury. Noncoding RNAs are a group of functional RNA molecules that are not translated into proteins, which can regulate gene expression in various manners. An increasing number of studies have shown that noncoding RNAs, especially miRNAs, lncRNAs, and circRNAs, can interfere with the progression of ferroptosis by modulating ferroptosis-related genes or proteins directly or indirectly. In this review, we summarize the basic mechanisms and regulations of ferroptosis and focus on the recent studies on the mechanism for different types of ncRNAs to regulate ferroptosis in different physiological and pathological conditions, which will deepen our understanding of ferroptosis regulation by noncoding RNAs and provide new insights into employing noncoding RNAs in ferroptosis-associated therapeutic strategies.
Collapse
Affiliation(s)
| | - Cen Zhang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China;
| |
Collapse
|
50
|
Zhang Q, Fan X, Zhang X, Ju S. Ferroptosis in tumors and its relationship to other programmed cell death: role of non-coding RNAs. J Transl Med 2023; 21:514. [PMID: 37516888 PMCID: PMC10387214 DOI: 10.1186/s12967-023-04370-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/17/2023] [Indexed: 07/31/2023] Open
Abstract
Programmed cell death (PCD) plays an important role in many aspects of individual development, maintenance of body homeostasis and pathological processes. Ferroptosis is a novel form of PCD characterized by the accumulation of iron-dependent lipid peroxides resulting in lethal cell damage. It contributes to tumor progression in an apoptosis-independent manner. In recent years, an increasing number of non-coding RNAs (ncRNAs) have been demonstrated to mediate the biological process of ferroptosis, hence impacting carcinogenesis, progression, drug resistance, and prognosis. However, the clear regulatory mechanism for this phenomenon remains poorly understood. Moreover, ferroptosis does not usually exist independently. Its interaction with PCD, like apoptosis, necroptosis, autophagy, pyroptosis, and cuproptosis, to destroy cells appears to exist. Furthermore, ncRNA seems to be involved. Here, we review the mechanisms by which ferroptosis occurs, dissect its relationship with other forms of death, summarize the key regulatory roles played by ncRNAs, raise relevant questions and predict possible barriers to its application in the clinic, offering new ideas for targeted tumour therapy.
Collapse
Affiliation(s)
- Qi Zhang
- Medical School of Nantong University, Nantong University, Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xinfeng Fan
- Medical School of Nantong University, Nantong University, Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xinyu Zhang
- Medical School of Nantong University, Nantong University, Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
- Department of Medical School of Nantong University, No.19, Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Shaoqing Ju
- Medical School of Nantong University, Nantong University, Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, No.20, Xisi Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|