1
|
Chen Q, Zhong G, Fang X, Lin C, Wang S, Li M. The multifaceted role of Sestrin 3 (SESN3) in oxidative stress, inflammation and tumorigenesis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119938. [PMID: 40174866 DOI: 10.1016/j.bbamcr.2025.119938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/07/2025] [Accepted: 03/20/2025] [Indexed: 04/04/2025]
Abstract
The pathogenesis of inflammation and tumors is a focal point of scientific inquiry, with oxidative stress often serving as the primary initiator. Within the human genome, the SESN3 gene encodes the SESN3 protein, a crucial antioxidant stress protein. Acting as a regulatory factor, SESN3 intricately modulates cellular oxidative stress, actively participating in cellular protection and repair mechanisms. Its functions span antioxidative, anti-aging, and anti-tumor properties. The expression of SESN3 is closely linked to cellular and oxidative stress, metabolic status, and specific signaling pathways. This review aims to delve into the origins and functions of SESN3, its role within signaling pathways, and its contributions to inflammation and tumorigenesis.
Collapse
Affiliation(s)
- Qiusan Chen
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Guoqiang Zhong
- Department of Gastroenterology, Guangzhou First People's Hospital, Guangzhou, Guangdong, China
| | - Xianmei Fang
- Department of Ultrasonography, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Chuangzhen Lin
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shanping Wang
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mingsong Li
- Department of Gastroenterology, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Yang C, Liao X, Zhou K, Yao Y, He X, Zhong W, Zheng D, Yang Y, Li M, Zhou M, Zhou Y, Li L, Bai Y, Shi K, Qian Z. Multifunctional nanoparticles and collagenase dual loaded thermosensitive hydrogel system for enhanced tumor-penetration, reversed immune suppression and photodynamic-immunotherapy. Bioact Mater 2025; 48:1-17. [PMID: 40028237 PMCID: PMC11870144 DOI: 10.1016/j.bioactmat.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 03/05/2025] Open
Abstract
Breast cancer is the most prevalent and lethal malignancy among females, with a critical need for safer and less invasive treatments. Photodynamic therapy (PDT) can effectively eliminate tumor cells with minimal side effects. Furthermore, the combination of PDT and immunotherapy using nanoparticles has shown promise in treating both primary and distant metastatic tumor cells. Therefore, this study proposes applying the PDT-immunotherapy combination to breast cancer treatment. However, the low immunogenicity characteristic of "cold" tumors in part of breast cancer significantly diminishes therapeutic efficacy. To address this challenge, here, a nano-gel system (designated as HCSC-gel) is constructed, which co-delivers a mitochondria-targeted photosensitizer and a STING agonist, capable of robustly activating "cold" tumor immunity. This system is further enhanced by collagenase (CN) to improve therapeutic outcomes. Upon injection into the primary tumor site, HCSC-gel rapidly forms a gel matrix, releasing CN to degrade the tumor extracellular matrix and facilitate the penetration of photosensitizers, STING agonists, and oxygen into the tumor tissue. Under laser irradiation, PDT and STING-mediated immune responses are activated, reversing the low immunogenicity of breast cancer and effectively treating both primary and metastatic lesions. This HCSC-gel nano hydrogel delivery platform is anticipated to provide novel insights for the clinical management of breast cancer and other low immunogenic "cold" tumors, offering significant benefits to patients.
Collapse
Affiliation(s)
- Chengli Yang
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Xukun Liao
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Kai Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Yongchao Yao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Xinlong He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Wen Zhong
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Dan Zheng
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yan Yang
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Ming Li
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Meng Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yadi Zhou
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Lin Li
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yang Bai
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| |
Collapse
|
3
|
Zhou M, Wu W, Wang Y, Zhang B, Zhao X, Zhou H, Cao Y, Wu P, Wang M, Wang J. Targeting COL5A1 enhances anoikis thus attenuating malignancy of glioblastoma via inhibiting the Wnt/β-catenin signaling pathway. J Neurooncol 2025:10.1007/s11060-025-05036-7. [PMID: 40402199 DOI: 10.1007/s11060-025-05036-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/03/2025] [Indexed: 05/23/2025]
Abstract
PURPOSE As one of the most prevalent primary brain tumors, glioblastoma (GBM) is characterized by its severe malignancy and extremely poor prognosis. Recent studies have demonstrated that targeting anoikis and malignancy showed impressed efficiency for treatment in a wide range of solid tumors, however, relevant research on GBM still remains unclarified. METHODS In this study, genes related with malignancy and anoikis of GBM were identified by utilizing the Cancer Genome Atlas (TCGA), the Chinese Glioma Genome Atlas (CGGA) and the Molecular Signatures Database (MSigDB). Subsequently, the role of the key gene was validated via proliferation, invasion and migration experiments both in conditions with and without attachment. Moreover, RNA sequencing analysis was employed to reveal further mechanisms. RESULTS Here, Type V collagen alpha 1 (COL5A1) was identified as a critical gene associated with anoikis and poor outcomes. Additionally, COL5A1 knockdown induced significant reduction in malignancy of GBM both in vitro and in vivo. Moreover, cell anoikis was remarkable enhanced by reduced expression of COL5A1 after low-attachment cell culture. Mechanically, RNA sequencing analysis revealed that the activity of the Wnt/β-catenin signaling pathway was diminished following COL5A1 knockdown, which indicated that COL5A1 reduced anoikis via regulating Wnt/β-catenin signaling pathway thus promoted malignancies of GBM cells. CONCLUSION These findings demonstrated the novel evidence that COL5A1 serves as an essential regulatory factor influencing both anoikis and malignancy of GBM cells by regulating Wnt/β-catenin signaling pathway, indicating that COL5A1 could be a novel prognosis-related biomarker and potential therapeutic target for GBM.
Collapse
Affiliation(s)
- Mingjing Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Wei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yichang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Beichen Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xuyan Zhao
- Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Haoyu Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yiyang Cao
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Pancheng Wu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Maode Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China.
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Jia Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China.
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
4
|
Xu M, Pan G, Zhang Q, Huang J, Wu Y, Ashan Y. FOXM1 boosts glycolysis by upregulating SQLE to inhibit anoikis in breast cancer cells. J Cancer Res Clin Oncol 2025; 151:162. [PMID: 40360780 PMCID: PMC12075401 DOI: 10.1007/s00432-025-06174-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/15/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Resisting anoikis is a prerequisite for cancer to spread and invade and a major cause of cancer-related deaths. Yet, the intricate mechanisms of how cancer cells evade anoikis remain largely unknown. There is a significant need to explore how these mechanisms play out in breast cancer (BC). METHODS Bioinformatics analysis revealed the expression levels of SQLE and FOXM1 in BC tissue, along with their correlation. The enrichment pathways of SQLE were also explored. qPCR detected the expression of SQLE and FOXM1 in BC cells. CCK-8 assessed cell viability, while flow cytometry measured anoikis. Western blot was employed to examine the protein expression of key genes in glycolytic metabolism and apoptosis-related proteins. Extracellular acidification rate was quantified, and corresponding kits evaluated glucose consumption, lactate production, and adenosine triphosphate levels in cells. Dual-luciferase reporter assays and chromatin immunoprecipitation tests unveiled the binding relationship between FOXM1 and SQLE. RESULTS SQLE was found to be highly expressed in BC and enriched in pathways associated with anoikis and glycolysis. SQLE curbed anoikis in BC via the aerobic glycolysis pathway. There was also a direct binding between FOXM1 and SQLE and a positive correlation between their expression. Recovery experiments substantiated that FOXM1 targeted SQLE to suppress anoikis in BC cells. CONCLUSION FOXM1 upregulates SQLE, which in turn mediates glycolysis to suppress anoikis in BC. The FOXM1/SQLE axis is a promising therapeutic target for BC treatment.
Collapse
Affiliation(s)
- Mei Xu
- Radiotherapy Department of Changji Hui Autonomous Prefecture People's Hospital, No. 303 Yan'an North Road, Changji City, Xinjiang Province, 831100, China.
| | - Guozhi Pan
- Cardiothoracic Surgery Department, Changji Hui Autonomous Prefecture People's Hospital, Changji, China
| | - Qian Zhang
- Radiotherapy Department of Changji Hui Autonomous Prefecture People's Hospital, No. 303 Yan'an North Road, Changji City, Xinjiang Province, 831100, China
| | - Jiangming Huang
- Department of Surgical Oncology, Changji People's Hospital, Changji, China
| | - Yehua Wu
- General Surgery Department of Changji Hui Autonomous Prefecture People's Hospital, Changji, China
| | - Yashengjiang Ashan
- Radiotherapy Department of Changji Hui Autonomous Prefecture People's Hospital, No. 303 Yan'an North Road, Changji City, Xinjiang Province, 831100, China
| |
Collapse
|
5
|
Zhang J, Cheng L, Jiang S, Zhu D. Machine learning based identification of anoikis related gene classification patterns and immunoinfiltration characteristics in diabetic nephropathy. Sci Rep 2025; 15:15271. [PMID: 40312440 PMCID: PMC12046048 DOI: 10.1038/s41598-025-99395-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: 07/06/2024] [Accepted: 04/21/2025] [Indexed: 05/03/2025] Open
Abstract
Anoikis and immune cell infiltration are pivotal factors in the pathophysiological mechanism of diabetic nephropathy (DN), yet a comprehensive understanding of the mechanism is lacking. This work aimed to pinpoint distinctive anoikis-related genes (ARGs) in DN and delve into their impact on the immune landscape. Three datasets (GSE30528, GSE47184, and GSE96804) were downloaded from the gene expression omnibus (GEO) dataset. Differentially expressed genes (DEGs) were identified using the "limma" package, while ARGs were obtained from GSEA, GeneCard, and Harmonizome datasets. The intersection of DEGs and ARGs was analyzed for Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. The CIBERSORT algorithm was employed to estimate the infiltration percentage of 22 immune cell types in DN renal tissue. Subsequently, the least absolute shrinkage and selection operator (LASSO), support vector machine recursive feature elimination (SVM-RFE), and random forest (RF) algorithms were adopted to screen key ARGs related to DN. After that, receiver operating characteristic (ROC) analysis was employed to assess the diagnostic accuracy of each gene and the real-time quantitative polymerase chain reaction (RT-qPCR) was adopted to quantitatively detect the expression of biomarkers in DN cell models. Finally, correlations between key genes and immune cell infiltration were analyzed, and a competitive endogenous ribonucleic acid (RNA) (ceRNA) network based on key genes was constructed. A total of 59 DEARGs were identified. GO functional annotation enrichment analysis revealed their involvement in kidney development, extracellular matrix (ECM), cytoplasmic vesicle cavity, immunoinflammatory response, and cytokine effect. KEGG pathway analysis indicated that MAPK, PI3K -Akt, IL -17, TNF, and HIF- 1 signaling pathways are critical for DN. In addition, seven key genes, including PDK4, S100A8, HTRA1, CHI3L1, WT1, CDKN1B, and EGF, were screened by machine learning algorithm. Most of these genes exhibited low expression in renal tissue of DN patients and positive correlation with neutrophils, and their expressions were verified in an external dataset cell model. The ceRNA analysis suggested potential regulatory pathways (H19/miR-15b-5p/PDK4 and KCNQ1T1/miR-1207-3p/WT1) influencing early DN progression. This work provided a comprehensive analysis of the role of DEARGs in DN for the first time, offering valuable insights for further understanding the disease mechanism and guiding clinical diagnosis, treatment, and research of DN.
Collapse
Affiliation(s)
- Jing Zhang
- The Third Department of Jiaozhou City Traditional Chinese Medicine Hospital, Jiaozhou, 266300, Shandong, China
| | - Lulu Cheng
- Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, China
| | - Shan Jiang
- Traditional Chinese Medicine Department of Linyi People's Hospital, Linyi, 276000, Shandong, China
| | - Duosheng Zhu
- The Third Department of Jiaozhou City Traditional Chinese Medicine Hospital, Jiaozhou, 266300, Shandong, China.
| |
Collapse
|
6
|
Yan W, Xiang S, Feng J, Zu X. Role of ubiquitin-specific proteases in programmed cell death of breast cancer cells. Genes Dis 2025; 12:101341. [PMID: 40083330 PMCID: PMC11904532 DOI: 10.1016/j.gendis.2024.101341] [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: 01/20/2024] [Revised: 03/26/2024] [Accepted: 04/11/2024] [Indexed: 03/16/2025] Open
Abstract
Breast cancer (BC) is the most common malignant tumor and the leading cause of cancer-related deaths among women worldwide. Great progress has been recently achieved in controlling breast cancer; however, mortality from breast cancer remains a substantial challenge, and new treatment mechanisms are being actively sought. Programmed cell death (PCD) is associated with the progression and treatment of many types of human cancers. PCD can be divided into multiple pathways including autophagy, apoptosis, mitotic catastrophe, necroptosis, ferroptosis, pyroptosis, and anoikis. Ubiquitination is a post-translational modification process in which ubiquitin, a 76-amino acid protein, is coupled to the lysine residues of other proteins. Ubiquitination is involved in many physiological events and promotes cancer development and progression. This review elaborates the role of ubiquitin-specific protease (USP) in programmed cell death, which is common in breast cancer cells, and lays the foundation for tumor diagnosis and targeted therapy.
Collapse
Affiliation(s)
| | | | - Jianbo Feng
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001 Hunan, China
| | - Xuyu Zu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001 Hunan, China
| |
Collapse
|
7
|
Kim SC, Kim GH, Park JH, Lee KH, Koh J, Kim TY, Lee DW, Kim YJ, Kim S, Park SY, Min A, Shin YK, Im SA, Ku JL. Establishment and characterization of 24 breast cancer cell lines and 3 breast cancer organoids reveals molecular heterogeneity and drug response variability in malignant pleural effusion-derived models. Breast Cancer Res 2025; 27:66. [PMID: 40312397 PMCID: PMC12044882 DOI: 10.1186/s13058-025-02032-7] [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: 11/18/2024] [Accepted: 04/22/2025] [Indexed: 05/03/2025] Open
Abstract
Intratumoral heterogeneity of breast cancer cells causes undesired drug resistance and predispose to disease recurrence. We investigate the molecular heterogeneity of breast cancer cells derived from malignant pleural effusions (MPE) to understand variations in drug resistance and cellular evolution. MPE provides a unique environment, with cells experiencing significant microenvironmental changes that promote intratumoral heterogeneity and therapeutic resistance. By establishing 24 cell lines and 3 organoids from MPE samples of breast cancer patients, we performed extensive genomic, transcriptomic, and drug sensitivity analyses. Our findings reveal substantial genetic and transcriptomic diversity among MPE-derived cell lines, highlighting dynamic alterations in driver mutations and signaling pathways that correlate with variable drug responses. Notably, temporal and spatial heterogeneity within patient-derived samples emphasized the adaptability of breast cancer cells in MPE, as different subclones displayed distinct drug sensitivities. This work underscores the critical role of molecular profiling in understanding treatment resistance in breast cancer, presenting MPE-derived cell lines as valuable preclinical models for exploring personalized therapies in aggressive cancer phenotypes.
Collapse
Grants
- 2021M3H9A1030151, NRF2022R1A5A102641311, 2022M3A9B6018217 National Research Foundation of Korea
- 2021M3H9A1030151, NRF2022R1A5A102641311, 2022M3A9B6018217 National Research Foundation of Korea
- 2021M3H9A1030151, NRF2022R1A5A102641311, 2022M3A9B6018217 National Research Foundation of Korea
- 2021M3H9A1030151, NRF2022R1A5A102641311, 2022M3A9B6018217 National Research Foundation of Korea
- 2021M3H9A1030151, NRF2022R1A5A102641311, 2022M3A9B6018217 National Research Foundation of Korea
- HA22C0111 National R&D Program for Cancer Control through the National Cancer Center (NCC) funded by the Ministry of Health & Welfare, Republic of Korea
- HA22C0111 National R&D Program for Cancer Control through the National Cancer Center (NCC) funded by the Ministry of Health & Welfare, Republic of Korea
- HA22C0111 National R&D Program for Cancer Control through the National Cancer Center (NCC) funded by the Ministry of Health & Welfare, Republic of Korea
- HA22C0111 National R&D Program for Cancer Control through the National Cancer Center (NCC) funded by the Ministry of Health & Welfare, Republic of Korea
- HA22C0111 National R&D Program for Cancer Control through the National Cancer Center (NCC) funded by the Ministry of Health & Welfare, Republic of Korea
- HA22C0111 National R&D Program for Cancer Control through the National Cancer Center (NCC) funded by the Ministry of Health & Welfare, Republic of Korea
- HA22C0111 National R&D Program for Cancer Control through the National Cancer Center (NCC) funded by the Ministry of Health & Welfare, Republic of Korea
- HA22C0111 National R&D Program for Cancer Control through the National Cancer Center (NCC) funded by the Ministry of Health & Welfare, Republic of Korea
- HA22C0111 National R&D Program for Cancer Control through the National Cancer Center (NCC) funded by the Ministry of Health & Welfare, Republic of Korea
- Korean Cell Line Research Foundation
- National R&D Program for Cancer Control through the National Cancer Center (NCC) funded by the Ministry of Health & Welfare, Republic of Korea
Collapse
Affiliation(s)
- Soon-Chan Kim
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
- Medical Research Center, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Division of Data Science, College of Information and Communication Technology, The University of Suwon, Hwaseong, 18323, Republic of Korea
| | - Ga-Hye Kim
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Jae-Hyeon Park
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Kyung-Hun Lee
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jiwon Koh
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
- Department of Pathology, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Tae-Yong Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Dae-Won Lee
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Yu-Jin Kim
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Seongyeong Kim
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Song-Yi Park
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Ahrum Min
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Young-Kyoung Shin
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Seock-Ah Im
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea.
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | - Ja-Lok Ku
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Medical Research Center, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
8
|
Mivehchi H, Eskandari-Yaghbastlo A, Emrahoglu S, Saeidpour Masouleh S, Faghihinia F, Ayoubi S, Nabi Afjadi M. Tiny messengers, big Impact: Exosomes driving EMT in oral cancer. Pathol Res Pract 2025; 268:155873. [PMID: 40022766 DOI: 10.1016/j.prp.2025.155873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/13/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Exosomes are indispensable extracellular vesicles that facilitate intercellular communication and are crucial for both healthy and pathological conditions, including cancer. The capacity of exosomes to echo the molecular characteristics of their cells of origin, including malignant cells, makes them indispensable tools for diagnosing and tracking disease progression in the field of oncology. Oral squamous cell carcinoma (OSCC), which has been identified as the sixth most prevalent cancer worldwide, has been linked to numerous risk factors, including tobacco use, alcohol consumption, human papillomavirus (HPV) infection, and inadequate oral hygiene. Exosomes pointedly influence the advancement of oral cancer via promoting tumor cell growth, invasion, angiogenesis, and immune evasion through the alteration of the tumor microenvironment. A critical apparatus in cancer metastasis is the epithelial-to-mesenchymal transition (EMT), during which cancer cells acquire improved migratory and invasive properties. EMT plays a role in metastasis, resistance to treatment, and evasion of the immune response. Exosomes facilitate EMT in oral cancer by delivering bioactive molecules that influence EMT signaling pathways. These exosomes inspire EMT in recipient cells, by this means enhancing tumor invasion and metastasis. This study aims to identify the specific exosomal components and signaling pathways that are tangled in EMT, in that way providing new avenues for targeted therapies designed to hinder the metastasis of oral cancer.
Collapse
Affiliation(s)
- Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | | | - Sahand Emrahoglu
- School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | | | - Farbod Faghihinia
- School of Dentistry, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Saminalsadat Ayoubi
- School of Dental Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
9
|
Tang Q, Ma C, Xie J, Zhang Q, Zhang B, Bian W, Lu Q, Wan Z, Wu W. Unraveling anoikis in glioblastoma: insights from single-cell sequencing and prognostic modeling. Cancer Cell Int 2025; 25:116. [PMID: 40140848 PMCID: PMC11948803 DOI: 10.1186/s12935-025-03752-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Despite advances, Glioblastoma (GBM) treatment remains challenging due to its rapid progression and resistance to therapies. OBJECTIVES This study aimed to investigate the role of anoikis-a mechanism by which cells evade programmed cell death upon detachment from the extracellular matrix-in GBM progression and prognosis. METHODS Utilizing single-cell sequencing and bulk-transcriptome sequencing data from TCGA, GEO, and CGGA databases, we performed comprehensive bioinformatics analyses. We identified anoikis-related genes, constructed a prognostic model using 101 machine learning algorithms, and validated its clinical utility across multiple cohorts.Finally, we also verified the expression of model genes and the function of key gene in clinical samples and cell lines. RESULTS Single-cell sequencing revealed heterogeneous expression of anoikis-related genes across distinct cell populations within GBM. MES-like Malignant cells and Myeloids exhibited higher enrichment of these genes, implicating their role in anoikis resistance and tumor aggressiveness. The prognostic model, based on identified genes, effectively stratified patients into high-risk and low-risk groups, demonstrating significant differences in survival outcomes. Mutation and tumor microenvironment analyses highlighted distinct genetic landscapes and immune cell infiltration patterns associated with different risk groups. SLC43A3 emerged as a key gene, showing significant upregulation in tumor tissues and correlating with poor prognosis in GBM. CONCLUSION This study provides insights into the molecular mechanisms of anoikis resistance in GBM, underscoring its critical role in tumor progression and patient prognosis. The developed prognostic model offers a promising tool for personalized treatment strategies and warrants further exploration of targeted therapies to improve outcomes for GBM patients.
Collapse
Affiliation(s)
- Qikai Tang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, People's Republic of China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Chenfeng Ma
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Jiaheng Xie
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Qixiang Zhang
- Department of Neurosurgery, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Bingtao Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, People's Republic of China
| | - Weiqi Bian
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Qingyu Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Zeyu Wan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Wei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, People's Republic of China.
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
10
|
Giron-Michel J, Padelli M, Oberlin E, Guenou H, Duclos-Vallée JC. State-of-the-Art Liver Cancer Organoids: Modeling Cancer Stem Cell Heterogeneity for Personalized Treatment. BioDrugs 2025; 39:237-260. [PMID: 39826071 PMCID: PMC11906529 DOI: 10.1007/s40259-024-00702-0] [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] [Accepted: 12/21/2024] [Indexed: 01/20/2025]
Abstract
Liver cancer poses a global health challenge with limited therapeutic options. Notably, the limited success of current therapies in patients with primary liver cancers (PLCs) may be attributed to the high heterogeneity of both hepatocellular carcinoma (HCCs) and intrahepatic cholangiocarcinoma (iCCAs). This heterogeneity evolves over time as tumor-initiating stem cells, or cancer stem cells (CSCs), undergo (epi)genetic alterations or encounter microenvironmental changes within the tumor microenvironment. These modifications enable CSCs to exhibit plasticity, differentiating into various resistant tumor cell types. Addressing this challenge requires urgent efforts to develop personalized treatments guided by biomarkers, with a specific focus on targeting CSCs. The lack of effective precision treatments for PLCs is partly due to the scarcity of ex vivo preclinical models that accurately capture the complexity of CSC-related tumors and can predict therapeutic responses. Fortunately, recent advancements in the establishment of patient-derived liver cancer cell lines and organoids have opened new avenues for precision medicine research. Notably, patient-derived organoid (PDO) cultures have demonstrated self-assembly and self-renewal capabilities, retaining essential characteristics of their respective in vivo tissues, including both inter- and intratumoral heterogeneities. The emergence of PDOs derived from PLCs serves as patient avatars, enabling preclinical investigations for patient stratification, screening of anticancer drugs, efficacy testing, and thereby advancing the field of precision medicine. This review offers a comprehensive summary of the advancements in constructing PLC-derived PDO models. Emphasis is placed on the role of CSCs, which not only contribute significantly to the establishment of PDO cultures but also faithfully capture tumor heterogeneity and the ensuing development of therapy resistance. The exploration of PDOs' benefits in personalized medicine research is undertaken, including a discussion of their limitations, particularly in terms of culture conditions, reproducibility, and scalability.
Collapse
Affiliation(s)
- Julien Giron-Michel
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France.
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France.
| | - Maël Padelli
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
- Department of Biochemistry and Oncogenetics, Paul Brousse Hospital, AP-HP, Villejuif, France
| | - Estelle Oberlin
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
| | - Hind Guenou
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
| | - Jean-Charles Duclos-Vallée
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
- INSERM UMR-S 1193, Paul Brousse Hospital, Villejuif, France
- Hepato-Biliary Department, Paul Brousse Hospital, APHP, Villejuif, France
- Fédération Hospitalo-Universitaire (FHU) Hepatinov, Villejuif, France
| |
Collapse
|
11
|
Jin L, Qin Y, Zhao Y, Zhou X, Zeng Y. Endothelial cytoskeleton in mechanotransduction and vascular diseases. J Biomech 2025; 182:112579. [PMID: 39938443 DOI: 10.1016/j.jbiomech.2025.112579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
The cytoskeleton is an important structural component that regulates various aspects of cell morphology, movement, and intracellular signaling. It plays a pivotal role in the cellular response to biomechanical stimuli, particularly in endothelial cells, which are critical for vascular homeostasis and the pathogenesis of cardiovascular diseases. Mechanical forces, such as shear and tension, activate intracellular signaling cascades that regulate transcription, translation, and cellular behaviors. Despite extensive research into cytoskeletal functions, the precise mechanisms by which the cytoskeleton transduces mechanical signals remain incompletely understood. This review focuses on the role of cytoskeletal components in membrane, cytoplasm, and nucleus in mechanotransduction, with an emphasis on their structure, mechanical and biological behaviors, dynamic interactions, and response to mechanical forces. The collaboration between membrane cytoskeleton, cytoplasmic cytoskeleton, and nucleoskeleton is indispensable for endothelial cells to respond to mechanical stimuli. Understanding their mechanoresponsive mechanisms is essential for advancing therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Linlu Jin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Yixue Qin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Yunran Zhao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Xintong Zhou
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China
| | - Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041 Sichuan, China.
| |
Collapse
|
12
|
Meng Q, Li Z, He X, Hu Y, Wu G, Huang J, Luo Z, Hu Y, Shen X. Anti-TNBC effects of Lappaol F by targeting epithelial-mesenchymal transition via regulation of GSK-3β/YAP/β-catenin and PI3K/AKT pathways. Front Pharmacol 2025; 16:1496511. [PMID: 39989901 PMCID: PMC11842333 DOI: 10.3389/fphar.2025.1496511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/17/2025] [Indexed: 02/25/2025] Open
Abstract
Purpose Lappaol F (LAF), a lignan extracted from Fructus Arctii, has a wide spectrum of anti-tumor effects, including inhibition of TNBC cell growth. However, the pharmacological mechanism of LAF targeting epithelial-mesenchymal transition (EMT) to inhibit Triple-negative breast cancer (TNBC) growth remains poorly understood. The present study aimed to reveal the potential mechanism of LAF against TNBC by in vivo and in vitro experiments. Methods In situ, transplantation-induced MDA-MB-231 solid tumor model in NCG mice and cultured MDA-MB-231 and Hs-578T cells were used to evaluate the anti-TNBC effect of LAF. Flow cytometry, wound healing, transwell assay, western blot, RT-PCR, and immunofluorescence analysis were carried out to investigate the pharmacological mechanism of LAF against TNBC. Results LAF significantly inhibited the growth of MDA-MB-231 tumors, with downregulated tumor level of vimentin and upregulated level of ZO-1. In MDA-MB-231 and Hs-578T cells, LAF markedly suppressed cell proliferation, migration and invasion, and promoted apoptosis. Similarly, LAF increased the expression of ZO-1 and occludin proteins in MDA-MB-231 cells, and inhibited the expression of vimentin, snail and slug proteins in MDA-MB-231 and Hs-578T cells, as well as the expression of N-caderin in Hs-578T cells. Moreover, LAF also inhibited the phosphorylation of GSK-3β, thereby inhibited the downstream nuclear translocation of β-catenin and the expression of YAP. Furthermore, LAF significantly inhibited the expression of PI3K and AKT, and the phosphorylation of downstream mTOR. Conclusion LAF showed anti-TNBC effect both in vitro and in vivo. Reversal of EMT by inhibiting GSK-3β/YAP/β-catenin signaling and PI3K/AKT/mTOR signaling contributes to the effect.
Collapse
Affiliation(s)
- Qiqi Meng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhiping Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaofeng He
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yuanhao Hu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guiyun Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiawen Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhuohui Luo
- Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou, Hainan, China
- Hainan Pharmaceutical Research and Development Science Park, Haikou, Hainan, China
| | - Yingjie Hu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaoling Shen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Sheng ZH, Gong XY, Huang PP, Xu QY, Zhang WJ, Sun FB, Song KY, Zeng DC. An Innovative Anoikis Signature With Inflammatory Infiltrates in Osteoarthritis. Int J Rheum Dis 2025; 28:e70093. [PMID: 39895467 DOI: 10.1111/1756-185x.70093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/15/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
AIM To explore the relationship between an innovative anoikis-related gene signature and inflammatory infiltrates in patients with osteoarthritis. METHODS Gene expression profiles (GSM1248759 and GSE200843) were curated from the Gene Expression Omnibus database, followed by the construction of a protein-protein interaction network. Functional and genomic enrichment analyses were conducted using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. The CIBERSORT method was employed to investigate immune cell infiltration differences between osteoarthritic and control tissues. Additionally, the ConsensusClusterPlus package in R software was utilized to identify distinct anoikis patterns (Cluster C1 and Cluster C2) and conduct molecular biological investigations. RESULTS Analysis revealed two distinct anoikis patterns (Cluster C1 and Cluster C2), with Cluster C2 patients exhibiting varying immune cell levels compared to Cluster C1 patients. Molecular investigations identified 84 DEGs enriched in specific pathways such as adipocytokine signaling, cytokine-cytokine receptor interaction, ECM-receptor interaction, and the PPAR signaling pathway. qPCR experiments confirmed the elevated expression levels of specific genes, including SOD2, MAPK14, CEACM3, LAMB3, COL13A1, TLR3, NOTCH3, and KLF12, in the IL-1β-induced group compared with the osteoarthritis group. CONCLUSION This study highlights the role of anoikis-related genes and immune infiltration differences in osteoarthritis, enhancing our understanding of its development.
Collapse
Affiliation(s)
- Ze-Hao Sheng
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xin-Yi Gong
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Peng-Peng Huang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qi-Yu Xu
- Department of Rehabilitation Medicine, Anhui no.2 Provincial People's Hospital, Hefei, Anhui, China
| | - Wen-Jie Zhang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Feng-Bao Sun
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kai-Yi Song
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Du-Chun Zeng
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Taha SR, Boulos F. E-cadherin staining in the diagnosis of lobular versus ductal neoplasms of the breast: the emperor has no clothes. Histopathology 2025; 86:327-340. [PMID: 39138705 PMCID: PMC11707503 DOI: 10.1111/his.15295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Categorizing breast neoplasia as ductal or lobular is a daily exercise that relies on a combination of histologic and immunohistochemical tools. The historically robust link between loss of the E-cadherin molecule and lobular neoplasia has rendered staining for E-cadherin by immunohistochemistry a staple of this diagnostic process. Unfortunately, discordances between E-cadherin expression and histomorphology, and variations in E-cadherin staining patterns and intensities abound in clinical practice, but are often neglected in favour of a binary interpretation of the E-cadherin result. In this article, we highlight the complexities of E-cadherin expression through a review of the E-cadherin protein and its associated gene (CDH1), the mechanisms leading to aberrant/absent E-cadherin expression, and the implications of these factors on the reliability of the E-cadherin immunohistochemical stain in the classification of ductal versus lobular mammary neoplasia.
Collapse
MESH Headings
- Female
- Humans
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/diagnosis
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Cadherins/metabolism
- Cadherins/analysis
- Carcinoma, Ductal, Breast/diagnosis
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Lobular/diagnosis
- Carcinoma, Lobular/metabolism
- Carcinoma, Lobular/pathology
- Immunohistochemistry
Collapse
Affiliation(s)
- Seyed R Taha
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMOUSA
| | - Fouad Boulos
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
15
|
Zhang Y, Yi S, Luan M. Advances in non-apoptotic regulated cell death: implications for malignant tumor treatment. Front Oncol 2025; 15:1519119. [PMID: 39949740 PMCID: PMC11821507 DOI: 10.3389/fonc.2025.1519119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Cell death mechanisms are broadly classified into accidental cell death (ACD) and regulated cell death (RCD). ACD such as necrosis, is an uncontrolled, accidental process, while RCD is tightly regulated by specific signaling pathways and molecular mechanisms. Tumor cells are characterized by their ability to evade cell death and sustain uncontrolled proliferation. The failure of programmed cell death is a key contributor to tumor initiation, progression, and resistance to cancer therapies. Traditionally, research has focused primarily on apoptosis as the dominant form of RCD in cancer. However, emerging evidence highlights the importance of other non-apoptotic forms of RCD, such as pyroptosis, ferroptosis, necroptosis, and parthanatos, in tumorigenesis and treatment response. These pathways are gaining attention for their potential roles in overcoming therapy resistance. In this review, we will discuss the recent advances in the study of non-apoptotic cell death pathways in malignant tumors and explore their therapeutic implications, offering insights into new targets for cancer treatment strategies.
Collapse
Affiliation(s)
- Yizheng Zhang
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Shiqi Yi
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, China
| | - Mingyuan Luan
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| |
Collapse
|
16
|
Liu H, Mei M, Zhong H, Lin S, Luo J, Huang S, Zhou J. Identification of Anoikis-Related Genes in Chronic Kidney Disease Based on Bioinformatics Analysis Combined with Experimental Validation. J Inflamm Res 2025; 18:973-994. [PMID: 39867944 PMCID: PMC11761547 DOI: 10.2147/jir.s498820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/09/2025] [Indexed: 01/28/2025] Open
Abstract
Background Chronic kidney disease (CKD) is a progressive condition that arises from diverse etiological factors, resulting in structural alterations and functional impairment of the kidneys. We aimed to establish the Anoikis-related gene signature in CKD by bioinformatics analysis. Methods We retrieved 3 datasets from the Gene Expression Omnibus (GEO) database to obtain differentially expressed genes (DEGs), followed by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA) of them, which were intersected with Anoikis-related genes (ARGs) to derive Anoikis-related differentially expressed genes (ARDEGs). Besides, we conducted weighted gene co-expression network analysis (WGCNA) to identify hub genes. And then, we adopted the quantitative real-time PCR (RT-qPCR) assay to validate the hub genes among several CKD animal models. Furthermore, we constructed a competitive endogenous RNA (ceRNA) network for the hub genes utilizing the ENCORI and miRDB databases, while also calculating Spearman correlation coefficients. Ultimately, we applied the CIBERSORTx algorithm to conduct immune infiltration analysis, classifying immune characteristics based on the abundance of 22 immune cell types. Results To summarize, we identified 13 ARDEGs. WGCNA yielded 6 hub genes, all of which demonstrated significant diagnostic potential in univariate logistic regression analysis (P<0.05). The principal pathways enriched were involved in cell cycle progression Toxoplasmosis, Cell adhesion molecules, Influenza A, Pathogenic Escherichia coli infection, Small cell lung cancer, Amoebiasis, TNF signaling pathway, and Leukocyte transendothelial migration. Notably, 6 immune cell types exhibited significant differences (P<0.05) across subgroups with distinct immune characteristics. Moreover, 2 hub genes showed significant variations (P<0.05) across these immune characteristic subtypes. Among the 4 types of CKD mouse models, the mRNA expression levels of LAMB3 and CDH3 were significantly (P<0.05) up-regulated in the model group. Conclusion We identified 6 hub genes that may serve as potential key biomarkers of Anoikis-related progression in CKD.
Collapse
Affiliation(s)
- Hong Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Manxue Mei
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Hua Zhong
- Department of Gerontology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People’s Republic of China
| | - Shuyin Lin
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Jiahui Luo
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Sirong Huang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Jiuyao Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| |
Collapse
|
17
|
Yang D, Chen H, Zhou Z, Guo J. ANXA5 predicts prognosis and immune response and mediates proliferation and migration in head and neck squamous cell carcinoma. Gene 2024; 931:148867. [PMID: 39168258 DOI: 10.1016/j.gene.2024.148867] [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: 01/04/2024] [Revised: 08/03/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a common malignancy that often develops unnoticed. Typically, these tumors are identified at advanced stages, resulting in a relatively low chance of successful treatment. Anoikis serves as a natural defense against the spread of tumor cells, meaning circumventing anoikis can effectively inhibit tumor metastasis. Nonetheless, studies focusing on anoikis in the context of HNSCC remain scarce. METHODS Anoikis-related genes (ARGs) were identified by using the GeneCards and Harmonizome databases. Expression data of these genes and relevant clinical features were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. A LASSO regression and a prognostic risk score model were developed to determine their prognostic significance. The analysis included the use of the CIBERSORT algorithm to quantify immune and stromal cell presence. Furthermore, in vitro and in vivo, we confirmed the expression and functional roles of proteins and mRNA of genes independently predictive of prognosis. RESULTS The study identified eight genes linked to prognosis (ANXA5, BAK1, CDKN2A, PPARG, CCR7, MAPK11, CRYAB, CRYBA1) and developed a prognostic model that effectively forecasts the survival outcomes for patients with HNSCC. A higher survival likelihood is associated with lower risk scores. In addition, a significant relationship was found between immune and risk score, and ANXA5 deletion promoted the killing of HNSCC cells by activated CD8+ T cells. During the screening process, 65 different chemotherapeutic drugs were found to have significant differences in IC50 values when comparing high- and low-risk categories. ANXA5 emerged as a gene with independent prognostic significance, exhibiting notably elevated protein and mRNA levels in HNSCC tissue compared to non-tumorous tissue. The suppression of ANXA5 gene activity resulted in a substantial decrease in both the growth and mobility of HNSCC cells. Animal model experiments demonstrated that inhibiting ANXA5 suppressed HNSCC growth and migration in vivo. CONCLUSION Through bioinformatics, a prognostic risk model of high precision was developed, offering valuable insights into the survival rates and immune responses in patients with HNSCC. ANXA5 is highlighted as a significant prognostic factor among the identified genes, indicating its promise as a potential therapeutic target for those with HNSCC.
Collapse
Affiliation(s)
- Donghui Yang
- Department of Otorhinolaryngology, Gaozhou People's Hospital, Gaozhou, China.
| | - Huikuan Chen
- Department of Otorhinolaryngology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zheng Zhou
- Department of Otolaryngology Head and Neck Surgery, Hunan Children's Hospital, Changsha, China
| | - Jinfei Guo
- Department of Otorhinolaryngology, Gaozhou People's Hospital, Gaozhou, China
| |
Collapse
|
18
|
Qin H, Wang Q, Xu J, Zeng H, Liu J, Yu F, Yang J. Integrative analysis of anoikis-related genes prognostic signature with immunotherapy and identification of CDKN3 as a key oncogene in lung adenocarcinoma. Int Immunopharmacol 2024; 143:113282. [PMID: 39383787 DOI: 10.1016/j.intimp.2024.113282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/01/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024]
Abstract
Anoikis, a form of programmed cell death induced by loss of cell contact, is closely associated with tumor invasion and metastasis, making it highly significant in lung cancer research. We examined the expression patterns and prognostic relevance of Anoikis-related genes (ARGs) in lung adenocarcinoma (LUAD) using the TCGA-LUAD database. This study identified molecular subtypes associated with Anoikis in LUAD and conducted functional enrichment analyses. We constructed an ARG risk score using univariate least absolute shrinkage and selection operator (LASSO) Cox regression, validated externally with GEO datasets and clinical samples. The clinical applicability of the prognostic model was evaluated using nomograms, calibration curves, decision curve analysis (DCA), and time-dependent AUC assessments. We identified four prognostically significant genes (PLK1, SLC2A1, CDKN3, PHLDA2) and two ARG-related molecular subtypes. ARGs were generally upregulated in LUAD and correlated with multiple pathways including the cell cycle and DNA replication. The prognostic model indicated that the low-risk group had better outcomes and significant correlations with clinicopathological features, tumor microenvironment, immune therapy responses, drug sensitivity, and pan-RNA epigenetic modification-related genes. Patients with low-risk LUAD were potential beneficiaries of immune checkpoint inhibitor (ICI) therapy. Prognostic ARGs' distribution and expression across various immune cell types were further analyzed using single-cell RNA sequencing. The pivotal role of CDKN3 in LUAD was confirmed through qRT-PCR and gene knockout experiments, demonstrating that CDKN3 knockdown inhibits tumor cell proliferation, migration, and invasion. Additionally, we constructed a ceRNA network involving CDKN3/hsa-miR-26a-5p/SNHG6, LINC00665, DUXAP8, and SLC2A1/hsa-miR-218-5p/RNASEH1-AS1, providing new insights for personalized and immune therapy decisions in LUAD patients.
Collapse
Affiliation(s)
- Haotian Qin
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Qichang Wang
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Juan Xu
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Hefei 238001, China
| | - Hui Zeng
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Jixian Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China.
| | - Fei Yu
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Shenzhen 518036, China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China.
| | - Jun Yang
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen 518036, China.
| |
Collapse
|
19
|
Lo Buglio G, Lo Cicero A, Campora S, Ghersi G. The Multifaced Role of Collagen in Cancer Development and Progression. Int J Mol Sci 2024; 25:13523. [PMID: 39769286 PMCID: PMC11678882 DOI: 10.3390/ijms252413523] [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: 11/20/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/05/2025] Open
Abstract
Collagen is a crucial protein in the extracellular matrix (ECM) essential for preserving tissue architecture and supporting crucial cellular functions like proliferation and differentiation. There are twenty-eight identified types of collagen, which are further divided into different subgroups. This protein plays a critical role in regulating tissue homeostasis. However, in solid tumors, the balance can be disrupted, due to an abundance of collagen in the tumor microenvironment, which significantly affects tumor growth, cell invasion, and metastasis. It is important to investigate the specific types of collagens in cancer ECM and their distinct roles in tumor progression to comprehend their unique contribution to tumor behavior. The diverse pathophysiological functions of different collagen types in cancers illustrate collagen's dual roles, offering potential therapeutic options and serving as prognostic markers.
Collapse
Affiliation(s)
- Gabriele Lo Buglio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Alessandra Lo Cicero
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
| | - Simona Campora
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (G.L.B.); (S.C.)
- Abiel srl, 90128 Palermo, Italy
| |
Collapse
|
20
|
Kennedy KV, Wang JX, McMillan E, Zhou Y, Teranishi R, Semeao A, Mirchandani L, Umeweni CN, Dhakal D, Baccarella A, Ishikawa S, Sasaki M, Itami T, Harman AC, Joannas L, Karakasheva TA, Nakagawa H, Muir AB. Lysyl Oxidase Mediates Proliferation and Differentiation in the Esophageal Epithelium. Biomolecules 2024; 14:1560. [PMID: 39766266 PMCID: PMC11674119 DOI: 10.3390/biom14121560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
In homeostatic conditions, the basal progenitor cells of the esophagus differentiate into a stratified squamous epithelium. However, in the setting of acid exposure or inflammation, there is a marked failure of basal cell differentiation, leading to basal cell hyperplasia. We have previously shown that lysyl oxidase (LOX), a collagen crosslinking enzyme, is upregulated in the setting of allergic inflammation of the esophagus; however, its role beyond collagen crosslinking is unknown. Herein, we propose a non-canonical epithelial-specific role of LOX in the maintenance of epithelial homeostasis using 3D organoid and murine models. We performed quantitative reverse transcriptase PCR, Western blot, histologic analysis, and RNA sequencing on immortalized non-transformed human esophageal epithelial cells (EPC2-hTERT) with short-hairpin RNA (shRNA) targeting LOX mRNA in both monolayer and 3D organoid culture. A novel murine model with a tamoxifen-induced Lox knockout specific to the stratified epithelium (K5CreER; Loxfl/fl) was utilized to further define the role of epithelial LOX in vivo. We found that LOX knockdown decreased the proliferative capacity of the esophageal epithelial cells in monolayer culture, and dramatically reduced the organoid formation rate (OFR) in the shLOX organoids. LOX knockdown was associated with decreased expression of the differentiation markers filaggrin, loricrin, and involucrin, with RNA sequencing analysis revealing 1224 differentially expressed genes demonstrating downregulation of pathways involved in cell differentiation and epithelial development. Mice with Lox knockout in their stratified epithelium demonstrated increased basaloid content of their esophageal epithelium and decreased Ki-67 staining compared to the vehicle-treated mice, suggesting reduced differentiation and proliferation in the Lox-deficient epithelium in vivo. Our results demonstrate, both in vivo and in vitro, that LOX may regulate epithelial homeostasis in the esophagus through the modulation of epithelial proliferation and differentiation. Understanding the mechanisms of perturbation in epithelial proliferation and differentiation in an inflamed esophagus could lead to the development of novel treatments that could promote epithelial healing and restore homeostasis.
Collapse
Affiliation(s)
- Kanak V. Kennedy
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Joshua X. Wang
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
| | - Emily McMillan
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
| | - Yusen Zhou
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
| | - Ryugo Teranishi
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
| | - Ann Semeao
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
| | - Leena Mirchandani
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
| | - Chizoba N. Umeweni
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
| | - Diya Dhakal
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
| | - Alyssa Baccarella
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Satoshi Ishikawa
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
| | - Masaru Sasaki
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
| | - Takefumi Itami
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
| | - Adele C. Harman
- Transgenic Core, CHOP Research Institute, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Leonel Joannas
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Tatiana A. Karakasheva
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
| | - Hiroshi Nakagawa
- Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA;
| | - Amanda B. Muir
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA (E.M.); (Y.Z.); (R.T.); (A.S.); (L.M.); (D.D.); (A.B.); (S.I.); (M.S.); (T.I.); (T.A.K.)
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
21
|
Lotfi MS, Rassouli FB. Navigating the complexities of cell death: Insights into accidental and programmed cell death. Tissue Cell 2024; 91:102586. [PMID: 39426124 DOI: 10.1016/j.tice.2024.102586] [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: 08/21/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Cell death is a critical biological phenomenon that can be categorized into accidental cell death (ACD) and programmed cell death (PCD), each exhibiting distinct signaling, mechanistic and morphological characteristics. This paper provides a comprehensive overview of seven types of ACD, including coagulative, liquefactive, caseous, fat, fibrinoid, gangrenous and secondary necrosis, discussing their pathological implications in conditions such as ischemia and inflammation. Additionally, we review eighteen forms of PCD, encompassing autophagy, apoptosis, necroptosis, pyroptosis, paraptosis, ferroptosis, anoikis, entosis, NETosis, eryptosis, parthanatos, mitoptosis, and newly recognized types such as methuosis, autosis, alkaliptosis, oxeiptosis, cuprotosis and erebosis. The implications of these cell death modalities for cellular processes, development, and disease-particularly in the context of neoplastic and neurodegenerative disorders-are also covered. Furthermore, we explore the crosstalk between various forms of PCD, emphasizing how apoptotic mechanisms can influence pathways like necroptosis and pyroptosis. Understanding this interplay is crucial for elucidating cellular responses to stress, as well as for its potential relevance in clinical applications and therapeutic strategies. Future research should focus on clarifying the molecular mechanisms that govern different forms of PCD and their interactions.
Collapse
Affiliation(s)
- Mohammad-Sadegh Lotfi
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Fatemeh B Rassouli
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
22
|
Luo JY, Zheng S, Yang J, Ma C, Ma XY, Wang XX, Fu XN, Mao XZ. Development and validation of biomarkers related to anoikis in liver cirrhosis based on bioinformatics analysis. World J Hepatol 2024; 16:1306-1320. [PMID: 39606164 PMCID: PMC11586749 DOI: 10.4254/wjh.v16.i11.1306] [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: 06/14/2024] [Revised: 09/29/2024] [Accepted: 10/20/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND According to study, anoikis-related genes (ARGs) have been demonstrated to play a significant impact in cirrhosis, a major disease threatening human health worldwide. AIM To investigate the relationship between ARGs and cirrhosis development to provide insights into the clinical treatment of cirrhosis. METHODS RNA-sequencing data related to cirrhosis were obtained from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) between cirrhotic and normal tissues were intersected with ARGs to derive differentially expressed ARGs (DEARGs). The DEARGs were filtered using the least absolute shrinkage and selection operator, support vector machine recursive feature elimination, and random forest algorithms to identify biomarkers for cirrhosis. These biomarkers were used to create a nomogram for predicting the prognosis of cirrhosis. The proportions of diverse immune cell subsets in cirrhotic vs normal tissues were compared using the CIBERSORT computational method. In addition, the linkage between immune cells and biomarkers was assessed, and a regulatory network of mRNA, miRNA, and transcription factors was constructed relying on the biomarkers. RESULTS The comparison of cirrhotic and normal tissue samples led to the identification of 635 DEGs. Subsequent intersection of the DEGs with ARGs produced a set of 26 DEARGs. Subsequently, three DEARGs, namely, ACTG1, STAT1, and CCR7, were identified as biomarkers using three machine-learning algorithms. The proportions of M1 and M2 macrophages, resting CD4 memory T cells, resting mast cells, and plasma cells significantly differed between cirrhotic and normal tissue samples. The proportions of M1 and M2 macrophages, resting CD4 memory T cells, and resting mast cells were significantly correlated with the expression of the three biomarkers. The mRNA-miRNA-TF network showed that ACTG1, CCR7, and STAT1 were regulated by 28, 42, and 35 miRNAs, respectively. Moreover, AR, MAX, EP300, and FOXA1 were found to regulate four miRNAs related to the biomarkers. CONCLUSION This study revealed ACTG1, STAT1, and CCR7 as biomarkers of cirrhosis, providing a reference for developing novel diagnostic and therapeutic strategies for cirrhosis.
Collapse
Affiliation(s)
- Jiang-Yan Luo
- Department of Gastroenterology, The Second Affiliated Hospital of Dali University, Kunming 650011, Yunnan Province, China
| | - Sheng Zheng
- Department of Gastroenterology, The Third People's Hospital of Yunnan Province, Kunming 650011, Yunnan Province, China.
| | - Juan Yang
- Department of Gastroenterology, The Third People's Hospital of Yunnan Province, Kunming 650011, Yunnan Province, China
| | - Chi Ma
- Department of Gastroenterology, The Second Affiliated Hospital of Dali University, Kunming 650011, Yunnan Province, China
| | - Xiao-Ying Ma
- Department of Gastroenterology, The Second Affiliated Hospital of Dali University, Kunming 650011, Yunnan Province, China
| | - Xing-Xing Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Dali University, Kunming 650011, Yunnan Province, China
| | - Xin-Nian Fu
- Department of Gastroenterology, The Second Affiliated Hospital of Dali University, Kunming 650011, Yunnan Province, China
| | - Xiao-Zhou Mao
- Department of Gastroenterology, The Second Affiliated Hospital of Dali University, Kunming 650011, Yunnan Province, China
| |
Collapse
|
23
|
Moreira MP, Franco EP, Barros BAF, Anjos BRD, Almada DDG, Barbosa INT, Braga LDC, Cassali GD, Silva LM. Standard chemotherapy impacts on in vitro cellular heterogeneity in spheroids enriched with cancer stem cells (CSCs) derived from triple-negative breast cancer cell line. Biochem Biophys Res Commun 2024; 734:150765. [PMID: 39357337 DOI: 10.1016/j.bbrc.2024.150765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
Triple-negative breast cancer is a heterogeneous disease with high recurrence and mortality, linked to cancer stem cells (CSCs). Our study characterized distinct cell subpopulations and signaling pathways to explore chemoresistance. We observed cellular heterogeneity among and within the cells regarding phenotyping and drug response. In untreated BT-549 cells, we noted plasticity properties in both CD44+/CD24+/CD146+ hybrid cells and CD44-/CD24+/CD146+ epithelial cells, enabling phenotypic conversion into CD44+/CD24-/CD146- epithelial-mesenchymal transition (EMT)-like like breast CSCs (BCSCs). Additionally, non-BCSCs may give rise to ALDH+ epithelial-like BCSCs. Enriched BCSCs demonstrated the potential to differentiation into CD44-/CD24-/CD146- cells and exhibited self-renewal capabilities. Similar phenotypic plasticity was not observed in untreated Hs 578T and HMT-3522 S1 cells. BT-549 cells were more resistant to paclitaxel/PTX than to doxorubicin/DOX, a phenomenon potentially linked to the presence of CD24+ cells prior to treatment. Under the CSCs-enriched spheroids model, BT-549 demonstrated extreme resistance to DOX, likely due to the enrichment of BCSCs CD44+/CD24-/CD146- and the tumor cells CD44-/CD24-/CD146-. Additionally, DOX treatment induced the enrichment of plastic and chemoresistant cells, further exacerbating resistance mechanisms. BT-549 exhibited high heterogeneity, leading to significant alterations in cell subpopulations under BCSCs enrichment, demonstrating increased phenotypic plasticity during EMT. This phenomenon appears to play a major role in DOX resistance, as indicated by the presence of the refractory cells CD44+/CD24-/CD146- BCSCs EMT-like, CD44-/CD24-/CD146- tumor cells, and elevated STAT3 expression. Gene expression data from BT-549 CSCs-enriched spheroids suggests that ferroptosis may be occurring via autophagic regulation triggered by RAB7A, highlighting this gene as a potential therapeutic target.
Collapse
Affiliation(s)
- Milene Pereira Moreira
- Serviço de Biologia Celular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias - Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, Minas Gerais, 30510-010, Brazil.
| | - Eliza Pereira Franco
- Serviço de Biologia Celular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias - Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, Minas Gerais, 30510-010, Brazil
| | - Bárbara Avelar Ferreira Barros
- Serviço de Biologia Celular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias - Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, Minas Gerais, 30510-010, Brazil; Programa de Pós-Graduação em Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais - Avenida Presidente Antônio Carlos 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Bianca Rocha Dos Anjos
- Serviço de Biologia Celular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias - Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, Minas Gerais, 30510-010, Brazil; Programa de Pós-Graduação em Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais - Avenida Presidente Antônio Carlos 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Daniela de Gouvêa Almada
- Serviço de Biologia Celular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias - Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, Minas Gerais, 30510-010, Brazil
| | - Isabela Nery Tavares Barbosa
- Serviço de Biologia Celular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias - Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, Minas Gerais, 30510-010, Brazil
| | - Letícia da Conceição Braga
- Serviço de Biologia Celular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias - Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, Minas Gerais, 30510-010, Brazil
| | - Geovanni Dantas Cassali
- Laboratório de Patologia Comparada, Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais - Avenida Presidente Antônio Carlos 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Luciana Maria Silva
- Serviço de Biologia Celular, Diretoria de Pesquisa e Desenvolvimento, Fundação Ezequiel Dias - Rua Conde Pereira Carneiro 80, Gameleira, Belo Horizonte, Minas Gerais, 30510-010, Brazil
| |
Collapse
|
24
|
Li L, Li L, Wang Y, Wu B, Guan Y, Chen Y, Zhao J. Integration of Machine Learning and Experimental Validation to Identify Anoikis-Related Prognostic Signature for Predicting the Breast Cancer Tumor Microenvironment and Treatment Response. Genes (Basel) 2024; 15:1458. [PMID: 39596658 PMCID: PMC11594124 DOI: 10.3390/genes15111458] [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: 10/21/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Anoikis-related genes (ANRGs) are crucial in the invasion and metastasis of breast cancer (BC). The underlying role of ANRGs in the prognosis of breast cancer patients warrants further study. Methods: The anoikis-related prognostic signature (ANRS) was generated using a variety of machine learning methods, and the correlation between the ANRS and the tumor microenvironment (TME), drug sensitivity, and immunotherapy was investigated. Moreover, single-cell analysis and spatial transcriptome studies were conducted to investigate the expression of prognostic ANRGs across various cell types. Finally, the expression of ANRGs was verified by RT-PCR and Western blot analysis (WB), and the expression level of PLK1 in the blood was measured by the enzyme-linked immunosorbent assay (ELISA). Results: The ANRS, consisting of five ANRGs, was established. BC patients within the high-ANRS group exhibited poorer prognoses, characterized by elevated levels of immune suppression and stromal scores. The low-ANRS group had a better response to chemotherapy and immunotherapy. Single-cell analysis and spatial transcriptomics revealed variations in ANRGs across cells. The results of RT-PCR and WB were consistent with the differential expression analyses from databases. NU.1025 and imatinib were identified as potential inhibitors for SPIB and PLK1, respectively. Additionally, findings from ELISA demonstrated increased expression levels of PLK1 in the blood of BC patients. Conclusions: The ANRS can act as an independent prognostic indicator for BC patients, providing significant guidance for the implementation of chemotherapy and immunotherapy in these patients. Additionally, PLK1 has emerged as a potential blood-based diagnostic marker for breast cancer patients.
Collapse
Affiliation(s)
- Longpeng Li
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Longhui Li
- School of Kinesiology and Health, Capital University of Physical Education and Sports, Beijing 100191, China
| | - Yaxin Wang
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Baoai Wu
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Yue Guan
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Yinghua Chen
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| | - Jinfeng Zhao
- Institute of Physical Education and Sport, Shanxi University, Taiyuan 030006, China; (L.L.)
| |
Collapse
|
25
|
Wang Y, Hu Q, Cao Y, Yao L, Liu H, Wen Y, Bao Y, Zhang S, Lv C, Zhao GS. FOSL1 promotes stem cell‑like characteristics and anoikis resistance to facilitate tumorigenesis and metastasis in osteosarcoma by targeting SOX2. Int J Mol Med 2024; 54:94. [PMID: 39219279 PMCID: PMC11374145 DOI: 10.3892/ijmm.2024.5418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Metastasis is the leading cause of cancer‑related death in osteosarcoma (OS). OS stem cells (OSCs) and anoikis resistance are considered to be essential for tumor metastasis formation. However, the underlying mechanisms involved in the maintenance of a stem‑cell phenotype and anoikis resistance in OS are mostly unknown. Fos‑like antigen 1 (FOSL1) is important in maintaining a stem‑like phenotype in various cancers; however, its role in OSCs and anoikis resistance remains unclear. In the present study, the dynamic expression patterns of FOSL1 were investigated during the acquisition of cancer stem‑like properties using RNA sequencing, PCR, western blotting and immunofluorescence. Flow cytometry, tumor‑sphere formation, clone formation assays, anoikis assays, western blotting and in vivo xenograft and metastasis models were used to further investigate the responses of the stem‑cell phenotype and anoikis resistance to FOSL1 overexpression or silencing in OS cell lines. The underlying molecular mechanisms were evaluated, focusing on whether SOX2 is crucially involved in FOSL1‑mediated stemness and anoikis in OS. FOSL1 expression was observed to be upregulated in OSCs and promoted tumor‑sphere formation, clone formation and tumorigenesis in OS cells. FOSL1 expression correlated positively with the expression of stemness‑related factors (SOX2, NANOG, CD117 and Stro1). Moreover, FOSL1 facilitated OS cell anoikis resistance and promoted metastases by regulating the expression of apoptosis related proteins BCL2 and BAX. Mechanistically, FOSL1 upregulated SOX2 expression by interacting with the SOX2 promoter and activating its transcription. The results also showed that SOX2 is critical for FOSL1‑mediated stem‑like properties and anoikis resistance. The current findings indicated that FOSL1 is an important regulator that promotes a stem cell‑like phenotype and anoikis resistance to facilitate tumorigenesis and metastasis in OS by regulating the transcription of SOX2. Thus, FOSL1 might represent an attractive target for therapeutic interventions in OS.
Collapse
Affiliation(s)
- Yang Wang
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610064, P.R. China
| | - Qin Hu
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610064, P.R. China
| | - Ya Cao
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400037, P.R. China
| | - Li Yao
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610064, P.R. China
| | - Haoran Liu
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610064, P.R. China
| | - Yafeng Wen
- Department of Spine Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yixi Bao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Shun Zhang
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610064, P.R. China
| | - Chuanzhu Lv
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610064, P.R. China
| | - Guo-Sheng Zhao
- Department of Spine Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
26
|
Dong B, Gu Y, Sun X, Wang X, Zhou Y, Rong Z, Zhang J, Shi X, Zhang Z, He X, Chen L, Xiong Q, Pang X, Cui Y. Targeting TUBB3 Suppresses Anoikis Resistance and Bone Metastasis in Prostate Cancer. Adv Healthc Mater 2024; 13:e2400673. [PMID: 38809199 DOI: 10.1002/adhm.202400673] [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: 02/24/2024] [Revised: 04/21/2024] [Indexed: 05/30/2024]
Abstract
Bone metastases occur in more than 70% of advanced prostate cancer (PCa) patients, leading to a poor prognosis. Resistance to detachment-induced apoptosis, also known as anoikis, plays a crucial role in the onset of tumor metastasis. Targeting anoikis resistance is of immense therapeutic significance in repression of metastatic spread. In this study, based on an anoikis-related prognostic risk model of PCa, this study identifies TUBB3 as a key anoikis-related prognostic gene that is highly expressed in bone metastatic PCa. TUBB3 expression is increased in anoikis-resistant PCa cells, and TUBB3 depletion significantly reverses anoikis resistance during extracellular matrix (ECM) detachment and inhibits anoikis-resistance-induced PCa cell invasion and migration as well as epithelial-mesenchymal transition (EMT) process. TUBB3 knockdown significantly reduces αvβ3/FAK/Src axis activation, blocking its downstream oncogenic signaling. In addition, this work develops bone-targeting lipid nanoparticles (BT-LNP) based on bisphosphonate-modified ionizable lipid for systemic delivery of siRNA targeting TUBB3 (siTUBB3). BT-LNP-delivered siTUBB3 therapy with localization in the bone microenvironment significantly attenuate PCa bone metastasis progression in vivo upon intravenous administration. These findings pinpoint that TUBB3, as a key regulator of anoikis resistance, is an effective therapeutic target in bone metastatic PCa and that BT-LNP-mediated systemic delivery of siTUBB3 can be developed as a novel therapeutic strategy for this disease.
Collapse
Affiliation(s)
- Bingqi Dong
- Department of General Surgery, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yanlun Gu
- Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, Beijing, 100191, China
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing, 100034, China
- School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Haidian District, Beijing, 100191, China
| | - Xiaojiao Sun
- School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Haidian District, Beijing, 100191, China
- Department of Urology Surgery, Peking University Third Hospital, Xueyuan Road 38, Haidian District, Beijing, 100191, China
| | - Xin Wang
- Department of General Surgery, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Ying Zhou
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Zhuona Rong
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Jixin Zhang
- Department of Pathology, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Xuedong Shi
- Department of Orthopedics, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Zhuo Zhang
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Xu He
- Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, Beijing, 100191, China
- School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Haidian District, Beijing, 100191, China
| | - Lin Chen
- Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, Beijing, 100191, China
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing, 100034, China
- School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Haidian District, Beijing, 100191, China
| | - Qingqing Xiong
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, P. R. China
| | - Xiaocong Pang
- Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, Beijing, 100191, China
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yimin Cui
- Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, Beijing, 100191, China
- School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Haidian District, Beijing, 100191, China
| |
Collapse
|
27
|
Fang X, Wei M, Liu X, Lu L, Liu G. Identification of anoikis-related long non-coding RNA signature as a novel prognostic model in lung adenocarcinoma. Transl Cancer Res 2024; 13:5458-5472. [PMID: 39525036 PMCID: PMC11543027 DOI: 10.21037/tcr-24-264] [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: 02/19/2024] [Accepted: 08/22/2024] [Indexed: 11/16/2024]
Abstract
Background Anoikis, as a specific form of programmed cell death, involves in tumor metastasis. However, there is still lacking of anoikis-related long non-coding RNA (lncRNA) risk signature in the diagnosis and prognosis of lung adenocarcinoma (LUAD). This study constructed a prognostic risk model by comprehensively analyzing anoikis-related lncRNAs which could effectively diagnose and predict the outcomes of LUAD patients. Methods A list of anoikis-related genes (ARGs) was retrieved from literatures. Anoikis-related lncRNAs were selected using co-expression analysis from The Cancer Genome Atlas (TCGA) database. Univariate and multivariate regression analyses were used to construct a prognostic model. The performance of the risk signature in predicting the prognosis and clinical significance were determined by Kaplan-Meier survival analysis, receiver operating characteristic (ROC) curves, univariate and multivariate regression analyses. Moreover, the differences of tumor immune microenvironment between the high- and low-risk groups were explored. Finally, a novel nomogram was developed by combining the signature and clinicopathological factors, and the association between lncRNAs and differential N6-methyladenosine (m6A) genes was analyzed by Spearman's analysis. Results A total of 1,694 anoikis-related lncRNAs were identified from 479 cases of LUAD. According to the univariate and multivariate Cox analyses, we established a prognostic risk model consisting of seven lncRNAs (AC026355.2, AL606489.1, AL031667.3, LINC02802, LINC01116, AC018529.1, and AP000844.2). This prognostic risk model could efficiently classify low- and high-risk patients. The area under the curve (AUC) value was 0.717, which indicated more powerful predictive capability than commonly used clinicopathological factors. The high- and low-risk groups demonstrated different immune microenvironment. Moreover, the nomogram also demonstrated good performance in predicting the prognosis. Twelve differential m6A regulators were identified, and RBM15 was found to be correlated positively with the hub lncRNA AL606489.1. Conclusions Our study constructed a prognostic risk model based on anoikis-related lncRNAs, which could provide novel perspective on the prognosis of LUAD patients.
Collapse
Affiliation(s)
- Xisheng Fang
- Department of Medical Oncology, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Medical Oncology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
| | - Mei Wei
- Department of Nursing, Guangzhou Health Science College, Guangzhou, China
| | - Xia Liu
- Department of Medical Oncology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
| | - Lin Lu
- Department of Medical Oncology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
| | - Guolong Liu
- Department of Medical Oncology, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Medical Oncology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
| |
Collapse
|
28
|
Zhu B, Xiang K, Li T, Li X, Shi F. The signature of extracellular vesicles in hypoxic breast cancer and their therapeutic engineering. Cell Commun Signal 2024; 22:512. [PMID: 39434182 PMCID: PMC11492701 DOI: 10.1186/s12964-024-01870-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: 07/03/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024] Open
Abstract
Breast cancer (BC) currently ranks second in the global cancer incidence rate. Hypoxia is a common phenomenon in BC. Under hypoxic conditions, cells in the tumor microenvironment (TME) secrete numerous extracellular vesicles (EVs) to achieve intercellular communication and alter the metabolism of primary and metastatic tumors that shape the TME. In addition, emerging studies have indicated that hypoxia can promote resistance to tumor treatment. Engineered EVs are expected to become carriers for cancer treatment due to their high biocompatibility, low immunogenicity, high drug delivery efficiency, and ease of modification. In this review, we summarize the mechanisms of EVs in the primary TME and distant metastasis of BC under hypoxic conditions. Additionally, we highlight the potential applications of engineered EVs in mitigating the malignant phenotypes of BC cells under hypoxia.
Collapse
Affiliation(s)
- Baiheng Zhu
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Kehao Xiang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Tanghua Li
- The First Clinical Medical School, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xin Li
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Fujun Shi
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
29
|
Zhang Z, Chen X, Li Y, Zhang F, Quan Z, Wang Z, Yang Y, Si W, Xiong Y, Ju J, Bian Y, Sun S. The resistance to anoikis, mediated by Spp1, and the evasion of immune surveillance facilitate the invasion and metastasis of hepatocellular carcinoma. Apoptosis 2024; 29:1564-1583. [PMID: 39066845 PMCID: PMC11416391 DOI: 10.1007/s10495-024-01994-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2024] [Indexed: 07/30/2024]
Abstract
Anoikis-Related Genes (ARGs) lead to the organism manifesting resistance to anoikis and are associated with unfavorable prognostic outcomes across various malignancies.Therefore, it is crucial to identify the pivotal target genes related to anoikis in HCC .We found that ARGs were significantly correlated with prognosis and immune responses in HCC. The core gene, SPP1, notably promoted anoikis resistance and metastasis in HCC through both in vivo and in vitro studies. The PI3K-Akt-mTOR pathway played a critical role in anoikis suppression within HCC contexts. Our research unveiled SPP1's role in enhancing PKCα phosphorylation, which in turn activated the PI3K-Akt-mTOR cascade. Additionally, SPP1 was identified as a key regulator of MDSCs and Tregs migration, directly affecting their immunosuppressive capabilities.These findings indicate that in HCC, SPP1 promoted anoikis resistance and facilitated immune evasion by modulating MDSCs and Tregs.
Collapse
Affiliation(s)
- Zhengwei Zhang
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Xiaoning Chen
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Yapeng Li
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Feng Zhang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin Medical University, Harbin, 150081, China
| | - Zhen Quan
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Zhuo Wang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin Medical University, Harbin, 150081, China
| | - Yang Yang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin Medical University, Harbin, 150081, China
| | - Wei Si
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin Medical University, Harbin, 150081, China
| | - Yuting Xiong
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin Medical University, Harbin, 150081, China
| | - Jiaming Ju
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin Medical University, Harbin, 150081, China.
| | - Yu Bian
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin Medical University, Harbin, 150081, China.
| | - Shibo Sun
- The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
30
|
Mei J, Jiang XY, Tian HX, Rong DC, Song JN, Wang L, Chen YS, Wong RCB, Guo CX, Wang LS, Wang LY, Wang PY, Yin JY. Anoikis in cell fate, physiopathology, and therapeutic interventions. MedComm (Beijing) 2024; 5:e718. [PMID: 39286778 PMCID: PMC11401975 DOI: 10.1002/mco2.718] [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: 03/28/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024] Open
Abstract
The extracellular matrix (ECM) governs a wide spectrum of cellular fate processes, with a particular emphasis on anoikis, an integrin-dependent form of cell death. Currently, anoikis is defined as an intrinsic apoptosis. In contrast to traditional apoptosis and necroptosis, integrin correlates ECM signaling with intracellular signaling cascades, describing the full process of anoikis. However, anoikis is frequently overlooked in physiological and pathological processes as well as traditional in vitro research models. In this review, we summarized the role of anoikis in physiological and pathological processes, spanning embryonic development, organ development, tissue repair, inflammatory responses, cardiovascular diseases, tumor metastasis, and so on. Similarly, in the realm of stem cell research focused on the functional evolution of cells, anoikis offers a potential solution to various challenges, including in vitro cell culture models, stem cell therapy, cell transplantation, and engineering applications, which are largely based on the regulation of cell fate by anoikis. More importantly, the regulatory mechanisms of anoikis based on molecular processes and ECM signaling will provide new strategies for therapeutic interventions (drug therapy and cell-based therapy) in disease. In summary, this review provides a systematic elaboration of anoikis, thus shedding light on its future research.
Collapse
Affiliation(s)
- Jie Mei
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Xue-Yao Jiang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Hui-Xiang Tian
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Ding-Chao Rong
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
| | - Jia-Nan Song
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
- School of Life Sciences Westlake University Hangzhou Zhejiang China
| | - Luozixian Wang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
- Centre for Eye Research Australia Royal Victorian Eye and Ear Hospital Melbourne Victoria Australia
- Ophthalmology Department of Surgery The University of Melbourne Melbourne Victoria Australia
| | - Yuan-Shen Chen
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Raymond C B Wong
- Centre for Eye Research Australia Royal Victorian Eye and Ear Hospital Melbourne Victoria Australia
- Ophthalmology Department of Surgery The University of Melbourne Melbourne Victoria Australia
| | - Cheng-Xian Guo
- Center of Clinical Pharmacology the Third Xiangya Hospital Central South University Changsha Hunan China
| | - Lian-Sheng Wang
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Lei-Yun Wang
- Department of Pharmacy Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan Hubei Province China
| | - Peng-Yuan Wang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| |
Collapse
|
31
|
Wen YC, Lin YW, Ho KH, Yang YC, Lai FR, Chu CY, Chen JQ, Lee WJ, Chien MH. The oncogenic ADAMTS1-VCAN-EGFR cyclic axis drives anoikis resistance and invasion in renal cell carcinoma. Cell Mol Biol Lett 2024; 29:126. [PMID: 39333870 PMCID: PMC11429190 DOI: 10.1186/s11658-024-00643-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Metastasis, the leading cause of renal cell carcinoma (RCC) mortality, involves cancer cells resisting anoikis and invading. Until now, the role of the matrix metalloproteinase (MMP)-related enzyme, A disintegrin and metalloprotease with thrombospondin motifs 1 (ADAMTS1), in RCC anoikis regulation remains unclear. METHODS The clinical significance of ADAMTS1 and its associated molecules in patients with RCC was investigated using data from the Gene Expression Omnibus (GEO) and TCGA datasets. Human phosphoreceptor tyrosine kinase (RTK) array, luciferase reporter assays, immunoprecipitation (IP) assays, western blotting, and real-time reverse-transcription quantitative polymerase chain reaction (RT-qPCR) were used to elucidate the underlying mechanisms of ADAMTS1. Functional assays, including anoikis resistance assays, invasion assays, and a Zebrafish xenotransplantation model, were conducted to assess the roles of ADAMTS1 in conferring resistance to anoikis in RCC. RESULTS This study found elevated ADAMTS1 transcripts in RCC tissues that were correlated with a poor prognosis. ADAMTS1 manipulation significantly affected cell anoikis through the mitochondrial pathway in RCC cells. Human receptor tyrosine kinase (RTK) array screening identified that epidermal growth factor receptor (EGFR) activation was responsible for ADAMTS1-induced anoikis resistance and invasion. Further investigations revealed that enzymatically active ADAMTS1-induced versican V1 (VCAN V1) proteolysis led to EGFR transactivation, which in turn, through positive feedback, regulated ADAMTS1. Additionally, ADAMTS1 can form a complex with p53 to influence EGFR signaling. In vivo, VCAN or EGFR knockdown reversed ADAMTS1-induced prometastatic characteristics of RCC. A clinical analysis revealed a positive correlation between ADAMTS1 and VCAN or the EGFR and patients with RCC with high ADAMTS1 and VCAN expression had the worst prognoses. CONCLUSIONS Our results collectively uncover a novel cyclic axis involving ADAMTS1-VCAN-EGFR, which significantly contributes to RCC invasion and resistance to anoikis, thus presenting a promising therapeutic target for RCC metastasis.
Collapse
Affiliation(s)
- Yu-Ching Wen
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
| | - Yung-Wei Lin
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
- International Master/PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuo-Hao Ho
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
| | - Yi-Chieh Yang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
- Department of Medical Research, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Feng-Ru Lai
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
| | - Chih-Ying Chu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
| | - Ji-Qing Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan
- Department of Cancer Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Wei-Jiunn Lee
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan.
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, 111 Xinglong Rd., Sec. 3, Wenshan Dist, Taipei, 11696, Taiwan.
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital Taipei, Taipei, Taiwan.
| |
Collapse
|
32
|
Yin L, Zhang Z, Yan Z, Yan Q. Multicenter cohort analysis of anoikis and EMT: implications for prognosis and therapy in lung adenocarcinoma. Discov Oncol 2024; 15:462. [PMID: 39298078 DOI: 10.1007/s12672-024-01293-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND Anoikis and epithelial-mesenchymal transition (EMT) are pivotal in the distant metastasis of lung adenocarcinoma (LUAD). A detailed understanding of their interplay and the identification of key genes is vital for effective therapeutic strategies against LUAD metastasis. METHODS Key prognostic genes related to anoikis and EMT were identified through univariate Cox regression analysis. We utilized ten machine learning algorithms to develop the Anoikis and EMT-Related Optimal Model (AEOM). The TCGA-LUAD dataset served as the training cohort, while six additional international multicenter LUAD datasets were employed as validation cohorts. The average concordance index (c-index) was used to evaluate model performance and identify the most effective model. Subsequent multi-omics analyses were conducted to explore differences in pathway enrichment, immune infiltration, and mutation landscapes between high and low AEOM groups. Experimental validation demonstrated that RHPN2, a key biomarker within the model, acts as an oncogene facilitating LUAD progression. RESULTS The AEOM displayed superior prognostic predictive performance for LUAD patients, outperforming numerous previously published LUAD signatures. Biologically, the AEOM was notably associated with immune features; the high AEOM group exhibited decreased immune activity and a tendency towards immune-cold tumors, as well as a higher tumor mutational burden (TMB). Subgroup analysis revealed that the low AEOM + high TMB group had the most favorable prognosis. The high AEOM group was primarily enriched in cell cycle-related pathways, promoting cancer cell proliferation. RHPN2, a crucial gene within the AEOM (correlation = 0.85, P < 0.05), was linked to poorer prognosis in LUAD patients with elevated RHPN2 expression. Further in vitro experiments showed that RHPN2 modulates LUAD cell proliferation and invasion. CONCLUSION The AEOM provides a robust prognostic model for LUAD, uncovering critical immune and biological pathways, with RHPN2 identified as a key oncogenic driver. These findings offer valuable insights for targeted therapies and enhanced patient outcomes.
Collapse
Affiliation(s)
- Lu Yin
- School of Mathmatic and Information, Nanjing Normal University of Special Education, Nanjing, China
| | - Zhanshuo Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhu Yan
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an Second People's Hospital, Huai'an, China.
| | - Qiuyue Yan
- Emergency Medicine Department, The Affiliated Huai'an Hospital of Yangzhou University, Huai'an Fifth People's Hospital, Huai'an, China.
| |
Collapse
|
33
|
Zhang T, Chen S, Qu S, Wang L. Anoikis-Related Genes Impact Prognosis and Tumor Microenvironment in Bladder Cancer. Mol Biotechnol 2024:10.1007/s12033-024-01255-x. [PMID: 39172330 DOI: 10.1007/s12033-024-01255-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/25/2024] [Indexed: 08/23/2024]
Abstract
Anoikis tolerance is an important biological process of tumor colonization and metastasis outside the primary tumor. Recent research has progressively elucidated the function and underlying mechanisms of anoikis in the metastasis of various solid tumors. Nevertheless, the specific mechanisms of anoikis in bladder cancer and its consequent effects on the tumor immune microenvironment remain ambiguous. In this study, we developed an anoikis score based on five genes (ETV7, NGF, SCD, LAMC1, and CASP6) and categorized subjects into high and low-risk groups using the median score from the TCGA database. Our findings indicate that SCD enhances the proliferation of bladder cancer cells in vitro. Furthermore, integrating the anoikis score with clinicopathological features to construct a prognostic nomogram demonstrated precision in assessing patient outcomes. Immune cell analysis revealed elevated infiltration levels of Treg cells and M2 macrophages in the high anoikis score group, whereas CD8+ T cell levels were reduced. This study highlights the importance of anoikis score in predicting patient prognosis, immune cell infiltration, and drug response, which may provide a treatment modality worth exploring in depth for the study of bladder cancer.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shaojun Chen
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shanna Qu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Longsheng Wang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
34
|
Liu Y, Ding L, Chen G, Wang P, Wang X. A thermo-sensitive hydrogel with prominent hemostatic effect prevents tumor recurrence via anti-anoikis-resistance. J Nanobiotechnology 2024; 22:496. [PMID: 39164723 PMCID: PMC11334358 DOI: 10.1186/s12951-024-02739-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
Tumor cells can survive when detached from the extracellular matrix (ECM) or lose cell-cell connections, a phenomenon known as anoikis-resistance (AR). AR is closely associated with tumor cell metastasis and recurrence, enabling tumor cells to disseminate, migrate, and invade after detachment. To address this issue, a novel intervention method combining intraoperative hemostasis with multifunctional nanozyme driven-enhanced chemodynamic therapy (ECDT) has been proposed, which holds the potential to weaken the AR capability of tumor cells and suppress tumor recurrence. Here, a nanocomposite containing a dendritic mesoporous nanoframework with Cu2+ was developed using an anion-assisted approach after surface PEG grafting and glucose oxidase (GOx) anchoring (DMSN-Cu@GOx/PEG). DMSN-Cu@GOx/PEG was further encapsulated in a thermal-sensitive hydrogel (H@DMSN-Cu@GOx/PEG). DMSN-Cu@GOx/PEG utilizes its high peroxidase (POD) activity to elevate intracellular ROS levels, thereby weakening the AR capability of bladder cancer cells. Additionally, through its excellent catalase (CAT) activity, DMSN-Cu@GOx/PEG converts the high level of hydrogen peroxide (H2O2) catalyzed by intracellular GOx into oxygen (O2), effectively alleviating tumor hypoxia, downregulating hypoxia-inducible factor-1α (HIF-1α) expression, inhibiting epithelial-mesenchymal transition (EMT) processes, and ultimately suppressing the migration and invasion of bladder cancer cells. Interestingly, in vivo results showed that the thermosensitive hydrogel H@DMSN-Cu@GOx/PEG could rapidly gel at body temperature, forming a gel film on wounds to eliminate residual tumor tissue after tumor resection surgery. Importantly, H@DMSN-Cu@GOx/PEG exhibited excellent hemostatic capabilities, effectively enhancing tissue coagulation during post-tumor resection surgery and mitigating the risk of cancer cell dissemination and recurrence due to surgical bleeding. Such hydrogels undoubtedly possess strong surgical application. Our developed novel nanosystem and hydrogel can inhibit the AR capability of tumor cells and prevent recurrence post-surgery. This study represents the first report of using dendritic mesoporous silica-based nanoreactors for inhibiting the AR capability of bladder cancer cells and suppressing tumor recurrence post-surgery, providing a new avenue for developing strategies to impede tumor recurrence after surgery.
Collapse
Affiliation(s)
- Yang Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
- Hubei Key Laboratory of Urological Diseases, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Lei Ding
- School of Rare Earths, University of Science and Technology of China, Hefei, 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou, 341000, People's Republic of China
| | - Gaojie Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
- Hubei Key Laboratory of Urological Diseases, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Peiyuan Wang
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research On the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, People's Republic of China.
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China.
- Hubei Key Laboratory of Urological Diseases, Wuhan University, Wuhan, 430071, People's Republic of China.
- Medical Research Institute, Wuhan University, Wuhan, 430071, People's Republic of China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China.
| |
Collapse
|
35
|
Sun M, Bai J, Wang H, Li M, Zhou L, Li S. Unraveling the relationship between anoikis-related genes and cancer-associated fibroblasts in liver hepatocellular carcinoma. Heliyon 2024; 10:e35306. [PMID: 39165997 PMCID: PMC11334810 DOI: 10.1016/j.heliyon.2024.e35306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
This study intended to determine the molecular subtypes of liver hepatocellular carcinoma (LIHC) on the strength of anoikis-related genes (ARGs) and to assess their prognostic value and prospective relationship with immune cell infiltration and cancer-associated fibroblasts (CAFs). Univariate Cox regression analysis yielded 66 prognosis-related ARGs and classified LIHC into two distinct subtypes, with subtype A demonstrating overexpression of most prognosis-related ARGs and a significant survival disadvantage. Furthermore, a reliable prediction model was developed using ARGs to evaluate the risk of LIHC patients. This model served as an independent prognostic indicator and a quantitative tool for clinical prognostic prediction. Additionally, subtype-specific differences in immune cell infiltration were observed, and the risk score was potentially linked to immune-related characteristics. Moreover, the study identified a significant association between CAF score and LIHC prognosis, with a low CAF score indicating a favorable patient prognosis. In conclusion, this study reveals the molecular mechanisms underlying the development and progression of LIHC and identifies potential therapeutic targets for the disease.
Collapse
Affiliation(s)
- Meng Sun
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Jiangtao Bai
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Haisong Wang
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Mei Li
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Long Zhou
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Shanfeng Li
- Department of Interventional Vascular Surgery, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
36
|
Li W, Huang M, Wu Z, Zhang Y, Cai Y, Su J, Xia J, Yang F, Xiao D, Yang W, Xu Y, Liu Z. mRNA-Lipid Nanoparticle-Mediated Restoration of PTPN14 Exhibits Antitumor Effects by Overcoming Anoikis Resistance in Triple-Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309988. [PMID: 39189475 PMCID: PMC11348215 DOI: 10.1002/advs.202309988] [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: 12/19/2023] [Revised: 06/11/2024] [Indexed: 08/28/2024]
Abstract
Triple-negative breast cancer (TNBC) poses a challenging prognosis due to early metastasis driven by anoikis resistance. Identifying crucial regulators to overcome this resistance is vital for improving patient outcomes. In this study, a genome-wide CRISPR/Cas9 knockout screen in TNBC cells has identified tyrosine-protein phosphatase nonreceptor type 14 (PTPN14) as a key regulator of anoikis resistance. PTPN14 expression has shown a progressive decrease from normal breast tissue to metastatic tumors. Overexpressing PTPN14 has induced anoikis and inhibited cell proliferation in TNBC cells, while normal human breast cells are unaffected. Mechanistically, PTPN14 is identified as a key factor in dephosphorylating breast cancer antiestrogen resistance 3, a novel substrate, leading to the subsequent inhibition of PI3K/AKT and ERK signaling pathways. Local delivery of in vitro transcribed PTPN14 mRNA encapsulated by lipid nanoparticles in a TNBC mouse model has effectively inhibited tumor growth and metastasis, prolonging survival. The study underscores PTPN14 as a potential therapeutic target for metastatic TNBC, with the therapeutic strategy based on mRNA expression of PTPN14 demonstrating clinical application prospects in alleviating the burden of both primary tumors and metastatic disease.
Collapse
Affiliation(s)
- Wei Li
- Department of Clinical PharmacologyHunan Key Laboratory of Pharmacogeneticsand National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008P. R. China
- Institute of Clinical PharmacologyEngineering Research Center for applied Technology of Pharmacogenomics of Ministry of EducationCentral South UniversityChangsha410078P. R. China
| | - Masha Huang
- Department of Biochemistry and Molecular Cell BiologyShanghai Key Laboratory for Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Zhaoping Wu
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangsha410008P. R. China
| | - Yu Zhang
- Department of Biochemistry and Molecular Cell BiologyShanghai Key Laboratory for Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Ying Cai
- Department of Biochemistry and Molecular Cell BiologyShanghai Key Laboratory for Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Juncheng Su
- Department of Gastrointestinal SurgeryRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127P. R. China
| | - Jia Xia
- Department of NephrologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127P. R. China
| | - Fan Yang
- Department of PhysiologySchool of Basic Medical SciencesShandong UniversityJinan250011P. R. China
| | - Desheng Xiao
- Department of PathologySchool of Basic MedicineXiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Wen Yang
- Department of Biochemistry and Molecular Cell BiologyShanghai Key Laboratory for Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Yingjie Xu
- Department of Biochemistry and Molecular Cell BiologyShanghai Key Laboratory for Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Zhaoqian Liu
- Department of Clinical PharmacologyHunan Key Laboratory of Pharmacogeneticsand National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008P. R. China
- Institute of Clinical PharmacologyEngineering Research Center for applied Technology of Pharmacogenomics of Ministry of EducationCentral South UniversityChangsha410078P. R. China
| |
Collapse
|
37
|
Nalla LV, Khairnar A. Empagliflozin drives ferroptosis in anoikis-resistant cells by activating miR-128-3p dependent pathway and inhibiting CD98hc in breast cancer. Free Radic Biol Med 2024; 220:288-300. [PMID: 38734268 DOI: 10.1016/j.freeradbiomed.2024.05.018] [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: 11/15/2023] [Revised: 02/18/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024]
Abstract
A tumour suppressor miRNA, miR-128-3p, is widely involved in various biological processes and has been found to get downregulated in breast cancer patients. We previously published that ectopically expressed miR-128-3p suppressed migration, invasion, cell cycle arrest, and breast cancer stem cells. In the present study, we explored the role of Empagliflozin (EMPA) as a miR-128-3p functionality-mimicking drug in inducing ferroptosis by inhibiting CD98hc. Given that CD98hc is one of the proteins critical in triggering ferroptosis, we confirmed that miR-128-3p and EMPA inhibited SP1, leading to inhibition of CD98hc expression. Further, transfection with siCD98hc, miR-128-3p mimics, and inhibitors was performed to assess their involvement in the ferroptosis of anoikis-resistant cells. We proved that anoikis-resistant cells possess high ROS and iron levels. Further, miR-128-3p and EMPA treatments induced ferroptosis by inhibiting GSH and enzymatic activity of GPX4 and also induced lipid peroxidation. Moreover, EMPA suppressed bioluminescence of 4T1-Red-FLuc induced thoracic cavity, peritoneal tumour burden and lung nodules in an in-vivo metastatic model of breast cancer. Collectively, we revealed that EMPA sensitized the ECM detached cells to ferroptosis by synergically activating miR-128-3p and lowering the levels of SP1 and CD98hc, making it a potential adjunct drug for breast cancer chemotherapy.
Collapse
Affiliation(s)
- Lakshmi Vineela Nalla
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat, India; Department of Pharmacology, GITAM School of Pharmacy, GITAM (Deemed to be University), Rushikonda, Visakhapatnam, Andhra Pradesh, 530045, India
| | - Amit Khairnar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat, India; International Clinical Research Center, St. Anne's University Hospital Brno, Brno, 602 00, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno, 62500, Czech Republic; International Clinical Research Center, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno, 6250, Czech Republic.
| |
Collapse
|
38
|
Zhang Y, Xue X, Li F, Zhang B, Zheng P, Mi Y. Integrative nomogram model based on anoikis-related genes enhances prognostic evaluation in colorectal cancer. Heliyon 2024; 10:e33637. [PMID: 39040248 PMCID: PMC11261108 DOI: 10.1016/j.heliyon.2024.e33637] [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: 02/27/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Background Revealing the role of anoikis resistance plays in CRC is significant for CRC diagnosis and treatment. This study integrated the CRC anoikis-related key genes (CRC-AKGs) and established a novel model for improving the efficiency and accuracy of the prognostic evaluation of CRC. Methods CRC-ARGs were screened out by performing differential expression and univariate Cox analysis. CRC-AKGs were obtained through the LASSO machine learning algorithm and the LASSO Risk-Score was constructed to build a nomogram clinical prediction model combined with the clinical predictors. In parallel, this work developed a web-based dynamic nomogram to facilitate the generalization and practical application of our model. Results We identified 10 CRC-AKGs and a risk-related prognostic Risk-Score was calculated. Multivariate COX regression analysis indicated that the Risk-Score, TNM stage, and age were independent risk factors that significantly associated with the CRC prognosis(p < 0.05). A prognostic model was built to predict the outcome with satisfied accuracy (3-year AUC = 0.815) for CRC individuals. The web interactive nomogram (https://yuexiaozhang.shinyapps.io/anoikisCRC/) showed strong generalizability of our model. In parallel, a substantial correlation between tumor microenvironment and Risk-Score was discovered in the present work. Conclusion This study reveals the potential role of anoikis in CRC and sets new insights into clinical decision-making in colorectal cancer based on both clinical and sequencing data. Also, the interactive tool provides researchers with a user-friendly interface to input relevant clinical variables and obtain personalized risk predictions or prognostic assessments based on our established model.
Collapse
Affiliation(s)
- Yuexiao Zhang
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Xia Xue
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Fazhan Li
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Bo Zhang
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Pengyuan Zheng
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Yang Mi
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| |
Collapse
|
39
|
Song W, Hu H, Yuan Z, Yao H. A prognostic model for anoikis-related genes in pancreatic cancer. Sci Rep 2024; 14:15200. [PMID: 38956290 PMCID: PMC11220081 DOI: 10.1038/s41598-024-65981-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
Anoikis, a distinct form of programmed cell death, is crucial for both organismal development and maintaining tissue equilibrium. Its role extends to the proliferation and progression of cancer cells. This study aimed to establish an anoikis-related prognostic model to predict the prognosis of pancreatic cancer (PC) patients. Gene expression data and patient clinical profiles were sourced from The Cancer Genome Atlas (TCGA-PAAD: Pancreatic Adenocarcinoma) and the International Cancer Genome Consortium (ICGC-PACA: Pancreatic Ductal Adenocarcinoma). Non-cancerous pancreatic tissue gene expression data were obtained from the Genotype-Tissue Expression (GTEx) project. The R package was used to construct anoikis-related PC prognostic models, which were later validated with the ICGC-PACA database. Survival analyses demonstrated a poorer prognosis for patients in the high-risk group, consistent across both TCGA-PAAD and ICGC-PACA datasets. A nomogram was designed as a predictive tool to estimate patient mortality. The study also analyzed tumor mutations and immune infiltration across various risk groups, uncovering notable differences in tumor mutation patterns and immune landscapes between high- and low-risk groups. In conclusion, this research successfully developed a prognostic model centered on anoikis-related genes, offering a novel tool for predicting the clinical trajectory of PC patients.
Collapse
Affiliation(s)
- Wenbin Song
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, 300052, People's Republic of China
| | - Haiyang Hu
- Department of Cardiac Critical Care Medicine, Affiliated Hospital of Jining Medical University, Jining, 272007, People's Republic of China
| | - Zhengbo Yuan
- School of Medicine, Xiamen University, No.4221 Xiangan South Road, Xiangan District, Xiamen, 361102, People's Republic of China.
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, No.55 Zhenghai load, Siming District, Xiamen, 361001, People's Republic of China.
| | - Hao Yao
- Department of Hepatological Surgery, The Second Hospital of Tianjin Medical University, No.23 Pingjiang Road, Hexi District, Tianjin, 300211, People's Republic of China.
| |
Collapse
|
40
|
Solyanik G, Kolesnik D, Prokhorova I, Yurchenko O, Pyaskovskaya O. Mitochondrial dysfunction significantly contributes to the sensitivity of tumor cells to anoikis and their metastatic potential. Heliyon 2024; 10:e32626. [PMID: 38994085 PMCID: PMC11237942 DOI: 10.1016/j.heliyon.2024.e32626] [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: 11/19/2023] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 07/13/2024] Open
Abstract
It is well-known that the survival of metastatic cells during their dissemination plays an important role in metastasis. However, does this mean that the final result of the metastatic cascade (the volume of metastatic damage to distant organs and tissues) depends on, or at least correlates with, the degree of resistance to anoikis (distinctive hallmarks of metastatic cells)? This question remains open.The aim of the work was to study in vitro the changes in the survival rates, proliferative activity, oxidative stress, and glycolysis intensity during three days of anchorage-dependent and anchorage-independent growth of two Lewis lung carcinoma cell lines (LLC and LLC/R9) and compare these changes with the status of mitochondria and metastatic potential of the cells in vivo. Methods The number and volume of lung metastases were estimated for each cell line after intramuscular inoculation of the cells in C57Bl/6 mice. For the in vitro study, the cells were seeded on Petri dishes pretreated with poly-HEMA or untreated dishes and then allowed to grow for 3 days. Cell viability, cell cycle progression, the level of reactive oxygen species (ROS), glucose consumption and lactate production rates were investigated daily in both growth conditions. An electron microscopy study of intracellular structures was carried out. Results The study showed (as far as we know for the first time) a correlation between the metastatic potential of cells (determined in vivo) and their sensitivity to anoikis (assessed in vitro). The transition of LLC/R9 cells with an inherently defective mitochondrial system to the conditions of anchorage-independent growth was characterized by a decrease in survival, a slowdown in growth rates, an increase in both glucose consumption rate and intracellular ROS levels and manyfold lower metastatic potential, compared to highly metastatic LLC cells with the normal mitochondrial system.
Collapse
Affiliation(s)
- G.I. Solyanik
- Laboratory of Molecular and Cellular Mechanisms of Metastasis, RE Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, The National Academy of Sciences of Ukraine, Kyiv, 03022, Ukraine
| | - D.L. Kolesnik
- Laboratory of Molecular and Cellular Mechanisms of Metastasis, RE Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, The National Academy of Sciences of Ukraine, Kyiv, 03022, Ukraine
| | - I.V. Prokhorova
- Laboratory of Molecular and Cellular Mechanisms of Metastasis, RE Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, The National Academy of Sciences of Ukraine, Kyiv, 03022, Ukraine
| | - O.V. Yurchenko
- Laboratory of Molecular and Cellular Mechanisms of Metastasis, RE Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, The National Academy of Sciences of Ukraine, Kyiv, 03022, Ukraine
| | - O.N. Pyaskovskaya
- Laboratory of Molecular and Cellular Mechanisms of Metastasis, RE Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, The National Academy of Sciences of Ukraine, Kyiv, 03022, Ukraine
| |
Collapse
|
41
|
Silva-Pavez E, Mendoza E, Morgado-Cáceres P, Ahumada-Castro U, Bustos G, Kangme-Encalada M, de Arbina AL, Puebla-Huerta A, Muñoz F, Cereceda L, Varas-Godoy M, Hidalgo Y, Cardenas JC. Mitochondrial division inhibitor (mdivi-1) induces extracellular matrix (ECM)-detachment of viable breast cancer cells by a DRP1-independent mechanism. Sci Rep 2024; 14:14178. [PMID: 38898058 PMCID: PMC11187114 DOI: 10.1038/s41598-024-64228-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024] Open
Abstract
Increasing evidence supports the hypothesis that cancer progression is under mitochondrial control. Mitochondrial fission plays a pivotal role in the maintenance of cancer cell homeostasis. The inhibition of DRP1, the main regulator of mitochondrial fission, with the mitochondrial division inhibitor (mdivi-1) had been associated with cancer cell sensitivity to chemotherapeutics and decrease proliferation. Here, using breast cancer cells we find that mdivi-1 induces the detachment of the cells, leading to a bulk of floating cells that conserved their viability. Despite a decrease in their proliferative and clonogenic capabilities, these floating cells maintain the capacity to re-adhere upon re-seeding and retain their migratory and invasive potential. Interestingly, the cell detachment induced by mdivi-1 is independent of DRP1 but relies on inhibition of mitochondrial complex I. Furthermore, mdivi-1 induces cell detachment rely on glucose and the pentose phosphate pathway. Our data evidence a novel DRP1-independent effect of mdivi-1 in the attachment of cancer cells. The generation of floating viable cells restricts the use of mdivi-1 as a therapeutic agent and demonstrates that mdivi-1 effect on cancer cells are more complex than anticipated.
Collapse
Affiliation(s)
- Eduardo Silva-Pavez
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Bellavista, Bellavista 7, Recoleta, Santiago, Chile.
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile.
| | - Elizabeth Mendoza
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Pablo Morgado-Cáceres
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Ulises Ahumada-Castro
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Galdo Bustos
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Matías Kangme-Encalada
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | | | - Andrea Puebla-Huerta
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Felipe Muñoz
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Lucas Cereceda
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Biomedical Research and Innovation Center (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Manuel Varas-Godoy
- Cancer Cell Biology Lab., Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago, Chile
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Avenida Del Valle Norte 725, Huechuraba, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santos Dumont 964, Independencia, Santiago, Chile
| | - Yessia Hidalgo
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Biomedical Research and Innovation Center (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - J Cesar Cardenas
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile.
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile.
- Buck Institute for Research on Aging, Novato, USA.
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, USA.
| |
Collapse
|
42
|
Zhang T, Wang X, Wang D, Lei M, Hu Y, Chen Z, Li Y, Luo Y, Zhang L, Zhu Y. Synergistic effects of photodynamic therapy and chemotherapy: Activating the intrinsic/extrinsic apoptotic pathway of anoikis for triple-negative breast cancer treatment. BIOMATERIALS ADVANCES 2024; 160:213859. [PMID: 38642515 DOI: 10.1016/j.bioadv.2024.213859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/23/2024] [Accepted: 04/12/2024] [Indexed: 04/22/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly invasive and metastatic subtype of breast cancer that often recurs after surgery. Herein, we developed a cyclodextrin-based tumor-targeted nano delivery system that incorporated the photosensitizer chlorin e6 (Ce6) and the chemotherapeutic agent lonidamine (LND) to form the R6RGD-CMβCD-se-se-Ce6/LND nanoparticles (RCC/LND NPS). This nanosystem could target cancer cells, avoid lysosomal degradation and further localize within the mitochondria. The RCC/LND NPS had pH and redox-responsive to control the release of Ce6 and LND. Consequently, the nanosystem had a synergistic effect by effectively alleviating hypoxia, enhancing the production of cytotoxic reactive oxygen species (ROS) and amplifying the efficacy of photodynamic therapy (PDT). Furthermore, the RCC/LND NPS + light weakened anoikis resistance, disrupted extracellular matrix (ECM), activated both the intrinsic apoptotic pathway (mitochondrial pathway) and extrinsic apoptotic pathway (receptor death pathway) of anoikis. In addition, the nanosystem showed significant anti-TNBC efficacy in vivo. These findings collectively demonstrated that RCC/LND NPS + light enhanced the anticancer effects, induced anoikis and inhibited tumor cell migration and invasion through a synergistic effect of chemotherapy and PDT. Overall, this study highlighted the promising potential of the RCC/LND NPS + light for the treatment of TNBC.
Collapse
Affiliation(s)
- Tianyu Zhang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Xueyuan Wang
- College of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Dongna Wang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Meng Lei
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Yixue Hu
- College of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Zhimeng Chen
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Yuting Li
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Yingnan Luo
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Liefeng Zhang
- College of Life Science, Nanjing Normal University, Nanjing 210023, China; School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| | - Yongqiang Zhu
- College of Life Science, Nanjing Normal University, Nanjing 210023, China; School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
43
|
Glotzbach A, Rohlf K, Gonscharow A, Lüke S, Demirci Ö, Begher-Tibbe B, Overbeck N, Reinders J, Cadenas C, Hengstler JG, Edlund K, Marchan R. EDI3 knockdown in ER-HER2+ breast cancer cells reduces tumor burden and improves survival in two mouse models of experimental metastasis. Breast Cancer Res 2024; 26:87. [PMID: 38816770 PMCID: PMC11138102 DOI: 10.1186/s13058-024-01849-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 05/23/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Despite progress understanding the mechanisms underlying tumor spread, metastasis remains a clinical challenge. We identified the choline-producing glycerophosphodiesterase, EDI3 and reported its association with metastasis-free survival in endometrial cancer. We also observed that silencing EDI3 slowed cell migration and other cancer-relevant phenotypes in vitro. Recent work demonstrated high EDI3 expression in ER-HER2+ breast cancer compared to the other molecular subtypes. Silencing EDI3 in ER-HER2+ cells significantly reduced cell survival in vitro and decreased tumor growth in vivo. However, a role for EDI3 in tumor metastasis in this breast cancer subtype was not explored. Therefore, in the present work we investigate whether silencing EDI3 in ER-HER2+ breast cancer cell lines alters phenotypes linked to metastasis in vitro, and metastasis formation in vivo using mouse models of experimental metastasis. METHODS To inducibly silence EDI3, luciferase-expressing HCC1954 cells were transduced with lentiviral particles containing shRNA oligos targeting EDI3 under the control of doxycycline. The effect on cell migration, adhesion, colony formation and anoikis was determined in vitro, and significant findings were confirmed in a second ER-HER2+ cell line, SUM190PT. Doxycycline-induced HCC1954-luc shEDI3 cells were injected into the tail vein or peritoneum of immunodeficient mice to generate lung and peritoneal metastases, respectively and monitored using non-invasive bioluminescence imaging. Metabolite levels in cells and tumor tissue were analyzed using targeted mass spectrometry and MALDI mass spectrometry imaging (MALDI-MSI), respectively. RESULTS Inducibly silencing EDI3 reduced cell adhesion and colony formation, as well as increased susceptibility to anoikis in HCC1954-luc cells, which was confirmed in SUM190PT cells. No influence on cell migration was observed. Reduced luminescence was seen in lungs and peritoneum of mice injected with cells expressing less EDI3 after tail vein and intraperitoneal injection, respectively, indicative of reduced metastasis. Importantly, mice injected with EDI3-silenced cells survived longer. Closer analysis of the peritoneal organs revealed that silencing EDI3 had no effect on metastatic organotropism but instead reduced metastatic burden. Finally, metabolic analyses revealed significant changes in choline and glycerophospholipid metabolites in cells and in pancreatic metastases in vivo. CONCLUSIONS Reduced metastasis upon silencing supports EDI3's potential as a treatment target in metastasizing ER-HER2+ breast cancer.
Collapse
Affiliation(s)
- Annika Glotzbach
- Leibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Katharina Rohlf
- Leibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Anastasia Gonscharow
- Leibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Simon Lüke
- Leibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Özlem Demirci
- Leibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
- Department of Biology, Science Faculty, Dicle University, Diyarbakir, Turkey
| | - Brigitte Begher-Tibbe
- Leibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Nina Overbeck
- Leibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Jörg Reinders
- Leibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Cristina Cadenas
- Leibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Karolina Edlund
- Leibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Rosemarie Marchan
- Leibniz Research Centre for Working Environment and Human Factors at the TU Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany.
| |
Collapse
|
44
|
Silva D, Quintas C, Gonçalves J, Fresco P. β 2-Adrenoceptor Activation Favor Acquisition of Tumorigenic Properties in Non-Tumorigenic MCF-10A Breast Epithelial Cells. Cells 2024; 13:262. [PMID: 38334654 PMCID: PMC10854540 DOI: 10.3390/cells13030262] [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: 11/28/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
Noradrenaline and adrenaline, and their cognate receptors, are currently accepted to participate in cancer progression. They may also participate in cancer initiation, although their role in this phase is much less explored. The aim of this work was to study the influence of adrenergic stimulation in several processes related to breast cancer carcinogenesis, using several adrenergic agonists in the MCF-10A non-tumorigenic breast cells. Activation of the β-adrenoceptors promoted an epithelial phenotype in MCF-10A cells, revealed by an increased expression of the epithelial marker E-cadherin and a decrease in the mesenchymal markers, N-cadherin and vimentin. MCF-10A cell motility and migration were also impaired after the β-adrenoceptors activation. Concomitant with this effect, β-adrenoceptors decrease cell protrusions (lamellipodia and filopodia) while increasing cell adhesion. Activation of the β-adrenoceptors also decreases MCF-10A cell proliferation. When the MCF-10A cells were cultured under low attachment conditions, activation the of β- (likely β2) or of α2-adrenoceptors had protective effects against cell death, suggesting a pro-survival role of these adrenoceptors. Overall, our results showed that, in breast cells, adrenoceptor activation (mainly through β-adrenoceptors) may be a risk factor in breast cancer by inducing some cancer hallmarks, providing a mechanistic explanation for the increase in breast cancer incidences that may be associated with conditions that cause massive adrenergic stimulation, such as stress.
Collapse
Affiliation(s)
- Dany Silva
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (D.S.); (C.Q.); (P.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal
| | - Clara Quintas
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (D.S.); (C.Q.); (P.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal
| | - Jorge Gonçalves
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (D.S.); (C.Q.); (P.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal
| | - Paula Fresco
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (D.S.); (C.Q.); (P.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
45
|
Deng H, Wei Z, Du J, Shen Z, Zhou C. Predicting the prognosis, immune response, and immunotherapy in head and neck squamous cell carcinoma using a novel risk model based on anoikis-related lncRNAs. Eur J Med Res 2023; 28:548. [PMID: 38017579 PMCID: PMC10683111 DOI: 10.1186/s40001-023-01521-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/09/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is an extremely heterogeneous and metastatic disease. Anoikis, which is a specific type of programmed apoptosis, is involved in tumor metastasis, tissue homeostasis, and development. Herein, we constructed an anoikis-related long non-coding RNA (lncRNA) signature to predict the prognosis, immune responses, and therapeutic effects in HNSCC patients. METHODS A total of 501 HNSCC samples were acquired from the TCGA database and randomly classified into the training and validation groups (1:1 ratio). Thereafter, the results derived from the training set were analyzed with the LASSO regression analysis, and a novel anoikis-related lncRNA risk model was constructed. Time-dependent ROC curves and Kaplan-Meier analysis were carried out to assess the diagnostic value and survival outcomes. A nomogram was utilized to predict the prognostic accuracy. Furthermore, we studied the tumor microenvironment, tumor mutation burden, enrichment pathways, and the response to chemotherapy and immunotherapy. RESULTS Seven anoikis-related lncRNAs (AC015878.1, CYTOR, EMSLR, LINC01503, LINC02084, RAB11B-AS1, Z97200.1) were screened to design a novel risk model, which was recognized as the independent prognostic factor for HNSCC patients. The findings implied that low-risk patients showed significantly longer OS, PFS, and DSS compared to those high-risk patients. The two groups that were classified using the risk model showed significant differences in their immune landscape. The risk model also predicted that low-risk HNSCC patients could attain a better response to immunotherapy, while high-risk patients would be more sensitive to gemcitabine, docetaxel, and cisplatin. CONCLUSIONS We constructed a novel risk model that could be employed for effectively predicting patient prognosis with a good independent prognostic value for HNSCC patients. Furthermore, this model could be used for designing new immunotherapeutic and chemotherapeutic strategies, and it helps clinicians establish personalized and detailed strategies for HNSCC patients.
Collapse
Affiliation(s)
- Hongxia Deng
- Department of Otolaryngology-Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, NingboZhejiang, 315040, China
| | - Zhengyu Wei
- Department of Otolaryngology-Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, NingboZhejiang, 315040, China
| | - Juan Du
- Health Science Center, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Zhisen Shen
- Department of Otolaryngology-Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, NingboZhejiang, 315040, China
| | - Chongchang Zhou
- Department of Otolaryngology-Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, NingboZhejiang, 315040, China.
| |
Collapse
|