1
|
Zhang J, Guo H, Gong C, Shen J, Jiang G, Liu J, Liang T, Guo L. Therapeutic targets in the Wnt signaling pathway: Treating cancer with specificity. Biochem Pharmacol 2025; 236:116848. [PMID: 40049295 DOI: 10.1016/j.bcp.2025.116848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 04/20/2025]
Abstract
The Wnt signaling pathway is a critical regulatory mechanism that governs cell cycle progression, apoptosis, epithelial-mesenchymal transition (EMT), angiogenesis, stemness, and the tumor immune microenvironment, while also maintaining tissue homeostasis. Dysregulated activation of this pathway is implicated in various cancers, closely linked to tumor initiation, progression, and metastasis. The Wnt/β-catenin axis plays a central role in the pathogenesis of common cancers, including colorectal cancer (CRC), breast cancer (BC), liver cancer, and lung cancer. Unlike traditional chemotherapy, targeted therapy offers a more precise approach to cancer treatment. As a key regulator of oncogenesis, the Wnt pathway represents a promising target for clinical interventions. This review provides a comprehensive analysis of the Wnt signaling pathway, exploring its roles in tumor biology and its implications in human malignancies. It further examines the molecular mechanisms and modes of action across different cancers, detailing how the Wnt pathway contributes to tumor progression through mechanisms such as metastasis promotion, immune modulation, drug resistance, and enhanced cellular proliferation. Finally, therapeutic strategies targeting Wnt pathway components are discussed, including inhibitors targeting extracellular members, as well as those within the cell membrane, cytoplasm, and nucleus. The potential of these targets in the development of novel therapeutic agents underscores the critical importance of intervening in the Wnt signaling pathway for effective cancer treatment.
Collapse
Affiliation(s)
- Jiaxi Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Haochuan Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Chengxuan Gong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Jie Shen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Guijie Jiang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Jiarui Liu
- State Key Laboratory of Flexible Electronics (LoFE), Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China.
| | - Li Guo
- State Key Laboratory of Flexible Electronics (LoFE), Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| |
Collapse
|
2
|
Shi C, Li Y, Zou X, Yao D, Wei J, Liu Z, Su Y, Yu B, Zhang X, Wang Z, Liang H, Gang H, Zhao Y, Gu M. AP2M1 is a prognostic marker associated with cell cycle arrest and the tumor immune microenvironment in acute myeloid leukemia. Leuk Lymphoma 2025:1-15. [PMID: 40386881 DOI: 10.1080/10428194.2025.2504161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/09/2025] [Accepted: 05/04/2025] [Indexed: 05/20/2025]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous clonal disease of hematopoietic progenitor cells and the most common malignant myeloid disease in adults. Although significant progress has been made in treatment, the outlook remains bleak, and new therapeutic targets need to be sought. AP-2 complex subunit mu (AP2M1) is a core component of the clathrin-mediated endocytic machinery, AP2M1 plays a critical role in cancer progression. However, its function in acute myeloid leukemia (AML) progression remains unclear. Our study reveals that AP2M1 is highly expressed in AML and is associated with poor prognosis. Mechanistic studies suggest that this effect may result through cell cycle arrest and is associated with the tumor microenvironment, and our findings suggest that AP2M1 is a potential oncogene and prognostic marker for AML.
Collapse
Affiliation(s)
- Ce Shi
- NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yang Li
- NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xing Zou
- NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Dayong Yao
- Department of Urology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jia Wei
- NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhiyu Liu
- Department of Laboratory Diagnostics, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yue Su
- NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Boqian Yu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Xin Zhang
- NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhenkun Wang
- NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hui Liang
- Heilongjiang Province Key Laboratory of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hao Gang
- Harbin Medical University, Harbin, Heilongjiang, China
| | - Yanhong Zhao
- Department of Hematology, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Mengmeng Gu
- NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
3
|
Sun MX, Zhu HC, Yu Y, Yao Y, Li HY, Feng FB, Wang QY, Liu RJ, Sun CG. Role of the Wnt signaling pathway in the complex microenvironment of breast cancer and prospects for therapeutic potential (Review). Int J Oncol 2025; 66:36. [PMID: 40145557 PMCID: PMC12068849 DOI: 10.3892/ijo.2025.5742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
The focus on breast cancer treatment has shifted from the cytotoxic effects of single drugs on tumor cells to multidimensional multi‑pathway synergistic intervention strategies targeting the tumor microenvironment (TME). The activation of the Wnt signaling pathway in the TME of breast cancer cells serves a key regulatory role in tissue homeostasis and is a key driver of the carcinogenic process. Modulating the crosstalk between the Wnt pathway and TME of breast cancer is key for understanding the biological behavior of breast cancer and advancing the development of novel antitumor drugs. The present review aimed to summarize the complex mechanisms of the Wnt signaling pathway in the breast cancer TME, interactions between the Wnt signaling pathway and components of the breast cancer TME and breast cancer‑associated genes, as well as the interactions between the Wnt signaling pathway and other signaling cascades at the molecular level. Furthermore, the present review aimed to highlight the unique advantages of the Wnt signaling pathway in the macro‑regulation of the TME and the current therapeutic strategies targeting the Wnt signaling pathway, their potential clinical value and future research directions in breast cancer treatment.
Collapse
Affiliation(s)
- Meng Xuan Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Han Ci Zhu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Yang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Hua Yao Li
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Fu Bin Feng
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Qing Yang Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Rui Juan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Chang Gang Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
4
|
Wu C, Gao Y, Jin Z, Huang Z, Wang H, Lu S, Guo S, Zhang F, Zhang J, Huang J, Tao X, Liu X, Zhang X, You L, Li Q, Wu J. PTPRG-AS1 regulates the KITLG/KIT pathway through the ceRNA axis to promote the malignant progression of gastric cancer and the intervention effect of Compound Kushen injection on it. Pharmacol Res 2025; 215:107743. [PMID: 40250508 DOI: 10.1016/j.phrs.2025.107743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/12/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
Gastric cancer (GC) is a common malignant tumor with high mortality, recurrence, and metastasis rates. Compound Kushen injection (CKI) combination chemotherapy has been clinically used for the treatment of GC in China for many years, but its underlying mechanisms of action remain unclear. Recent reports have highlighted the important role of the competing endogenous RNA (ceRNA) mechanism of noncoding RNA (ncRNA) and messenger RNA (mRNA) formation in GC and other tumors. This study aimed to investigate the effects of CKI on GC from the ceRNA perspective. We confirmed the inhibitory effect of CKI on GC in mouse models and cell lines. By examining the GC cell lines sensitive to CKI treatment, we developed the CNScore method to analyze the ceRNA network, revealing that the CKI-GC ceRNA network promotes GC proliferation and metastasis through the PTPRG-AS1/hsa-miR-421/KITLG axis. Finally, we constructed GC cell models with PTPRG-AS1 overexpression or knockdown and GC liver metastasis models and found that PTPRG-AS1 can sponge hsa-miR-421, releasing KITLG and promoting GC proliferation and metastasis through the KITLG/KIT pathway. Taken together, CKI can suppress these malignant phenotypes by regulating the PTPRG-AS1/hsa-miR-421/KITLG axis.
Collapse
Affiliation(s)
- Chao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yifei Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhengsen Jin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhihong Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Haojia Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shan Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Siyu Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Fanqin Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jingyuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiaqi Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaoyu Tao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xinkui Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Xiaomeng Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Leiming You
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Qinglin Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang Province 310022, China.
| | - Jiarui Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
5
|
Xun Y, Chen G, Tang G, Zhang C, Zhou S, Fong TL, Chen Y, Xiong R, Wang N, Feng Y. Traditional Chinese medicine and natural products in management of hepatocellular carcinoma: Biological mechanisms and therapeutic potential. Pharmacol Res 2025; 215:107733. [PMID: 40209965 DOI: 10.1016/j.phrs.2025.107733] [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: 10/09/2024] [Revised: 03/27/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
Hepatocellular carcinoma (HCC), originating from hepatocytes, is the most common type of primary liver cancer. HCC imposes a significant global health burden with high morbidity and mortality, making it a critical public concern. Surgical interventions, including hepatectomy and liver transplantation, are pivotal in achieving long-term survival for patients with HCC. Additionally, ablation therapy, endovascular interventional therapy, radiotherapy, and systemic anti-tumor therapies are commonly employed. However, these treatment modalities are often associated with considerable challenges, including high postoperative recurrence rates and adverse effects. Traditional Chinese medicine (TCM) and natural products have been utilized for centuries as a complementary approach in managing HCC and its complications, demonstrating favorable clinical outcomes. Various bioactive compounds derived from TCM and natural products have been identified and purified, and their mechanisms of action have been extensively investigated. This review aims to provide a comprehensive and up-to-date evaluation of the clinical efficacy of TCM, natural products and their active constituents in the treatment and management of HCC. Particular emphasis is placed on elucidating the potential molecular mechanisms and therapeutic targets of these agents, including their roles in inhibiting HCC cell proliferation, inducing apoptosis and pyroptosis, suppressing tumor invasion and metastasis, and restraining angiogenesis within HCC tissues.
Collapse
Affiliation(s)
- Yunqing Xun
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Guang Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Guoyi Tang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Shichen Zhou
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Tung-Leong Fong
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Yue Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Ruogu Xiong
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong.
| |
Collapse
|
6
|
Xing G, Cui Y, Guo Z, Han B, Zhao G. Progress on the mechanism of intestinal microbiota against colorectal cancer. Front Cell Infect Microbiol 2025; 15:1565103. [PMID: 40357397 PMCID: PMC12066583 DOI: 10.3389/fcimb.2025.1565103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/24/2025] [Indexed: 05/15/2025] Open
Abstract
The intestinal microbiota plays a crucial role in the occurrence and development of colorectal cancer, and its anti - colorectal cancer mechanism has become a research hotspot. This article comprehensively expounds on the molecular mechanisms of the intestinal microbiota in anti - colorectal cancer, including aspects such as immune regulation, activation of carcinogenic signaling pathways (it should be noted that it is more reasonable to be "inhibition of carcinogenic signaling pathways"), metabolite - mediated effects, and maintenance of intestinal barrier function. At the same time, it explores the roles and potential mechanisms of intervention methods such as probiotic supplementation therapy, immunotherapy, and fecal microbiota transplantation. In addition, it analyzes the impact of the intestinal flora on the therapeutic efficacy of colorectal cancer. The existing research results are summarized, and the future research directions are prospected, with the aim of providing new theoretical bases and treatment ideas for the prevention and treatment of colorectal cancer.
Collapse
Affiliation(s)
- Guoqiang Xing
- Department of General Surgery, Tianjin Fifth Central Hospital, Tianjin, China
| | - Yu Cui
- Department of General Surgery, Tianjin Fifth Central Hospital, Tianjin, China
| | - Zhiyue Guo
- College of Integrated Traditional Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bing Han
- Department of General Surgery, Tianjin Fifth Central Hospital, Tianjin, China
| | - Guogang Zhao
- Department of General Surgery, Tianjin Fifth Central Hospital, Tianjin, China
| |
Collapse
|
7
|
Lan X, Li W, Zhao K, Wang J, Li S, Zhao H. Revisiting the role of cancer-associated fibroblasts in tumor microenvironment. Front Immunol 2025; 16:1582532. [PMID: 40313969 PMCID: PMC12043473 DOI: 10.3389/fimmu.2025.1582532] [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: 02/24/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are integral components of the tumor microenvironment playing key roles in tumor progression, metastasis, and therapeutic resistance. However, challenges persist in understanding their heterogeneity, origin, and functional diversity. One major obstacle is the lack of standardized naming conventions for CAF subpopulations, with current systems failing to capture their full complexity. Additionally, the identification of CAFs is hindered by the absence of specific biomarkers, limiting the precision of diagnostic and therapeutic strategies. In vitro culture conditions often fail to maintain the in vivo characteristics of CAFs, which complicates their study and the translation of findings to clinical practice. Although current detection methods, such as antibodies, mRNA probes, and single-cell transcriptomics, offer insights into CAF biology, they lack standardization and fail to provide reliable quantitative measures. Furthermore, the dynamic interactions between CAFs, tumor cells, and immune cells within the TME remain insufficiently understood, and the role of CAFs in immune evasion and therapy resistance is an area of ongoing research. Understanding how CAFs influence drug resistance and the immune response is essential for developing more effective cancer therapies. This review aims to provide an in-depth analysis of the challenges in CAF research, propose future research directions, and emphasize the need for improved CAF-targeted therapeutic strategies. By addressing these gaps, it seeks to highlight the potential of CAFs as targets for overcoming therapeutic resistance and enhancing the efficacy of cancer treatments.
Collapse
Affiliation(s)
| | | | | | | | | | - Hai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
8
|
Tao S, Gao Y, Wang X, Wu C, Zhang Y, Zhu H, Li J. CAF-derived exosomal LINC01711 promotes breast cancer progression by activating the miR-4510/NELFE axis and enhancing glycolysis. FASEB J 2025; 39:e70471. [PMID: 40172996 DOI: 10.1096/fj.202402024rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 04/04/2025]
Abstract
Breast cancer (BRCA) is among the most prevalent malignancies in women, characterized by a complex tumor microenvironment significantly influenced by cancer-associated fibroblasts (CAFs). CAFs contribute to tumor progression by secreting exosomes that can modulate cancer cell behavior. This study highlights how CAF-derived exosomes transmit the long non-coding RNA (lncRNA) LINC01711, which activates TXN through the miR-4510/NELFE axis, thereby enhancing glycolysis in BRCA cells. Utilizing BRCA single-cell sequencing data from the GEO database, the study employed dimensionality reduction, clustering, and cell annotation techniques to uncover the central role of NELFE in BRCA. Experimental findings revealed that LINC01711 is highly expressed in CAF-derived exosomes, which upregulate TXN via the miR-4510/NELFE axis, promoting the glycolytic pathway and subsequently increasing the proliferation, migration, and invasion potential of BRCA cells. These results shed light on a novel molecular mechanism underlying BRCA progression and suggest potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Shuang Tao
- Wujin Hospital Affiliated with Jiangsu University, Changzhou, People's Republic of China
- The Wujin Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yali Gao
- Ningxia Medical University, Yinchuan, People's Republic of China
| | - Xiang Wang
- Wujin Hospital Affiliated with Jiangsu University, Changzhou, People's Republic of China
- The Wujin Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Chunxia Wu
- Wujin Hospital Affiliated with Jiangsu University, Changzhou, People's Republic of China
- The Wujin Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yi Zhang
- Wujin Hospital Affiliated with Jiangsu University, Changzhou, People's Republic of China
- The Wujin Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Hong Zhu
- Wujin Hospital Affiliated with Jiangsu University, Changzhou, People's Republic of China
- The Wujin Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Jinping Li
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan, People's Republic of China
| |
Collapse
|
9
|
Deng H, Wu D, He Y, Yu X, Liu J, Zhang Y, Leng B, Yuan X, Xiao L. E2F1-driven EXOSC10 transcription promotes hepatocellular carcinoma growth and stemness: a potential therapeutic target. Hereditas 2025; 162:60. [PMID: 40221814 PMCID: PMC11992873 DOI: 10.1186/s41065-025-00430-7] [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: 01/14/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND E2F Transcription Factor 1 (E2F1) is a transcription factor that plays a crucial role in the growth of many cancers, including hepatocellular carcinoma (HCC). Herein, this study probed the functions and underlying mechanisms of E2F1 in HCC tumorigenesis. METHODS The expression profiles of E2F1 and Exosome Component 10 (EXOSC10) were detected using qRT-PCR and western blotting. Functional experiments were carried out using 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, tube formation, and sphere formation assays in vitro, as well as xenograft experiments in vivo, respectively. Stemness-related proteins were assayed using western blotting. The interaction between E2F1 and EXOSC10 was verified using bioinformatics analysis and dual-luciferase reporter assay. RESULTS E2F1 was highly expressed in HCC tissues and cells, and was associated with advanced TNM stage, distant metastasis, and short survival rate. Functionally, knockdown of E2F1 suppressed HCC cell proliferation, angiogenesis, and stemness, and induced cell apoptosis. Mechanistically, E2F1 directly bound to the promoter region of EXOSC10 to up-regulate its expression. EXOSC10 silencing impaired HCC cell proliferation, angiogenesis, and stemness. Moreover, the anticancer effects of E2F1 knockdown were reversed by EXOSC10 elevation. In vivo assay, E2F1 deficiency suppressed HCC tumor growth and eliminated cancer stemness, while these effects were abolished by EXOSC10 up-regulation. CONCLUSION E2F1 promotes EXOSC10 transcription and then facilitates HCC growth and cancer stemness, revealing a potential target for HCC therapy.
Collapse
Affiliation(s)
- Haoyue Deng
- Department of Pathology, Suining Central Hospital, Suining, 629000, Sichuan, China
| | - Dingyong Wu
- Department of Oncology, Songshan General Hospital, Chongqing, 401120, China
| | - Yongpeng He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Inaffiliationidualized Treatment, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, 400030, China
| | - Xiaolei Yu
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China
| | - Jifei Liu
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China
| | - Yanrui Zhang
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China
| | - Bing Leng
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China
| | - Xiaofeng Yuan
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China.
| | - Liguo Xiao
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China.
| |
Collapse
|
10
|
Chen Q, Jin J, Li P, Wang X, Wang Q. Navigating Glioma Complexity: The Role of Abnormal Signaling Pathways in Shaping Future Therapies. Biomedicines 2025; 13:759. [PMID: 40149733 PMCID: PMC11940491 DOI: 10.3390/biomedicines13030759] [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: 02/21/2025] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
Gliomas are a type of highly heterogeneous and invasive central nervous system tumor. Traditional treatment methods have limited efficacy, and the prognosis for patients remains poor. Recent studies have revealed the crucial roles of several abnormal signaling pathways in the pathogenesis of gliomas, including the Receptor Tyrosine Kinase/Rat Sarcoma Virus Oncogene/Phosphatidylinositol-3-Kinase (RTK/RAS/PI3K) pathway, the Wingless-Related Integration Site/β-Catenin (Wnt/β-Catenin) pathway, the Hippo/YAP (Hippo/Yes-associated protein) pathway, and the Slit/Robo (Slit Guidance Ligands/Roundabout) signaling pathway. These pathways play extremely vital roles in tumor proliferation, invasion, and treatment resistance. This article comprehensively and systematically reviews the molecular mechanisms of these signaling pathways, deeply summarizing the research progress of various treatment strategies, including targeted inhibitors, gene therapy, and nanomedicine against them. Moreover, the combination of targeted therapy and personalized treatment regimens is expected to overcome the current treatment bottleneck and provide a more favorable survival prognosis for glioblastoma patients.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Pharmacy, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China;
| | - Jin Jin
- Department of Rehabilitation, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China;
| | - Pian Li
- Liyuan Cardiovascular Center, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China;
| | - Xiuping Wang
- Department of Pharmacy, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China;
| | - Qianyan Wang
- Liyuan Cardiovascular Center, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China;
| |
Collapse
|
11
|
Xu Y, Xie K, Li L, Li Z, Lu Q, Feng J. FOXN3 Downregulation in Colorectal Cancer Enhances Tumor Cell Stemness by Promoting EP300-Mediated Epigenetic Upregulation of SOX12. Mol Carcinog 2025; 64:410-424. [PMID: 39607349 DOI: 10.1002/mc.23852] [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: 06/12/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024]
Abstract
Cancer stemness plays a crucial role in promoting the progression of colorectal cancer (CRC). Forkhead box N3 (FOXN3) is a tumor suppressor protein. Herein, we investigated the role of FOXN3 in the regulation of CRC cell stemness. Cell viability, proliferation, migration, and invasion were assessed utilizing cell counting kit-8 assay, 5-ethynyl-20-deoxyuridine assay, and Transwell assay, respectively. Cell-sphere formation was assessed using a sphere-forming assay. The enrichment of H3K27ac modifications at the SRY-related HMG-box 12 (SOX12) promoter, interactions among FOXN3, SOX12, and E1A binding protein p300 (EP300) were analyzed using chromatin immunoprecipitation or dual luciferase reporter assays. We found that FOXN3 overexpression inhibited CRC cell proliferation, migration, invasion, stemness, and tumor formation in mice by inactivating the Wnt/β-catenin signaling, while these effects of FOXN3 overexpression were reversed by the overexpression of SOX12. Mechanistically, EP300 increased SOX12 expression in CRC cells by promoting H3K27ac enrichment in the SOX12 promoter. In addition, FOXN3 transcriptionally inhibited EP300 expression in CRC cells by binding to the EP300 promoter. As expected, EP300 overexpression weakened the inhibitory effect of FOXN3 overexpression on CRC cell stemness. Collectively, FOXN3 upregulation inhibited CRC cell stemness by suppressing EP300-mediated epigenetic upregulation of SOX12.
Collapse
Affiliation(s)
- Yanjie Xu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ke Xie
- Department of Gastrointestinal Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ling Li
- Department of Gastrointestinal Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhong Li
- Department of Gastrointestinal Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Qicheng Lu
- Department of Gastrointestinal Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jin Feng
- Department of Gastrointestinal Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
12
|
Liu J, Hu C, Jin Y. ALDOB suppresses the activity of CD8 + T cells in colorectal cancer via the WNT signaling pathway. Immunol Cell Biol 2025; 103:307-316. [PMID: 39909069 DOI: 10.1111/imcb.12853] [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: 08/26/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/07/2025]
Abstract
The glycolytic enzyme, fructose-1,6-bisphosphate aldolase B (ALDOB), is recognized for its key role in shaping tthe umor immune microenvironment. However, the precise ways in which it influences the CD8+ T cell immune response in colorectal cancer (CRC) are still largely unknown. This study is designed to elucidate the interplay between ALDOB and the immune system in CRC. We analyzed the high expression of ALDOB in CRC tissues and cells through bioinformatics, clinical samples and in vitro experiments, finding that it promoted tumor progression. Its high expression was negatively correlated with CD8 expression and positively correlated with PDL1 expression. Further cell experiments revealed that ALDOB overexpression enhanced the expression of WNT signaling pathway-related proteins (β-catenin and c-myc), which in turn promoted PDL1 expression in CRC cells, inhibiting the proliferation and killing effect of CD8+ T cells in co-culture systems. Our findings disclose how ALDOB influences CD8+ T cell recruitment and antitumor immune function, proposing it as a potential target for the treatment of CRC.
Collapse
Affiliation(s)
- Jinwei Liu
- Department of Gastrointestinal Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Chao Hu
- Department of Reproductive Medicine, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Yuan Jin
- Department of Gastrointestinal Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| |
Collapse
|
13
|
Clevenger AJ, Collier CA, Gorley JPM, Colijn S, McFarlin MK, Solberg SC, Kopetz ES, Stratman AN, Raghavan SA. Oncogenic KRAS Mutations Confer a Unique Mechanotransduction Response to Peristalsis in Colorectal Cancer Cells. Mol Cancer Res 2025; 23:128-142. [PMID: 39485528 PMCID: PMC11802306 DOI: 10.1158/1541-7786.mcr-24-0624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/04/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024]
Abstract
Colorectal cancer tumors start as polyps on the inner lining of the colorectum, in which they are exposed to the mechanics of peristalsis. Our previous work leveraged a custom-built peristalsis bioreactor to demonstrate that colonic peristalsis led to cancer stem cell enrichment in colorectal cancer cells. However, this malignant mechanotransductive response was confined to select colorectal cancer lines that harbored an oncogenic mutation in the Kirsten rat sarcoma virus (KRAS) gene. In this study, we explored the involvement of activating KRAS mutations on peristalsis-associated mechanotransduction in colorectal cancer. Peristalsis enriched cancer stem cell marker Leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) in KRAS mutant lines in a Wnt ligand-independent manner. Conversely, LGR5 enrichment in wild-type KRAS lines exposed to peristalsis were minimal. LGR5 enrichment downstream of peristalsis translated to increased tumorigenicity in vivo. Differences in mechanotransduction were apparent via unbiased gene set enrichment analysis, in which many unique pathways were enriched in wild-type versus mutant lines. Peristalsis also triggered β-catenin nuclear localization independent of Wnt ligands, particularly in KRAS mutant lines. The involvement of KRAS was validated via gain and loss of function strategies. Peristalsis-induced β-catenin activation and LGR5 enrichment depended on the activation of the MEK/ERK cascade. Taken together, our results demonstrated that oncogenic KRAS mutations conferred a unique peristalsis-associated mechanotransduction response to colorectal cancer cells, resulting in cancer stem cell enrichment and increased tumorigenicity. These mechanosensory connections can be leveraged in improving the sensitivity of emerging therapies that target oncogenic KRAS. Implications: Oncogenic KRAS empowers colorectal cancer cells to harness the mechanics of colonic peristalsis for malignant gain independent of other cooperating signals.
Collapse
Affiliation(s)
| | - Claudia A. Collier
- Department of Biomedical Engineering, Texas A&M University, College Station, TX
| | - John Paul M. Gorley
- Department of Biomedical Engineering, Texas A&M University, College Station, TX
| | - Sarah Colijn
- Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Maygan K. McFarlin
- Department of Biomedical Engineering, Texas A&M University, College Station, TX
| | - Spencer C. Solberg
- Department of Biomedical Engineering, Texas A&M University, College Station, TX
| | - E. Scott Kopetz
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX
| | - Amber N. Stratman
- Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Shreya A. Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX
| |
Collapse
|
14
|
Chen L, Tang J, Chang Y, Hang D, Ji J, Chen G. SMURF1 leads to the β-catenin signaling-mediated progression of esophageal squamous carcinoma by losing PATZ1-induced CCNG2 transcription. Biochem Pharmacol 2025; 232:116688. [PMID: 39617210 DOI: 10.1016/j.bcp.2024.116688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 11/10/2024] [Accepted: 11/28/2024] [Indexed: 12/16/2024]
Abstract
Cyclin G2 (CCNG2), a known inhibitor of cell cycle progression, has been identified as a suppressor for the canonical β-catenin pathway. This study explores the impact of CCNG2 on β-catenin activity and malignant characteristics of esophageal squamous cell carcinoma (ESCC) cells, and the mechanism behind CCNG2 dysregulation. In ESCC tissues and cells, CCNG2 was under-expressed and associated with poor clinical outcomes, whereas β-catenin showed an opposite trend. Inducing CCNG2 overexpression in ESCC cells led to a reduction in β-catenin levels, which in turn suppressed proliferation, cell cycle progression, migration, invasion, stemness, and tumorigenesis. Additionally, it enhanced the cytotoxicity and proliferation of T cells in co-culture systems. However, these beneficial effects were negated by the Wnt signaling agonist BML-284. Furthermore, PATZ1 was found as a transcription factor promoting CCNG2 transcription. However, the PATZ1 protein in ESCC cells was degraded by SMURF1. Silencing of SMURF1 restored CCNG2 expression and inhibited β-catenin, thereby suppressing the malignant phenotype of ESCC cells and reducing T cell exhaustion. Yet, these effects were blocked by further silencing of PATZ1. In summary, this research demonstrates that SMURF1 activates β-catenin signaling by suppressing the PATZ1/CCNG2 axis, thereby promoting the progression of ESCC.
Collapse
Affiliation(s)
- Lingling Chen
- Department of Gastroenterology, Pudong New Area People's Hospital, Shanghai 201299, PR China
| | - Jie Tang
- Department of Gastroenterology, Jiangwan Hospital, Hongkou District, Shanghai 200434, PR China
| | - Yunli Chang
- Department of Gastroenterology, Pudong New Area People's Hospital, Shanghai 201299, PR China
| | - Dongyun Hang
- Department of Gastroenterology, Pudong New Area People's Hospital, Shanghai 201299, PR China
| | - Jieru Ji
- Department of Gastroenterology, Pudong New Area People's Hospital, Shanghai 201299, PR China.
| | - Guoyu Chen
- Department of Gastroenterology, Pudong New Area People's Hospital, Shanghai 201299, PR China.
| |
Collapse
|
15
|
Duan Y, Xu Y, Dou Y, Xu D. Helicobacter pylori and gastric cancer: mechanisms and new perspectives. J Hematol Oncol 2025; 18:10. [PMID: 39849657 PMCID: PMC11756206 DOI: 10.1186/s13045-024-01654-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Gastric cancer remains a significant global health challenge, with Helicobacter pylori (H. pylori) recognized as a major etiological agent, affecting an estimated 50% of the world's population. There has been a rapidly expanding knowledge of the molecular and pathogenetic mechanisms of H. pylori over the decades. This review summarizes the latest research advances to elucidate the molecular mechanisms underlying the H. pylori infection in gastric carcinogenesis. Our investigation of the molecular mechanisms reveals a complex network involving STAT3, NF-κB, Hippo, and Wnt/β-catenin pathways, which are dysregulated in gastric cancer caused by H. pylori. Furthermore, we highlight the role of H. pylori in inducing oxidative stress, DNA damage, chronic inflammation, and cell apoptosis-key cellular events that pave the way for carcinogenesis. Emerging evidence also suggests the effect of H. pylori on the tumor microenvironment and its possible implications for cancer immunotherapy. This review synthesizes the current knowledge and identifies gaps that warrant further investigation. Despite the progress in our previous knowledge of the development in H. pylori-induced gastric cancer, a comprehensive investigation of H. pylori's role in gastric cancer is crucial for the advancement of prevention and treatment strategies. By elucidating these mechanisms, we aim to provide a more in-depth insights for the study and prevention of H. pylori-related gastric cancer.
Collapse
Affiliation(s)
- Yantao Duan
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yonghu Xu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Dou
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dazhi Xu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
16
|
Xu Y, Yang L, Li G, Rao C. The Role of NF-κB/MIR155HG in Regulating the Stemness and Radioresistance in Breast Cancer Stem Cells. FRONT BIOSCI-LANDMRK 2025; 30:25810. [PMID: 39862080 DOI: 10.31083/fbl25810] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Breast cancer stem cells (BCSCs) are instrumental in treatment resistance, recurrence, and metastasis. The development of breast cancer and radiation sensitivity is intimately pertinent to long non-coding RNA (lncRNA). This work is formulated to investigate how the lncRNA MIR155HG affects the stemness and radioresistance of BCSCs. METHODS Effects of MIR155HG knockdown on BCSCs were gauged in MCF-7 and MDA-MB-231 cell lines. MIR155HG expression was manipulated in cells, followed by an assessment of stemness, DNA damage repair, apoptosis, cell cycle, and the Wnt signaling pathway under radiation conditions. The interaction between nuclear factor kappa B (NF-κB) subunit RelA and MIR155HG was examined using a dual-luciferase reporter assay. To examine the binding interaction between RelA and MIR155HG promoter, chromatin immunoprecipitation was performed. RESULTS Breast cancer-derived stem cells exhibited a high level of MIR155HG. Knockdown of MIR155HG reduced stemness, enhanced radiosensitivity, induced apoptosis, and arrested cells in the G1 phase. Mechanistically, MIR155HG knockdown repressed Wnt/β-catenin signaling and mediated apoptosis-related protein expressions. NF-κB subunit RelA transcriptionally activated MIR155HG, thereby contributing to radioresistance in BCSCs. CONCLUSION NF-κB regulates MIR155HG transcriptionally to activate the Wnt pathway, thus enhancing stemness and radioresistance in BCSCs. Targeting MIR155HG may enhance the susceptibility of cancer stem cells to radiation-induced cell death, potentially improving therapeutic outcomes. These findings underscore MIR155HG as a promising therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Yunbao Xu
- Department of Chemoradiotherapy, Ningbo No 2 Hospital, 315000 Ningbo, Zhejiang, China
| | - Lu Yang
- Department of Chemoradiotherapy, Ningbo No 2 Hospital, 315000 Ningbo, Zhejiang, China
| | - Guangming Li
- Department of Breast Surgery, Ningbo No 2 Hospital, 315000 Ningbo, Zhejiang, China
| | - Chuangzhou Rao
- Department of Chemoradiotherapy, Ningbo No 2 Hospital, 315000 Ningbo, Zhejiang, China
| |
Collapse
|
17
|
Zhang H, Kim H, Yuan T, Zhang Z, Kaul SC, Wadhwa R. Molecular Characterization of Cancer Preventive and Therapeutic Potential of Three Antistress Compounds, Triethylene Glycol, Withanone, and Withaferin A. Int J Mol Sci 2025; 26:493. [PMID: 39859209 PMCID: PMC11764651 DOI: 10.3390/ijms26020493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The molecular link between stress and carcinogenesis and the positive outcomes of stress intervention in cancer therapy have recently been well documented. Cancer stem cells (CSCs) facilitate cancer malignancy, drug resistance, and relapse and, hence, have emerged as a new therapeutic target. Here, we aimed to investigate the effect of three previously described antistress compounds (triethylene glycol, TEG; Withanone, Wi-N, and Withaferin A, Wi-A) on the stemness and differentiation characteristics of cancer cells. Breast carcinoma, glioblastoma, and neuroblastoma cells were treated with a non-toxic concentration of TEG (0.1%), Wi-N (5 µM), and Wi-A (0.1 µM) in 2D and 3D cultures. The results demonstrated that TEG, Wi-N, and Wi-A suppressed the stemness properties, which was linked with their inhibition of epithelial-mesenchymal transition (EMT) signaling. In particular, Wi-N and TEG caused a stronger reduction in the self-renewal capability of CSCs than Wi-A, as evidenced by a tumor spheroid formation assay and analyses of stemness-related genes (ALDH1, CD44, NANOG, CD133, SOX2). Furthermore, TEG and Wi-N caused the differentiation of cancer cells. Each of these was supported by (i) the upregulation of KRT18, KRT19, E-cadherin, and downregulation of vimentin in breast carcinoma; (ii) increased levels of GFAP, MAP2, and PSD-95 in astrocytoma; and (iii) increased NeuN, GAP-43, and NF200 levels in neuroblastoma. Furthermore, a reduction in cancer progression-related proteins (PI3K, N-myc) was recorded in treated cells. Our results suggest that TEG and Wi-N may be recruited to target cancer cell stemness and differentiation therapy.
Collapse
Affiliation(s)
- Huayue Zhang
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki 305-8575, Japan; (H.Z.); (T.Y.); (Z.Z.)
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Central 4-1, Tsukuba 305-8565, Japan; (H.K.); (S.C.K.)
| | - Hyonchol Kim
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Central 4-1, Tsukuba 305-8565, Japan; (H.K.); (S.C.K.)
| | - Tian Yuan
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki 305-8575, Japan; (H.Z.); (T.Y.); (Z.Z.)
| | - Zhenya Zhang
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki 305-8575, Japan; (H.Z.); (T.Y.); (Z.Z.)
| | - Sunil C. Kaul
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Central 4-1, Tsukuba 305-8565, Japan; (H.K.); (S.C.K.)
| | - Renu Wadhwa
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Central 4-1, Tsukuba 305-8565, Japan; (H.K.); (S.C.K.)
| |
Collapse
|
18
|
Menon NA, Kumar CD, Ramachandran P, Blaize B, Gautam M, Cordani M, Lekha Dinesh Kumar. Small-molecule inhibitors of WNT signalling in cancer therapy and their links to autophagy and apoptosis. Eur J Pharmacol 2025; 986:177137. [PMID: 39551337 DOI: 10.1016/j.ejphar.2024.177137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Cancer represents an intricate and heterogeneous ailment that evolves from a multitude of epigenetic and genetic variations that disrupt normal cellular function. The WNT/β-catenin pathway is essential in maintaining the balance between cell renewal and differentiation in various tissues. Abnormal activation of this pathway can lead to uncontrolled cell growth and initiate cancer across a variety of tissues such as the colon, skin, liver, and ovary. It enhances characteristics that lead to cancer progression, including angiogenesis, invasion and metastasis. Processes like autophagy and apoptosis which regulate cell death and play a crucial role in maintaining cellular equilibrium are also intimately linked with WNT/ β-catenin pathway. Thus, targeting WNT pathway has become a key strategy in developing antitumor therapies. Employing small molecule inhibitors has emerged as a targeted therapy to improve the clinical outcome compared to conventional cancer treatments. Many strategies using small molecule inhibitors for modulating the WNT/β-catenin pathway, such as hindering WNT ligands' secretion or interaction, disrupting receptor complex, and blocking the nuclear translocation of β-catenin have been investigated. These interventions have shown promise in both preclinical and clinical settings. This review provides a comprehensive understanding of the role of WNT/β-catenin signalling pathway's role in cancer, emphasizing its regulation of autophagy and apoptosis. Our goal is to highlight the potential of specific small molecule inhibitors targeting this pathway, fostering the development of novel, tailored cancer treatments.
Collapse
Affiliation(s)
- Nayana A Menon
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad, 500007, Telangana, India
| | - Chethana D Kumar
- Department of Surgical ICU, Christian Medical College, IDA Scudder Road, Vellore, 632004, Tamil Nadu, India
| | - Pournami Ramachandran
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad, 500007, Telangana, India
| | - Britny Blaize
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad, 500007, Telangana, India
| | - Mridul Gautam
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad, 500007, Telangana, India
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040, Madrid, Spain.
| | - Lekha Dinesh Kumar
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad, 500007, Telangana, India.
| |
Collapse
|
19
|
Shankari G, Raji R, Prabhu D, Jeyakanthan J, Gopinath SCB. Progressive Dynamics of Cancer Stem Cells in Oral Squamous Cell Carcinoma. Curr Cancer Drug Targets 2025; 25:113-117. [PMID: 39279115 DOI: 10.2174/0115680096340994240906111400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 09/18/2024]
Affiliation(s)
- Gopalakrishnan Shankari
- Center for Bioinformatics, Karpagam Academy of Higher Education, Karpagam, India
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, 641021, India
| | - Rajmichael Raji
- Structural Biology and Biocomputing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, 630003, India
| | - Dhamodharan Prabhu
- Center for Bioinformatics, Karpagam Academy of Higher Education, Karpagam, India
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, 641021, India
| | - Jeyaraman Jeyakanthan
- Structural Biology and Biocomputing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, 630003, India
| | - Subash C B Gopinath
- Center for Global Health Research, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, 602105, Tamil Nadu, India
- Faculty of Chemical Engineering & Technology and Institute of Nano Electronic Engineering, 02600 Arau, Perlis, Malaysia
- Department of Technical Sciences, Western Caspian University, Baku, AZ 1075, Azerbaijan
| |
Collapse
|
20
|
Yi K, Chen W, Zhou X, Xie C, Zhong C, Zhu J. Bisphenol S exposure promotes stemness of triple-negative breast cancer cells via regulating Gli1-mediated Sonic hedgehog pathway. ENVIRONMENTAL RESEARCH 2025; 264:120293. [PMID: 39505130 DOI: 10.1016/j.envres.2024.120293] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/09/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
Bisphenol S (BPS), one of the most common alternatives for bisphenol A (BPA), has been implied to increase the risk of breast cancer. Triple-negative breast cancer (TNBC) is a highly aggressive type of breast cancer with a poor prognosis. However, the association between BPS and TNBC remains unclear. Cancer stem cells (CSCs) have a crucial role in breast cancer initiation, metastasis, and recurrence. Here, we proposed that BPS, equivalent to the human internal exposure and the environmental concentrations, enhanced CSC-like properties by upregulating sphere formation, self-renewal, the percentage of CD44+/CD24- cells, and the expression of CSC markers. Moreover, BPS promoted the migration, invasion, and epithelial-mesenchymal transition (EMT) in TNBC cells. Mechanistically, BPS activated the Sonic Hedgehog (SHH) signaling pathway in TNBC cells. Molecular docking analysis further showed that BPS upregulated SHH signaling pathway via directly binding Gli1 protein. Furthermore, inhibitor of SHH pathway or Gli1 siRNA attenuated the promoting effects of BPS on stemness, invasion, and migration of TNBC cells. In summary, our data firstly provide evidence that environmentally relevant BPS concentration treatment significantly enhanced TNBC malignant phenotype by activating the Sonic Hedgehog/Gli1 signaling pathway, raising high concerns about the potential population biology hazards of BPS.
Collapse
Affiliation(s)
- Kefan Yi
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiyi Chen
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xu Zhou
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunfeng Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jianyun Zhu
- Department of Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China.
| |
Collapse
|
21
|
Su G, Wang J, Liu S, Fu X, Li Y, Pan G. Identification and Validation of Epithelial Cell Centre Regulatory Transcription Factors in the Gastric Cancer Microenvironment. Int J Gen Med 2024; 17:6567-6584. [PMID: 39759895 PMCID: PMC11697670 DOI: 10.2147/ijgm.s496006] [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: 10/12/2024] [Accepted: 12/13/2024] [Indexed: 01/07/2025] Open
Abstract
Purpose To identify the epithelial cell centre regulatory transcription factors in the gastric cancer (GC) microenvironment and provide a new strategy for the diagnosis and treatment of GC. Methods The GC single-cell dataset was downloaded from the Gene Expression Omnibus (GEO) database. The regulatory mechanisms of transcription factors in both pan-cancer and GC microenvironments were analysed using the Cancer Genome Atlas (TGCA) database. Real-time quantitative PCR (RT-qPCR) was used to determine the mRNA expression levels of Prospero homeobox gene 1 (PROX1) and Endothelial PAS domain-containing protein 1 (EPAS1) in the human gastric mucosal normal epithelial cell line (GES-1) and the GC cell line (AGS). Immunohistochemistry (IHC) was used to determine the amounts of PROX1 and EPAS1 protein expression in GC and adjacent tissues. GC patients' overall survival (OS) was tracked through outpatient, Inpatient case inquiry, or phone follow-up. Results The single-cell data from GSE184198 was re-annotated, resulting in nine cell subsets: T cells (13364), NK cells (606), B cells (2525), Epithelial cells (2497), DC cells (1167), Fibroblast cells (372), Endothelial cells (271), Neutrophils cells (246) and Macrophage cells (420). Analysis of cell subgroup signalling pathways revealed that communication intensity between epithelial cells and smooth muscle cells was highest. Transcription factors PROX1 and EPAS1 were notably active in epithelial cells. Cell communication analysis indicated that IFNG may interact with IFNGR1/2 and LIF with IL6ST and LIFR to regulate the downstream PROX1 and EPAS1. PROX1 and EPAS1 were upregulated and negatively correlated with tumour mutation burden (TMB). They also exhibited high positive correlations with immune checkpoints CTLA4 and PDCD1LG2, as well as with chemokines CCL24 and CXCL12 and their receptors CCR3 and CCR4. Additionally, PROX1 and EPAS1 were positively correlated with immunosuppressive factors ADORA2A, CD160, IL10, TGFBR1, KDR and CSF1R, as well as with immunostimulators CD276, PVR, TNFRSF25, ULBP1, CXCL12 and ENTPD1. In GC tissues and AGS, PROX1 and EPAS1 were both substantially expressed. In the meantime, they showed a positive correlation with clinicopathological features such TNM stage and degree of differentiation. In GC patients, the up-regulated group's PROX1 and EPAS1 prognosis was noticeably poorer than the down-regulated group's. Conclusion PROX1 and EPAS1 are likely central regulatory transcription factors in the epithelial cells of the GC environment, regulated by IFNG and LIF. They may contribute to GC progression by modulating the tumour's immune microenvironment.
Collapse
Affiliation(s)
- Guomiao Su
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yun Nan, People’s Republic of China
| | - Juan Wang
- Clinical Laboratory, Yunnan Province Third People’s Hospital, Kunming, Yun Nan, People’s Republic of China
| | - Shiyue Liu
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yun Nan, People’s Republic of China
| | - Xiaonan Fu
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yun Nan, People’s Republic of China
| | - Yanxi Li
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yun Nan, People’s Republic of China
| | - Guoqing Pan
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yun Nan, People’s Republic of China
| |
Collapse
|
22
|
Li J, Ma Y, Wu Q, Ping P, Li J, Xu X. The potential role of HPV oncoproteins in the PD-L1/PD-1 pathway in cervical cancer: new perspectives on cervical cancer immunotherapy. Front Oncol 2024; 14:1488730. [PMID: 39735605 PMCID: PMC11671370 DOI: 10.3389/fonc.2024.1488730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/28/2024] [Indexed: 12/31/2024] Open
Abstract
Cervical cancer (CC) is a common malignant tumour of the female reproductive system that is highly harmful to women's health. The efficacy of traditional surgery, radiotherapy and chemotherapy is limited, especially for recurrent and metastatic CC. With continuous progress in diagnostic and treatment technology, immunotherapy has become a new approach for treating CC and has become a new therapy for recurrent and metastatic CC. However, immunotherapy is not effective for all patients with CC. Therefore, factors related to immunotherapy efficacy in CC patients have become the focus of researchers. High-risk human papillomavirus (HPV) infection is an important factor that drives CC development and affects its progression and prognosis. Increasing attention has been given to the mechanism of the E5, E6 and E7 proteins, which are encoded by the HPV gene, in the occurrence and development of CC and their interaction with programmed cell death ligand-1/programmed cell death-1 (PD-L1/PD-1). Although some preliminary studies have been conducted on these topics, a comprehensive and systematic review of these topics is not available. This review comprehensively summarizes related articles from journals with impact factors greater than 3 and published in the past 5 years; it also reviews studies on the mechanism of HPV and CC, the mechanism of PD-L1/PD-1 axis regulation in CC, and the mechanism by which the interaction between HPV-related oncoproteins and the PD-L1/PD-1 pathway affects the development and prognosis of CC. This study provides theoretical support for the use of immunotherapies for CC, provides a basis for the selection of specific medications that target different HPV-related proteins, and provides a new perspective for the discovery of new immunotherapy targets for CC.
Collapse
Affiliation(s)
| | | | | | | | - Juan Li
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Dalian Medical
University, Dalian, China
| | - Xiaoying Xu
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Dalian Medical
University, Dalian, China
| |
Collapse
|
23
|
Li L, Huang W, Ren X, Wang Z, Ding K, Zhao L, Zhang J. Unlocking the potential: advancements and future horizons in ROR1-targeted cancer therapies. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2603-2616. [PMID: 39145866 DOI: 10.1007/s11427-024-2685-9] [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: 02/02/2024] [Accepted: 07/11/2024] [Indexed: 08/16/2024]
Abstract
While receptor tyrosine kinase-like orphan receptor 1 (ROR1) is typically expressed at low levels or absent in normal tissues, its expression is notably elevated in various malignant tumors and conditions, including chronic lymphocytic leukemia (CLL), breast cancer, ovarian cancer, melanoma, and lung adenocarcinoma. This distinctive feature positions ROR1 as an attractive target for tumor-specific treatments. Currently, several targeted drugs directed at ROR1 are undergoing clinical development, including monoclonal antibodies, antibody-drug conjugates (ADCs), and chimeric antigen receptor T-cell therapy (CAR-T). Additionally, there are four small molecule inhibitors designed to bind to ROR1, presenting promising avenues for the development of PROTAC degraders targeting ROR1. This review offers updated insights into ROR1's structural and functional characteristics, embryonic development implications, cell survival signaling pathways, and evolutionary targeting strategies, all of which have the potential to advance the treatment of malignant tumors.
Collapse
Affiliation(s)
- Lin Li
- State Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Weixue Huang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Xiaomei Ren
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
| | - Linxiang Zhao
- State Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Jinwei Zhang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
| |
Collapse
|
24
|
Liu S, Liu Y, Zhou Y, Xia G, Liu H, Zeng Y, Pei Z, Cao J, Jing G, Zou H, Liao C. NSUN5 promotes tumorigenic phenotypes through the WNT signaling pathway and immunosuppression of CD8+ T cells in gastric cancer. Cell Signal 2024; 124:111475. [PMID: 39428025 DOI: 10.1016/j.cellsig.2024.111475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/07/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
NSUN5, a key member of the M5C methylation family, plays a significant role in fundamental biological processes like cell proliferation and differentiation. However, its specific function and mechanisms in gastric cancer remain insufficiently understood. Initially, we examined NSUN5's differential expression in gastric cancer versus normal tissues, along with survival trends, associated signaling pathways, and immune infiltration using the TCGA database. Subsequently, we conducted immunohistochemistry experiments to assess NSUN5 expression in gastric cancer tissues. Gain-and loss-of-function experiments were carried out to investigate NSUN5's impact on the proliferation, stemness, and migratory capabilities of gastric cancer cells, as well as the expression of vital proteins in pertinent signaling pathways. Our findings demonstrate that NSUN5 is not only overexpressed in gastric cancer tissues, but also positively associated with tumor stage and inversely linked with patient prognosis. NSUN5 promotes the in vitro proliferation, stemness, and migration of gastric cancer cells, and the in vivo growth of these cells, chiefly through the activation of the WNT/β-catenin signaling pathway. Additionally, NSUN5 appears to diminish the infiltration of CD8+ T cells in gastric cancer, contributing to immune evasion. In conclusion, NSUN5 functions as a proto-oncogene in the progression of gastric cancer and may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Shuhao Liu
- Gastrointestinal Hernia Surgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No.152 Aiguo Road, Nanchang City 330006, Jiangxi Province, PR China
| | - Yong Liu
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, PR China.; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, PR China
| | - Yijun Zhou
- School of Medicine, Sun Yat-sen University (Shenzhen), Shenzhen 518107, China
| | - Gaoshui Xia
- Nanchang Medical College. No. 689, Huiren Avenue, Nanchang Xiaolan Economic And Technological Development Zone, Nanchang City 330052, Jiangxi Province, PR China
| | - Haibo Liu
- Gastrointestinal Hernia Surgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No.152 Aiguo Road, Nanchang City 330006, Jiangxi Province, PR China
| | - Yu Zeng
- Gastrointestinal Hernia Surgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No.152 Aiguo Road, Nanchang City 330006, Jiangxi Province, PR China; Jiangxi Medical College, Nanchang University, No. 461, Bayi Avenue, Nanchang City 330006, Jiangxi Province, PR China
| | - Zhihui Pei
- Gastrointestinal Hernia Surgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No.152 Aiguo Road, Nanchang City 330006, Jiangxi Province, PR China; Jiangxi Medical College, Nanchang University, No. 461, Bayi Avenue, Nanchang City 330006, Jiangxi Province, PR China
| | - Jing Cao
- Gastrointestinal Hernia Surgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No.152 Aiguo Road, Nanchang City 330006, Jiangxi Province, PR China
| | - Guifang Jing
- Gastrointestinal Hernia Surgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No.152 Aiguo Road, Nanchang City 330006, Jiangxi Province, PR China
| | - Hailin Zou
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, PR China.
| | - Chuanwen Liao
- Gastrointestinal Hernia Surgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No.152 Aiguo Road, Nanchang City 330006, Jiangxi Province, PR China
| |
Collapse
|
25
|
Yang T, Dong Y, Wang G, Guan X. SOX13-mediated transcription of LRP11 enhances malignant properties of tumor cells and CD8 + T cell inactivation in breast cancer through the β-catenin/PD-L1 axis. Cell Signal 2024; 124:111383. [PMID: 39243917 DOI: 10.1016/j.cellsig.2024.111383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/22/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND High expression of low-density lipoprotein receptor related protein 11 (LRP11) has been associated with unfavorable prognosis of breast cancer (BC). This study explores the exact roles of LRP11 in BC progression and investigates the associated mechanism. METHODS LRP11 expression in BC tissues and cells was determined by immunohistochemistry or RT-qPCR. LRP11 upregulation was induced in two human BC cell lines to investigate its impact on cell proliferation, migration, and invasion. Its regulation on immune activity was assessed by detecting PD-L1 protein levels and generating a co-culture system of cancer cells and CD8+ T cells. Mouse allograft tumor models were generated to analyze the function of LRP11 in tumorigenesis and immune activity in vivo. Gain-of-function assays of SRY-box transcription factor 13 (SOX13) were performed to investigate its function in development and immunosuppression of BC. RESULTS LRP11 was found to be highly expressed in BC tissues and cells, presenting an association with unfavorable prognosis of patients. Artificial upregulation of LRP11 in BC cells triggered malignant properties of cells, enhancing β-catenin-mediated transcriptional activation of PD-L1, thus decreasing immune activity of the co-cultured CD8+ T cells. Consistently, LRP11 upregulation in mouse 4 T1 cells and promoted tumorigenesis and immune evasion in mice. SOX13 was found to bind the LRP11 promoter for transcriptional activation. Upregulation of SOX13 similarly promoted growth of BC cells and immunosuppression, with its oncogenic effects blocked by the additional LRP11 knockdown. CONCLUSION This study demonstrates that SOX13 is responsible for LRP11 transcription activation, leading to increased malignant phenotype of BC cells and diminished activity CD8+ T cells. This evidence highlights SOX13 and LRP11 as promising novel therapeutic targets to reduce malignant phenotype of BC cells and overcome immunosuppression.
Collapse
Affiliation(s)
- Tingting Yang
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, Jilin, PR China
| | - Yi Dong
- The Second Breast Surgery Department, Jilin Cancer Hospital, Changchun 130012, Jilin, PR China
| | - Guoxiang Wang
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, Jilin, PR China
| | - Xin Guan
- Breast Surgery Department, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin, PR China.
| |
Collapse
|
26
|
Czarnogórski MC, Czernicka A, Koper K, Petrasz P, Pokrywczyńska M, Juszczak K, Kowalski F, Drewa T, Adamowicz J. Cancer stem cells and their role in metastasis. Cent European J Urol 2024; 78:40-51. [PMID: 40371432 PMCID: PMC12073518 DOI: 10.5173/ceju.2024.0144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/30/2024] [Indexed: 05/16/2025] Open
Abstract
Introduction Cancer, next to cardiovascular diseases, remains the primary concern of modern medicine in developed countries. Despite the unprecedented progress in targeted therapies and personalised medicine, including immunotherapy and gene therapy, we are still unable to efficiently treat many malignancies. One of the major obstacles to treating cancer is its ability to metastasise. Hence, a better understanding of cancer biology with emphasis on the metastasis formation may hold the key to further ameliorating cancer treatment. Nowadays, there is a growing body of evidence for the common denominator of neoplasia, which seems to be universal - cancer stem cells which are being found in a growing number of cancers. Material and methods We conducted a Web of Science and Medline database search using the terms "cancer stem cells", "carcinogenesis", and "stem cells" in conjunction with "metastasis", without setting time limits. Results The existence of cancer stem cells was proven both in animal models and in humans. We know beyond doubt that cancer stem cells may be found in bladder cancer, breast cancer, and colon cancer, among others. The cancer stem cells in the aforementioned cancers may initiate tumour formation ex vivo and thus theoretically lead to tumour recurrence. Their role in the formation of metastases, however, is still under investigation. Conclusions Although their exact role is yet to be identified, it is now obvious that cancer stem cells give rise to primary mass in solid tumours and differentiated cancer cells in leukaemias. However, the role of cancer stem cells in metastasis is still obscure.
Collapse
Affiliation(s)
- Michał C. Czarnogórski
- Department and Chair of Urology and Andrology, Ludwik Rydygier’s Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Aleksandra Czernicka
- Department and Chair of Urology and Andrology, Ludwik Rydygier’s Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Krzysztof Koper
- Department of Oncology, Ludwik Rydygier’s Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Piotr Petrasz
- Department of Urology and Urological Oncology, Multidisciplinary Regional Hospital in Gorzow Wielkopolski, Poland
| | - Marta Pokrywczyńska
- Department of Regenerative Medicine, Chair of Urology, Ludwik Rydygier’s Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Kajetan Juszczak
- Department and Chair of Urology and Andrology, Ludwik Rydygier’s Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Filip Kowalski
- Department and Chair of Urology and Andrology, Ludwik Rydygier’s Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Tomasz Drewa
- Department and Chair of Urology and Andrology, Ludwik Rydygier’s Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Jan Adamowicz
- Department and Chair of Urology and Andrology, Ludwik Rydygier’s Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| |
Collapse
|
27
|
Schöpe PC, Torke S, Kobelt D, Kortüm B, Treese C, Dumbani M, Güllü N, Walther W, Stein U. MACC1 revisited - an in-depth review of a master of metastasis. Biomark Res 2024; 12:146. [PMID: 39580452 PMCID: PMC11585957 DOI: 10.1186/s40364-024-00689-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024] Open
Abstract
Cancer metastasis remains the most lethal characteristic of tumors mediating the majority of cancer-related deaths. Identifying key molecules responsible for metastasis, understanding their biological functions and therapeutically targeting these molecules is therefore of tremendous value. Metastasis Associated in Colon Cancer 1 (MACC1), a gene first described in 2009, is such a key driver of metastatic processes, initiating cellular proliferation, migration, invasion, and metastasis in vitro and in vivo. Since its discovery, the value of MACC1 as a prognostic biomarker has been confirmed in over 20 cancer entities. Additionally, several therapeutic strategies targeting MACC1 and its pro-metastatic functions have been developed. In this review, we will provide a comprehensive overview on MACC1, from its clinical relevance, towards its structure and role in signaling cascades as well as molecular networks. We will highlight specific biological consequences of MACC1 expression, such as an increase in stem cell properties, its immune-modulatory effects and induced therapy resistance. Lastly, we will explore various strategies interfering with MACC1 expression and/or its functions. Conclusively, this review underlines the importance of understanding the role of individual molecules in mediating metastasis.
Collapse
Affiliation(s)
- Paul Curtis Schöpe
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Sebastian Torke
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Dennis Kobelt
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Benedikt Kortüm
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Christoph Treese
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Malti Dumbani
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Nazli Güllü
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Wolfgang Walther
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
- German Cancer Consortium (DKTK), Berlin and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
28
|
Bian Z, Chen B, Shi G, Yuan H, Zhou Y, Jiang B, Li L, Su H, Zhang Y. Single-cell landscape identified SERPINB9 as a key player contributing to stemness and metastasis in non-seminomas. Cell Death Dis 2024; 15:812. [PMID: 39528470 PMCID: PMC11555415 DOI: 10.1038/s41419-024-07220-5] [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: 05/29/2024] [Revised: 10/27/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Embryonal carcinoma (EC), characterized by a high degree of stemness similar to that of embryonic stem cells, is the most malignant subtype within non-seminomatous testicular germ cell tumors (TGCTs). However, the mechanisms underlying its malignancy remain unknown. In this study, we employed single-cell RNA sequencing to analyze four non-seminoma samples. Our differential expression analysis revealed high expression of SERPINB9 in metastatic EC cells. We conducted in vitro experiments to further investigate SERPINB9's role in the progression of EC. Functionally, the knockdown of SERPINB9 in NCCIT and NTERA-2 leads to a diminished migratory capability and decreased cis-platin resistance, as demonstrated by Transwell migration assay and drug sensitivity assay. Moreover, embryoid bodies showed reduced size and lower OCT4 expression, alongside heightened expression of differentiation markers AFP, ACTA2, and CD57 in shSERPINB9 cells. In vivo, the role of SERPINB9 in maintaining cancer stemness was validated by the limiting dilution assay. Mechanistically, Bulk RNA-seq further showed downregulation of ERK1/2 signaling and WNT signaling pathways with concomitant upregulation of differentiation pathways subsequent to SERPINB9 knockdown. Additionally, the analysis indicated increased levels of cytokines linked to tertiary lymphoid structures (TLS), such as IL6, IL11, IL15, CCL2, CCL5, and CXCL13 in shSERPINB9 cells, which were further validated by ELISA. Our research indicates that SERPINB9 plays a key role in driving tumor progression by enhancing tumor stemness and suppressing TLS. This study stands as the first to elucidate the molecular signature of non-seminomas at a single-cell level, presenting a wealth of promising targets with substantial potential for informing the development of future therapeutic interventions.
Collapse
Affiliation(s)
- Zhouliang Bian
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, PR China
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, PR China
| | - Biying Chen
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, PR China
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, PR China
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Haihua Yuan
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, PR China
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, PR China
| | - Yue Zhou
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Bin Jiang
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, PR China.
| | - Long Li
- Department of Urology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.
| | - Hengchuan Su
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China.
| | - Yanjie Zhang
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, PR China.
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, PR China.
| |
Collapse
|
29
|
Katoh M, Katoh M. Claudin 1, 4, 6 and 18 isoform 2 as targets for the treatment of cancer (Review). Int J Mol Med 2024; 54:100. [PMID: 39301632 DOI: 10.3892/ijmm.2024.5424] [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: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
The 24 claudin (CLDN) genes in the human genome encode 26 representative CLDN family proteins. CLDNs are tetraspan‑transmembrane proteins at tight junctions. Because several CLDN isoforms, such as CLDN6 and CLDN18.2, are specifically upregulated in human cancer, CLDN‑targeting monoclonal antibodies (mAbs), antibody‑drug conjugates (ADCs), bispecific antibodies (bsAbs) and chimeric antigen receptor (CAR) T cells have been developed. In the present review, CLDN1‑, 4‑, 6‑ and 18.2‑targeting investigational drugs in clinical trials are discussed. CLDN18.2‑directed therapy for patients with gastric and other types of cancer is the most advanced area in this field. The mouse/human chimeric anti‑CLDN18.2 mAb zolbetuximab has a single‑agent objective response rate (ORR) of 9%, and increases progression‑free and overall survival in combination with chemotherapy. The human/humanized anti‑CLDN18.2 mAb osemitamab, and ADCs AZD0901, IBI343 and LM‑302, with single‑agent ORRs of 28‑60%, have been tested in phase III clinical trials. In addition, bsAbs, CAR T cells and their derivatives targeting CLDN4, 6 or 18.2 are in phase I and/or II clinical trials. AZD0901, IBI343, zolbetuximab and the anti‑CLDN1 mAb ALE.C04 have been granted fast track designation or priority review designation by the US Food and Drug Administration.
Collapse
Affiliation(s)
- Masuko Katoh
- Department of Global Network, M & M Precision Medicine, Tokyo 113‑0033, Japan
| | - Masaru Katoh
- Department of Global Network, M & M Precision Medicine, Tokyo 113‑0033, Japan
| |
Collapse
|
30
|
Zhang Y, Ren J, Liao Z, Li X, Zhang C, Huang B, Cao Y, Chen J. Downregulating LKB1 in bone marrow mesenchymal stem cells could inhibit CD4 + T cell proliferation via the PD-1/PD-L1 signaling pathway. Immunobiology 2024; 229:152856. [PMID: 39369651 DOI: 10.1016/j.imbio.2024.152856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Our previous research has shown that LKB1 in amniotic mesenchymal stem cells (MSCs) serves as a vital regulator of regulatory T cell differentiation and T cell proliferation, which may have a similar role in bone marrow MSCs (BMMSCs). Therefore, we investigated the role of LKB1 in BMMSCs for regulating CD4+ T cell proliferation in the bone micro-environment of AML. METHODS RT-PCR was used to assessed LKB1 expression in BMMSCs derived from AML patients and healthy controls. Subsequently, LKB1 was knocked down in the BMMSCs line HS-5 (HS-5-LKB1KD). Co-cultures in vitro were established to analyze the effect of HS-5-LKB1KD on CD4+ T cell. Flow cytometry was employed to measure PD-L1 and CD4+ T cell proliferation levels. Western blot was utilized to detect related proteins. RESULTS The expression of LKB1 in BMMSCs derived from AML patients was decreased. Knockdown of LKB1 in HS-5 resulted in upregulation of PD-L1 expression. Co-culture of peripheral blood CD4+ T cell with HS-5-LKB1KD exhibited reduced CD4+ T cell proliferation compared to co-culture with HS-5-LKB1con. Furthermore, blocking PD-L1 in the co-culture conditions could restore the reduced CD4+ T cell proliferation. Additionally, it was found that upregulation of the Wnt signaling pathway-related proteins following LKB1 knockdown in HS-5, indicating that downregulating LKB1 could promote PD-L1 expression through activation of the Wnt signaling pathway. CONCLUSIONS The decreased expression of LKB1 in BMMSCs may activate the Wnt signaling pathway, leading to increased PD-L1 expression. This inhibited CD4+ T cell proliferation, which might lead to impaired anti-tumor immunity in AML patients and promote AML progression.
Collapse
Affiliation(s)
- Yaqin Zhang
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Jingyi Ren
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Zhongxian Liao
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Xiaoyu Li
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Chunying Zhang
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Bihan Huang
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Yingping Cao
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China.
| | - Jiadi Chen
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China.
| |
Collapse
|
31
|
Hou Y, Zhao Z, Li P, Cao Y, Zhang Y, Guo C, Nie X, Hou J. Combination therapies with Wnt signaling inhibition: A better choice for prostate cancer treatment. Biochim Biophys Acta Rev Cancer 2024; 1879:189186. [PMID: 39332651 DOI: 10.1016/j.bbcan.2024.189186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024]
Abstract
The intractability and high mortality rate of castration-resistant prostate cancer (CRPC) remain the most challenging problems in the field of prostate cancer (PCa). Emerging evidence has shown that the dysregulation of Wnt signaling pathways, which are highly conserved cascades that regulate embryonic development and maintain tissue homeostasis, is involved in various stages of PCa occurrence and progression. In this review, we systemically discuss the mechanisms by which the androgen receptor (AR) signaling pathway and Wnt signaling pathways participate in the occurrence of PCa and its progression to CRPC. Specifically, we elaborate on how Wnt signaling pathways induce the malignant transformation of prostate cells, promote the malignant progression of PCa and establish an immunosuppressive prostate tumor microenvironment through interaction with the AR pathway or in an AR-independent manner. We also discuss how Wnt signaling pathways enhances the stemness characteristics of prostate cancer stem cells (PCSCs) to induce the occurrence and metastasis of CPPC. Additionally, we discuss the latest progress in the use of different types of drugs that inhibit the Wnt signaling pathways in the treatment of PCa. We believe that the combination of Wnt signaling-based drugs with endocrine and other therapies is necessary and may enhance the clinical efficacy in the treatment of all types of PCa.
Collapse
Affiliation(s)
- Yifan Hou
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng 475003, China
| | - Zhenhua Zhao
- Ma'anshan 86 Hospital, China RongTong Medical Healthcare Group Co. Ltd, Ma'anshan 243100, China
| | - Pan Li
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Yujia Cao
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Yi Zhang
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Changsheng Guo
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng 475003, China
| | - Xiaobo Nie
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Junqing Hou
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng 475003, China.
| |
Collapse
|
32
|
Winnard PT, Vesuna F, Bol GM, Gabrielson KL, Chenevix-Trench G, Ter Hoeve ND, van Diest PJ, Raman V. Targeting RNA helicase DDX3X with a small molecule inhibitor for breast cancer bone metastasis treatment. Cancer Lett 2024; 604:217260. [PMID: 39306228 DOI: 10.1016/j.canlet.2024.217260] [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: 07/01/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 10/03/2024]
Abstract
Patients who present with breast cancer bone metastasis only have limited palliative treatment strategies and efficacious drug treatments are needed. In breast cancer patient data, high levels of the RNA helicase DDX3 are associated with poor overall survival and bone metastasis. Consequently, our objective was to target DDX3 in a mouse breast cancer bone metastasis model using a small molecule inhibitor of DDX3, RK-33. Histologically confirmed live imaging indicated no bone metastases in the RK-33 treated cohort, as opposed to placebo-treated mice. We generated a cell line from a bone metastatic lesion in mouse and found that it along with a patient-derived bone metastasis cell line gained resistance to conventional chemotherapeutics but not to RK-33. Finally, differential levels of DDX3 were observed in breast cancer patient metastatic bone samples. Overall, this study indicates that DDX3 is a relevant clinical target in breast cancer bone metastasis and that RK-33 can be a safe and effective treatment for these patients.
Collapse
Affiliation(s)
- Paul T Winnard
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Farhad Vesuna
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Guus M Bol
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Medical Oncology, University Medical Center Utrecht Cancer Center, GA, Utrecht, the Netherlands
| | - Kathleen L Gabrielson
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Georgia Chenevix-Trench
- QIMR Berghofer Medical Research Institute, Royal Brisbane Hospital, Locked Bag 2000, Brisbane, QLD, 4029, Australia
| | - Natalie D Ter Hoeve
- Department of Pathology, University Medical Center Utrecht Cancer Center, 3508 GA, Utrecht, the Netherlands
| | - Paul J van Diest
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Pathology, University Medical Center Utrecht Cancer Center, 3508 GA, Utrecht, the Netherlands
| | - Venu Raman
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Pathology, University Medical Center Utrecht Cancer Center, 3508 GA, Utrecht, the Netherlands; Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
33
|
Tuoheti M, Li J, Zhang C, Gao F, Wang J, Wu Y. MiR-124-3p inhibits cell stemness in glioblastoma via targeting EPHA2 through ALKBH5-mediated m6A modification. Hum Cell 2024; 38:10. [PMID: 39460871 DOI: 10.1007/s13577-024-01129-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
Glioblastoma (GBM) is the most aggressive form of glioma, characterized by high mortality and poor prognosis. Dysregulation of microRNAs (miRNAs) plays a critical role in the progression and metastasis of GBM. This study aimed to investigate the role and molecular mechanism of miR-124-3p in GBM. Levels of miR-124-3p, EPHA2, and ALKBH5 were measured using quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation, migration, invasion, and stemness were assessed using the Cell Counting Kit-8 (CCK-8), colony formation, Transwell, and sphere formation assays, respectively. Bioinformatics prediction, dual-luciferase reporter assays, and RNA pull-down experiments were employed to validate the target of miR-124-3p. RNA binding protein immunoprecipitation (RIP) and methylated RNA immunoprecipitation (Me-RIP) were utilized to evaluate the regulation of miR-124-3p maturation by ALKBH5. The results indicated that overexpression of miR-124-3p inhibited the proliferation, migration, invasion, and stemness of GBM cells. EPHA2 was identified as a direct downstream target of miR-124-3p, and its overexpression reversed the inhibitory effects of miR-124-3p on cellular functions. Furthermore, miR-124-3p targeted EPHA2 to inactivate the Wnt/β-catenin pathway. Additionally, ALKBH5 negatively regulated miR-124-3p by impeding its processing. In conclusion, knockdown of ALKBH5 promoted the processing of pri-miR-124-3p, increasing mature miR-124-3p levels, which inhibited the malignant behaviors of GBM cells by targeting EPHA2. These findings highlight the importance of the ALKBH5/miR-124-3p/EPHA2 axis in GBM.
Collapse
Affiliation(s)
- Maimaitiyiming Tuoheti
- Department of Neurosurgery, The People's Hospital of Xinjiang Uygur Autonomous Region, #91 Tianchi Road, Tianshan District, Urumqi, 830001, Xinjiang, China
| | - Jinxian Li
- Department of Rehabilitative Medicine, The People's Hospital of Xinjiang Uygur Autonomous Region, #91 Tianchi Road, Tianshan District, Urumqi, 830001, Xinjiang, China
| | - Cheng Zhang
- Department of Neurosurgery, The People's Hospital of Xinjiang Uygur Autonomous Region, #91 Tianchi Road, Tianshan District, Urumqi, 830001, Xinjiang, China
| | - Feng Gao
- Department of Neurosurgery, The People's Hospital of Xinjiang Uygur Autonomous Region, #91 Tianchi Road, Tianshan District, Urumqi, 830001, Xinjiang, China
| | - Jichao Wang
- Department of Neurosurgery, The People's Hospital of Xinjiang Uygur Autonomous Region, #91 Tianchi Road, Tianshan District, Urumqi, 830001, Xinjiang, China
| | - Yonggang Wu
- Department of Neurosurgery, The People's Hospital of Xinjiang Uygur Autonomous Region, #91 Tianchi Road, Tianshan District, Urumqi, 830001, Xinjiang, China.
| |
Collapse
|
34
|
Yang Y, Yan F, Gao Z, Li H, Wen S, Li Q, Li J, Huang N, Zhao W. The NEDD4/FLRT2 axis regulates NSCLC cell stemness. Front Pharmacol 2024; 15:1459978. [PMID: 39444619 PMCID: PMC11496253 DOI: 10.3389/fphar.2024.1459978] [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: 07/05/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION Lung cancer is the leading cause of cancer-related death worldwide. The treatment for lung cancer, particularly for non-small cell lung cancer (NSCLC), remains a clinical challenge. Cancer stem cells are vital for lung cancer development. This study aimed to determine the influence of the neuronally expressed developmentally downregulated 4-fibronectin leucine-rich transmembrane 2 (NEDD4-FLRT2) axis on cancer cell stemness in NSCLC. METHODS FLRT2 expression in NSCLC tissues and stem cells was investigated using western blot and RT-qPCR. The sphere formation assay and the abundance of stemness markers were employed to confirm the stemness of NSCLC stem cells. The CCK-8, colony formation, and Trans-well assays, as well as flow cytometry, were used to determine NSCLC stem cell growth, metastasis, and apoptosis, respectively. The Co-IP assay was used to confirm the binding between NEDD4 and FLRT2. Xenograft tumor mouse models were used to investigate tumorigenesis in vivo. RESULTS Here, we reported that FLRT2 expression was reduced in NSCLC tissues, cells, and NSCLC stem cells. FLRT2 upregulation inhibited NSCLC stem cell proliferation, sphere formation, and drug resistance and promoted drug-resistant cell apoptosis. Furthermore, FLRT2 overexpression demonstrated antitumor effects in a xenograft tumor mouse model. Mechanically, FLRT2 was ubiquitinated and degraded by E3 ligase NEDD4. NEDD4 overexpression significantly abolished the inhibitory effects of FLRT2 on NSCLC stemness, as evidenced by in vitro and in vivo experiments. DISCUSSION This study revealed that FLRT2 acted as a tumor suppressor by inhibiting cancer cell stemness in NSCLC. NEDD4 promoted ubiquitination degradation of FLRT2 protein. NEDD4 counteracted the inhibitory effects of FLRT2 on NSCLC stem cell tumorigenesis.
Collapse
Affiliation(s)
- Yuping Yang
- Department of Respiratory and Critical Care Medicine, School of Clinical Medicine and The First Affiliated Hospital of Chengdu Medical College, School of Clinical Medicine, of Chengdu Medical College, Chengdu, China
- Key Laboratory of Geriatic Respiratory Diseases of Sichuan Higher Education Institutes, School of Clinical Medicine and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Fei Yan
- Department of Medical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Ziwei Gao
- Department of Respiratory and Critical Care Medicine, School of Clinical Medicine and The First Affiliated Hospital of Chengdu Medical College, School of Clinical Medicine, of Chengdu Medical College, Chengdu, China
- Key Laboratory of Geriatic Respiratory Diseases of Sichuan Higher Education Institutes, School of Clinical Medicine and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Houke Li
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Shengke Wen
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Qi Li
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Jiayuan Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Na Huang
- Department of Respiratory and Critical Care Medicine, School of Clinical Medicine and The First Affiliated Hospital of Chengdu Medical College, School of Clinical Medicine, of Chengdu Medical College, Chengdu, China
- Key Laboratory of Geriatic Respiratory Diseases of Sichuan Higher Education Institutes, School of Clinical Medicine and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wei Zhao
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Clinical Laboratory, School of Clinical Medicine and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
35
|
Yin H, Sun L, Yuan Y, Zhu Y. PPIC-labeled CAFs: Key players in neoadjuvant chemotherapy resistance for gastric cancer. Transl Oncol 2024; 48:102080. [PMID: 39116799 PMCID: PMC11362775 DOI: 10.1016/j.tranon.2024.102080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/23/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the fourth leading cause of cancer deaths, with advanced cases having a median survival of less than one year. Neoadjuvant chemotherapy (NCT) is vital but faces drug resistance issues, partly due to cancer-associated fibroblasts (CAFs). Yet, specific CAF subpopulations contributing to resistance are poorly understood. METHODS Differentially expressed genes (DEGs) between chemosensitive and resistant GC patients were identified using GEO2R. Single-cell sequencing (scRNA-seq) identified CAF-related genes. Immunohistochemistry verified key genes in NCT-treated GC samples, analyzing their correlation with tumor regression grade (TRG) and clinicopathological characteristics. RESULTS PPIC as a gene highly expressed in CAFs was closely associated with NCT resistance in gastric cancer. Immunohistochemistry results revealed positivity for the expression of cyclophilin C (CypC), encoded by PPIC, in the 5-fluorouracil and cisplatin NCT resistant and -sensitive groups of gastric cancer patients at rates of 69.7 % (76/109) and 43.6 % (24/55), respectively (p < 0.001). The high expression of CypC in CAFs was positively correlated to tumor size (p = 0.025), T stage (p = 0.004), TNM stage (p = 0.004), and vascular invasion (p = 0.027). In cancer cells the expression of CypC was associated with OS (p = 0.026). However, in CAFs, CypC expression was not related to OS (p = 0.671). CONCLUSIONS PPIC-labeled CAF subgroups are related to NCT resistance and poor prognosis in GC and they may cause drug resistance through signaling pathways such as glucose metabolism and extracellular matrix remodeling. However, the exact mechanism behind the involvement of PPIC-labeled CAF in drug resistance of GC requires further study.
Collapse
Affiliation(s)
- Honghao Yin
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Lili Sun
- Departments of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China; Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China.
| | - Yanmei Zhu
- Departments of Pathology, Affiliated Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University), Shenyang, China.
| |
Collapse
|
36
|
Croft W, Pounds R, Jeevan D, Singh K, Balega J, Sundar S, Williams A, Ganesan R, Kehoe S, Ott S, Zuo J, Yap J, Moss P. The chromatin landscape of high-grade serous ovarian cancer metastasis identifies regulatory drivers in post-chemotherapy residual tumour cells. Commun Biol 2024; 7:1211. [PMID: 39341888 PMCID: PMC11438996 DOI: 10.1038/s42003-024-06909-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Disease recurrence following chemotherapy is a major clinical challenge in ovarian cancer (OC), but little is known regarding how the tumour epigenome regulates transcriptional programs underpinning chemoresistance. We determine the single cell chromatin accessibility landscape of omental OC metastasis from treatment-naïve and neoadjuvant chemotherapy-treated patients and define the chromatin accessibility profiles of epithelial, fibroblast, myeloid and lymphoid cells. Epithelial tumour cells display open chromatin regions enriched with motifs for the oncogenic transcription factors MEIS and PBX. Post chemotherapy microenvironments show profound tumour heterogeneity and selection for cells with accessible chromatin enriched for TP53, TP63, TWIST1 and resistance-pathway-activating transcription factor binding motifs. An OC chemoresistant tumour subpopulation known to be present prior to treatment, and characterised by stress-associated gene expression, is enriched post chemotherapy. Nuclear receptors RORa, NR2F6 and HNF4G are uncovered as candidate transcriptional drivers of these cells whilst closure of binding sites for E2F2 and E2F4 indicate post-treated tumour having low proliferative capacity. Delineation of the gene regulatory landscape of ovarian cancer cells surviving chemotherapy treatment therefore reveals potential core transcriptional regulators of chemoresistance, suggesting novel therapeutic targets for improving clinical outcome.
Collapse
Affiliation(s)
- W Croft
- Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK.
| | - R Pounds
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, UK
| | - D Jeevan
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - K Singh
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, UK
| | - J Balega
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, UK
| | - S Sundar
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, UK
| | - A Williams
- Histopathology Department, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - R Ganesan
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
- Histopathology Department, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - S Kehoe
- Department of Gynaecological Oncology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - S Ott
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - J Zuo
- Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - J Yap
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
- Pan-Birmingham Gynaecological Cancer Centre, City Hospital, Birmingham, UK
| | - P Moss
- Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK.
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Birmingham, UK.
- National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, Birmingham, UK.
| |
Collapse
|
37
|
Kumar A, Saha L. Colorectal cancer cell dormancy: An insight into pathways. World J Gastroenterol 2024; 30:3810-3817. [PMID: 39351431 PMCID: PMC11438629 DOI: 10.3748/wjg.v30.i33.3810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 09/02/2024] Open
Abstract
Cancer cell dormancy (CCD) in colorectal cancer (CRC) poses a significant challenge to effective treatment. In CRC, CCD contributes to tumour recurrence, drug resistance, and amplifying the disease's burden. The molecular mechanisms governing CCD and strategies for eliminating dormant cancer cells remain largely unexplored. Therefore, understanding the molecular mechanisms governing dormancy is crucial for improving patient outcomes and developing targeted therapies. This editorial highlights the complex interplay of signalling pathways and factors involved in colorectal CCD, emphasizing the roles of Hippo/YAP, pluripotent transcription factors such as NANOG, HIF-1α signalling, and Notch signalling pathways. Additionally, ERK/p38α/β/MAPK pathways, AKT signalling pathway, and Extracellular Matrix Metalloproteinase Inducer, along with some potential less explored pathways such as STAT/p53 switch and canonical and non-canonical Wnt and SMAD signalling, are also involved in promoting colorectal CCD. Highlighting their clinical significance, these findings may offer the potential for identifying key dormancy regulator pathways, improving treatment strategies, surmounting drug resistance, and advancing personalized medicine approaches. Moreover, insights into dormancy mechanisms could lead to the development of predictive biomarkers for identifying patients at risk of recurrence and the tailoring of targeted therapies based on individual dormancy profiles. It is essential to conduct further research into these pathways and their modulation to fully comprehend CRC dormancy mechanisms and enhance patient outcomes.
Collapse
Affiliation(s)
- Anil Kumar
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Lekha Saha
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
38
|
Guo T, Xu J. Cancer-associated fibroblasts: a versatile mediator in tumor progression, metastasis, and targeted therapy. Cancer Metastasis Rev 2024; 43:1095-1116. [PMID: 38602594 PMCID: PMC11300527 DOI: 10.1007/s10555-024-10186-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/31/2024] [Indexed: 04/12/2024]
Abstract
Tumor microenvironment (TME) has been demonstrated to play a significant role in tumor initiation, progression, and metastasis. Cancer-associated fibroblasts (CAFs) are the major component of TME and exhibit heterogeneous properties in their communication with tumor cells. This heterogeneity of CAFs can be attributed to various origins, including quiescent fibroblasts, mesenchymal stem cells (MSCs), adipocytes, pericytes, endothelial cells, and mesothelial cells. Moreover, single-cell RNA sequencing has identified diverse phenotypes of CAFs, with myofibroblastic CAFs (myCAFs) and inflammatory CAFs (iCAFs) being the most acknowledged, alongside newly discovered subtypes like antigen-presenting CAFs (apCAFs). Due to these heterogeneities, CAFs exert multiple functions in tumorigenesis, cancer stemness, angiogenesis, immunosuppression, metabolism, and metastasis. As a result, targeted therapies aimed at the TME, particularly focusing on CAFs, are rapidly developing, fueling the promising future of advanced tumor-targeted therapy.
Collapse
Affiliation(s)
- Tianchen Guo
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Junfen Xu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
39
|
Cao JZ, Wang CQ, Shi Z, Song JY, Tustumi F, de Castria TB, Wu J, Pocha C, Tsilimigras DI, Wang ZH, Tao T, Lu J. NOVA2 regulates the properties of liver cancer stem cells and lenvatinib resistance in hepatocellular carcinoma via the Wnt pathway. J Gastrointest Oncol 2024; 15:1674-1685. [PMID: 39279925 PMCID: PMC11399850 DOI: 10.21037/jgo-24-145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/16/2024] [Indexed: 09/18/2024] Open
Abstract
Background The regulation of cancer stem cells (CSCs) is influenced by RNA-binding proteins (RBPs). The present study sought to investigate the role of NOVA2 in the processes of self-renewal, carcinogenesis, and lenvatinib resistance in liver CSCs. Methods Neuro-oncological ventral antigen 2 (NOVA2) expression in liver CSCs was examined by real-time polymerase chain reaction (PCR). In vitro experiments were used to assess the effects of NOVA2 on liver CSC expansion and lenvatinib resistance. Results In our study, the expression of the RBP NOVA2 was higher in CSCs. NOVA2 also increased the capacity for self-renewal and carcinogenesis of the liver CSCs via the Wnt pathway. Further, suppressing the Wnt pathway leads to desensitization of the hepatocellular carcinoma (HCC) cells that overexpress NOVA2 to apoptosis caused by lenvatinib. Analyzing patient data confirmed reduced levels of NOVA2 and therefore we speculate that NOVA2 may serve as a potential indicator for response to lenvatinib in patients with HCC. Methyltransferase-like 3 (METTL3) and YTH N6-methyladenosine RNA-binding protein 1 (YTHDF1)-dependent N6-methyladenosine (m6A) methylation were linked to upregulation of NOVA2 in HCC. Furthermore, it was shown that the expression of METTL3 was elevated in cellular models of type 2 diabetes mellitus (T2DM). Conclusions NOVA2 is involved in the process of liver CSC self-renewal and carcinogenesis. In addition, NOVA2 expression may help identify patients with a higher chance of benefiting from lenvatinib treatment and can be a promising therapeutic target for HCC.
Collapse
Affiliation(s)
- Jing-Zhu Cao
- Department of Endocrinology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chao-Qun Wang
- Department of Endocrinology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zheng Shi
- Department of Gastrointestinal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jing-Yun Song
- Department of Endocrinology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Francisco Tustumi
- Department of Gastroenterology, Digestive Surgery Division, University of São Paulo Medical School, Sao Paulo, Brazil
| | - Tiago Biachi de Castria
- Moffitt Cancer Center, Tampa, FL, USA
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jennifer Wu
- Division of Hematology and Oncology, Perlmutter Cancer Center of NYU Langone Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Christine Pocha
- Sanford School of Medicine, University of South Dakota, Avera Hepatology and Transplant, Sioux Falls, SD, USA
| | - Diamantis I Tsilimigras
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Zhi-Hong Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Tao Tao
- Department of Endocrinology and Metabolism, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jin Lu
- Department of Endocrinology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
40
|
Limonta P, Chiaramonte R, Casati L. Unveiling the Dynamic Interplay between Cancer Stem Cells and the Tumor Microenvironment in Melanoma: Implications for Novel Therapeutic Strategies. Cancers (Basel) 2024; 16:2861. [PMID: 39199632 PMCID: PMC11352669 DOI: 10.3390/cancers16162861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Cutaneous melanoma still represents a significant health burden worldwide, being responsible for the majority of skin cancer deaths. Key advances in therapeutic strategies have significantly improved patient outcomes; however, most patients experience drug resistance and tumor relapse. Cancer stem cells (CSCs) are a small subpopulation of cells in different tumors, including melanoma, endowed with distinctive capacities of self-renewal and differentiation into bulk tumor cells. Melanoma CSCs are characterized by the expression of specific biomarkers and intracellular pathways; moreover, they play a pivotal role in tumor onset, progression and drug resistance. In recent years, great efforts have been made to dissect the molecular mechanisms underlying the protumor activities of melanoma CSCs to provide the basis for novel CSC-targeted therapies. Herein, we highlight the intricate crosstalk between melanoma CSCs and bystander cells in the tumor microenvironment (TME), including immune cells, endothelial cells and cancer-associated fibroblasts (CAFs), and its role in melanoma progression. Specifically, we discuss the peculiar capacities of melanoma CSCs to escape the host immune surveillance, to recruit immunosuppressive cells and to educate immune cells toward an immunosuppressive and protumor phenotype. We also address currently investigated CSC-targeted strategies that could pave the way for new promising therapeutic approaches for melanoma care.
Collapse
Affiliation(s)
- Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences “R. Paoletti”, Università degli Studi di Milano, 20133 Milan, Italy
| | - Raffaella Chiaramonte
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy;
| | - Lavinia Casati
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy;
| |
Collapse
|
41
|
Zhang Q, Zhu F, Tong Y, Shi D, Zhang J. CHD4 R975H mutant activates tumorigenic pathways and promotes stemness and M2-like macrophage polarization in endometrial cancer. Sci Rep 2024; 14:18617. [PMID: 39127769 PMCID: PMC11316823 DOI: 10.1038/s41598-024-69233-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Endometrial cancer (EC), one of the most prevalent carcinomas in females, is associated with increasing mortality. We identified the CHD4 R975H mutation as a high-frequency occurrence in EC patients through a comprehensive survey of EC databases. Computational predictions suggest that this mutation profoundly impacts the structural and functional integrity of CHD4. Functional assays revealed that the CHD4 R975H mutation enhances EC cell invasion, proliferation, and colony formation, promoting a cancer stem cell (CSC)-like phenotype. RNA-seq analysis of cells expressing CHD4 R975H mutant revealed a transcriptomic landscape marked by the activation of several cancer-promoting signaling pathways, including TNF-α signaling via NF-κB, KRAS, P53, mTOR, TGF-β, EGFR, Myc and growth factor signaling. Validation assays confirmed the activation of these pathways, further demonstrating that CHD4 R975H mutation induces stemness in EC cells and M2-like polarization of tumor-associated macrophages (TAMs). Our study elucidated the oncogenic role of CHD4 R975H mutation, highlighting its dual impact on facilitating cancer stemness and transforming TAMs into an immunosuppressive subtype. These findings contribute valuable insights into the molecular mechanisms driving EC progression and open avenues for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Qinglin Zhang
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Fengzhi Zhu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Yin Tong
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China
| | - Diwen Shi
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Jiangwen Zhang
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
42
|
Aceto GM, Pagotto S, Del Pizzo FD, Saoca C, Selvaggi F, Visone R, Cotellese R, Aguennouz M, Lattanzio R, Catalano T. Differential Regulation of Wingless-Wnt/c-Jun N-Terminal Kinase Crosstalk via Oxidative Eustress in Primary and Metastatic Colorectal Cancer Cells. Biomedicines 2024; 12:1816. [PMID: 39200280 PMCID: PMC11351841 DOI: 10.3390/biomedicines12081816] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
In the tumor microenvironment (TME), ROS production affects survival, progression, and therapy resistance in colorectal cancer (CRC). H2O2-mediated oxidative stress can modulate Wnt/β-catenin signaling and metabolic reprogramming of the TME. Currently, it is unclear how mild/moderate oxidative stress (eustress) modulates Wnt/β-catenin/APC and JNK signaling relationships in primary and metastatic CRC cells. In this study, we determined the effects of the H2O2 concentration inducing eustress on isogenic SW480 and SW620 cells, also in combination with JNK inhibition. We assessed cell viability, mitochondrial respiration, glycolysis, and Wnt/β-catenin/APC/JNK gene and protein expression. Primary CRC cells were more sensitive to H2O2 eustress combined with JNK inhibition, showing a reduction in viability compared to metastatic cells. JNK inhibition under eustress reduced both glycolytic and respiratory capacity in SW620 cells, indicating a greater capacity to adapt to TME. In primary CRC cells, H2O2 alone significantly increased APC, LEF1, LRP6, cMYC and IL8 gene expression, whereas in metastatic CRC cells, this effect occurred after JNK inhibition. In metastatic but not in primary tumor cells, eustress and inhibition of JNK reduced APC, β-catenin, and pJNK protein. The results showed differential cross-regulation of Wnt/JNK in primary and metastatic tumor cells under environmental eustress conditions. Further studies would be useful to validate these findings and explore their therapeutic potential.
Collapse
Affiliation(s)
- Gitana Maria Aceto
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.P.); (R.V.); (R.C.)
| | - Sara Pagotto
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.P.); (R.V.); (R.C.)
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.D.D.P.); (R.L.)
| | - Francesco Domenico Del Pizzo
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.D.D.P.); (R.L.)
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Concetta Saoca
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.S.); (M.A.); (T.C.)
| | - Federico Selvaggi
- Unit of General Surgery, Ospedale Clinicizzato SS Annunziata of Chieti, 66100 Chieti, Italy;
| | - Rosa Visone
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.P.); (R.V.); (R.C.)
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.D.D.P.); (R.L.)
| | - Roberto Cotellese
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.P.); (R.V.); (R.C.)
- Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy
| | - M’hammed Aguennouz
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.S.); (M.A.); (T.C.)
| | - Rossano Lattanzio
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.D.D.P.); (R.L.)
- Department of Innovative Technologies in Medicine & Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Teresa Catalano
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.S.); (M.A.); (T.C.)
| |
Collapse
|
43
|
Shi R, Yu R, Lian F, Zheng Y, Feng S, Li C, Zheng X. Targeting HSP47 for cancer treatment. Anticancer Drugs 2024; 35:623-637. [PMID: 38718070 DOI: 10.1097/cad.0000000000001612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Heat shock protein 47 (HSP47) serves as an endoplasmic reticulum residing collagen-specific chaperone and plays an important role in collagen biosynthesis and structural assembly. HSP47 is encoded by the SERPINH1 gene, which is located on chromosome 11q13.5, one of the most frequently amplified regions in human cancers. The expression of HSP47 is regulated by multiple cellular factors, including cytokines, transcription factors, microRNAs, and circular RNAs. HSP47 is frequently upregulated in a variety of cancers and plays an important role in tumor progression. HSP47 promotes tumor stemness, angiogenesis, growth, epithelial-mesenchymal transition, and metastatic capacity. HSP47 also regulates the efficacy of tumor therapies, such as chemotherapy, radiotherapy, and immunotherapy. Inhibition of HSP47 expression has antitumor effects, suggesting that targeting HSP47 is a feasible strategy for cancer treatment. In this review, we highlight the function and expression of regulatory mechanisms of HSP47 in cancer progression and point out the potential development of therapeutic strategies in targeting HSP47 in the future.
Collapse
Affiliation(s)
- Run Shi
- School of Medicine, Pingdingshan University, Pingdingshan, China
| | | | | | | | | | | | | |
Collapse
|
44
|
Pan Z, Ke C, Zheng H, Guo X, Gao W, Huang X, Chen C, Xiong Y, Zheng S, Zheng F, Hu W. FERMT1 suppression induces anti-tumor effects and reduces stemness in glioma cancer cells. J Cancer Res Clin Oncol 2024; 150:338. [PMID: 38976072 PMCID: PMC11231014 DOI: 10.1007/s00432-024-05859-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/17/2024] [Indexed: 07/09/2024]
Abstract
OBJECTIVE Glioma is a leading cause of mortality worldwide, its recurrence poses a major challenge in achieving effective treatment outcomes. Cancer stem cells (CSCs) have emerged as key contributors to tumor relapse and chemotherapy resistance, making them attractive targets for glioma cancer therapy. This study investigated the potential of FERMT1 as a prognostic biomarker and its role in regulating stemness through cell cycle in glioma. METHODS Using data from TCGA-GBM, GSE4290, GSE50161 and GSE147352 for analysis of FERMT1 expression in glioma tissues. Then, the effects of FERMT1 knockdown on cell cycle, proliferation, sphere formation ability, invasion and migration were investigated. The influences of FERMT1 on expression of glycolysis-related proteins and levels of ATP, glucose, lactate and G6PDH were also explored. Furthermore, the effects of FERMT1 knockdown on cellular metabolism were evidenced. RESULTS Significant upregulation of FERMT1 in glioma tissues was observed. Silencing FERMT1 not only affected the cell cycle but also led to a notable reduction in proliferation, invasion and migration. The expression of glycolysis-associated proteins including GLUT1, GLUT3, GLUT4, and SCO2 were reduced by FERMT1 knockdown, resulted in increased ATP and glucose as well as decreased lactic acid and G6PDH levels. FERMT1 knockdown also inhibited cellular metabolism. Moreover, FERMT1 knockdown significantly reduced sphere diameter, along with inhibiting the expression of transcription factors associated with stemness in glioma cells. CONCLUSION These findings demonstrated that FERMT1 could be an ideal target for the advancement of innovative strategies against glioma treatment via modulating cellular process involved in stemness regulation and metabolism.
Collapse
Affiliation(s)
- Zhigang Pan
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, 34# zhongshan North Road, Quanzhou, Fujian, 362000, China
| | - Chuhan Ke
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, 34# zhongshan North Road, Quanzhou, Fujian, 362000, China
| | - Hanlin Zheng
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, 34# zhongshan North Road, Quanzhou, Fujian, 362000, China
| | - Xiumei Guo
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, 34# zhongshan North Road, Quanzhou, Fujian, 362000, China
| | - Wen Gao
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, 34# zhongshan North Road, Quanzhou, Fujian, 362000, China
| | - Xinyue Huang
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, 34# zhongshan North Road, Quanzhou, Fujian, 362000, China
| | - Chunhui Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, 34# zhongshan North Road, Quanzhou, Fujian, 362000, China
| | - Yu Xiong
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, 34# zhongshan North Road, Quanzhou, Fujian, 362000, China
| | - Shuni Zheng
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, 34# zhongshan North Road, Quanzhou, Fujian, 362000, China.
| | - Feng Zheng
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, 34# zhongshan North Road, Quanzhou, Fujian, 362000, China.
| | - Weipeng Hu
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, 34# zhongshan North Road, Quanzhou, Fujian, 362000, China.
| |
Collapse
|
45
|
ISHIKAWA K, CHAMBERS JK, UCHIDA K. Activation of the Wnt/β-catenin signaling pathway and CTNNB1 mutations in canine intestinal epithelial proliferative lesions. J Vet Med Sci 2024; 86:748-755. [PMID: 38811188 PMCID: PMC11251820 DOI: 10.1292/jvms.24-0125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024] Open
Abstract
Nuclear expression of β-catenin has been reported in canine intestinal epithelial tumors (IETs) and colorectal inflammatory polyps (CIPs) with dysplastic epithelia. However, the role of the Wnt/β-catenin signaling pathway in these lesions remains unclear. To investigate the association between the nuclear β-catenin expression and the activation of the Wnt/β-catenin signaling pathway, immunohistochemistry and mutation analyses were conducted on 64 IETs and 20 CIPs. IETs and CIPs with β-catenin nuclear and/or cytoplasm immunolabeling were classified as β-catenin (+). The immunostaining scores of c-Myc and Cyclin D1 and Ki-67 index were significantly higher in β-catenin (+) cases than in β-catenin (-) cases. Identical APC mutations (p.E154D and p.K155X) were detected in 6/41 β-catenin (+) IETs; all 6 of IETs with APC mutations were Jack Russell Terriers. CTNNB1 mutations were detected in 29/42 β-catenin (+) IETs, 3/11 β-catenin (+) CIPs, and 2/22 β-catenin (-) IETs, most of which were hotspots associated with human colorectal carcinoma. In one Miniature Dachshund diagnosed with a CIP that subsequently developed into an IET, the same CTNNB1 mutation was detected in both lesions. The immunohistochemical results suggest that cell proliferative activity in β-catenin (+) cases may be associated with activation of the Wnt/β-catenin signaling pathway. The mutation analysis results suggest that CTNNB1 mutations may be associated with cytoplasmic β-catenin accumulation in IET and CIP. Furthermore, the dysplastic epithelium in CIP may progress to IET through the activation of the Wnt/β-catenin signaling pathway by the CTNNB1 mutation.
Collapse
Affiliation(s)
- Kento ISHIKAWA
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - James K CHAMBERS
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyuki UCHIDA
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
46
|
Dai G, Sun Y. Knockdown of GNL3 inhibits LUAD cell growth by regulating Wnt-β-catenin pathway. Allergol Immunopathol (Madr) 2024; 52:46-52. [PMID: 38970264 DOI: 10.15586/aei.v52i4.1117] [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: 04/26/2024] [Accepted: 05/26/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a leading cause of tumor-associated mortality, and it is needed to find new target to combat this disease. Guanine nucleotide-binding -protein-like 3 (GNL3) mediates cell proliferation and apoptosis in several cancers, but its role in LUAD remains unclear. OBJECTIVE To explore the expression and function of Guanine nucleotide-binding protein-like 3 (GNL3) in lung adenocarcinoma (LUAD) and its potential mechanism in inhibiting the growth of LUAD cells. METHODS We evaluated the expression of GNL3 in LUAD tissues and its association with patient prognosis using databases and immunohistochemistry. Cell proliferation was assessed by CCK-8 assay as well as colony formation, while apoptosis was evaluated by FCM. The effect of GNL3 knockdown on the Wnt/β-catenin axis was investigated by Immunoblot analysis. RESULTS GNL3 is overexpressed in LUAD tissues and is correlated with poor prognosis. Knockdown of GNL3 significantly inhibited the growth as well as induced apoptosis in A549 as well as H1299 cells. Furthermore, we found that the inhibitory effect of GNL3 knockdown on LUAD cell growth is associated with the downregulation of the Wnt/β-catenin axis. CONCLUSION GNL3 is key in the progression of LUAD by metiating Wnt/β-catenin axis. Targeting GNL3 may represent a novel therapeutic method for LUAD treatment.
Collapse
Affiliation(s)
- Guihong Dai
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu Province China
| | - Yuejun Sun
- Department of Pathology, Affiliated Jiangyin Clinical College of Xuzhou Medical University, Jiangyin, Jiangsu Province, China;
| |
Collapse
|
47
|
He T, Hu C, Li S, Fan Y, Xie F, Sun X, Jiang Q, Chen W, Jia Y, Li W. The role of CD8 + T-cells in colorectal cancer immunotherapy. Heliyon 2024; 10:e33144. [PMID: 39005910 PMCID: PMC11239598 DOI: 10.1016/j.heliyon.2024.e33144] [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: 10/27/2023] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024] Open
Abstract
Immunotherapy has been an advanced and effective approach to treating various types of solid tumors in recent years, and the most successful strategy is immune checkpoint inhibitors (ICIs), which have shown beneficial effects in patients with colorectal cancer (CRC). Drug resistance to ICIs is usually associated with CD8+ T-cells targeting tumor antigens; thus, CD8+ T-cells play an important role in immunotherapy. Unfortunately, Under continuous antigen stimulation, tumor microenvironment(TME), hypoxia and other problems it leads to insufficient infiltration of CD8+ T-cells, low efficacy and mechanism exhaustion, which have become obstacles to immunotherapy. Thus, this article describes the relationship between CRC and the immune system, focuses on the process of CD8+ T-cells production, activation, transport, killing, and exhaustion, and expounds on related mechanisms leading to CD8+ T-cells exhaustion. Finally, this article summarizes the latest strategies and methods in recent years, focusing on improving the infiltration, efficacy, and exhaustion of CD8+ T-cells, which may help to overcome the barriers to immunotherapy.
Collapse
Affiliation(s)
- Tao He
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Chencheng Hu
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Shichao Li
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Yao Fan
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Fei Xie
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Xin Sun
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Qingfeng Jiang
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Weidong Chen
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Yingtian Jia
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Wusheng Li
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou City, Sichuan Province, China
| |
Collapse
|
48
|
Fu L, Li F, Xue X, Xi H, Sun X, Hu R, Wen H, Liu S. Exploring the potential of thiophene derivatives as dual inhibitors of β-tubulin and Wnt/β-catenin pathways for gastrointestinal cancers in vitro. Heliyon 2024; 10:e32241. [PMID: 38912446 PMCID: PMC11190604 DOI: 10.1016/j.heliyon.2024.e32241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/25/2024] Open
Abstract
Background Gastrointestinal cancer poses a considerable global health risk, encompassing a heterogeneous spectrum of malignancies that afflict the gastrointestinal tract. It is significant to develop efficacious therapeutic agents, as they are indispensable for both the treatment and prevention of this formidable disease. Methods In this study, we synthesized a novel thiophene derivative, designated as compound 1312. An assessment was performed to investigate its anti-proliferative activity in several cancer cell lines (GES-1, EC9706, SGC7901, and HT-29). Furthermore, we performed molecular biology techniques to investigate the inhibitory impact of compound 1312 on gastrointestinal cell lines SGC-7901 and HT-29. Results Our findings reveal that compound 1312 exhibits significant efficacy in suppressing colony formation of cancer cells. Notably, it triggers cell cycle arrest at the G2/M phase in gastrointestinal cell lines SGC7901 and HT-29. Compound 1312 was confirmed to exert inhibitory effects on cell migration and invasion in SGC7901. Additionally, the compound elicits apoptotic cell death through the activation of the DNA repair enzyme poly (ADP-ribose) polymerase (PARP) and the caspase signaling cascade. Furthermore, in vitro experiments revealed that compound 1312 effectively suppresses both the β-tubulin cytoskeletal network and the Wnt/β-catenin signaling pathway. These multifaceted anti-cancer activities highlight the potential of compound 1312 as a promising therapeutic agent for the treatment of gastrointestinal malignancies. Conclusion This study indicates the promising potential of compound 1312 as a prospective candidate agent for gastrointestinal cancer treatment. Further comprehensive investigations are needed to explore its therapeutic efficacy in greater detail.
Collapse
Affiliation(s)
- Lina Fu
- School of Medicine, Huanghe Science and Technology College, Zhengzhou 450063, Henan, China
| | - Fuhao Li
- The First Clinical Medicine College of Zhejiang Chinese Medical University, Zhejiang 310053, Hangzhou, China
| | - Xia Xue
- Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Huayuan Xi
- Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiangdong Sun
- Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ruoyu Hu
- Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Huijuan Wen
- School of Medicine, Huanghe Science and Technology College, Zhengzhou 450063, Henan, China
- Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Simeng Liu
- School of Medicine, Huanghe Science and Technology College, Zhengzhou 450063, Henan, China
- Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| |
Collapse
|
49
|
Han R, Yang J, Zhu Y, Gan R. Wnt signaling in gastric cancer: current progress and future prospects. Front Oncol 2024; 14:1410513. [PMID: 38952556 PMCID: PMC11216096 DOI: 10.3389/fonc.2024.1410513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/13/2024] [Indexed: 07/03/2024] Open
Abstract
Levels of the Wnt pathway components are abnormally altered in gastric cancer cells, leading to malignant cell proliferation, invasion and metastasis, poor prognosis and chemoresistance. Therefore, it is important to understand the mechanism of Wnt signaling pathway in gastric cancer. We systematically reviewed the molecular mechanisms of the Wnt pathway in gastric cancer development; and summarize the progression and the challenges of research on molecular agents of the Wnt pathway.
Collapse
Affiliation(s)
- Ruyue Han
- Cancer Research Institute, Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jing Yang
- Department of Gastroenterology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yingying Zhu
- Cancer Research Institute, Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Runliang Gan
- Cancer Research Institute, Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
50
|
Busico A, Gasparini P, Rausa E, Cattaneo L, Bozzi F, Silvestri M, Capone I, Conca E, Tamborini E, Perrone F, Vitellaro M, Ricci MT, Casanova M, Chiaravalli S, Bergamaschi L, Massimino M, Milione M, Sozzi G, Pruneri G, Ferrari A, Signoroni S. Molecular profiling of pediatric and young adult colorectal cancer reveals a distinct genomic landscapes and potential therapeutic avenues. Sci Rep 2024; 14:13138. [PMID: 38849509 PMCID: PMC11161608 DOI: 10.1038/s41598-024-64149-7] [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: 12/14/2023] [Accepted: 06/05/2024] [Indexed: 06/09/2024] Open
Abstract
Colorectal cancer (CRC) is a global health concern, and the incidence of early onset (EO) CRC, has an upward trend. This study delves into the genomic landscape of EO-CRC, specifically focusing on pediatric (PED) and young adult (YA) patients, comparing them with adult (AD) CRC. In this retrospective monocentric investigation, we performed targeted next-generation sequencing to compare the mutational profile of 38 EO-CRCs patients (eight PED and 30 YA) to those of a 'control group' consisting of 56 AD-CRCs. Our findings reveal distinct molecular profiles in EO-CRC, notably in the WNT and PI3K-AKT pathways. In pediatrics, we observed a significantly higher frequency of RNF43 mutations, whereas APC mutations were more prevalent in adult cases. These observations suggest age-related differences in the activation of the WNT pathway. Pathway and copy number variation analysis reveal that AD-CRC and YA-CRC have more similarities than the pediatric patients. PED shows a peculiar profile with CDK6 amplification and the enrichment of lysine degradation pathway. These findings may open doors for personalized therapies, such as PI3K-AKT pathway inhibitors or CDK6 inhibitors for pediatric patients. Additionally, the distinct molecular signatures of EO-CRC underscore the need for age-specific treatment strategies and precision medicine. This study emphasizes the importance of comprehensive molecular investigations in EO-CRCs, which can potentially improve diagnostic accuracy, prognosis, and therapeutic decisions for these patients. Collaboration between the pediatric and adult oncology community is fundamental to improve oncological outcomes for this rare and challenging pediatric tumor.
Collapse
Affiliation(s)
- A Busico
- Department of Diagnostic Innovation, Pathology Unit 2, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - P Gasparini
- sc Epigenomics and Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy.
| | - E Rausa
- Unit of Hereditary Digestive Tract Tumors, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - L Cattaneo
- Department of Diagnostic Innovation, Biobank, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - F Bozzi
- Department of Diagnostic Innovation, Pathology Unit 2, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - M Silvestri
- Department of Diagnostic Innovation, Pathology Unit 2, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - I Capone
- Department of Diagnostic Innovation, Pathology Unit 2, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - E Conca
- Department of Diagnostic Innovation, Pathology Unit 2, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - E Tamborini
- Department of Diagnostic Innovation, Pathology Unit 2, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - F Perrone
- Department of Diagnostic Innovation, Pathology Unit 2, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - M Vitellaro
- Unit of Hereditary Digestive Tract Tumors, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
- Department of Diagnostic Innovation, Biobank, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
- Colorectal Surgery Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - M T Ricci
- Unit of Hereditary Digestive Tract Tumors, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - M Casanova
- Colorectal Surgery Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - S Chiaravalli
- sc Pediatric Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - L Bergamaschi
- sc Pediatric Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - M Massimino
- sc Pediatric Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - M Milione
- Department of Diagnostic Innovation, Pathology Unit 1, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - G Sozzi
- sc Epigenomics and Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - G Pruneri
- Department of Diagnostic Innovation, Pathology Unit 2, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - A Ferrari
- sc Pediatric Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| | - S Signoroni
- Unit of Hereditary Digestive Tract Tumors, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milano, Italy
| |
Collapse
|