1
|
Liang H, Zhou B, Li P, Zhang X, Zhang S, Zhang Y, Yao S, Qu S, Chen J. Stemness regulation in prostate cancer: prostate cancer stem cells and targeted therapy. Ann Med 2025; 57:2442067. [PMID: 39711287 DOI: 10.1080/07853890.2024.2442067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Increasing evidence indicates that cancer stem cells (CSCs) and cancer stem-like cells form a special subpopulation of cells that are ubiquitous in tumors. These cells exhibit similar characteristics to those of normal stem cells in tissues; moreover, they are capable of self-renewal and differentiation, as well as high tumorigenicity and drug resistance. In prostate cancer (PCa), it is difficult to kill these cells using androgen signaling inhibitors and chemotherapy drugs. Consequently, the residual prostate cancer stem cells (PCSCs) mediate tumor recurrence and progression. OBJECTIVE This review aims to provide a comprehensive and up-to-date overview of PCSCs, with a particular emphasis on potential therapeutic strategies targeting these cells. METHODS After searching in PubMed and Embase databases using 'prostate cancer' and 'cancer stem cells' as keywords, studies related were compiled and examined. RESULTS In this review, we detail the origin and characteristics of PCSCs, introduce the regulatory pathways closely related to CSC survival and stemness maintenance, and discuss the link between epithelial-mesenchymal transition, tumor microenvironment and tumor stemness. Furthermore, we introduce the currently available therapeutic strategies targeting CSCs, including signaling pathway inhibitors, anti-apoptotic protein inhibitors, microRNAs, nanomedicine, and immunotherapy. Lastly, we summarize the limitations of current CSC research and mention future research directions. CONCLUSION A deeper understanding of the regulatory network and molecular markers of PCSCs could facilitate the development of novel therapeutic strategies targeting these cells. Previous preclinical studies have demonstrated the potential of this treatment approach. In the future, this may offer alternative treatment options for PCa patients.
Collapse
Affiliation(s)
- Hao Liang
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Bin Zhou
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Peixin Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyi Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shijie Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Yaozhong Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shengwen Yao
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Sifeng Qu
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Jun Chen
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| |
Collapse
|
2
|
Zhang S, Miao L, Tian X, Yang B, Luo B. Opportunities and challenges of immuno-oncology: A bibliometric analysis from 2014 to 2023. Hum Vaccin Immunother 2025; 21:2440203. [PMID: 39885669 PMCID: PMC11792843 DOI: 10.1080/21645515.2024.2440203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 02/01/2025] Open
Abstract
The emergence of immuno-oncology (IO) has led to revolutionary changes in the field of cancer treatment. Despite notable advancements in this field, a thorough exploration of its full depth and extent has yet to be performed. This study provides a comprehensive overview of publications pertaining to IO. Publications on IO from 2014 to 2023 were retrieved by searching the Web of Science Core Collection database (WoSCC). VOSviewer software and Citespace software were used for the visualized analysis. A total of 1,874 articles have been published in the IO domain. The number of publications and citations has been increasing annually. This study also examines the primary research directions within the field of IO. In conclusion, this study offers a comprehensive overview of the opportunities and challenges associated with IO, illuminating the current status of research and indicating potential future trajectories in this rapidly progressing field. This study provides a comprehensive survey of the current research status and hot spots within the field of IO. It will assist researchers in comprehending the current research emphasis and development trends in this field and offers guidance for future research directions.
Collapse
Affiliation(s)
- Siqi Zhang
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| | - Lina Miao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoxia Tian
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Bingxu Yang
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Baoping Luo
- School of Clinical Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, China
- Department of Oncology, Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
3
|
Li W, Xiao L, Li H, Cui W. Global research trends of immunosenescence and immunotherapy: A bibliometric study. Hum Vaccin Immunother 2025; 21:2469403. [PMID: 39992200 PMCID: PMC11853558 DOI: 10.1080/21645515.2025.2469403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/31/2025] [Accepted: 02/15/2025] [Indexed: 02/25/2025] Open
Abstract
Immunosenescence refers to the gradual decline in immune system function with age, increasing susceptibility to infections and cancer in the elderly. The advent of novel immunotherapies has revolutionized the field of cancer treatment. However, the majority of patients exhibit poor re-sponses to immunotherapy, with immunosenescence likely playing a significant role. In recent years, significant progress has been made in understanding the interplay between immunosenescence and immunotherapy. Our research aims to explore the prospects and development trends in the field of immunosenescence and immunotherapy using a bibliometric analysis. Relevant articles were collected from the Web of Science Core Collection (WoSCC) (retrieved on July 20, 2024). Primary bibliometric characteristics were analyzed using the R package "Biblio-metrix," and keyword co-occurrence analysis and visualization were conducted using VOSviewer. A total of 213 English-language original research and review articles spanning 35 years were re-trieved for bibliometric analysis. There was a surge in publications in this field starting in 2017. The United States and China contributed the most articles. Frontiers in Immunology was the most productive journal, while the University of California System was the highest contributing institution. Besse Benjamin from France emerged as the most influential researcher in this field. Popular keywords included "nivolumab," "T cells," "dendritic cells," and "regulatory T cells." The "immunosenescence-associated secretory phenotype" has become a new hotspot, with immune checkpoint inhibitors remaining a central theme in this domain. The field of immunosenescence and immunotherapy is entering a phase of rapid development and will continue to hold significant value in future research.
Collapse
Affiliation(s)
- Wendi Li
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Xiao
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiyang Li
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Cui
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Zheng XQ, Wang DB, Jiang YR, Song CL. Gut microbiota and microbial metabolites for osteoporosis. Gut Microbes 2025; 17:2437247. [PMID: 39690861 DOI: 10.1080/19490976.2024.2437247] [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: 07/11/2024] [Revised: 11/13/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Osteoporosis is an age-related bone metabolic disease. As an essential endocrine organ, the skeletal system is intricately connected with extraosseous organs. The crosstalk between bones and other organs supports this view. In recent years, the link between the gut microecology and bone metabolism has become an important research topic, both in preclinical studies and in clinical trials. Many studies have shown that skeletal changes are accompanied by changes in the composition and structure of the gut microbiota (GM). At the same time, natural or artificial interventions targeting the GM can subsequently affect bone metabolism. Moreover, microbiome-related metabolites may have important effects on bone metabolism. We aim to review the relationships among the GM, microbial metabolites, and bone metabolism and to summarize the potential mechanisms involved and the theory of the gut‒bone axis. We also describe existing bottlenecks in laboratory studies, as well as existing challenges in clinical settings, and propose possible future research directions.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Ding-Ben Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Yi-Rong Jiang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| |
Collapse
|
5
|
Ma Y, Boycott C, Zhang J, Gomilar R, Yang T, Stefanska B. SIRT1/DNMT3B-mediated epigenetic gene silencing in response to phytoestrogens in mammary epithelial cells. Epigenetics 2025; 20:2473770. [PMID: 40029260 PMCID: PMC11881848 DOI: 10.1080/15592294.2025.2473770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025] Open
Abstract
We performed an integrated analysis of genome-wide DNA methylation and expression datasets in normal cells and healthy animals exposed to polyphenols with estrogenic activity (i.e. phytoestrogens). We identified that phytoestrogens target genes linked to disrupted cellular homeostasis, e.g. genes limiting DNA break repair (RNF169) or promoting ribosomal biogenesis (rDNA). Existing evidence suggests that DNA methylation may be governed by sirtuin 1 (SIRT1) deacetylase via interactions with DNA methylating enzymes, specifically DNMT3B. Since SIRT1 was reported to be regulated by phytoestrogens, we test whether phytoestrogens suppress genes related to disrupted homeostasis via SIRT1/DNMT3B-mediated transcriptional silencing. Human MCF10A mammary epithelial cells were treated with phytoestrogens, pterostilbene (PTS) or genistein (GEN), followed by analysis of cell growth, DNA methylation, gene expression, and SIRT1/DNMT3B binding. SIRT1 occupancy at the selected phytoestrogen-target genes, RNF169 and rDNA, was accompanied by consistent promoter hypermethylation and gene downregulation in response to GEN, but not PTS. GEN-mediated hypermethylation and SIRT1 binding were linked to a robust DNMT3B enrichment at RNF169 and rDNA promoters. This was not observed in cells exposed to PTS, suggesting a distinct mechanism of action. Although both SIRT1 and DNMT3B bind to RNF169 and rDNA promoters upon GEN, the two proteins do not co-occupy the regions. Depletion of SIRT1 abolishes GEN-mediated decrease in rDNA expression, suggesting SIRT1-dependent epigenetic suppression of rDNA by GEN. These findings enhance our understanding of the role of SIRT1-DNMT3B interplay in epigenetic mechanisms mediating the impact of phytoestrogens on cell biology and cellular homeostasis.
Collapse
Affiliation(s)
- Yuexi Ma
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Cayla Boycott
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Jiaxi Zhang
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Rekha Gomilar
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Tony Yang
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Barbara Stefanska
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
6
|
Huang FJ, Fang YY, Wen JY, Li JJ, Lin Q, Su QY, Chen YY, Wang L, Zeng JJ, Chi BT, He RQ, Qin DY, Yang LH, Chen G. From PD-1/PD-L1 to tertiary lymphoid structures: Paving the way for precision immunotherapy in cholangiocarcinoma treatment. Hum Vaccin Immunother 2025; 21:2444697. [PMID: 39757910 DOI: 10.1080/21645515.2024.2444697] [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: 09/17/2024] [Revised: 12/07/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a highly malignant hepatobiliary tumor characterized by limited treatment options and poor prognosis. The recent rise of immunotherapy has significantly influenced research in this field. This study presents a bibliometric analysis of 416 articles retrieved from the WOSCC, Wan fang Data, CNKI and VIP databases, spanning contributions from 32 countries, 589 institutions and 3,200 authors. The analysis identified "PD-L1," "PD-1" and "pembrolizumab" as central research foci, while "immune checkpoint inhibitors," "tumor immune microenvironment," "tertiary lymphoid structures" and "durvalumab" emerged as key areas of interest. These findings emphasize the pivotal role of immunotherapy in improving survival outcomes for CCA, and they highlight the significance of tertiary lymphoid structures within the tumor microenvironment as a promising target for future research. This study offers a strategic overview of the evolving landscape of CCA immunotherapy, providing valuable insights to guide future scientific endeavors in this domain.
Collapse
Affiliation(s)
- Fang-Ju Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Ye-Ying Fang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jia-Ying Wen
- Department of Radiotherapy, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jian-Jun Li
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Qian Lin
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Qin-Yan Su
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Yi-Yang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Lei Wang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jian-Jia Zeng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Bang-Teng Chi
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Di-Yuan Qin
- Department of Computer Science and Technology, School of Computer and Electronic Information, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Li-Hua Yang
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| |
Collapse
|
7
|
Kim HS, Woo W, Choi YG, Bharat A, Chae YK. Novel association between graft rejection and post-transplant malignancy in solid organ transplantation. World J Transplant 2025; 15:102384. [DOI: 10.5500/wjt.v15.i2.102384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/10/2024] [Accepted: 01/09/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Advancements in immunosuppressive therapies have improved graft survival by enhancing graft tolerance and preventing organ rejection. However, the risk of malignancy associated with prolonged immunosuppression remains a concern, as it can adversely affect recipients’ quality of life and survival. While the link between immunosuppression and increased cancer risk is well-documented, the specific interactions between graft rejection and post-transplant malignancy (PTM) remain poorly understood. Addressing this knowledge gap is crucial for devising immunosuppressive strategies that balance rejection prevention with cancer risk reduction.
AIM To investigate whether immunosuppression in PTM reduces rejection risk, while immune activation during rejection protects against malignancy.
METHODS We analyzed data from the United Network for Organ Sharing’s Organ Procurement and Transplantation Network database (1987–2023) on adult, first-time, single-organ transplant recipients with no prior history of malignancy (in donors or recipients). Landmark analyses at 1, 2, 3, 5, 10, 15, and 20 years post-transplant, Kaplan–Meier analyses, and time-dependent Cox proportional hazards regression models, each incorporating the temporal dimension of outcomes, assessed the association between rejection-induced graft failure (RGF) and PTM. Multivariate models were adjusted for clinical and immunological factors, including immunosuppression regimens.
RESULTS The cohort included 579905 recipients (kidney: 386878; liver: 108390; heart: 45046; lung: 37643; pancreas: 1948) with a mean follow-up of 7.3 years and a median age of 50.6 ± 13.2 years. RGF was associated with a reduction in PTM risk across all time points [hazard ratio (HR) = 0.07-0.20, P < 0.001], even after excluding mortality cases. Kidney transplant recipients exhibited the most pronounced reduction (HR = 0.22, P < 0.001). Conversely, among recipients with PTM, RGF risk decreased across all time points up to 15 years after excluding mortality cases (HR = 0.49–0.80, P < 0.001). This risk reduction was observed in kidney, liver, heart, and lung transplants (HRs = 0.90, 0.21, 0.21, and 0.18, respectively; P < 0.001) but not in pancreas transplants.
CONCLUSION RGF reduces PTM risk, particularly in kidney transplants, while PTM decreases RGF risk in kidney, liver, heart, and lung transplants.
Collapse
Affiliation(s)
- Hye Sung Kim
- Department of Medicine, Temple University Hospital, Philadelphia, PA 19140, United States
| | - Wongi Woo
- Department of Medicine, St. Joseph Medical Center, Stockton, CA 95204, United States
| | - Young-Geun Choi
- Department of Mathematics Education, Sungkyunkwan University, Seoul 03063, South Korea
| | - Ankit Bharat
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Young Kwang Chae
- Department of Medicine, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, United States
| |
Collapse
|
8
|
Zhang X, Wang Z, Yang J, Li Y, Lu C, Hao Y, He G, Zhang Y, Song Q, Long J, Liang J, Tang Y. Smartphone-based urine colourimetric assay for home self-screening of HPV infection. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 334:125923. [PMID: 39983278 DOI: 10.1016/j.saa.2025.125923] [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: 10/08/2024] [Revised: 01/26/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
Currently, producing NPSH after HPV infection of cells has been confirmed. These NPSH-containing substances accumulate around the urethral opening and are subsequently washed out with urine. Therefore, indirect detection of HPV infection by assessing NPSH levels in urine is feasible, but it has not been reported in detail so far. Here, an assay using phosphotungstic acid to oxidise and produce colour changes by NPSH in urine was developed. This assay enabled the rapid, non-invasive identification of HPV infection by detecting the metabolic byproduct NPSH produced by HPV-infected cells. Employing a smartphone-based device, developed using an ambient light sensor, reduces the cost and simplifies the operation associated with the colourimetric assay. The colourimetric assay was used to detect L-cysteine and L-ascorbic acid standard substance (as NPSH mimics), the limited of detection were 0.12 mM and 31.25 μM, respectively, with high reproducibility and stability. When this colourimetric assay was used to evaluate urine samples from individuals suspected of HPV infection, along with other at-home self-screening methods for HPV nucleic acid detection in urine, showed comparable sensitivity and specificity. Compared with nucleic acid detection in urine, this colourimetric assay is cost-effective, user-friendly, amenable to self-sampling, and enables testing at one's convenience and location of choice, which is more suitable for home self-testing or population self-screening.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Guangdong Province Engineering Research Center of Antibody Drug and Immunoassay, Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zhaoguang Wang
- Guangdong Province Engineering Research Center of Antibody Drug and Immunoassay, Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Jing Yang
- Guangdong Province Engineering Research Center of Antibody Drug and Immunoassay, Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yingying Li
- Center of Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Cheng Lu
- Guangdong Province Engineering Research Center of Antibody Drug and Immunoassay, Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yanqiang Hao
- Guangdong Women and Children Hospital, Guangzhou 510000, China
| | - Guanbo He
- Guangdong Zhongxin Biotech Limited, Guangzhou 510000, China
| | | | - Qifang Song
- Guangdong Province Engineering Research Center of Antibody Drug and Immunoassay, Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Jun Long
- Center of Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Jiajie Liang
- Guangdong Province Engineering Research Center of Antibody Drug and Immunoassay, Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; Research Center of Cancer Diagnosis and Therapy, Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China; Foshan Holly Biotech Co., Ltd, Foshan 528300, China; Guangdong Zhongxin Biotech Limited, Guangzhou 510000, China.
| | - Yong Tang
- Guangdong Province Engineering Research Center of Antibody Drug and Immunoassay, Department of Biological Sciences and Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; Research Center of Cancer Diagnosis and Therapy, Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China; Guangdong Zhongxin Biotech Limited, Guangzhou 510000, China.
| |
Collapse
|
9
|
Wen J, Cao X, Zhou B, Yang F, Wang X, Li Y, Zhao X, Mei J, Zhu W, Sun L, Huang F, Wang M. GC-MSCs transcriptionally upregulate SALL4 in gastric cancer through miR-4669/TIMP3/β-catenin signaling. Cell Signal 2025; 130:111668. [PMID: 39965736 DOI: 10.1016/j.cellsig.2025.111668] [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: 08/16/2024] [Revised: 12/07/2024] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUNDS Gastric cancer-associated mesenchymal stem cells (GC-MSCs) as integral components of the tumor microenvironment potentiate gastric cancer growth and metastasis. SALL4 is aberrantly upregulated in gastric cancer and pivotal for malignant progression. Whether GC-MSCs is responsible for SALL4 upregulation and the underlying mechanisms remains elusive. METHODS Cancer growth and metastasis capacities were assessed by cell colony formation assay, transwell assay, and epithelial-mesenchymal transition protein detection in vitro as well as subcutaneous xenograft and peritoneal metastasis models in vivo. SALL4 was measured by qPCR, western blot and immunohistochemistry staining. Gain- and loss-functional analysis were performed for miRNA and target gene. β-catenin signaling was assessed by immunofluorescence staining and Top/FopFlash luciferase assay. Transcriptional regulation was conducted using chemicals, luciferase reporter and ChIP assay. Clinical tissues and TCGA-STAD database were included for expression profile, correlation and clinical relevance analysis. RESULTS GC-MSCs promoted gastric cancer growth and metastasis along with elevation of SALL4 and miR-4669 in cancer cells and tissues. Overexpression of miR-4669 mimicked GC-MSC effects, while miR-4669 knockdown eliminated their oncogenic roles. TIMP3 was identified as a target of miR-4669 and mediated its functions. TIMP3 overexpression counteracted GC-MSC-induced cancer progression and SALL4 expression. GC-MSCs activated SALL4 transcription through the miR-4669/TIMP3/β-catenin pathway. The regulatory axis was aberrantly expressed in gastric cancer tissues, correlated with each other in certain cancer tissues and associated with lymph node metastasis. CONCLUSIONS GC-MSCs transcriptionally upregulate SALL4 to facilitate gastric cancer cell growth and metastasis via miR-4669/TIMP3/β-catenin pathway, highlighting the crucial role of GC-MSCs in the aberrant upregulation of SALL4.
Collapse
Affiliation(s)
- Jing Wen
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Xiaoli Cao
- Department of Laboratory Medicine, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu Province 226321, China
| | - Baocheng Zhou
- Department of Medical Laboratory, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu Province 222000, China
| | - Fang Yang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Xiang Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Yuanyuan Li
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Xinlan Zhao
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Jingyu Mei
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Wei Zhu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Li Sun
- Department of Clinical Laboratory, Kunshan First People's Hospital, Affiliated to Jiangsu University, Kunshan 215300, China
| | - Feng Huang
- Department of Clinical Laboratory, Kunshan First People's Hospital, Affiliated to Jiangsu University, Kunshan 215300, China; Department of Clinical Laboratory, Maternal and Child Health Care Hospital of Kunshan, Suzhou, Jiangsu Province, China
| | - Mei Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China.
| |
Collapse
|
10
|
Gou M, Zhang H, Qian N, Zhang Y, Sun Z, Li G, Wang Z, Dai G. Deep learning radiomics analysis for prediction of survival in patients with unresectable gastric cancer receiving immunotherapy. Eur J Radiol Open 2025; 14:100626. [PMID: 39807092 PMCID: PMC11728962 DOI: 10.1016/j.ejro.2024.100626] [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: 10/28/2024] [Revised: 12/03/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
Objective Immunotherapy has become an option for the first-line therapy of advanced gastric cancer (GC), with improved survival. Our study aimed to investigate unresectable GC from an imaging perspective combined with clinicopathological variables to identify patients who were most likely to benefit from immunotherapy. Method Patients with unresectable GC who were consecutively treated with immunotherapy at two different medical centers of Chinese PLA General Hospital were included and divided into the training and validation cohorts, respectively. A deep learning neural network, using a multimodal ensemble approach based on CT imaging data before immunotherapy, was trained in the training cohort to predict survival, and an internal validation cohort was constructed to select the optimal ensemble model. Data from another cohort were used for external validation. The area under the receiver operating characteristic curve was analyzed to evaluate performance in predicting survival. Detailed clinicopathological data and peripheral blood prior to immunotherapy were collected for each patient. Univariate and multivariable logistic regression analysis of imaging models and clinicopathological variables was also applied to identify the independent predictors of survival. A nomogram based on multivariable logistic regression was constructed. Result A total of 79 GC patients in the training cohort and 97 patients in the external validation cohort were enrolled in this study. A multi-model ensemble approach was applied to train a model to predict the 1-year survival of GC patients. Compared to individual models, the ensemble model showed improvement in performance metrics in both the internal and external validation cohorts. There was a significant difference in overall survival (OS) among patients with different imaging models based on the optimum cutoff score of 0.5 (HR = 0.20, 95 % CI: 0.10-0.37, P < 0.001). Multivariate Cox regression analysis revealed that the imaging models, PD-L1 expression, and lung immune prognostic index were independent prognostic factors for OS. We combined these variables and built a nomogram. The calibration curves showed that the C-index of the nomogram was 0.85 and 0.78 in the training and validation cohorts. Conclusion The deep learning model in combination with several clinical factors showed predictive value for survival in patients with unresectable GC receiving immunotherapy.
Collapse
Affiliation(s)
- Miaomiao Gou
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Hongtao Zhang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Niansong Qian
- Department of Thoracic Oncology, The Eighth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Yong Zhang
- Department of Medical Oncology, The Second Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Zeyu Sun
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Guang Li
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Zhikuan Wang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Guanghai Dai
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| |
Collapse
|
11
|
Javali PS, Thirumurugan K. Structural characterization of Aurora kinase B modulation by Epigallocatechin gallate: Insights from docking and dynamics simulations. J Mol Graph Model 2025; 136:108973. [PMID: 39914083 DOI: 10.1016/j.jmgm.2025.108973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 01/04/2025] [Accepted: 01/31/2025] [Indexed: 02/26/2025]
Abstract
Aurora Kinase B (AURKB) is crucial for chromosome alignment, segregation, and cytokinesis, phosphorylating essential proteins for accurate cell division. Mutations and overexpression of AURKB are common in various cancers. Inhibiting AURKB reduces therapy resistance, making it a promising therapeutic target. Synthetic inhibitors like AZD1152 and ZM447439 show selectivity for AURKB but often lack specificity due to high homology within the aurora kinase family. Conversely, natural molecules such as flavonoids offer selectivity, lower toxicity, and potential synergy with existing chemotherapies. Investigating natural AURKB inhibitors could lead to safer and more effective cancer treatments. Epigallocatechin-3-gallate (EGCG), a catechin ester in green tea, inhibits glioma cell line proliferation by inducing spontaneous apoptosis and reduces cancer cell invasiveness by decreasing metalloproteinase, cytokine, and chemokine activities. Additionally, EGCG inhibits several kinases, including PI3K, mTOR, EGFR, and AKT, acting as an effective ATP-competitive inhibitor. Thus, EGCG may enhance the efficacy of anti-cancer therapies as an AURKB inhibitor. This study used in silico tools to predict EGCG's pharmacodynamics and pharmacokinetics, and employed AutoDock for molecular docking with AURKB. The ligand-protein complex and Apo form of AURKB were simulated for 100 ns with GROMACS using the CHARM36 force field. Free energy surface analysis and MMPBSA methods confirmed the stability and spontaneity of EGCG binding to AURKB. The conformational dynamics of the DFG (Asp-Phe-Gly) motif in AURKB upon EGCG binding revealed significant changes crucial for ATP binding and kinase activity. The distance between the phenylalanine residue of the DFG motif and the αC helix in holo AURKB increased from 14.80 Å to 23.62 Å in the lowest free energy structure, indicating a shift from the DFG-in to the DFG-out state, affecting ATP binding. The study also noted transitions in the overall protein secondary structures, such as turn to coil, coil to sheet, and coil to helix, contributing to a stable structure upon EGCG binding. These findings highlight the complex interplay between EGCG and AURKB, providing insights into the conformational dynamics and structural alterations induced by this interaction, which has implications for reducing glioma cell chemosensitivity to therapeutic drugs.
Collapse
Affiliation(s)
- Prashanth S Javali
- Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India
| | - Kavitha Thirumurugan
- Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
12
|
Yang D, Yang C, Huang L, Guan M, Song C. Role of ubiquitination-driven metabolisms in oncogenesis and cancer therapy. Semin Cancer Biol 2025; 110:17-35. [PMID: 39929409 DOI: 10.1016/j.semcancer.2025.02.004] [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: 01/17/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025]
Abstract
Ubiquitination represents one of the most critical post-translational modifications, comprising a multi-stage enzyme process that plays a pivotal role in a myriad of cellular biological activities. The deregulation of the processes of ubiquitination and deubiquitination is associated with the development of cancers and other diseases. This typescript reviews the impact of ubiquitination on metabolic processes, elucidating the regulatory functions of ubiquitination on pivotal enzymes within metabolic pathways in pathological contexts. It underscores the role of ubiquitination-driven metabolism disorders in the etiology of cancers, and oncogenesis, and highlights the potential therapeutic efficacy of targeting ubiquitination-driven enzymes in cancer metabolism, their combination with immune checkpoint inhibitors, and their clinical applications.
Collapse
Affiliation(s)
- Dongqin Yang
- Department of Laboratory Medicine of Huashan Hospital, Fudan University, Shanghai 200040, China; Central Laboratory, Huashan Hospital, Fudan University, 12 Middle Urumuqi Road, Shanghai 200040, China
| | - Can Yang
- Department of Laboratory Medicine of Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Linlin Huang
- Central Laboratory, Huashan Hospital, Fudan University, 12 Middle Urumuqi Road, Shanghai 200040, China
| | - Ming Guan
- Department of Laboratory Medicine of Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Chunhua Song
- Division of Hematology, The Ohio State University Wexner Medical Center, the James Cancer Hospital, Columbus, OH 43210, USA.
| |
Collapse
|
13
|
Abu-Lafi S, Falah M, Zeidan M, Rayan M, Rayan A. Expanding paclitaxel's therapeutic window: Investigating the pharmacokinetic, clinical formulation and mechanistic aspects of paclitaxel‑lipoate conjugate. Oncol Lett 2025; 29:216. [PMID: 40093869 PMCID: PMC11907689 DOI: 10.3892/ol.2025.14962] [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: 03/21/2024] [Accepted: 08/14/2024] [Indexed: 03/19/2025] Open
Abstract
Paclitaxel (PTX) is among the most widely used antimicrotubular chemotherapy agents available from natural sources. It has a wide range of antitumor effectiveness, particularly against breast, ovarian and lung malignancies. IDD-1040 is a novel anticancer chemical conjugate that combines lipoic acid with PTX and demonstrates an anticancer efficiency superior to that of PTX alone. The aim of the present study was to investigate the analytical, formulation and pharmacokinetic aspects of IDD-1040, shedding light on its pharmacological behavior and the possible mechanisms underlying its enhanced anticancer activity. IDD-1040 was administered to mice as an intravenous bolus, and the pharmacokinetic parameters were determined over the following 7 days. The results revealed a total clearance of 1.689 l/h.kg, volume of distribution of 1.93 l/kg, average half-life of 1.14 h and terminal half-life of 8.64 h. Notably, the area under the curve of IDD-1040 was >14-fold higher than that of PTX, suggesting slower metabolism and that prodrug itself may have antitumor activity. An in vitro tubulin polymerization assay revealed distinct tubulin-binding characteristics for IDD-1040 compared with PTX. Due to the poor water solubility of IDD-1040, a formulation development experiment was conducted. In total, 31 formulations were prepared that became transparent when diluted with water. In addition, some formulations achieved a relatively high drug content (12 mg/g) without the use of surfactants. Moreover, they included fewer excipients compared with the formulations diluted with water, suggesting a promising approach for drug formulation. In summary, IDD-1040 exhibited extended circulation, efficient tissue distribution and reduced metabolite formation in vitro, warranting further exploration of its mechanisms of action and therapeutic potential. Future studies are recommended to assess the stability, pharmacokinetics and pharmacodynamics of these refined IDD-1040 formulations to gauge their suitability for clinical application.
Collapse
Affiliation(s)
- Saleh Abu-Lafi
- Faculty of Pharmacy, Al-Quds University, Abu-Deis 144, Palestine
| | - Mizied Falah
- Research Center, Holy Family Hospital, Nazareth 1600000, Israel
| | - Mahmoud Zeidan
- Molecular Genetics and Virology Laboratory, Qasemi Research Center, Faculty of Science, Al-Qasemi Academic College, Baka El-Garbiah 3010000, Israel
| | - Mouhammad Rayan
- Geriatric Department, Holy Family Hospital, Nazareth 16000, Israel
| | - Anwar Rayan
- Department of Science and Technology, Faculty of Science, Al-Qasemi Academic College, Baka El-Garbiah 3010000, Israel
| |
Collapse
|
14
|
Ferrato LJ, Ruiz TFR, de Souza LG, Brito-Filho GE, Colleta SJ, Leonel ECR, Taboga SR. Bisphenol A exposure alters hormonal modulation and responsivity in the prostate of aged female gerbils. Mol Cell Endocrinol 2025; 600:112507. [PMID: 39988023 DOI: 10.1016/j.mce.2025.112507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
The female prostate is a gland regulated by steroid hormones for homeostasis. Bisphenol A (BPA) is an endocrine disruptor related to the progression of malignant lesions in prostate. The aim of this study was to analyze the hormonal modulation and responsiveness of the prostate of aged female gerbils previously exposed to BPA. Females were exposed to 50 μg/kg/day during pregnancy and lactation, and the prostate was analyzed at 18 months of age. Control groups were included to normalize the analysis. The samples were analyzed using histological and immunohistochemical techniques for the expression of androgen (AR), estrogen (ERα and ERβ), prolactin (PRL), and progesterone (PR) receptors, as well as steroidogenic enzymes (5α-reductase and aromatase) and the proliferation (PHH3) and epigenetic (EZH2) markers. Protein quantification was also performed for the receptors and enzymes described, as well as morphological and morphometric analyses of the gland. The results showed an increase in the epithelial expression of AR and ERα and a decrease in the expression of ERβ in this compartment. In addition, a decrease in epithelial expression was observed for the 5α-reductase and aromatase in the prostate epithelium and an increase in the expression of PHH3 and EZH2. By Western blot analyses, significant differences were observed in the protein quantification of the receptors and enzymes described. Thus, the current study showed the role of BPA in intraprostatic hormonal modulation, through alterations in the expression of hormone receptors and the conversion of enzymes in the female prostate, which can lead to the progression of malignant lesions in this tissue.
Collapse
Affiliation(s)
- Luara Jesus Ferrato
- Microscopy and Microanalysis Center, Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University - UNESP, SP, Brazil.
| | - Thalles Fernando Rocha Ruiz
- Microscopy and Microanalysis Center, Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University - UNESP, SP, Brazil; Department of Structural and Functional Biology, Institute of Biology, State University of Campinas -UNICAMP, SP, Brazil.
| | - Lorena Gabriela de Souza
- Microscopy and Microanalysis Center, Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University - UNESP, SP, Brazil
| | - Gervásio Evangelista Brito-Filho
- Microscopy and Microanalysis Center, Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University - UNESP, SP, Brazil
| | - Simone Jacovaci Colleta
- Microscopy and Microanalysis Center, Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University - UNESP, SP, Brazil
| | - Ellen Cristina Rivas Leonel
- Department of Histology, Embryology and Cell Biology, Institute of Biological Sciences (ICB III), Federal University of Goiás - UFG, Goiânia, GO, Brazil; Animal Molecular and Cellular Biology, Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Sebastião Roberto Taboga
- Microscopy and Microanalysis Center, Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University - UNESP, SP, Brazil.
| |
Collapse
|
15
|
Qi SJ, Liu KX, Liu HY, Zhang GQ, Zheng HB, Sun JY, Sun B, Lou HX. Discovery of potent quinone oxidoreductase 2 inhibitors to overcome TRAIL resistance of non-small cell lung cancer. Eur J Med Chem 2025; 288:117382. [PMID: 39987833 DOI: 10.1016/j.ejmech.2025.117382] [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: 12/11/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 02/25/2025]
Abstract
Resistance to tumor-necrosis-factor-related apoptosis-inducing ligand (TRAIL) of cancer cells is a main obstacle for the chemotherapy. NRH: quinone oxidoreductase 2 (NQO2), known as a chemopreventive target, has emerged as a promising therapeutic target for overcoming TRAIL resistance in non-small cell lung cancer (NSCLC). Here we report the design, synthesis and evaluation of resveratrol analogues as novel selective NQO2 inhibitors, and analogue 20b, with potent NQO2 inhibitory activity (IC50 = 95 nM) and relatively low cytotoxicity, displayed synergistic lethal effects in combination with TRAIL on TRAIL-resistant NSCLC cells. In addition, mechanistic studies revealed that 20b sensitized TRAIL-resistant A549 cells to apoptosis through the generation of reactive oxygen species (ROS) and the upregulation of death receptor 5 (DR5). Furthermore, 20b showed no acute toxicity in the healthy mice at a single dose of 2000 mg/kg. Molecular docking confirmed the binding mode of 20b within the NQO2 active site, highlighting key interactions responsible for its enhanced potency. This study provided novel molecular templates for development of NQO2 inhibitor, and laid a foundation for developing agents against TRAIL-resistant cancers for targeting NQO2.
Collapse
Affiliation(s)
- Si-Jie Qi
- National Glycoengineering Research Center, Shandong University, Qingdao, 266237, PR China
| | - Ke-Xin Liu
- National Glycoengineering Research Center, Shandong University, Qingdao, 266237, PR China
| | - Hao-Yu Liu
- National Glycoengineering Research Center, Shandong University, Qingdao, 266237, PR China
| | - Gao-Qiang Zhang
- National Glycoengineering Research Center, Shandong University, Qingdao, 266237, PR China
| | - Hong-Bo Zheng
- Key Laboratory of Natural Products & Chemical Biology, Ministry of Education, Shandong University, Jinan, 250012, PR China
| | - Jia-Yu Sun
- National Glycoengineering Research Center, Shandong University, Qingdao, 266237, PR China
| | - Bin Sun
- National Glycoengineering Research Center, Shandong University, Qingdao, 266237, PR China.
| | - Hong-Xiang Lou
- Key Laboratory of Natural Products & Chemical Biology, Ministry of Education, Shandong University, Jinan, 250012, PR China.
| |
Collapse
|
16
|
Watling CZ, Kelly RK, Watts EL, Graubard BI, Petrick JL, Matthews CE, McGlynn KA. Total testosterone, sex hormone-binding globulin, and free testosterone concentrations and risk of primary liver cancer: A prospective analysis of 200,000 men and 180,000 postmenopausal women. Int J Cancer 2025; 156:1518-1528. [PMID: 39499225 PMCID: PMC11826138 DOI: 10.1002/ijc.35244] [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: 08/27/2024] [Revised: 10/07/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024]
Abstract
In most countries, males have ~2-3 times higher incidence of primary liver cancer than females. Sex hormones have been hypothesized to contribute to these differences, but the evidence remains unclear. Using data from the UK Biobank, which included ~200,000 males and ~180,000 postmenopausal females who provided blood samples at recruitment, we estimated hazard ratios (HR2) and 95% confidence intervals (CI) for a doubling in hormone concentration from multivariable adjusted Cox regression for circulating total testosterone, sex-hormone binding globulin (SHBG), and free testosterone concentrations and risk of primary liver cancer. After a median of 11.8 years of follow-up, 531 cases of primary liver cancer were observed, of which 366 occurred in males and 165 occurred in females. Total testosterone and SHBG were shown to be positively associated with liver cancer risk in both males and females (Total testosterone HR2: 3.42, 95% CI:2.42-4.84 and 1.29, 0.97-1.72, respectively; SHBG HR2: 5.44, 4.42-6.68 and 1.52, 1.09-2.12, respectively). However, free testosterone was inversely associated with primary liver cancer in males (HR2: 0.42, 0.32-0.55) and no association was observed in females. When analyses compared two main liver cancer subtypes, hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC), there was evidence of heterogeneity; associations for total testosterone and SHBG concentrations were only positively associated with HCC in both males (HR2: 3.56, 2.65-4.79 and 7.72, 6.12-9.73, respectively) and females (HR2: 1.65, 1.20-2.27 and 6.74, 3.93-11.5, respectively) but not with ICC. Further research understanding the mechanisms of how sex-steroids may influence liver cancer risk is needed.
Collapse
Affiliation(s)
- Cody Z. Watling
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Rebecca K. Kelly
- The George Institute for Global HealthUniversity of New South WalesSydneyNew South WalesAustralia
- School of Medicine, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Eleanor L. Watts
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Barry I. Graubard
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | | | - Charles E. Matthews
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Katherine A. McGlynn
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| |
Collapse
|
17
|
Li C, Cheng S, Yu J, Zheng Q, Yu G, Xu M, Meng X, Zeng X, Liu K, Xu B, Luo H, Xu G. Hit to lead optimization of the 4-trifluoromethylquinoline derivatives as novel SGK1 inhibitors with potent anti-prostate cancer activity. Eur J Med Chem 2025; 287:117336. [PMID: 39908792 DOI: 10.1016/j.ejmech.2025.117336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/19/2025] [Accepted: 01/25/2025] [Indexed: 02/07/2025]
Abstract
Prostate cancer (PCa) remains a significant health concern for males, and serum/glucocorticoid-regulated kinase-1 (SGK1) plays a crucial role in its pathogenesis. This provides a promising target for the development of novel therapies against PCa. Herein, we reported the structural optimization of the hit compound H1, which was discovered in our previous work as an SGK1 inhibitor. Based on docking research for the active binding conformation of compound H1, a series of novel 4-trifluoromethyl quinoline derivatives were developed by replacing the 6-methoxy group in the quinoline skeleton of compound H1 with a larger aryl ring to occupy the hinge region of SGK1. Among them, compound 12f showed the strongest SGK1 inhibitory potency, with an IC50 value of 0.39 μM, representing a 7.8-fold improvement over compound H1. Molecular docking studies revealed that the 6-methoxyphenylamine moiety of compound 12f effectively extends into the hinge region of SGK1, establishing a crucial hydrogen bonding interaction with Glu183 that enhances its biological potency. In vivo, compound 12f effectively suppressed tumor growth in the PC3 xenograft model in BALB/c nude mice without inducing any observable toxicity. Moreover, mechanistic studies showed that compound 12f hindered PC3 cell migration and invasion, improved the thermal stability of SGK1 protein in PC3 cells, decreased SGK1 protein levels in tumor tissues, and effectively inhibited the phosphorylation of SGK1 and its substrates in PC3 cells in a dose- and time-dependent manner. In summary, the results of this study highlight the potential of 12f as a lead compound for further optimization in the development of new therapies against PCa targeting SGK1.
Collapse
Affiliation(s)
- Cheng Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China; Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Sha Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Qian Zheng
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Gang Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Mei Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Xueling Meng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Xiaoping Zeng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Kun Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Bixue Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China.
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China.
| | - Guangcan Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, China.
| |
Collapse
|
18
|
Qi J, Cheng H, Su L, Li J, Cheng F. A novel exosome-related prognostic risk model for thyroid cancer. Asia Pac J Clin Oncol 2025; 21:190-198. [PMID: 38577908 DOI: 10.1111/ajco.14063] [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: 12/24/2023] [Revised: 02/13/2024] [Accepted: 03/20/2024] [Indexed: 04/06/2024]
Abstract
AIM The aim was to build an exosome-related gene (ERG) risk model for thyroid cancer (TC) patients. METHODS Note that, 510 TC samples from The Cancer Genome Atlas database and 121 ERGs from the ExoBCD database were obtained. Differential gene expression analysis was performed to get ERGs in TC (TERGs). Functional enrichment analyses including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were conducted on the TERGs. Then we constructed a model based on LASSO Cox regression analysis. Kaplan-Meier survival analysis was applied and a Nomogram model was also built. The immune landscape was evaluated by CIBERSORT. RESULTS Thirty-eight TERGs were identified and their functions were enriched on 591 GO terms and 30 KEGG pathways. We built a Risk Score model based on FGFR3, ADRA1B, and POSTN. Risk Scores were significantly higher in T4 than in other stages, meanwhile, it didn't significantly differ in genders and TNM N or M classifications. The nomogram model could reliably predict the overall survival of TC patients. The mutation rate of BRAF and expression of cytotoxic T-lymphocyte-associated protein 4 were significantly higher in the high-risk group than in the low-risk group. The risk score was significantly correlated to the immune landscape. CONCLUSION We built a Risk Score model using FGFR3, ADRA1B, and POSTN which could reliably predict the prognosis of TC patients.
Collapse
Affiliation(s)
- Junfeng Qi
- Department of Ultrasound, Wuwei People's Hospital, Wuwei, China
| | - Hanshan Cheng
- Department of Ultrasound, Wuwei People's Hospital, Wuwei, China
| | - Long Su
- Department of Ultrasound, Wuwei People's Hospital, Wuwei, China
| | - Jun Li
- Department of Ultrasound, Wuwei People's Hospital, Wuwei, China
| | - Fei Cheng
- Department of Surgical Oncology, Wuwei People's Hospital, Wuwei, China
| |
Collapse
|
19
|
Lorenzoni S, Rodríguez-Nogales C, Blanco-Prieto MJ. Targeting tumor microenvironment with RGD-functionalized nanoparticles for precision cancer therapy. Cancer Lett 2025; 614:217536. [PMID: 39924081 DOI: 10.1016/j.canlet.2025.217536] [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: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
The need for precision therapies arises from the complexities associated with high-risk types of cancer, due to their aggressiveness and resistance to treatment. These diseases represent a global issue that requires transversal strategies involving cooperation among oncology specialists and experts from related fields, including nanomedicine. Nanoparticle-mediated active targeting of tumors has proven to be a revolutionary approach to address the most challenging neoplasms by overcoming the poor permeation at tumor site of untargeted, and nowadays questioned, strategies that rely solely on Enhanced Permeability and Retention (EPR) effects. The decoration of nanoparticles with Arg-Gly-Asp (RGD) peptides, which selectively target integrins on the cell membrane, marks a turning point in tumor microenvironment (TME) targeted strategies, enabling precision and efficiency in the delivery of chemotherapeutics. This review delves into the intricacies of the TME's features and targetable components (i.e. integrins), and the development of RGDs for nanoparticles' functionalization for active TME targeting. It provides a translational perspective on the integration of RGD-functionalized nanoparticles in oncology, highlighting their potential to overcome current therapeutic challenges, particularly in precision medicine. The current landscape of targeted nanomedicines in the clinic, and the development of RGD-nanomedicine for pediatric cancers are also discussed.
Collapse
Affiliation(s)
- Sara Lorenzoni
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, Pamplona, 31008, Pamplona, Spain; Cancer Center Clínica Universidad de Navarra (CCUN), Avenida Pio XII 36, 31008, Pamplona, Spain
| | - Carlos Rodríguez-Nogales
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, Pamplona, 31008, Pamplona, Spain; Cancer Center Clínica Universidad de Navarra (CCUN), Avenida Pio XII 36, 31008, Pamplona, Spain.
| | - María J Blanco-Prieto
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, Pamplona, 31008, Pamplona, Spain; Cancer Center Clínica Universidad de Navarra (CCUN), Avenida Pio XII 36, 31008, Pamplona, Spain.
| |
Collapse
|
20
|
Yang J. Unveiling the multifaceted roles of long non-coding RNA CTBP1-DT in human diseases: Special attention to its microprotein-encoding potential. Pathol Res Pract 2025; 268:155870. [PMID: 40020329 DOI: 10.1016/j.prp.2025.155870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/30/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
C-terminal binding protein 1 divergent transcript (CTBP1-DT) is a novel long non-coding RNA (lncRNA) located on human chromosome 4p16.3. Numerous studies have shown that CTBP1-DT plays a critical regulatory role in various human malignancies and non-malignant diseases. In several cancers, the expression of CTBP1-DT is upregulated, closely associated with the risk of 12 types of cancer, and strongly correlated with the clinical pathological features and poor prognosis of 10 of these cancers. Mechanistically, CTBP1-DT is stimulated by the transcription factors ETV5 and Sp1, or methylated by YTHDC1. By competitively inhibiting 12 microRNAs, it activates 3 signaling pathways that influence malignant behaviors of tumor cells, including proliferation, apoptosis, cell cycle arrest, migration, invasion, immune evasion, and chemoresistance. Importantly, it also encodes the microprotein DNA damage up-regulated protein (DDUP), which mediates cisplatin resistance through sustained response to DNA damage signals. Furthermore, CTBP1-DT has been implicated in the progression of non-malignant diseases such as diabetes and related conditions, cardiovascular diseases, and osteoarthritis. This review summarizes the latest research on the RNA and protein functions of CTBP1-DT in human diseases, outlines various molecular regulatory networks centered around CTBP1-DT, and discusses the opportunities and challenges of its clinical applications.
Collapse
Affiliation(s)
- Jingjie Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, China.
| |
Collapse
|
21
|
Wang L, ChenLiu Z, Wang D, Tang D. Cross-talks of GSH, mitochondria, RNA m6A modification, NRF2, and p53 between ferroptosis and cuproptosis in HCC: A review. Int J Biol Macromol 2025; 302:140523. [PMID: 39894098 DOI: 10.1016/j.ijbiomac.2025.140523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor with high morbidity and mortality, as well as poor prognosis. Therefore, it is imperative to explore alternative therapeutic targets for HCC treatment. Ferroptosis and cuproptosis have recently been identified as metal-dependent cell death mechanisms that play significant roles in HCC treatment. This study identified potential cross-talk between ferroptosis and cuproptosis, including the common hub glutathione, common site of occurrence, mitochondria, shared epigenetic modification mode, RNA N6 methyladenosine modification, mutual inhibitor, nuclear factor erythroid 2-related factor 2, and dual regulator, p53. These findings provide a theoretical foundation for the joint induction of HCC cell death and effective inhibition of HCC progression. However, some immune cells are susceptible to ferroptosis or cuproptosis, which may impair or enhance anti-cancer immune function. We propose strategies to target specific targets molecules such as tripartite motif containing 25, ferroptosis suppressor protein 1, and peroxisome proliferator-activated receptor gamma or exploit the unique acidic environment surrounding cancer cells to precisely induce ferroptosis in cancer cells. This approach aims to advance the development of precision medicine for HCC treatment.
Collapse
Affiliation(s)
- Leihan Wang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Zhenni ChenLiu
- Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou School of Clinical Medicine of Dalian Medical University, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou 225000, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou School of Clinical Medicine of Dalian Medical University, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou 225000, China.
| |
Collapse
|
22
|
Liang X, Tan X, Pei L, Dong C. circDHX33 suppresses glycolysis, malignant proliferation, and metastasis in prostate cancer by interacting with RNA-binding protein IGF2BP2 to destabilize its protein. Cytotechnology 2025; 77:56. [PMID: 39927135 PMCID: PMC11802939 DOI: 10.1007/s10616-025-00718-6] [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: 11/20/2024] [Accepted: 01/20/2025] [Indexed: 02/11/2025] Open
Abstract
Prostate cancer (PCa) is a malignant tumor characterized by dependence on androgens and enhanced glycolytic processes in response to the energy demands of rapid proliferation. This study delved into the role of circDHX33 interacting with IGF2BP2 in regulating the malignant behavior of PCa. circRNA expression data from PCa tissues and normal tissues were selected from the GEO database, and differentially expressed circRNAs were screened out. circDHX33 expression in clinical PCa samples was verified by RT-qPCR. Cellular experiments included cell culture, RNA interference and overexpression assays, as well as the use of Transwell migration invasion assay and EdU cell proliferation assay to assess the effect of circDHX33 on the proliferation and migration of PC-3 cells. In addition, its regulatory effect on energy metabolism in tumor cells was assessed by glycolysis assay. FISH assay, RNA pull-down, silver staining assay, and RIP were used to evaluate the interaction between circDHX33 and IGF2BP2. circDHX33 expression was significantly reduced in PCa tissues relative to normal tissues. Overexpression of circDHX33 significantly inhibited the glycolytic activity, proliferative capacity, and migratory and invasive abilities of PC-3 cells, and this effect was closely related to its reduction of IGF2BP2 protein stability. Knockdown of IGF2BP2 could reverse the malignant behavior of cells enhanced by circDHX33 knockdown. In addition, the direct intracellular interaction between circDHX33 and IGF2BP2 was verified. circDHX33 inhibits glycolysis and malignant proliferation in PCa through interaction with IGF2BP2, suggesting its potential as a potential therapeutic target.
Collapse
Affiliation(s)
- XiangDong Liang
- Department of Urology Surgery, The Fourth Hospital of Hebei Medical University, No. 12, JianKang Road, Chang’an, Shijiazhuang, 050000 Hebei China
| | - XiaoLiang Tan
- Department of Urology Surgery, The Fourth Hospital of Hebei Medical University, No. 12, JianKang Road, Chang’an, Shijiazhuang, 050000 Hebei China
| | - Long Pei
- Department of Urology Surgery, The Fourth Hospital of Hebei Medical University, No. 12, JianKang Road, Chang’an, Shijiazhuang, 050000 Hebei China
| | - ChunHui Dong
- Department of Urology Surgery, The Fourth Hospital of Hebei Medical University, No. 12, JianKang Road, Chang’an, Shijiazhuang, 050000 Hebei China
| |
Collapse
|
23
|
Chan N, Madani D, Allan RS, Sy J, Satgunaseelan L. Hidden identities in plurihormonal pituitary neuroendocrine tumours: expanding the spectrum of the 'silent corticogonadotroph adenoma'. Pathology 2025; 57:385-387. [PMID: 39632231 DOI: 10.1016/j.pathol.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/18/2024] [Accepted: 08/25/2024] [Indexed: 12/07/2024]
Affiliation(s)
- Noni Chan
- Department of Neuropathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Daniel Madani
- Department of Neurosurgery, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Rodney S Allan
- Department of Neurosurgery, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Joanne Sy
- Department of Neuropathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Laveniya Satgunaseelan
- Department of Neuropathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
24
|
Chai Y, Zhang J, Shao W, Zhang Z. Single-cell insights into HNSCC tumor heterogeneity and programmed cell death pathways. Transl Oncol 2025; 54:102341. [PMID: 40068384 DOI: 10.1016/j.tranon.2025.102341] [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: 09/30/2024] [Revised: 01/23/2025] [Accepted: 02/27/2025] [Indexed: 03/18/2025] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a prevalent malignancy often diagnosed in advanced stages. Despite advancements in therapy, it retains a high mortality rate and significant recurrence risk. This study utilizes single-cell sequencing (scRNA-seq) to unravel HNSCC's complexity, identify therapeutic targets, and refine prognostic models. METHODS Pseudotime trajectory and stemness analyses were performed on HNSCC tumor subpopulations, focusing on the C2 MALAT1+ Tumors subpopulation, which had the lowest CytoTRACE Score and represented the Lineage 2 endpoint in Slingshot analysis. The study examined programmed death and metabolic pathways in each subpopulation and developed a novel prognostic model using LASSO regression. RESULTS The C2 MALAT1+ Tumors subpopulation exhibited reduced expression of programmed death pathways (e.g., Entotic cell death, Apoptosis, Pyroptosis) and metabolic pathways (e.g., Riboflavin metabolism, Glycolysis/Gluconeogenesis). Key transcription factors included LEF1, RFX3, CREM, MZF1, and ZNF202. Prognostic models based on the MALAT1 Tumors Risk Score (MTRS) revealed worse survival and higher tumor purity in the high MTRS group. Risk genes included ADM, RPL31, EIF5B, and TAF7. Additionally, activated CD4 memory T cells were enriched in the high MTRS group, which also showed greater sensitivity to Cisplatin, Docetaxel, and Paclitaxel. CONCLUSIONS ScRNA-seq revealed the heterogeneity of HNSCC subpopulations, highlighting the unique features of the C2 MALAT1+ Tumors subpopulation. This study identified novel prognostic markers and therapeutic targets, offering insights into HNSCC progression, drug resistance, and potential treatments.
Collapse
Affiliation(s)
- Yuanhao Chai
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; University of Southern California, Los Angeles, USA
| | - Jianlin Zhang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenwen Shao
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ziwei Zhang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
25
|
Fu Z, Xin X, Zhan Y, Fan X, Li X, Chen T, Wang X. GPER agonist G-1 activates YAP to induce apoptosis in breast cancer cells. J Steroid Biochem Mol Biol 2025; 248:106693. [PMID: 39914681 DOI: 10.1016/j.jsbmb.2025.106693] [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: 10/20/2024] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 02/26/2025]
Abstract
G-1, a G protein-coupled estrogen receptor (GPER)-specific agonist, exhibits anticancer potential in breast cancer cells. This study aims to explore the molecular basis of apoptosis induced by G-1 in MCF-7 and MDA-MB-231 breast cancer cells. Here, we found that G-1 induced cytotoxicity and GPER-dependent apoptosis with PARP cleavage and mitochondrial membrane potential (MMP) loss, as well as nuclear condensation. Fluorescence resonance energy transfer (FRET) analysis in living cells indicated that G-1 effectively disrupted the interaction between large tumor suppressor 1/2 (LATS1/2) and Yes-associated protein (YAP). Furthermore, G-1 reduced YAP phosphorylation levels and promoted its nuclear accumulation. Notably, knockdown of YAP attenuated G-1-induced apoptosis, highlighting the crucial role of YAP in this process. Additionally, FRET analysis revealed that G-1 enhanced the binding of YAP to p73, leading to an increase in Bcl-2-associated X protein (Bax) expression and an induction of apoptosis. In summary, our findings demonstrate that G-1 induces apoptosis through the GPER/YAP/p73-mediated pathway.
Collapse
Affiliation(s)
- Ze Fu
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Xin Xin
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yongtong Zhan
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Xuhong Fan
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Xin Li
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Tongsheng Chen
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Xiaoping Wang
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| |
Collapse
|
26
|
Huang M, Zhu X, Xu W, Zhu J, Xun X, Su B, Chen H. TTC7A-ALK, a novel ALK fusion variant identified in a patient with metastatic lung adenocarcinoma, exhibits excellent response to crizotinib. Transl Oncol 2025; 54:102345. [PMID: 40054123 DOI: 10.1016/j.tranon.2025.102345] [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: 08/28/2024] [Revised: 02/20/2025] [Accepted: 03/02/2025] [Indexed: 03/18/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. ALK gene rearrangement has been identified in 3 % to 5 % of the patients with NSCLC. Thanks to the advancements in second-generation sequencing technology, an increasing number of novel fusion partners have been identified. In our research, we discovered a rare ALK fusion, TTC7A-ALK, in a patient with advanced lung adenocarcinoma using targeted next-generation sequencing (NGS). After being diagnosed with advanced lung adenocarcinoma with TTC7A-ALK fusion, the patient received crizotinib treatment and achieved a progression-free survival of 29 months. Additonanlly, we conducted further functional analyses on this fusion protein to assess its oncogenic potential. Similar to EML4-ALK, the TTC7A-ALK fusion protein can promote the growth of Ba/F3 cells under IL-3-independent conditions in vitro. In vivo studies demonstrate that the TTC7A-ALK fusion protein could enhance the tumorigenesis of NIH3T3 cells in nude mice, which can be suppressed by crizotinib. Mechanistic studies indicated that the ectopic expression of TTC7A-ALK in 293T cells led to the hyperactivation of downstream MAPK and PI3K/Akt pathways, which can be inhibited by crizotinib. Furthermore, upon tumor progression, the patient transitioned to alectinib, which provided rapid symptom relief and controlled the majority of lesions. Conclusionly, we identified and validated TTC7A-ALK as a oncogenic fusion in NSCLC. The patient demonstrated a significant clinical benefit from sequential treatment with crizotinib and alectinib, highlighting TTC7A-ALK as a novel therapeutic target for ALK inhibitors. These findings extend the spectrum of actionable ALK fusions and promote the inclusion of rare fusion detection in clinical diagnostic processes and treatment strategies.
Collapse
Affiliation(s)
- Meijin Huang
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China
| | - Xiangqing Zhu
- Department of Basic Medical Laboratory, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China
| | - Wenmang Xu
- Department of Pathology, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China
| | - Jun Zhu
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China
| | - Xin Xun
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China
| | - Bin Su
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China
| | - Hong Chen
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China.
| |
Collapse
|
27
|
Yang H, Xiong R, Zhang R, Sun S, Pan Y, Zhao Q, Bie J, Luo Y, Song G, Liu K. LINC01305 and LAD1 Co-Regulate CTTN and N-WASP Phosphorylation, Mediating Cytoskeletal Reorganization to Promote ESCC Metastasis. Mol Carcinog 2025; 64:756-768. [PMID: 39835575 PMCID: PMC11890417 DOI: 10.1002/mc.23885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/29/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is prone to metastasis and is a leading cause of mortality. The cytoskeleton is closely related to cell morphology and movement; however, little research has been conducted on ESCC metastasis. In this study, we found that the anchoring filament protein ladinin 1 (LAD1) specifically binds to LINC01305 for co-regulating the level of modulating cortactin proteins (CTTN) and neuronal Wiskott-Aldrich syndrome protein (N-WASP) phosphorylation, which mediates cytoskeletal reorganization and affects the metastasis of ESCC cells. Additionally, LINC01305 and LAD1 jointly promoted the epithelial-mesenchymal transition (EMT) process by activating the phosphoinositide-3-kinase-protein kinase B (PI3K/AKT) signaling pathway. Moreover, LINC01305 and LAD1 were related to the late clinical stage and lymph node metastasis of ESCC. Our study demonstrated that LINC01305 and LAD1 are major determinants of ESCC dissemination and revealed a novel molecular mechanism of cytoskeletal reorganization that controls ESCC metastasis. Trial Registration: N/A.
Collapse
Grants
- This work was supported by the National Institutes of the National Natural Science Foundation of China (82203851), the Sichuan Science and Technology Program (2024NSFSC1973, 2023YFS0473, MZGC20240072, MZGC20240071), the Nanchong Science and Technology Program (22SXQT0336, 20SXQT0328, 22SXQT0087, 22SXQT0340, 20SXQT0181), the Sichuan Medical Association Science and Technology Program (S2024009), and the North Sichuan Medical College Science and Technology Program (CBY24-KP03, CBY22-QDA01).
Collapse
Affiliation(s)
- Hang Yang
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical CollegeNanchongChina
| | - Rong Xiong
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Ruolan Zhang
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Shan Sun
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical CollegeNanchongChina
| | - Yingjie Pan
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical CollegeNanchongChina
| | - Quanneng Zhao
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Jun Bie
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Yi Luo
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Guiqin Song
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical CollegeNanchongChina
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| |
Collapse
|
28
|
Laplante P, Rosa R, Nebot-Bral L, Goulas J, Pouvelle C, Nikolaev S, Silvin A, Kannouche PL. Effect of MisMatch repair deficiency on metastasis occurrence in a syngeneic mouse model. Neoplasia 2025; 62:101145. [PMID: 39985912 PMCID: PMC11905862 DOI: 10.1016/j.neo.2025.101145] [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: 12/20/2024] [Revised: 02/08/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Mismatch repair deficiency leads to high mutation rates and microsatellite instability (MSI-H), associated with immune infiltration and responsiveness to immunotherapies. In early stages, MSI-H tumors generally have a better prognosis and lower metastatic potential than microsatellite-stable (MSS) tumors, especially in colorectal cancer. However, in advanced stages, MSI-H tumors lose this survival advantage for reasons that remain unclear. We developed a syngeneic mouse model of MSI cancer by knocking out the MMR gene Msh2 in the metastatic 4T1 breast cancer cell line. This model mirrored genomic features of MSI-H cancers and showed reduction in metastatic incidence compared to their MSS counterparts. In MSI-H tumors, we observed an enrichment of immune gene-signatures that negatively correlated with metastasis incidence. A hybrid epithelial-mesenchymal signature, related to aggressiveness was detected only in metastatic MSI-H tumors. Interestingly, we identified immature myeloid cells at primary and metastatic sites in MSI-H tumor-bearing mice, suggesting that MMR deficiency elicits specific immune responses beyond T-cell activation.
Collapse
Affiliation(s)
- Pierre Laplante
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Reginaldo Rosa
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Laetitia Nebot-Bral
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Jordane Goulas
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Caroline Pouvelle
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Sergey Nikolaev
- Paris-Saclay Université, Inserm-U981, Gustave Roussy, Villejuif, France
| | - Aymeric Silvin
- Paris-Saclay Université, Inserm-U1015, Gustave Roussy, Villejuif, France
| | - Patricia L Kannouche
- Paris-Saclay Université, CNRS-UMR9019, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France.
| |
Collapse
|
29
|
Zhang S, Li X, Xiahou Z, Chen A, Sun R, Liu C, Yuan J. Discovering the Potential Role of the C2 DUSP2+ MCs Subgroup in Lung Adenocarcinoma. Transl Oncol 2025; 54:102295. [PMID: 40014976 PMCID: PMC11910677 DOI: 10.1016/j.tranon.2025.102295] [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: 11/10/2024] [Revised: 12/19/2024] [Accepted: 01/21/2025] [Indexed: 03/01/2025] Open
Abstract
OBJECTIVE In both industrialized and developing nations worldwide, lung adenocarcinoma is one of the deadliest malignant tumors and the primary cause of cancer-related deaths. Its cellular heterogeneity is unclear to the fullest extent, although in recent years, its prevalence in younger individuals has increased. Therefore, it is urgent to deepen the understanding of lung adenocarcinoma and explore new therapeutic methods. METHODS CytoTRACE, Monocle, SCENIC, and enrichment analysis were used to analyze the single cell RNA data, we characterized the biological characteristics of mast cells (MCs) in lung adenocarcinoma patient samples. CellChat was used to analyze and validate the interaction between MCs and tumor cells in lung adenocarcinoma. Prognostic models were used to evaluate and predict the development trend and outcome of a patient's disease, such as the survival time of cancer patients. The python package SCENIC was used to evaluate the enrichment of transcription factors and the activity of regulators in lung adenocarcinoma cell subgroups. CCK-8 assay could validate the activity of a specific cell subgroup sequenced in single cell sequencing to confirm the role of this cell subgroup in tumor proliferation. RESULTS Our analysis identified seven major cell types, further grouping MCs within them and identifying four distinct subgroups, including MCs with high DUSP2 expression, which showed some tumor-related characteristics. In addition, we identified the key signaling receptor EGFR and validated it through in vitro knockdown experiments, demonstrating its role in promoting cancer. In addition, we established an independent prognostic indicator, the DUSP2+ MCs risk score, which showed an association between groups with high risk scores and poor outcomes. CONCLUSION These findings shed light on the complex interactions in the lung adenocarcinoma tumor microenvironment and suggest that targeting specific MCs subgroups, particularly through the EGFR signaling pathway, may provide new therapeutic strategies.
Collapse
Affiliation(s)
- Shengyi Zhang
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
| | - Xinhan Li
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Zhikai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China.
| | - Ailing Chen
- Quality Control Department, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
| | - Renfang Sun
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
| | - Chao Liu
- Department of Orthopaedics, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jie Yuan
- Sijing Town Community Healthcare Center, Shanghai, China.
| |
Collapse
|
30
|
Varlamova EG. Roles of selenium-containing glutathione peroxidases and thioredoxin reductases in the regulation of processes associated with glioblastoma progression. Arch Biochem Biophys 2025; 766:110344. [PMID: 39956249 DOI: 10.1016/j.abb.2025.110344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Glioblastoma remains the most common and aggressive primary tumor of the central nervous system in adults. Current treatment options include standard surgical resection combined with radiation/chemotherapy, but such protocol most likely only delays the inevitable. Therefore, the problem of finding therapeutic targets to prevent the occurrence and development of this severe oncological disease is currently acute. It is known that the functions of selenoproteins in the regulation of carcinogenesis processes are not unambiguous. Either they exhibit cytotoxic activity on cancer cells, or cytoprotective. A special place in the progression of oncological diseases of various etiologies is occupied by proteins of the thioredoxin and glutathione systems. These are two cellular antioxidant systems that regulate redox homeostasis, counteracting the increased production of reactive oxygen species in cells. The review reflects the latest data on the role of key enzymes of these redox systems in the regulation of processes associated with the progression of glioblastoma. A thorough consideration of these issues will expand fundamental knowledge about the functions of selenium-containing thioredoxin reductases and glutathione peroxidases in the therapy of glioblastomas and provide an understanding of the prospects for the treatment of this aggressive oncological disease.
Collapse
Affiliation(s)
- Elena G Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", St. Institutskaya 3, Pushchino, 142290, Russia.
| |
Collapse
|
31
|
Chen Y, Zhang B, Wang X, Chen Y, Anwar M, Fan J, Ma B. Prognostic value of preoperative modified Glasgow prognostic score in predicting overall survival in breast cancer patients: A retrospective cohort study. Oncol Lett 2025; 29:180. [PMID: 39990808 PMCID: PMC11843409 DOI: 10.3892/ol.2025.14926] [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: 10/22/2024] [Accepted: 01/07/2025] [Indexed: 02/25/2025] Open
Abstract
The modified Glasgow prognostic score (mGPS), based on C-reactive protein and albumin levels, is an inflammation-based prognostic tool used in various cancers. However, related research in breast cancer is limited. The present study evaluated the prognostic value of the preoperative mGPS in predicting overall survival (OS) of patients with breast cancer undergoing surgery. A retrospective cohort study was conducted involving 300 patients with breast cancer with up to 10 years of follow-up. Patients were categorized into three groups based on mGPS scores of 0, 1 and 2, and their clinical and pathological data were collected. Kaplan-Meier survival analysis and Cox proportional hazards models were used to assess survival outcomes and identify risk factors associated with higher mGPS scores. A prognostic nomogram was developed based on multivariate analysis to predict 5- and 10-year OS. Patients with high mGPS scores showed significantly poor survival outcomes. The 5- and 10-year survival rates for mGPS 0, 1 and 2 were 80, 70 and 55%, and 71, 55 and 22%, respectively (P<0.001). Multivariate Cox analysis identified the mGPS, age, smoking, PAM50 and TNM stage as independent predictors of OS. The nomogram based on the mGPS demonstrated good predictive accuracy (concordance index: 0.81) and calibration. The preoperative mGPS is an independent prognostic factor for OS of patients with breast cancer. It is a simple, cost-effective tool that can aid in risk stratification and guide treatment strategies. Further validation in larger cohorts is recommended.
Collapse
Affiliation(s)
- Yi Chen
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| | - Boxiang Zhang
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| | - Xiaoli Wang
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| | - Yanyan Chen
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| | - Munawar Anwar
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| | - Jingjing Fan
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| | - Binlin Ma
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Oncology, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Urumqi, Xinjiang Uygur Autonomous Region 830011, P. R. China
| |
Collapse
|
32
|
El Saftawy E, Aboulhoda BE, Alghamdi MA, Abd Elkhalek MA, AlHariry NS. Heterogeneity of modulatory immune microenvironment in bladder cancer. Tissue Cell 2025; 93:102679. [PMID: 39700733 DOI: 10.1016/j.tice.2024.102679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Urinary bladder cancer (UBC) is the ninth most common cancer worldwide. The intra-tumor heterogeneity of the UBC microenvironment explains the variances in response to therapy among patients. Tumor immune microenvironment (TIME) is based on the balance between anti-tumor and pro-tumorigenic immunity that eventually determines the tumor fate. This review addresses the recent insights of the cytokines, immune checkpoints, receptors, enzymes, proteins, RNAs, cancer stem cells (CSCs), tissue-resident cells, growth factors, epithelial-mesenchymal transition, microbiological cofactor, and paracrine action of cancer cells that mutually cross-talk within the TIME. In-depth balance and alteration of these factors influence the TIME and the overall tumor progression. This, in turn, highlights the prospects of the new era of manipulating these co-factors for improving the diagnosis, prognosis, and treatment of UBC. CONCLUSION: The heterogenic architecture of the TIME orchestrates the fate of the tumor. Nevertheless, recognizing the mutual cross-talk between these key players seems useful in prognostic and therapeutic approaches.
Collapse
Affiliation(s)
- Enas El Saftawy
- Department of Medical Parasitology, Faculty of Medicine, Cairo University, Cairo, Egypt; Department of Medical Parasitology, Armed Forces College of Medicine, Cairo, Egypt.
| | - Basma Emad Aboulhoda
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Mansour A Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia; Genomics and Personalized Medicine Unit, The Centre for Medical and Heath Research, King Khalid University, Abha 62529, Saudi Arabia
| | - Marwa Ali Abd Elkhalek
- Department of Medical Biochemistry& Molecular Biology, Armed Forces College of Medicine, Cairo, Egypt; Medical Biochemistry & Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | |
Collapse
|
33
|
Metsälä O, Wahlström G, Goel N, Miihkinen M, Taimen P, Schleutker J. Spatial profiling of ANO7 in prostate tissue: links to AR-signalling-associated lipid metabolism and inflammation. J Pathol 2025; 265:518-531. [PMID: 39978863 DOI: 10.1002/path.6405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/18/2024] [Accepted: 01/15/2025] [Indexed: 02/22/2025]
Abstract
Prostate cancer (PrCa) is highly prevalent in the Western world. Currently, however, there are many unmet needs in PrCa care, for example in distinguishing between clinically significant and indolent cases in early phases of the disease. ANO7 is a prostate-specific gene associated with PrCa risk and prognosis, but its exact function in the prostate remains unclear. This study investigates the role of ANO7 in benign prostatic epithelium using spatial transcriptomics by examining differences between ANO7-expressing and non-expressing epithelial regions and their corresponding stromal compartments. A total of 18,676 protein-coding genes were assessed from prostatectomy samples collected from patients with localised prostate cancer. In the collected sample cohort, ANO7 exhibited a distinct, heterogeneous, on-off epithelial expression pattern, enabling an in-depth analysis of ANO7-dependent processes. ANO7-positive epithelium was predominantly enriched with luminal epithelial cells and a specific NK cell subtype, CD56bright. In contrast, ANO7-negative regions were characterised by enrichment of club cells, inflammation, and features of proliferative inflammatory atrophy. Gene-set enrichment analysis revealed that ANO7 expression is associated with androgen receptor (AR) signalling and lipid metabolism. A detailed analysis of differentially expressed genes identified an ANO7- signature, which consisted of genes co-expressed with ANO7 in luminal cells, that demonstrated high consistency in bulk RNA-sequencing (RNA-seq) data. The ANO7-signature was enriched for AR-regulated genes, which highlighted lipid metabolism processes, particularly arachidonic acid metabolism, as a key metabolic feature of the ANO7-positive epithelium. Furthermore, the ANO7-signature demonstrated clinical significance in low-grade PrCa, correlating with a better response to therapy. In summary, these results highlight the potential role of ANO7 in regulating lipid metabolism associated with androgen signalling in benign luminal cells and low-grade cancer, reinforcing the hypothesis that ANO7 functions as a tumour suppressor. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Olli Metsälä
- Institute of Biomedicine, University of Turku, Turku, Finland
- FICAN West Cancer Center, University of Turku and Turku University Hospital, Turku, Finland
| | - Gudrun Wahlström
- Institute of Biomedicine, University of Turku, Turku, Finland
- FICAN West Cancer Center, University of Turku and Turku University Hospital, Turku, Finland
| | - Neha Goel
- Institute of Biomedicine, University of Turku, Turku, Finland
- FICAN West Cancer Center, University of Turku and Turku University Hospital, Turku, Finland
| | - Mitro Miihkinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pekka Taimen
- Institute of Biomedicine, University of Turku, Turku, Finland
- FICAN West Cancer Center, University of Turku and Turku University Hospital, Turku, Finland
- Department of Pathology, Laboratory Division, Turku University Hospital, Turku, Finland
| | - Johanna Schleutker
- Institute of Biomedicine, University of Turku, Turku, Finland
- FICAN West Cancer Center, University of Turku and Turku University Hospital, Turku, Finland
- Department of Genomics, Laboratory Division, Turku University Hospital, Turku, Finland
| |
Collapse
|
34
|
Ray SK, Mukherjee S. New insights into reductive stress responses and its clinical relation in cancer. Tissue Cell 2025; 93:102736. [PMID: 39826384 DOI: 10.1016/j.tice.2025.102736] [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: 08/22/2024] [Revised: 01/08/2025] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Cells are susceptible to both oxidative and reductive stresses, with reductive stress being less studied and potentially therapeutic in cancer. Reductive stress, characterized by an excess of reducing equivalents exceeding the activity of endogenous oxidoreductases, can lead to an imbalance in homeostasis, causing an increase in reactive oxygen species induction, affecting cellular antioxidant load and flux. Unlike oxidative stress, reductive stress has been understudied and poorly understood, and there is still much to learn about its mechanisms in cancer, its therapeutic potential, and how cancer cells react to it. Changes in redox balance and interference with redox signaling are linked to cancer cell growth, metastasis, and resistance to chemotherapy and radiation. Overconsumption of reducing equivalents can reduce metabolism, alter protein disulfide bond formation, disrupt mitochondrial homeostasis, and disrupt cancer cell signaling pathways. Novel approaches to delivering or using cancer medicines and techniques to influence redox biology have been discovered. Under reductive stress, cancer cells may coordinate separate pools of redox pairs, potentially impacting biology.
Collapse
Affiliation(s)
- Suman Kumar Ray
- Independent Researcher, Bhopal, Madhya Pradesh 462020, India
| | - Sukhes Mukherjee
- Department of Biochemistry. All India Institute of Medical Sciences. Bhopal, Madhya Pradesh 462020, India.
| |
Collapse
|
35
|
Xu X, Luo S, Wang Q, Zhang E, Liu J, Duan L. Allosteric site engagement and cooperativity mechanism by PHI1 for BRAF V600E kinase inhibition. Int J Biol Macromol 2025; 302:140475. [PMID: 39884594 DOI: 10.1016/j.ijbiomac.2025.140475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
With the ability to reveal allosteric sites, Ponatinib and Ponatinib Hybrid Inhibitor 1 (PHI1) are novel inhibitors of BRAF, a potent oncogene that activates the MAPK pathway. PHI1 also exhibits unique positive cooperativity, with enhanced inhibition on the other monomer when one monomer of the BRAFV600E dimer bound to an inhibitor. The abovementioned properties lack rigorous theoretical verification, so this study compared the interaction mechanisms of four inhibitor types and explored the source of the cooperativity of PHI1 via various computational methods. Results revealed that residues on the αC-helix formed hydrogen bonds with inhibitors, shifting the position of the αC-helix. PHI1 induced binding pocket contraction through contact with allosteric sites. Entropy contributions were considerably weakened when both BRAFV600E monomers were occupied, thereby increasing the binding ability of PHI1, indicating that entropy contributions were the main source of PHI1 cooperativity. The change in overall motion intensity tightened the binding pocket, increasing the binding abilities of hotspot residues, including Arg575 and Leu567. Moreover, three key hydrogen bonds formed between PHI1 and BRAFV600E in the dimer system were conducive to the binding. The insights derived from this study are expected to advance the development of inhibitors targeting BRAFV600E kinase.
Collapse
Affiliation(s)
- Xiaole Xu
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Song Luo
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Qihang Wang
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Enhao Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Jinxin Liu
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Lili Duan
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China.
| |
Collapse
|
36
|
Majumder T, Khot B, Suriyaarachchi H, Nathan A, Liu G. MYC regulation of the miR-92-Robo1 axis in Slit-mediated commissural axon guidance. Mol Biol Cell 2025; 36:ar50. [PMID: 40020181 DOI: 10.1091/mbc.e24-12-0534] [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: 03/15/2025] Open
Abstract
In the developing spinal cord, translational repression of Robo1 expression by microRNA-92 (miR-92) in precrossing commissural axons (CAs) inhibits Slit/Robo1-mediated repulsion facilitating commissural axon projection and midline crossing; however, the regulatory mechanisms governing miR-92 expression in the developing commissural neurons are currently lacking. Here, we propose that the transcription factor MYC regulates miR-92 expression in the developing spinal cord (of either sex) to control Robo1 levels in precrossing CAs, modulating Slit/Robo1-mediated repulsion and midline crossing. MYC, miR-92, and Robo1 are differentially expressed in the developing chicken spinal cord. MYC binds to the promoter region upstream of the gga-miR-92 gene in vitro. MYC knockdown dramatically decreases miR-92 expression and increases chicken Robo1 (cRobo1) levels. In contrast, overexpression of MYC significantly induces miR-92 expression and reduces cRobo1 levels. MYC knockdown or overexpression results in significant inhibition or induction of miR-92 activity in the developing chicken spinal cord, respectively. Disruption of the MYC-dependent regulation of the miR-92-cRobo1 axis affects Slit2-mediated CA growth cone collapse in vitro and impairs CA projection and midline crossing in vivo. These results elucidate the role of the MYC-miR-92-cRobo1 axis in Slit2/Robo1-mediated CA repulsion and midline crossing.
Collapse
Affiliation(s)
- Tanushree Majumder
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| | - Bhakti Khot
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| | | | - Anagaa Nathan
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| |
Collapse
|
37
|
Qin Z, Ye F, Wang J, Jiang J, Zhang X, Li H, Feng L. BUB1B promotes cisplatin resistance in gastric cancer via Rad51-mediated DNA damage repair. Transl Oncol 2025; 54:102334. [PMID: 40056529 DOI: 10.1016/j.tranon.2025.102334] [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: 11/28/2024] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Cisplatin resistance significantly impedes the treatment of gastric cancer (GC). This work examined the possible therapeutic target status and function of BUB1B in controlling cisplatin resistance. METHODS Following the identification of differentially expressed genes (DEGs), protein-protein interaction (PPI) network analysis was conducted using datasets from the Cancer Genome Atlas-stomach adenocarcinoma (TCGA-STAD), GSE51575, and GSE79973. Functional tests assessed the effect of BUB1B overexpression and knockdown on the GC cells. Enrichment analysis and RNA-seq identified pathways linked to BUB1B. Additionally, the function of BUB1B in GC cells resistant to cisplatin in regulating DNA repair was examined, as its relationship with Rad51 inhibitor (B02) in regulating cell cycle, proliferation, and apoptosis. The combined effects of Rad51 suppression and BUB1B overexpression on tumor development in cisplatin-resistant GC cells were further validated in vivo xenograft models. RESULTS Significant overexpression of six critical overlapping genes was seen in GC tissues. The GC cell invasion, migration, and proliferation processes were improved by BUB1B overexpression, whereas BUB1B knockdown prevented these outcomes. Genes involved in DNA repair were downregulated by BUB1B knockdown, according to an RNA-seq study. BUB1B overexpression boosted cell survival via modulating cell cycle proteins, but BUB1B knockdown hampered DNA repair and increased death in cisplatin-resistant GC cells. Overexpression of BUB1B enhanced tumor development in vivo and counteracted the inhibitory effects of B02 on cell growth. CONCLUSION BUB1B enhances cisplatin resistance in gastric cancer by regulating DNA repair and cell cycle progression, suggesting that targeting BUB1B may be a feasible therapeutic strategy.
Collapse
Affiliation(s)
- Zhe Qin
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Fangzhou Ye
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Jiayi Wang
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Jun Jiang
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Xiaohong Zhang
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Huanqing Li
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Li Feng
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China.
| |
Collapse
|
38
|
Huang Y, Zhu H, Liang Z, Wei W, Yang H, Wang Q, Huang H, He H, Mo R, Ye J, Dai Q, Zhong W, Liang Y. Development and validation of a kinase-related gene signature as a novel diagnostic and prognostic model for prostate cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167722. [PMID: 39965532 DOI: 10.1016/j.bbadis.2025.167722] [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: 07/31/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Prostate cancer (PCa) is a prevalent malignant tumor in men worldwide. Kinases play a key role in the development of multiple tumors. Nevertheless, the role of kinases in PCa remains largely unclear. METHODS A kinase-related gene signature was constructed by LASSO Cox regression analysis using the TCGA_PRAD cohort. The diagnostic and prognostic values of the signature were then evaulated. Furthermore, a loss-of-function assay was carried out to explore the function of NEK5 in PCa. RESULTS A signature of 13 kinase-related genes (NEK5, FRK, STK39, STYK1, IGF1R, RPS6KC1, TTK, CDK1, NEK2, PTK6, DAPK1, MELK and EPHA10) was constructed. The PCa patients presenting a high-risk score according to the signature demonstrated poorer disease-free survival compared to those with a low score. Additionally, TMB was found to be remarkably increased in patients categorized as high-risk relative to low-risk patients. Moreover, the 13-gene signature may also have good predictive value for PCa diagnosis. Furthermore, NEK5 expression was remarkably elevated in PCa tissues relative to benign tissues. NEK5 deficiency significantly inhibited PCa cell growth and suppressed mitochondrial OXPHOS. CONCLUSION The 13-gene signature constructed in this study may exhibit good performance in PCa diagnosis and prognosis evaluation. We identified the oncogenic role of NEK5 in PCa. NEK5 may serve as a therapeutic target for treatting PCa.
Collapse
Affiliation(s)
- Yaqiang Huang
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China
| | - Haiying Zhu
- Department of Endocrinology and Metabolism, The Affiliated Shunde Hospital of Jinan University, Foshan 528305, Guangdong, China
| | - Zhenguo Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, Guangdong, China
| | - Weiyang Wei
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China
| | - Hao Yang
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China
| | - Qi Wang
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China
| | - Hongxing Huang
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China
| | - Huichan He
- Guangzhou national laboratory, Guangzhou 510120, Guangdong, China
| | - Rujun Mo
- Department of Urology, The Tenth Affiliated Hospital of Southern Medical University, Dongguan People's Hospital, Dongguan 523018, Guangdong, China
| | - Jianheng Ye
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510120, Guangdong, China
| | - Qishan Dai
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510120, Guangdong, China
| | - Weide Zhong
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510120, Guangdong, China; Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau.
| | - Yingke Liang
- Department of Urology, Zhongshan City People's Hospital, Zhongshan 528400, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510120, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510120, Guangdong, China.
| |
Collapse
|
39
|
Yao TT, Chen L, Du Y, Jiang ZY, Cheng Y. MicroRNAs as Regulators, Biomarkers, and Therapeutic Targets in Autism Spectrum Disorder. Mol Neurobiol 2025; 62:5039-5056. [PMID: 39503812 DOI: 10.1007/s12035-024-04582-x] [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: 07/28/2024] [Accepted: 10/22/2024] [Indexed: 03/05/2025]
Abstract
The pathogenesis of autism spectrum disorder (ASD) is complex and is mainly influenced by genetic and environmental factors. Some research has indicated that environmental aspects may interplay with genetic aspects to enhance the risk, and microRNAs (miRNAs) are probably factors in explaining this link between heredity and the environment. MiRNAs are single-stranded noncoding RNAs that can regulate gene expression at the posttranscriptional level. Some research has indicated that miRNAs are closely linked to neurological diseases. Many aberrantly expressed miRNAs have been observed in autism, and these dysregulated miRNAs are expected to be potential biomarkers and provide new strategies for the treatment of this disease. This article reviews the research progress of miRNAs in autism, including their biosynthesis and function. It is found that some miRNAs show aberrant expression patterns in brain tissue and peripheral blood of autistic patients, which may serve as biomarkers of the disease. In addition, the article explores the novel role of exosomes as carriers of miRNAs with the ability to cross the blood-brain barrier and unique expression profiles, offering new possibilities for diagnostic and therapeutic interventions in ASD. The potential of miRNAs in exosomes as diagnostic markers for ASD is specifically highlighted, as well as the prospect of using engineered exosome-encapsulated miRNAs for targeted therapies.
Collapse
Affiliation(s)
- Tong-Tong Yao
- Center On Translational Neuroscience, Institute of National Security, Minzu University of China, 27th South Zhongguancun Avenue, Beijing, 100081, China
- School of Ethnology and Sociology, Minzu University of China, Beijing, China
| | - Lei Chen
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China
| | - Yang Du
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China
| | - Zhong-Yong Jiang
- Department of Medical Laboratory, Affiliated Cancer Hospital of Chengdu Medical College, Chengdu Seventh People's Hospital, Chengdu, China.
| | - Yong Cheng
- Center On Translational Neuroscience, Institute of National Security, Minzu University of China, 27th South Zhongguancun Avenue, Beijing, 100081, China.
- Center On Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China.
| |
Collapse
|
40
|
Musa FB, Moore KN, Podder V, Slomovitz BM. State of the art endocrine treatments for patients diagnosed with endometrial cancer in 2025. Curr Opin Obstet Gynecol 2025; 37:111-119. [PMID: 39773665 DOI: 10.1097/gco.0000000000001008] [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: 01/11/2025]
Abstract
PURPOSE OF REVIEW Endometrial cancer (EC) is rising in incidence, particularly in younger, premenopausal women, due to increasing rates of obesity and delayed childbearing. This review evaluates current and emerging endocrine therapies, with a focus on fertility-preserving approaches for early-stage EC and treatment options for advanced or recurrent disease. RECENT FINDINGS Fertility-sparing endocrine therapies, such as medroxyprogesterone acetate, megestrol acetate, and levonorgestrel-releasing intrauterine devices, achieve high response rates but carry recurrence risks. Biomarkers, including progesterone receptor status and molecular subtyping, are improving patient selection and outcomes. In advanced EC, single-agent and combination endocrine therapies with agents like selective estrogen receptor modulators, selective estrogen receptor down-regulators (SERDs), and aromatase inhibitors show efficacy, especially in hormone receptor-positive disease. Newer agents, including next-generation SERDs and proteolysis-targeting chimeras, hold potential for treating resistant cases. SUMMARY Endocrine therapy offers a well tolerated alternative to chemotherapy in selected EC patients, particularly those with hormone-sensitive tumors. Advances in molecular profiling and the development of novel endocrine agents are refining treatment strategies, supporting endocrine therapy's continued role in managing EC across various stages.
Collapse
Affiliation(s)
| | | | - Vivek Podder
- Mount Sinai Medical Center, Miami Beach, Florida, USA
| | | |
Collapse
|
41
|
Nathani P, Sharma P. Role of Artificial Intelligence in the Detection and Management of Premalignant and Malignant Lesions of the Esophagus and Stomach. Gastrointest Endosc Clin N Am 2025; 35:319-353. [PMID: 40021232 DOI: 10.1016/j.giec.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
The advent of artificial intelligence (AI) and deep learning algorithms, particularly convolutional neural networks, promises to address pitfalls, bridging the care for patients at high risk with improved detection (computer-aided detection [CADe]) and characterization (computer-aided diagnosis [CADx]) of lesions. This review describes the available artificial intelligence (AI) technology and the current data on AI tools for screening esophageal squamous cell cancer, Barret's esophagus-related neoplasia, and gastric cancer. These tools outperformed endoscopists in many situations. Recent randomized controlled trials have demonstrated the successful application of AI tools in clinical practice with improved outcomes.
Collapse
Affiliation(s)
- Piyush Nathani
- Department of Gastroenterology, University of Kansas School of Medicine, Kansas City, KS, USA.
| | - Prateek Sharma
- Department of Gastroenterology, University of Kansas School of Medicine, Kansas City, KS, USA; Kansas City Veteran Affairs Medical Center, Kansas City, MO, USA
| |
Collapse
|
42
|
Lee D, Green J, Crane J, Taylor DR, Reynolds S, Ng W, Jacob K, Whitelaw B, Aylwin S, Galata G, Lewis D, Seager M. Adrenal oncocytoma: a rare presentation of a benign 18 F-fluorodeoxyglucose PET avid virilising adrenal tumour. Nucl Med Commun 2025; 46:367-372. [PMID: 39604271 DOI: 10.1097/mnm.0000000000001932] [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: 11/29/2024]
Abstract
A woman in her 70s presented with features of hyperandrogenism including clitoral enlargement and deepening of her voice. Biochemical investigations revealed raised plasma androgens and urinary androgen metabolites and imaging findings showed a highly F-18 fluorodeoxyglucose (FDG)-PET avid left adrenal tumour initially suspected to be a malignant adrenocortical carcinoma (ACC). She subsequently underwent an uncomplicated laparoscopic adrenalectomy where complete resection of her tumour was achieved. Histopathological analysis demonstrated a benign adrenal oncocytoma with no evidence of malignancy. This case illustrates a rare presentation of a functioning virilising adrenal oncocytoma as a benign mimic of ACC.
Collapse
Affiliation(s)
- Danielle Lee
- King's College Hospital NHS Foundation Trust,
- Faculty of Life Sciences and Medicine, King's College London
| | | | - James Crane
- King's College Hospital NHS Foundation Trust,
| | - David R Taylor
- Department of Clinical Biochemistry (Synnovis), King's College Hospital NHS Foundation Trust, London and
| | | | - Wen Ng
- King's College Hospital NHS Foundation Trust,
| | - Koshy Jacob
- Eastbourne District General Hospital, Eastbourne, UK
| | | | | | | | - Dylan Lewis
- King's College Hospital NHS Foundation Trust,
| | | |
Collapse
|
43
|
Lu W, Yang S. METTL3/IGF2BP1 promotes the development of triple-negative breast cancer by mediating m6A methylation modification of PRMT7. Tissue Cell 2025; 93:102690. [PMID: 39709713 DOI: 10.1016/j.tice.2024.102690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND PRMT7 is upregulated in breast cancer and promotes tumor metastasis. Here we aimed to explore the function and mechanism of PRMT7 in triple-negative breast cancer (TNBC). METHODS The expression of PRMT7, METTL3 and IGF2BP1 was detected by immunohistochemistry (IHC), qRT-PCR and western blot. Cell viability and proliferation were measured using MTT and EdU assay. Flow cytometry and TUNEL assays were used to evaluate apoptosis. Invasion and migration were assessed by transwell and wound healing assays, respectively. Glucose consumption and lactate production were measured to assess glycolysis. In addition, the interaction between METTL3 and PRMT was verified by methylated RNA immunoprecipitation. The roles of METTL3 and PRMT in vivo were investigated through a xenograft model. RESULTS PRMT7 was upregulated in TNBC tissues and cells, and the knockdown of PRMT7 inhibited cell proliferation, invasion, migration and glycolysis, but induced apoptosis in TNBC cells. METTL3/IGF2BP1 enhanced PRMT7 expression by mediating the m6A methylation modification of PRMT7. Besides, METTL3 knockdown suppressed the progression of TNBC cells and regulated the WNT/β-catenin pathway via PRMT7. Moreover, silencing METTL3 restrained TNBC tumor growth in vivo through regulating PRMT7. CONCLUSION METTL3/IGF2BP1 facilitates the progression of TNBC by mediating m6A methylation modification of PRMT7.
Collapse
Affiliation(s)
- Wanli Lu
- Department of General Surgery, Qinghai University Affiliated Hospital, Xining 810000, China
| | - Shenghu Yang
- Department of General Surgery, Qinghai University Affiliated Hospital, Xining 810000, China.
| |
Collapse
|
44
|
Bai J, Chen P, Zhou Q, Tie X, Xia X, Wang Y, Jin L. KPNA2/KPNB1 promotes the malignant progression of gastric cancer induced by M2 macrophage polarization. Tissue Cell 2025; 93:102714. [PMID: 39765137 DOI: 10.1016/j.tice.2024.102714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/25/2024] [Accepted: 12/27/2024] [Indexed: 03/05/2025]
Abstract
Macrophages in the tumor microenvironment (TME) regulated gastric cancer progression, but the mechanism of macrophage polarization in gastric cancer progression remained unclear. This study mainly explored the molecular mechanism of macrophage polarization in the tumor microenvironment and its impact on the progression of gastric cancer. KPNA2 and KPNB1 expressions in cancer tissues and adjacent non-cancerous tissues were quantified via RT-qPCR and western blot. A correlation analysis was conducted between KPNA2 and KPNB1 expressions, utilizing the GEPIA2 database to link them with macrophage polarization. KPNA2-KPNB1 interaction was investigated on STRING, verified by Co-IP and IF assays. Raw246.7 cells were transfected with KPNA2 overexpression with or without si-KPNB1 plasmids. Then, M1/M2 macrophage markers and the proportion of M2 macrophages were measured by RT-qPCR, western blot, and IF. Co-culturing transfected Raw246.7 with MFC cells showed gastric cancer cell proliferation, apoptosis, migration, and invasion via CCK-8, flow cytometry, and transwell assays. KPNA2 and KPNB1 in gastric cancer tissues were elevated, exhibiting a positive correlation between them. KPNA2 overexpression facilitated the differentiation of macrophages into M2 type. KPNA2 overexpression in macrophages co-cultured with MFC cells stimulated MFC cells proliferation, repressed apoptosis, and enhanced migration/invasion. The interaction between KPNA2 and KPNB1 was confirmed through Co-IP and IF assays. Si-KPNB1 reversed the effects of KPNA2 overexpression on macrophages and gastric cancer cells. KPNA2 promoted the M2 polarization of macrophages by upregulating KPNB1, thereby inducing the proliferation and metastasis of gastric cancer.
Collapse
Affiliation(s)
- Juan Bai
- Department of Oncology, Wuzhong People's Hospital Affiliated to Ningxia Medical University, China
| | - Ping Chen
- The second Department of Oncology, Tumor Hospital, General Hospital of Ningxia Medical University, China.
| | - Qingxia Zhou
- Department of Oncology, Wuzhong People's Hospital Affiliated to Ningxia Medical University, China
| | - Xiaojun Tie
- Department of Oncology, Wuzhong People's Hospital Affiliated to Ningxia Medical University, China
| | - Xiao Xia
- Department of Oncology, Wuzhong People's Hospital Affiliated to Ningxia Medical University, China
| | - Yan Wang
- Department of Oncology, Wuzhong People's Hospital Affiliated to Ningxia Medical University, China
| | - Ling Jin
- Department of Oncology, Wuzhong People's Hospital Affiliated to Ningxia Medical University, China
| |
Collapse
|
45
|
Chong ZX, Ho WY, Yeap SK. Deciphering the roles of non-coding RNAs in liposarcoma development: Challenges and opportunities for translational therapeutic advances. Noncoding RNA Res 2025; 11:73-90. [PMID: 39736850 PMCID: PMC11683247 DOI: 10.1016/j.ncrna.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/29/2024] [Accepted: 11/13/2024] [Indexed: 01/01/2025] Open
Abstract
Liposarcoma is one of the most prevalent forms of soft tissue sarcoma, and its prognosis is highly dependent on its molecular subtypes. Non-coding RNAs (ncRNAs) like microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) can bind various cellular targets to regulate carcinogenesis. By affecting the expressions and activities of their downstream targets post-transcriptionally, dysregulations of miRNAs can alter different oncogenic signalling pathways, mediating liposarcoma progression. On the contrary, lncRNAs can sponge miRNAs to spare their downstream targets from translational repression, indirectly affecting miRNA-regulated oncogenic activities. In the past 15 years, multiple fundamental and clinical research has shown that different ncRNAs play essential roles in modulating liposarcoma development. Yet, there is a lack of an effective review report that could summarize the findings from various studies. To narrow this literature gap, this review article aimed to compare the findings from different studies on the tumour-regulatory roles of ncRNAs in liposarcoma and to understand how ncRNAs control liposarcoma progression mechanistically. Additionally, the reported findings were critically reviewed to evaluate the translational potentials of various ncRNAs in clinical applications, including employing these ncRNAs as diagnostic and prognostic biomarkers or as therapeutic targets in the management of liposarcoma. Overall, over 15 ncRNAs were reported to play essential roles in modulating different cellular pathways, including apoptosis, WNT/β-catenin, TGF-β/SMAD4, EMT, interleukin, and YAP-associated pathways to influence liposarcoma development. 28 ncRNAs were reported to be upregulated in liposarcoma tissues or circulation, whereas 11 were downregulated, making them potential candidates as liposarcoma diagnostic biomarkers. Among these ncRNAs, measuring the tissues or circulating levels of miR-155 and miR-195 was reported to help detect liposarcoma, differentiate liposarcoma subtypes, and predict the survival and treatment response of liposarcoma patients. Overall, except for a few ncRNAs like miR-155 and miR-195, current evidence to support the use of discussed ncRNAs as biomarkers and therapeutic targets in managing liposarcoma is mainly based on a single-center study with relatively small sample sizes or cell-based studies. Hence, more large-scale multi-center studies should be conducted to further confirm the sensitivity, specificity, and safety of ncRNAs as biomarkers and therapeutic targets. Instead of furthering investigation to confirm the translational values of all the discussed ncRNAs, which can be time- and cost-consuming, it would be more practical to focus on a few ncRNAs, including miR-155 and miR-195, to evaluate if they are sensitive and safe to be used as liposarcoma biomarkers and therapeutic agents or targets.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, 117599, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, 117599, Singapore
| | - Wan Yong Ho
- Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900, Sepang, Selangor, Malaysia
| |
Collapse
|
46
|
Mivehchi H, Eskandari-Yaghbastlo A, Emrahoglu S, Saeidpour Masouleh S, Faghihinia F, Ayoubi S, Nabi Afjadi M. Tiny messengers, big Impact: Exosomes driving EMT in oral cancer. Pathol Res Pract 2025; 268:155873. [PMID: 40022766 DOI: 10.1016/j.prp.2025.155873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/13/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Exosomes are indispensable extracellular vesicles that facilitate intercellular communication and are crucial for both healthy and pathological conditions, including cancer. The capacity of exosomes to echo the molecular characteristics of their cells of origin, including malignant cells, makes them indispensable tools for diagnosing and tracking disease progression in the field of oncology. Oral squamous cell carcinoma (OSCC), which has been identified as the sixth most prevalent cancer worldwide, has been linked to numerous risk factors, including tobacco use, alcohol consumption, human papillomavirus (HPV) infection, and inadequate oral hygiene. Exosomes pointedly influence the advancement of oral cancer via promoting tumor cell growth, invasion, angiogenesis, and immune evasion through the alteration of the tumor microenvironment. A critical apparatus in cancer metastasis is the epithelial-to-mesenchymal transition (EMT), during which cancer cells acquire improved migratory and invasive properties. EMT plays a role in metastasis, resistance to treatment, and evasion of the immune response. Exosomes facilitate EMT in oral cancer by delivering bioactive molecules that influence EMT signaling pathways. These exosomes inspire EMT in recipient cells, by this means enhancing tumor invasion and metastasis. This study aims to identify the specific exosomal components and signaling pathways that are tangled in EMT, in that way providing new avenues for targeted therapies designed to hinder the metastasis of oral cancer.
Collapse
Affiliation(s)
- Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | | | - Sahand Emrahoglu
- School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | | | - Farbod Faghihinia
- School of Dentistry, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Saminalsadat Ayoubi
- School of Dental Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
47
|
Guo Y, Dong W, Sun D, Zhao X, Huang Z, Liu C, Sheng Y. Bacterial metabolites: Effects on the development of breast cancer and therapeutic efficacy (Review). Oncol Lett 2025; 29:210. [PMID: 40070782 PMCID: PMC11894516 DOI: 10.3892/ol.2025.14956] [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: 11/09/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
Evidence suggests that various gut metabolites significantly impact breast cancer (BC) and its treatment. However, the underlying mechanisms remain poorly understood and require further investigation. In the present study, the current literature was reviewed to evaluate the roles of microbial metabolites in the development of BC and its response to treatment. Microbial metabolites, including secondary bile acids, short-chain fatty acids, amino acid metabolites, lipopolysaccharide, nisin and pyocyanin, serve crucial roles in the BC microenvironment. Microbial metabolites promote BC invasion, metastasis and recurrence by facilitating cellular movement, epithelial-mesenchymal transition, cancer stem cell function and diapedesis. Furthermore, certain metabolites, such as trimethylamine N-oxide and L-norvaline, can alter the pharmacokinetics of chemotherapeutic drugs. The present review highlights the possible involvement of microbial metabolites and bacteriocins in BC carcinogenesis, development and metastasis. These metabolites could provide new insights for BC treatment strategies and are considered potential therapeutic targets.
Collapse
Affiliation(s)
- Yan Guo
- Department of Endocrinology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Wenyan Dong
- Department of Thyroid and Breast Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Dezheng Sun
- Department of Thyroid and Breast Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Xiang Zhao
- Department of Thyroid and Breast Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Zhiping Huang
- Department of Hepatobiliary Surgery and Organ Transplantation, General Hospital of Southern Theater Command of People's Liberation Army, Guangzhou, Guangdong 51000, P.R. China
| | - Chaoqian Liu
- Department of Thyroid and Breast Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Yuan Sheng
- Department of Thyroid and Breast Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
48
|
Pu X, Zhang C, Jin J, Jin Y, Ren J, Zhou S, Patel H, Chen J, Wu B, Chen L, Qian H, Lin T. Phase separation of EEF1E1 promotes tumor stemness via PTEN/AKT-mediated DNA repair in hepatocellular carcinoma. Cancer Lett 2025; 613:217508. [PMID: 39884379 DOI: 10.1016/j.canlet.2025.217508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/15/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
This study aimed to investigate the associations of liquid-liquid phase separation (LLPS) and tumor stemness in hepatocellular carcinomas (HCC). LLPS-related genes were extracted from DrLLPS, LLPSDB and PhaSepDB databases. Stemness index (mRNAsi) was calculated based on the data from TCGA and Progenitor Cell Biology Consortium. Through some series of bioinformatics methods, we first found that stemness index mRNAsi was associated with worse survival outcomes, immune infiltration and therapy sensitivity in HCC. G2M checkpoint and DNA repair pathways were significantly activated with high mRNAsi. Totally, 71 differentially expressed LLPS genes in HCC were correlated with mRNAsi, and a mRNAsi-associated LLPS gene signature (KPNA2, EEF1E1 and ATIC) was identified to predict prognosis for HCC patients. mRNAsi-associated LLPS genes contributed to cluster HCC patients into four molecular clusters that markedly differed on survival, immune infiltration and therapy sensitivity. Further in vivo and in vitro experiments showed that EEF1E1 was highly expressed in HepG2 and HCCLM3 cells, and EEF1E1 silencing observably inhibited tumor cell growth, liver cancer stem cells (CSCs) markers (CD133, EpCAM and SOX2) expression, enhanced DNA damage marker γH2AX expression by activating PTEN/AKT pathway. EEF1E1 could undergo LLPS condensates, and roles of EEF1E1 on tumor cells were partly reversed after inhibiting LLPS using 1, 6-hexanediol. In conclusion, EEF1E1 was identified as a phase separation protein and involves in tumor stemness and DNA damage repair in HCC. EEF1E1 and its LLPS condensate may be novel targets to elaborate the underlying mechanisms of CSCs propagation in HCC.
Collapse
Affiliation(s)
- Xiaofan Pu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Chaolei Zhang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Junbin Jin
- Department of Hepatobiliary Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China
| | - Yifeng Jin
- Department of Hepatobiliary Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China
| | - Jianghao Ren
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Senhao Zhou
- Department of Otolaryngology Head and Neck Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Harsh Patel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY 11439, USA
| | - Jingyun Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Bicheng Wu
- The First School of Medicine, School of Information and Engieering, Wenzhou Medical University, Wenzhou, 325000, China
| | - Leyi Chen
- School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Haoran Qian
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| | - Tianyu Lin
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| |
Collapse
|
49
|
Chen Y, Wu J, Cai K, Xiao X, Chen Y, Zhang X, Deng S, Pei C, Chen Y, Xie Z, Li P, Liao Q. Bifidobacterium longum subsp. longum XZ01 delays the progression of colon cancer in mice through the interaction between the microbial spatial distribution and tumour immunity. Int Immunopharmacol 2025; 150:114283. [PMID: 39955918 DOI: 10.1016/j.intimp.2025.114283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/25/2025] [Accepted: 02/08/2025] [Indexed: 02/18/2025]
Abstract
Studies have shown that the colonisation of active microorganisms is more conducive to the development of tumour immunotherapy, but intuitive evidence regarding shaping of the tumour immune microenvironment is lacking. In this study, we used Bifidobacterium longum subsp. longum (XZ01) to intervene in a colon cancer mouse model and found that its mechanism may be related to the interaction between the spatial distribution of microorganisms and tumour immunity. Through the visualisation method we established, for the first time, we showed that harmful active bacteria such as Streptococcus and Rhodococcus specifically accumulate in the middle and upper layers of tumour tissue. These bacteria likely participate in signalling pathways that affect macrophages by directly contacting or invading the macrophages, leading to a nondifferentiated state in macrophages and the loss of some immune functions. Furthermore, the accumulation of Streptococcus and Rhodococcus fragments in the deep layer of tumour tissue likely upregulates the expression of IL-10 in tumour tissue and inhibits other immune cells, such as CD8+ T cells, DC and NK cells. In contrast, XZ01 can specifically compete for the growth sites of Streptococcus and Rhodococcus in the middle and upper layers of tumour tissue and probably protects macrophages from being invaded by harmful bacteria. XZ01 directly regulates the polarisation of M0 macrophages towards the M1 phenotype by upregulating IFN-γ, thus activating tumour immunity to inhibit the growth of tumour cells. This study revealed that the influence of active microorganisms on the tumour immune microenvironment is crucial for effective immunotherapy intervention, potentially offering new targets for improving patient prognosis.
Collapse
Affiliation(s)
- Ying Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; School of Pharmaceutical Sciences, Guangdong Yunfu Vocational College of Chinese Medicine, Yunfu 527300, China
| | - Jinyun Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Kaiwei Cai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaoyi Xiao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Ye Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xingyuan Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Song Deng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Chaoying Pei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yanlong Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518000, China
| | - Pei Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Qiongfeng Liao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
50
|
Niu YR, Xiang MD, Yang WW, Fang YT, Qian HL, Sun YK. NAD+/SIRT1 pathway regulates glycolysis to promote oxaliplatin resistance in colorectal cancer. World J Gastroenterol 2025; 31:100785. [DOI: 10.3748/wjg.v31.i11.100785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/13/2024] [Accepted: 02/13/2025] [Indexed: 03/13/2025] Open
Abstract
BACKGROUND Glycolysis provides growth advantages and leads to drug resistance in colorectal cancer (CRC) cells. SIRT1, an NAD+-dependent deacetylase, regulates various cellular processes, and its upregulation results in antitumor effects. This study investigated the role of SIRT1 in metabolic reprogramming and oxaliplatin resistance in CRC cells.
AIM To investigate the role of SIRT1 in metabolic reprogramming and overcoming oxaliplatin resistance in CRC cells.
METHODS We performed transcriptome sequencing of human CRC parental cells and oxaliplatin-resistant cells to identify differentially expressed genes. Key regulators were identified via the LINCS database. NAD+ levels were measured by flow cytometry, and the effects of SIRT1 on oxaliplatin sensitivity were assessed by MTS assays, colony formation assays, and xenograft models. Glycolytic function was measured using Western blot and Seahorse assays.
RESULTS Salermide, a SIRT1 inhibitor, was identified as a candidate compound that enhances oxaliplatin resistance. In oxaliplatin-resistant cells, SIRT1 was downregulated, whereas γH2AX and PARP were upregulated. PARP activation led to NAD+ depletion and SIRT1 inhibition, which were reversed by PARP inhibitor treatment. The increase in SIRT1 expression overcame oxaliplatin resistance, and while SIRT1 inhibition increased glycolysis, the increase in SIRT1 inhibited glycolysis in resistant CRC cells, which was characterized by reduced expression of the glycolytic enzymes PKM2 and LDHA, as well as a decreased extracellular acidification rate. The PKM2 inhibitor shikonin inhibited glycolysis and reversed oxaliplatin resistance induced by SIRT1 inhibition.
CONCLUSION SIRT1 expression is reduced in oxaliplatin-resistant CRC cells due to PARP activation, which in turn increases glycolysis. Restoring SIRT1 expression reverses oxaliplatin resistance in CRC cells, offering a promising therapeutic strategy to overcome drug resistance.
Collapse
Affiliation(s)
- Ya-Ru Niu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Mi-Dan Xiang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wen-Wei Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu-Ting Fang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hai-Li Qian
- National Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yong-Kun Sun
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|