1
|
Song Y, Zhang K, Zhang J, Li Q, Huang N, Ma Y, Hou N, Han F, Kan C, Sun X. Epigenetic regulation of nuclear receptors: Implications for endocrine-related diseases and therapeutic strategies. Genes Dis 2025; 12:101481. [PMID: 40290121 PMCID: PMC12022648 DOI: 10.1016/j.gendis.2024.101481] [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: 05/07/2024] [Revised: 10/20/2024] [Accepted: 11/03/2024] [Indexed: 04/30/2025] Open
Abstract
The expression and function of the receptor are controlled by epigenetic changes, such as DNA methylation, histone modification, and noncoding RNAs. These modifications play a pivotal role in receptor activity and can lead to or exacerbate endocrine-related diseases. This review examines the epigenetic alterations of nuclear receptors and their significant impact on conditions such as diabetes, thyroid disorders, and endocrine-related tumors. It highlights current therapies targeting these epigenetic mechanisms, including gene editing, epigenetic drugs, and various other therapeutic approaches. This review offers fresh insight into the mechanisms of endocrine-associated disorders, highlighting the latest progress in the development of novel epigenetic therapies that can be used to address receptor-endocrine interactions.
Collapse
Affiliation(s)
- Yixin Song
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Qinying Li
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Na Huang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Yujie Ma
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Fang Han
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261031, China
| |
Collapse
|
2
|
Li F, Jiang Y, Wang X, Gao Y, Lo CS, Su S, Wu Z, Jiang B, Zhao Z, Lin S, Xie Y, Chen J, Guo Q, Dong Z, Zhu J. Titanium nitride meta-biosensors targeting extracellular vesicles for high-sensitivity prostate cancer detection. Biosens Bioelectron 2025; 277:117288. [PMID: 39985905 DOI: 10.1016/j.bios.2025.117288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Disposable plasmonic metasurfaces with high biosensing performance are urgently sought for clinical label-free detection. Low-cost aluminum (Al) and titanium nitride (TiN) offer promising alternatives to noble metals for constructing these metasurfaces. However, Al suffers from limited chemical stability, and TiN exhibits weak plasmonic effects, both of which hinder their application in meta-biosensing. Here we integrate their complementary advantages and propose the TiN/Al meta-biosensors. They not only empower the unique near-field enhancement for sensing by TiN/Al hybrid plasmonic modes, but also construct a robust TiN armor against external wear, heat, moisture and corrosion during the bio-detection process. Compared to traditional gold-based counterparts, our meta-biosensors offer superior optical sensitivity at a much lower cost and with fewer pretreatment steps. The excellent biosensing performance facilitates the development of a high-throughput detection system for serum small extracellular vesicles (sEVs), aiding in the diagnosis and follow-up of prostate cancer. The sEVs meta-biosensing demonstrates a diagnostic sensitivity of 100% for significantly distinguishing early cancer, breaking through the conventional testing limitation. Moreover, it doubles the prediction accuracy of cancer recurrence risk following surgery. Our research highlights the potential for large-scale development of powerful meta-biosensors based on non-noble materials, opening up significant opportunities in cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Fajun Li
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China; Xiamen Dili-Chip Technology Co. Ltd., Xiamen, 361000, China; School of Electronic Science and Engineering, National Model Microelectronics College, Xiamen University, Xiamen, 361100, China
| | - Yuanyuan Jiang
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Xuegang Wang
- Department of Urology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Yuan Gao
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Ching Shu Lo
- School of Electronic Science and Engineering, National Model Microelectronics College, Xiamen University, Xiamen, 361100, China
| | - Shengdong Su
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Zhilin Wu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Bingliang Jiang
- Department of Urology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Zhongjie Zhao
- Department of Urology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Shaowei Lin
- Department of Nuclear Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Yinong Xie
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China
| | - Junjie Chen
- Analysis and Measurement Center, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Qiwei Guo
- Department of Central Laboratory, Department of Obstetrics and Gynecology, Fujian Key Clinical Specialty of Laboratory Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Zhaogang Dong
- Quantum Innovation Centre, Agency for Science Technology and Research, 2 Fusionopolis Way, Innovis #08-03, Singapore, 138634, Republic of Singapore
| | - Jinfeng Zhu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361100, China; Xiamen Dili-Chip Technology Co. Ltd., Xiamen, 361000, China; School of Electronic Science and Engineering, National Model Microelectronics College, Xiamen University, Xiamen, 361100, China.
| |
Collapse
|
3
|
Fang B, Lu Y, Li X, Wei Y, Ye D, Wei G, Zhu Y. Targeting the tumor microenvironment, a new therapeutic approach for prostate cancer. Prostate Cancer Prostatic Dis 2025; 28:260-269. [PMID: 38565910 DOI: 10.1038/s41391-024-00825-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND A growing number of studies have shown that in addition to adaptive immune cells such as CD8 + T cells and CD4 + T cells, various other cellular components within prostate cancer (PCa) tumor microenvironment (TME), mainly tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs) and myeloid-derived suppressor cells (MDSCs), have been increasingly recognized as important modulators of tumor progression and promising therapeutic targets. OBJECTIVE In this review, we aim to delineate the mechanisms by which TAMs, CAFs and MDSCs interact with PCa cells in the TME, summarize the therapeutic advancements targeting these cells and discuss potential new therapeutic avenues. METHODS We searched PubMed for relevant studies published through December 10 2023 on TAMs, CAFs and MDSCs in PCa. RESULTS TAMs, CAFs and MDSCs play a critical role in the tumorigenesis, progression, and metastasis of PCa. Moreover, they substantially mediate therapeutic resistance against conventional treatments including anti-androgen therapy, chemotherapy, and immunotherapy. Therapeutic interventions targeting these cellular components have demonstrated promising effects in preclinical models and several clinical trials for PCa, when administrated alone, or combined with other anti-cancer therapies. However, the lack of reliable biomarkers for patient selection and incomplete understanding of the mechanisms underlying the interactions between these cellular components and PCa cells hinder their clinical translation and utility. CONCLUSION New therapeutic strategies targeting TAMs, CAFs, and MDSCs in PCa hold promising prospects. Future research endeavors should focus on a more comprehensive exploration of the specific mechanisms by which these cells contribute to PCa, aiming to identify additional drug targets and conduct more clinical trials to validate the safety and efficacy of these treatment strategies.
Collapse
Affiliation(s)
- Bangwei Fang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Ying Lu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaomeng Li
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Gonghong Wei
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| |
Collapse
|
4
|
Zhou M, Li R, Lian G, Yang M, Li L, Yin Z, Li G, Zhao J, Tan R. Tetrahydrocurcumin alleviates colorectal tumorigenesis by modulating the SPP1/CD44 axis and preventing M2 tumor-associated macrophage polarization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156674. [PMID: 40220425 DOI: 10.1016/j.phymed.2025.156674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Recent studies show that secreted phosphoprotein 1 (SPP1) is linked to the progression of various cancers, including colorectal cancer (CRC). SPP1 also promotes M2 macrophage polarization, contributing to immune evasion in the tumor microenvironment. Tetrahydrocurcumin (THC) has been reported to alleviate CRC, but the mechanism remains unclear. PURPOSE The study aimed to explore how THC modulated the SPP1/CD44 axis to inhibit M2 polarization and suppress CRC development. METHODS Azoxymethane/dextran sulfate sodium (AOM/DSS)-induced mouse model was used to assess the anti-CRC effects of THC. Transcriptome sequencing was conducted to identify the key targets of THC in CRC. The effects of THC on CRC cells were evaluated by CCK-8, colony formation, migration assays, immunofluorescence staining and flow cytometry. Human monocytic cells, THP-1, and colon cancer cell line, HCT116, were co-cultured, both directly or indirectly, to mimic the tumor-macrophage interactions, and investigate the role of SPP1/CD44 axis and the intervention effect of THC. RESULTS THC significantly inhibited CRC carcinogenesis in mice and improved pathologic symptoms, serum inflammatory markers, and intestinal barrier integrity. THC inhibited CRC cell proliferation, migration and colony formation, while promoting apoptosis. Transcriptome analysis identified SPP1 as a key target of THC against CRC. SPP1 facilitated CRC progression by activating the ERK signaling pathway and maintaining the M2-like phenotype of macrophage, which further exacerbated this response. THC inhibited CRC development by targeting the SPP1/CD44 axis, rather than the integrin pathway. CONCLUSIONS SPP1 played a crucial role in maintaining the M2 phenotype of macrophage and promoting CRC cells proliferation. THC inhibited the activation of ERK signals in CRC cells and phenotypic transformation of M2-like macrophages through the SPP1/CD44 axis, thereby regulating the tumor immune microenvironment to exert anti-CRC effect.
Collapse
Affiliation(s)
- Mengting Zhou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China; Translational Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Rui Li
- Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610000, China.
| | - Guiyun Lian
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Mengni Yang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China; Translational Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Li
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
| | - Zhujun Yin
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
| | - Guiyu Li
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
| | - Junning Zhao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China; National Key Laboratory of Drug Regulatory Science, National Medical Products Administration (NMPA), Beijing 100038, China; National Center for Nanoscience and Technology, Beijing 100190, China.
| | - Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China; Translational Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
5
|
Mortezaee K. EZH2 regulatory roles in cancer immunity and immunotherapy. Pathol Res Pract 2025; 270:155992. [PMID: 40306002 DOI: 10.1016/j.prp.2025.155992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
Enhancer of zeste homolog 2 (EZH2) is a polycomb repressor complex 2 (PRC2) subunit that is responsible for silencing expression of target genes through generation of lysine 27 trimethylation on histone H3 (H3K27Me3). EZH2 is an oncogene aberrantly expressed in human cancers, and its overexpression favors immune escape and metastasis. Immune escape occurs via the impact of EZH2 on hampering antigen expression machinery, stabilizing FOXP3 in regulatory T cells (Tregs), inhibiting recruitment and activity of natural killer (NK) and CD8+ T cells, and inducing recruitment and activity of myeloid-derived suppressor cells (MDSCs). Besides, EZH2 also promotes intra-tumoral recruitment of tumor-associated macrophages (TAMs). A point is that pharmacologic EZH2 inhibition (not knockdown) seemingly promotes polarization of macrophages toward pro-tumor M2 phenotype, which defines resistance mechanism. Besides, increased EZH2 expression after anti-cytotoxic T lymphocyte associated antigen-4 (CTLA-4) and a rise in the tumoral expression of programmed death-ligand 1 (PD-L1) after EZH2 inhibition account for secondary immunosuppression in tumor ecosystem after immunotherapy, indicating the applicability of using EZH2 targeted therapies as a combinatory approach with anti-programmed death-1 (PD-1) or anti-CTLA-4 therapy. Such combination reinvigorates anti-tumor immunity and presumably hampers T cell exhaustion and acting as a promising regimen for retarding cancer growth.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran; Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
6
|
Liu Z, Hu J, Han X, Li L, Niu H, Zhang X, Wang N, Shi X, Sang L, Zhang Q, Qian X. SLAMF8 regulates Fc receptor-mediated phagocytosis in mouse macrophage cells through PI3K-Akt signaling. Immunol Lett 2025; 273:106990. [PMID: 39983459 DOI: 10.1016/j.imlet.2025.106990] [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: 08/11/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Emerging studies have demonstrated that phagocytosis checkpoints, which promote tumor-mediated immune evasion, are potential targets for cancer immunotherapy. In this study, the TCGA colorectal cancer (CRC) dataset and our RNA sequencing dataset suggested that SLAMF8 expression is significantly positively correlated with the expression levels of multiple phagocytosis checkpoint molecules. In vitro, we confirmed that SLAMF8 significantly regulated the phagocytosis of mouse CRC cells. RNA sequencing revealed that the expression of genes that promote Fc receptor (FcR)-mediated phagocytosis, such as FCGR1, FCGR3, FCGR2b, FCGR4, and ITGAM, was significantly upregulated after SLAMF8 knockdown. The Kyoto Encyclopedia of Genes and Genomes (KEGG) results suggested that the significantly enriched signaling pathways after SLAMF8 knockdown or overexpression included the PI3K-Akt signaling pathway. The protein expression levels of p-PI3K and p-Akt were significantly increased after SLAMF8 knockdown. When PI3K inhibitors and Fc blockers were added after SLAMF8 knockdown, mouse macrophage phagocytosis, and FcR expression decreased. Our results suggest that SLAMF8 may impair FcR-mediated phagocytosis through the PI3K-Akt signaling pathway and negatively regulate the antitumor immune response.
Collapse
Affiliation(s)
- Zhihao Liu
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing 210008, PR China; Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Jing Hu
- Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Xingzhi Han
- Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Li Li
- Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Haiqing Niu
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - Xin Zhang
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - Ning Wang
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - Xiao Shi
- Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Liuqi Sang
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing 210008, PR China
| | - Qun Zhang
- Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China.
| | - Xiaoping Qian
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing 210008, PR China; Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China; Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, PR China.
| |
Collapse
|
7
|
Zhang H, Du Y, Liu X, Gao R, Xu X, Hou X, Liu Z, Xin P, Wu W, Liu T, Zhao Y, Zhou H, Jiang Y. Enhancing curcumol delivery through PD-1 targeted nanocarriers: A novel therapeutic approach for prostate cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156595. [PMID: 40220412 DOI: 10.1016/j.phymed.2025.156595] [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: 10/27/2024] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Prostate cancer is a prevalent form of cancer that impacts men on a global scale, and its treatment faces challenges such as tumor metastasis, immune resistance, and epigenetic abnormalities. Most current research focuses on nanocarriers with a single function, but the dual mechanism of action-enhancing immune response and regulating EZH2 epigenetic modification-has not been reported. PURPOSE This study is the first to construct an engineered outer membrane vesicle (OMV) delivery system loaded with PD-1 antibody and Curcumol, combining two cutting-edge approaches: tumor immunotherapy and epigenetic regulation. We developed a nanocarrier system based on engineered OMVs (OMV-PD-1) to deliver the natural anticancer compound Curcumol, aiming to regulate epigenetic modifications and enhance tumor immune responses, thereby effectively inhibiting the proliferation and metastasis of prostate cancer cells. METHODS OMV-PD-1 was prepared using recombinant technology, and its characteristics were identified through the application of liquid chromatography-mass spectrometry (LC-MS), dynamic light scattering (DLS), and transmission electron microscopy (TEM). We assessed its antitumor activity against prostate cancer cells (PC3) in vitro and in vivo and explored its molecular mechanisms through RNA sequencing and gene set enrichment analysis (GSEA). RESULTS An outstanding encapsulation efficiency and a delayed drug release profile were evident in OMV-PD-1/Curcumol. In vitro experiments demonstrated that the system significantly inhibited PC3 cell migration (77.25 % inhibition) and invasion (73.03 % inhibition), and regulated histone methylation modifications (such as H3K9 and H3K27) by downregulating EZH2 gene expression. In vivo experiments confirmed its excellent tumor targeting in a humanized mouse model, significantly inhibiting tumor growth and enhancing immune responses, such as increased NK cell infiltration and elevated pro-inflammatory cytokine levels. CONCLUSION The OMV-PD-1/Curcumol delivery system developed in this study not only hinders the aggressive actions of prostate cancer cells by regulating epigenetic modifications but also significantly stimulates antitumor immune responses, offering a unique and readily implementable therapeutic avenue.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Yang Du
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Xujia Liu
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, 110134, China
| | - Ruxu Gao
- Department of Urology, The Cancer Hospital of Dalian University of Technology&Liaoning Cancer Hospital and institute, Shenyang, Liaoning Province 110042, China
| | - Xiaoxiao Xu
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Xiaolong Hou
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Zhenghua Liu
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Peng Xin
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Weiwei Wu
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Tao Liu
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Yue Zhao
- Department of Cell Biology, Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang City, Liaoning Province 110122, China.
| | - Hongxu Zhou
- Department of Anesthesiology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China.
| | - Yuanjun Jiang
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China.
| |
Collapse
|
8
|
Zhuang Y, Liao X, Niu F, Li M, Yan Y, He C, Wu X, Tian R, Gao G. Single-nucleus and spatial signatures of the brainstem in normal brain and mild traumatic brain injury in male mice. Nat Commun 2025; 16:5082. [PMID: 40450008 DOI: 10.1038/s41467-025-59856-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 05/07/2025] [Indexed: 06/03/2025] Open
Abstract
The mammalian brainstem is particularly vulnerable to mild traumatic brain injury (mTBI), which is associated with prolonged autonomic dysfunction and coma. The spatial cellular connections within the brainstem or the mechanisms underlying its response to injury have been underestimated. Here, we performed single-nucleus RNA sequencing with spatial transcriptome sequencing in both normal and mTBI brainstems in male mice, revealing thirty-five neuron and non-neuron clusters. Typically, we identified subtypes of neurons that co-release multiple neurotransmitters, especially in the sagittal midline of the brainstem. Spatially adjacent neurons sharing similar gene expression patterns. The brainstem's response to mTBI has two features: (1) Oligodendrocytes around the fourth ventricle exhibit widespread disconnection at 1-h post-injury, and (2) Injury-related noradrenergic neurons, particularly in their interaction with neurons located in theIRt and the Sol. These findings provides a reference for further integrative investigations of cellular and circuit functions of brainstem.
Collapse
Affiliation(s)
- Yuan Zhuang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xixian Liao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fei Niu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Ming Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yu Yan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chuanhang He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiang Wu
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Runfa Tian
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Guoyi Gao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
9
|
Liu X, Jiang R, Xu Y, Xu X, Fang L, Gao G, Han L, Chen Y, Du H, Cai Y, Zhu F, Chen M, Wang K, Li H, Wang G, Quan C. Dual cytokine-engineered macrophages rejuvenate the tumor microenvironment and enhance anti-PD-1 therapy in renal cell carcinoma. Int Immunopharmacol 2025; 156:114725. [PMID: 40294469 DOI: 10.1016/j.intimp.2025.114725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025]
Abstract
Despite advances in PD-1 blockade therapy, the immunosuppressive tumor microenvironment (TME) limits its efficacy in renal cell carcinoma (RCC). Here, we developed dual-cytokine-engineered macrophages co-delivering IL-12 and CXCL-9 to reprogram TME and enhance anti-PD-1 responsiveness. Single-cell RNA sequencing revealed that RCC harbor abundant M2-like tumor-associated macrophages (TAMs), which correlate with T-cell exhaustion. In vitro, engineered macrophages polarized M2-like TAMs to antitumor M1 phenotypes, secreted CXCL-9 to recruit cytotoxic T cells, and released IL-12 to amplify T/NK cell activation. In vivo, intravenously administered engineered macrophages homed to tumors, reshaped the TME by increasing CD8+ T cells, dendritic cells, and NK cells while reducing immunosuppressive Tregs and MDSCs. This approach synergized with PD-1 blockade, resulting in a 2.5-fold greater tumor growth inhibition compared to anti-PD-1 monotherapy. This dual-cytokine macrophage platform offers a novel strategy to overcome resistance to checkpoint inhibitors in RCC by delivering cytokine and remodeling TME, with implications for clinical translation.
Collapse
Affiliation(s)
- Xin Liu
- Tianjin institute of urology,Tianjin Medical University Second Hospital, Tianjin, China; Department of Urology,The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ranran Jiang
- Department of Oncology,The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yujun Xu
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaodi Xu
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lin Fang
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ge Gao
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lulu Han
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuxin Chen
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongwei Du
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ying Cai
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fei Zhu
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingjing Chen
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kaidi Wang
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hailong Li
- Department of Urology,The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Gang Wang
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Changyi Quan
- Tianjin institute of urology,Tianjin Medical University Second Hospital, Tianjin, China.
| |
Collapse
|
10
|
Qin J, Liao K, Huang L, Qiu R, Tang N, Wu J. Proteome-driven transcriptomic dissection of EMT networks in bladder cancer based on the VIM and CDH2 protein macromolecules influence: From molecular-protein subtyping to therapeutic target prioritization. Int J Biol Macromol 2025; 315:144585. [PMID: 40419050 DOI: 10.1016/j.ijbiomac.2025.144585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 05/05/2025] [Accepted: 05/21/2025] [Indexed: 05/28/2025]
Abstract
Bladder cancer progression is intrinsically linked to epithelial-mesenchymal transition (EMT), a protein-centric process driving metastasis and therapy resistance. This study systematically dissected bladder cancer transcriptomes through the lens of VIM (vimentin) and CDH2 (N-cadherin) protein networks, integrating datasets from GEO via cross-platform harmonization. Non-negative matrix factorization (NMF) resolved two molecular subtypes with distinct EMT-related protein expression profiles, characterized by asymmetric transcriptomic dysregulation. Functional enrichment revealed protein-driven pathways-including TGF-β signaling, Wnt/β-catenin activation, and ECM remodeling-as hallmarks of aggressive subtypes. LASSO regression identified 384 transcriptional drivers, while PPI network analysis prioritized 10 hub proteins (CALML5, THBS1, SMAD7, TAGLN, ICAM1, CEBPB, CNN1, TNFAIP3, TNFRSF1A, EFEMP2) via maximum clique centrality (MCC). Critically, TAGLN, CNN1, THBS1, and SMAD7 exhibited significant co-expression with VIM and CDH2 (correlation coefficients >0.1), implicating their roles in cytoskeletal protein assembly (TAGLN, CNN1), matricellular signaling (THBS1), and TGF-β pathway regulation (SMAD7). Functional validation confirmed these hub proteins as central to EMT plasticity, with TAGLN-VIM/CDH2 co-activation (r = 0.55 and 0.24, respectively) driving actin polymerization and protein-mediated invasion. SMAD7 further modulated TNFRSF1A-TNFAIP3 crosstalk to sustain mesenchymal phenotypes. This multi-omics framework delineates VIM/CDH2-centric protein interactomes as therapeutic vulnerabilities, proposing TAGLN/THBS1-targeted strategies to disrupt EMT-driven metastasis. By anchoring molecular subtyping and drug discovery in protein network topology, this work advances precision oncology for bladder cancer, bridging transcriptomic heterogeneity to actionable protein targets.
Collapse
Affiliation(s)
- Junkai Qin
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China
| | - Kecheng Liao
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China
| | - Lu Huang
- Department of Urology, Baidong Hospital, Affiliated Hospital of Youjiang Medical University for Nationalities and Key Laboratory of Molecular Pathology in Tumors of Baise, Baise 533000,China
| | - Rong Qiu
- Department of Urology, Baidong Hospital, Affiliated Hospital of Youjiang Medical University for Nationalities and Key Laboratory of Molecular Pathology in Tumors of Baise, Baise 533000,China
| | - Ning Tang
- Hepatobiliary Surgery Division, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang 524045, China
| | - Jun Wu
- Department of Urology, Baidong Hospital, Affiliated Hospital of Youjiang Medical University for Nationalities and Key Laboratory of Molecular Pathology in Tumors of Baise, Baise 533000,China.
| |
Collapse
|
11
|
Yang Q, Zhang Y, Zhang H, Yang Z, Feng Y, Ye B, Gong P, Qian G, Li D. Advanced N-glycoproteomics and proteomics approach revealed sexually dimorphic molecular signatures in primary mouse hepatocyte. Anal Bioanal Chem 2025:10.1007/s00216-025-05912-1. [PMID: 40410350 DOI: 10.1007/s00216-025-05912-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/25/2025]
Abstract
Sexual dimorphism plays a critical role in disease pathophysiology, but the subtlety and complexity of these differences, along with a lack of precise comparative methods, hinder the advancement of precision medicine and drug development. This limitation is particularly evident in metabolic dysfunction-associated steatotic liver disease (MASLD), where sex-specific molecular mechanisms remain insufficiently understood. To address this gap, we employed an advanced integrative N-glycoproteomics and proteomics approach to systematically analyze sex-biased molecular signatures in primary mouse hepatocytes (PMHs) under healthy and MASLD conditions. Our analysis identified 280 sex-biased proteins and 39 sex-biased N-glycosites, and KEGG enrichment revealed that female-biased molecules were primarily involved in lipid metabolism, while male-biased molecules were associated with inflammation and cytoskeletal remodeling. A combined dataset of 302 sex-biased molecules was further analyzed using protein-protein interaction (PPI) analysis and Rc value calculations, resulting in the identification of 21 hub proteins and 2 hub N-glycosites as MASLD-associated sex-biased signatures. Notably, MASLD amplified proteomic sex differences while attenuating them in N-glycosylation. Western blot validation of key signatures, including female-biased MVK and male-biased LGALS3, highlighted distinct molecular adaptations between the sexes in MASLD progression. Our study introduced an advanced analytical framework for high-resolution comparative molecular profiling by integrating N-glycoproteomics with proteomics, providing valuable insights into sex-biased molecular signatures, enhancing preclinical model development, and advancing sex-specific therapeutic strategies in MASLD research and broader biological systems.
Collapse
Affiliation(s)
- Qian Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yong Zhang
- Department of Nephrology, Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - He Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zi Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yanruyu Feng
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- Ninth People's Hospital of Zhengzhou, Zhengzhou, 45000, China
| | - Bengui Ye
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- Medical College of Tibet University, Lhasa, 850002, China
| | - Puyang Gong
- College of Pharmacy, Southwest Minzu University, Chengdu, 610041, China
| | - Guangsheng Qian
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Dapeng Li
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
12
|
Tang H, Zhang Y, Zhao D, Guo M, Yuan X, Wang X. Unlocking the lipid code: SREBPs as key drivers in gastrointestinal tumour metabolism. Lipids Health Dis 2025; 24:190. [PMID: 40413517 DOI: 10.1186/s12944-025-02612-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 05/15/2025] [Indexed: 05/27/2025] Open
Abstract
In recent years, metabolic reprogramming has emerged as a significant breakthrough in elucidating the onset and progression of gastrointestinal (GI) malignancies. As central regulatory hubs for lipid metabolism, sterol regulatory element binding proteins (SREBPs) integrate dietary metabolic signals and carcinogenic stimuli through subtype-specific mechanisms, thereby promoting malignant tumour phenotypes. In this review, we first present the molecular background, structural characteristics, and posttranscriptional regulatory networks associated with SREBPs. We subsequently describe a systematic analysis of the distinct activation patterns of SREBPs in liver, gastric, colorectal, and other gastrointestinal cancers. Furthermore, we explore targeted intervention strategies for different SREBP subtypes, including small molecule inhibitors (such as fatostatin, which inhibits SREBP cleavage), natural compounds (such as berberine, which modulates the AMPK/mTOR pathway), and statin-mediated inhibition of the mevalonic acid pathway. These strategies may enhance tumour cell sensitivity to chemotherapeutic agents (such as 5-FU, gezil, and tabine) and improve the response to synergistic chemoradiotherapy by reversing adaptive metabolic resistance driven by the tumour microenvironment. Through this review, we hope to provide new insights into precise interventions targeting various subtypes of the SREBP molecule.
Collapse
Affiliation(s)
- Haowen Tang
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuting Zhang
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Danni Zhao
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Minjie Guo
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiao Yuan
- Cancer Institute of Jiangsu University, Zhenjiang, China.
| | - Xu Wang
- Department of Thoracic Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
13
|
Xu J, Lu J, Gielzak M, Lilly Zheng S, Lu L, Wei J, Cornell B, Shi Z, Wang Q, Tran H, Engelmann V, Ashworth A, Lin K, Ross AE, Walsh PC, Marshall C, Luo J, Isaacs WB, Helfand BT, Pavlovich CP. Germline Testing for Prostate Cancer Patients: Evidence-Based Evaluation of Genes Recommended by NCCN Guidelines. Prostate 2025. [PMID: 40405569 DOI: 10.1002/pros.24918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/24/2025]
Abstract
BACKGROUND Approximately 50% of prostate cancer (PCa) patients meet the National Comprehensive Cancer Network (NCCN) guidelines for germline testing at diagnosis. However, the selection of genes for testing, their supporting evidence, and clinical interpretation remain poorly understood. METHODS An evidence-based evaluation of the recommended genes was conducted using data from the UK Biobank and Johns Hopkins School of Medicine, including 22,052 PCa patients and 191,055 unaffected controls. Association of germline pathogenic/likely pathogenic (P/LP) variants in each gene was tested using logistic regression, adjusting for age and genetic background. RESULTS Among the 11 NCCN-recommended PCa-related genes, significant associations (p < 0.0045) were identified between germline P/LP variants of five genes (HOXB13, BRCA2, ATM, CHEK2, and MSH2) and PCa risk. Additionally, BRCA2 and ATM variants were significantly associated with PCa aggressiveness. Of the 19 NCCN-recommended genes related to PARPi sensitivity, consistent evidence supported an enhanced response to PARPi therapy in patients with BRCA2 alterations, with weaker evidence for BRCA1, and limited supporting evidence for the remaining genes. Germline P/LP variants in BRCA2 and BRCA1 were observed in 0.77% and 0.14% of unselected PCa patients, respectively. Notably, no published study specifically assessed the efficacy of germline alterations, which were considerably rarer than somatic mutations. CONCLUSION Supporting statistical evidence is available for only a subset of the NCCN-recommended genes for germline testing. This evidence-based analysis may aid urologists-particularly those without specialized genetics training-in understanding germline testing for PCa risk assessment, prognosis, and treatment decision-making in clinical practice.
Collapse
Affiliation(s)
- Jianfeng Xu
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
- Division of Urology, Department of Surgery, Endeavor Health, Evanston, Illinois, USA
- University of Chicago Pritzker School of Medicine, Chicago, Illinois, USA
| | - Jim Lu
- GoPath Lab, LLC, Buffalo Grove, Illinois, USA
| | - Marta Gielzak
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Maryland, USA
| | - S Lilly Zheng
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
| | - Lucy Lu
- GoPath Lab, LLC, Buffalo Grove, Illinois, USA
| | - Jun Wei
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
| | | | - Zhuqing Shi
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
| | - Qiang Wang
- GoPath Lab, LLC, Buffalo Grove, Illinois, USA
| | - Huy Tran
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
| | | | - Annabelle Ashworth
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
| | - Kirk Lin
- Arizona Urology Specialists, Phoenix, Arizona, USA
| | - Ashley E Ross
- Department of Urology, Northwestern University Feinberg School of Medicine Northwestern, Chicago, Illinois, USA
| | - Patrick C Walsh
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Maryland, USA
| | | | - Jun Luo
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Maryland, USA
| | - William B Isaacs
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Maryland, USA
| | - Brian T Helfand
- Program for Genomic Translational Research, Endeavor Health, Evanston, Illinois, USA
- Division of Urology, Department of Surgery, Endeavor Health, Evanston, Illinois, USA
- University of Chicago Pritzker School of Medicine, Chicago, Illinois, USA
| | - Christian P Pavlovich
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Maryland, USA
| |
Collapse
|
14
|
Feng E, Feng E, Berg T, Nguyen IS, Nguyen LG, Chen W, Zhang M, Quigley D, Sharifi M, Li H, Coleman I, Nelson PS, Sjöström M, Zhao SG. Identifying prognostic targets in metastatic prostate cancer beyond AR. FEBS Open Bio 2025. [PMID: 40405591 DOI: 10.1002/2211-5463.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 05/01/2025] [Accepted: 05/13/2025] [Indexed: 05/24/2025] Open
Abstract
Genome-wide screens using CRISPR/RNAi can identify new therapeutic vulnerabilities in prostate cancer. In this study, we combine DepMap functional screen data with a large gene expression database (N = 1012) and clinical outcomes to identify potentially druggable targets. Eight genes (CYC, CYP51A1, DHFR, EBP, KIF15, PPM1D, SQLE, and UMPS) demonstrated strong dependency in cell lines and were also associated with worse prognosis clinically, representing potential therapeutic targets in metastatic prostate cancer. Four of these (DHFR, EBP, KIF15, and PPM1D) demonstrated higher expression in neuroendocrine prostate cancer. Furthermore, all but one (KIF15) were not significantly decreased from pretreatment to posttreatment, suggesting that they may remain targetable postabiraterone therapy. All eight genes showed evidence of protein expression in prostate cancers or cell lines. These potentially druggable targets associated with prostate cancer cell line dependency and worse clinical outcomes have also demonstrated literature support as potential targets, supporting further research into their potential clinical relevance as therapeutic targets in prostate cancer.
Collapse
Affiliation(s)
- Emily Feng
- Department of Radiation Oncology, University of California San Francisco, CA, USA
| | - Eric Feng
- Department of Radiation Oncology, University of California San Francisco, CA, USA
| | - Tracy Berg
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Isabella S Nguyen
- Department of Radiation Oncology, University of California San Francisco, CA, USA
| | - Lilac G Nguyen
- Department of Radiation Oncology, University of California San Francisco, CA, USA
| | - William Chen
- Department of Radiation Oncology, University of California San Francisco, CA, USA
| | - Meng Zhang
- Department of Radiation Oncology, University of California San Francisco, CA, USA
| | - David Quigley
- Department of Radiation Oncology, University of California San Francisco, CA, USA
| | - Marina Sharifi
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Haolong Li
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ilsa Coleman
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Peter S Nelson
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Martin Sjöström
- Department of Radiation Oncology, University of California San Francisco, CA, USA
- Lund University, Sweden
| | - Shuang G Zhao
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
15
|
Zang PD, Da Silva DM, Liu ZX, Kandukuri S, Tsao-Wei D, D’Souza A, Kast WM, Pal SK, Kefauver C, Juanqueira M, Yang L, Quinn DI, Dorff TB. Immune Modulation During Treatment with Enzalutamide Alone or with Radium-223 in Patients with Castration Resistant Prostate Cancer. Cancers (Basel) 2025; 17:1730. [PMID: 40427227 PMCID: PMC12110403 DOI: 10.3390/cancers17101730] [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: 03/29/2025] [Revised: 05/15/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
INTRODUCTION Prostate cancer has been generally resistant to immunotherapy approaches. Radiation can be immunostimulatory, but the extent to which standard prostate cancer treatments induce immune activation has not been well described. The bone-targeted radiopharmaceutical Radium223 (Ra223) has been proposed to enrich immune function, but clinical studies have not fully delineated whether this is true, or by what mechanisms. Enzalutamide has been shown to increase PD-L1 expression on dendritic cells, which could impact immune activation, though the extent to which this is associated with other evidence of immune activation remains uncertain, and combination strategies remain of interest. We performed a randomized phase II trial to evaluate whether Radium223 (Ra223) added to enzalutamide would induce greater immune activation and clinical responses compared to enzalutamide alone in men with metastatic castration-resistant prostate cancer (mCRPC). METHODS Eligible patients were randomized 2:1 to Arm A (enzalutamide 160 mg PO daily + Ra223 55 kBq/kg IV q4 weeks × 6 doses) or Arm B (enzalutamide 160 mg PO daily). Blood was collected at treatment start and during treatment to measure soluble immune checkpoint biomarkers (BTLA, TIM3, HVEM, GITR, LAG3, PD-1, CTLA-4, PD-L1, PD-L2, ICOS). Immunophenotyping by mass cytometry time of flight (CyTOF) was performed to measure peripheral blood mononuclear cell populations before and after treatment. CyTOF was used to determine changes in circulating immune cell population subsets before and after treatment. Biopsies were performed of an active bone metastatic lesion prior to study treatment and after at least 3 months. IHC was subsequently performed to examine changes in immune cell population subsets before and after treatment, and changes in pSTAT3 levels. RESULTS In total, 30 patients were enrolled, with median age 68. The median duration of follow up was 36 months. PSA responses, PFS, and OS were not significantly different between the two arms; however, the study was not powered for clinical endpoints. Peripheral blood and bone biopsy specimens were analyzed for immune correlatives. Soluble receptor concentrations showed significantly increased expression of PDL-2 in the combination arm, but this was not seen on CyTOF. Otherwise, there were no significant differences in markers of immune activation/exhaustion or immune cell population subsets in the combination arm and enzalutamide monotherapy arm. IHC also did not show a significant difference in immune cell population subsets in bone biopsy specimens before and after treatment in both arms. However, treatment with the combination arm did show significantly increased levels of pSTAT3 (p = 0.04), which was not seen in the enzalutamide monotherapy arm. CONCLUSIONS Our study showed an overall lack of evidence for immune activation or cytokine induction with the combination, which does not make a strong case for combinatorial immunotherapy approaches. However, the combination did induce higher levels of pSTAT3, which has been implicated in radio-resistance. Therefore, the addition of a STAT3 inhibitor to the combination may be of interest to improve efficacy.
Collapse
Affiliation(s)
- Peter D. Zang
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Diane M. Da Silva
- Department of Obstetrics & Gynecology, USC/Norris Comprehensive Cancer Center, 1450 Biggy Street, Los Angeles, CA 90033, USA
- Beckman Center for Immune Monitoring, USC/Norris Comprehensive Cancer Center, 1450 Biggy Street, Los Angeles, CA 90033, USA
| | - Zhang-Xu Liu
- Beckman Center for Immune Monitoring, USC/Norris Comprehensive Cancer Center, 1450 Biggy Street, Los Angeles, CA 90033, USA
| | - Shivani Kandukuri
- Clinical Pathology, USC/Norris Comprehensive Cancer Center, 1500 San Pablo Street, Los Angeles, CA 90033, USA
| | - Denice Tsao-Wei
- Department of Preventative Medicine, USC/Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Los Angeles, CA 90089, USA
| | - Anishka D’Souza
- Clinical Medicine, Department of Medical Oncology, USC/Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Los Angeles, CA 90089, USA
| | - W. Martin Kast
- Beckman Center for Immune Monitoring, USC/Norris Comprehensive Cancer Center, 1450 Biggy Street, Los Angeles, CA 90033, USA
- Department of Molecular Microbiology & Immunology and Urology, USC/Norris Comprehensive Cancer Center, 1450 Biggy Street, Los Angeles, CA 90033, USA
| | - Sumanta K. Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Cheryl Kefauver
- USC/Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Los Angeles, CA 90089, USA
| | - Maribel Juanqueira
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Lixin Yang
- Department of Pathology, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - David I. Quinn
- USC/Norris Comprehensive Cancer Center, 1450 Biggy Street, Los Angeles, CA 90033, USA
- Abbvie, 1000 Gateway Boulevard, South San Francisco, CA 94080, USA
| | - Tanya B. Dorff
- Department of Medical Oncology and Therapeutics Research, City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| |
Collapse
|
16
|
Cao Y, Zhao Z, Fang J, Lu Y, Huang Z, Wu G, Gao Q, Li R, Xu L, Xu X. Dual-Responsive Immunomodulatory RNAi Nanoplatform for Effective Immune Checkpoint Blockade and Enhanced Cancer Immunotherapy. Adv Healthc Mater 2025:e2500646. [PMID: 40394949 DOI: 10.1002/adhm.202500646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/24/2025] [Indexed: 05/22/2025]
Abstract
Immune checkpoint blockade (ICB) therapy has become the first-line treatment for cancer patients. However, the low response rate remains a clinical pain-point. Anti-hyperglycemic drug metformin has shown remarkable anticancer effect with the unique characteristic of modulating tumor immune microenvironment (TIME). Therefore, combining ICB with metformin could be a promising strategy for enhanced cancer immunotherapy, which however remains challenged due to the low bioavailability and severe adverse effects of metformin. This work herein designs an amphiphilic reduction-responsive metformin prodrug, which could complex small interfering RNA (siRNA) and then co-assemble with an endosomal pH-responsive PEGylated polymer to form a dual-responsive immunomodulatory RNAi nanoplatform. Using the orthotopic and metastatic breast cancer (BCa) tumor models, this work demonstrates that this RNAi nanoplatform could silence PD-L1 expression on BCa cells and suppress their proliferation via activating AMP-activated protein kinase (AMPK). Moreover, this AMPK activation could suppress the secretion of tumor-derived transforming growth factor β (TGF-β) and interleukin 6 (IL-6), which could enhance the maturation of dendritic cells (DCs) and activation of CD8+ T cells and impair the tumor infiltration of regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs), ultimately achieving the goal of enhanced cancer immunotherapy and significant inhibition of BCa tumor growth.
Collapse
Affiliation(s)
- Yuan Cao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Zixuan Zhao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, Institute of Pharmacy and Pharmacology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Junyue Fang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Yanan Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Zhuoshan Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Guo Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Qiyuan Gao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Rong Li
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, Institute of Pharmacy and Pharmacology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Lei Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, Institute of Pharmacy and Pharmacology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| |
Collapse
|
17
|
Zhang ZC, Shen Y, Lin YS, Yang B, Cao J, Li J, Zhao WB. Peptide-MHC I regulatory mechanisms and intervention strategies in anti-tumor T cell immunity. Acta Pharmacol Sin 2025:10.1038/s41401-025-01574-y. [PMID: 40379886 DOI: 10.1038/s41401-025-01574-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/22/2025] [Indexed: 05/19/2025]
Abstract
T cell immune responses are triggered by antigenic peptides presented through major histocompatibility complex class Is (pMHC-Is), which play an important role in the genesis, development, and therapy of tumors. The capacity of a specific pMHC-I to elicit T cell responses is deeply influenced by its expression level (quantity) and its immunogenicity (quality). Tumor cells can evade T cell immunity by down-regulating the quantity of pMHC-Is or selectively eliminating highly immunogenic antigenic peptides. Augmenting the quantity or quality of pMHC-Is is essential for tumor immunotherapy. However, the complexity of pMHC-I regulation and tumor heterogeneity pose challenges to clinical strategies. Consequently, developing approaches grounded in comprehensive analyses of pMHC-I regulatory mechanisms remains a focal point in the research of T cell immunity. In this review, we discuss how tumors modulate their surface pMHC-Is through genetic, epigenetic, and proteomic mechanisms and summarize potential therapeutic strategies targeting these mechanisms, which may provide a valuable reference for the development of novel tumor immunotherapies based on pMHC-I modulation. Tumor cells can achieve immune escape by interfering with the quantity and quality of pMHC-Is, and corresponding immunotherapy can also be achieved by the regulation of pMHC-Is.
Collapse
Affiliation(s)
- Zhi-Chao Zhang
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ying Shen
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310018, China
| | - Yu-Shen Lin
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Yang
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310018, China
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Ji Cao
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China.
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310018, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| | - Jun Li
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, 310009, China.
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, 310000, China.
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Wen-Bin Zhao
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China.
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310018, China.
| |
Collapse
|
18
|
Tamarindo GH, Ribeiro CF, Rodrigues S, Góes RM, Loda M. DHA suppresses hormone-sensitive and castration-resistant prostate cancer growth by decreasing de novo lipogenesis. Biochim Biophys Acta Mol Cell Biol Lipids 2025:159634. [PMID: 40383250 DOI: 10.1016/j.bbalip.2025.159634] [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: 02/25/2025] [Revised: 05/06/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
OBJECTIVE De novo lipogenesis (DNL) is associated with prostate cancer (PCa) progression, while fatty acid synthase (FASN) overexpression is a hallmark of DNL. Palmitate, its main product, is a saturated fatty acid that supports PCa growth. Polyunsaturated fatty acids (PUFAs), which can be acquired from the microenvironment, undergo peroxidation more readily and affect membrane fluidity. Docosahexaenoic acid (DHA) is a prototype PUFA omega-3 produced inefficiently in human cells. Its levels are low in PCa cells compared to normal cells. We hypothesize that excess DHA may reprogram lipid metabolism and induce cell growth suppression. METHODS Androgen-responsive LNCaP, castration-resistant cells C4-2 and 22Rv1, human PCa castration-resistant organoids, and prostate cancer xenografts were exposed to DHA. RESULTS DHA accumulated into lipid droplets as triacylglycerols and cholesterol esters, led to increased phospholipid acyl chain unsaturation and altered phospholipid ratio, a known trigger of endoplasmic reticulum (ER) stress. DHA caused a decrease in sterol regulatory element-binding protein (SREBP) transcriptional program, which, in turn, led to decreased expression of FASN. The subsequent reduction in DNL caused downregulation of the androgen receptor (AR) and its splice variant AR-V7. In addition, β-oxidation was enhanced, and DHA was preferentially oxidized over palmitate. Glucose oxidation also increased in the presence of DHA. Finally, DHA led to ROS overproduction, oxidative damage, and ER stress. CONCLUSIONS DHA reduces the growth of hormone-sensitive and castration-resistant PCa both in vitro and in vivo via deregulation of lipid metabolism.
Collapse
Affiliation(s)
- G H Tamarindo
- Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil; Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil; Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São Paulo, Brazil
| | - C F Ribeiro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, United States of America
| | - S Rodrigues
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, United States of America
| | - R M Góes
- Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil; Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São Paulo, Brazil
| | - M Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, United States of America; Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States of America; Nuffield Department of Surgical Sciences, Lincoln College, University of Oxford, Oxford, UK.
| |
Collapse
|
19
|
Cao Y, Wang J, Luo H, Wang Y, Cai X, Zhang T, Liao Y, Wang D. Role of exosomes in castration-resistant prostate cancer. Front Oncol 2025; 15:1498733. [PMID: 40438694 PMCID: PMC12116360 DOI: 10.3389/fonc.2025.1498733] [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: 09/26/2024] [Accepted: 04/18/2025] [Indexed: 06/01/2025] Open
Abstract
Prostate cancer (PCa) is one of the most common urological malignancies in older male patients. Castration-resistant prostate cancer (CRPC) is an aggressive and refractory stage of PCa and is the leading cause of PCa-related deaths. Exosomes are small spherical vesicles with a lipid bilayer membrane structure, secreted by cells, which carry large amounts of nucleic acids, proteins, lipids, and various important reactive small molecules. Numerous studies have demonstrated that exosomes are involved in the development of CRPC by delivering various biomolecules that regulate biological processes in recipient cells. Despite the advancement in treatments, CRPC remains poorly managed, underscoring the urgent need for novel treatment strategies.As research into exosomes continues, they have shown significant potential in the diagnosis and treatment of CRPC.Unlike previous reviews,this review not only provides an overview of exosomes but also comprehensively explores their role in the CRPC tumor microenvironment, angiogenesis, immune escape, metastasis, and drug resistance, with a focus on the potential value of exosomes in the diagnosis and treatment of CRPC.The literature review includes studies published up to June 2024, and the search strategy involved exosomes, CRPC, diagnosis,and treatment using Pubmed.
Collapse
Affiliation(s)
- Yuan Cao
- Department of Urology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Jianjun Wang
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- Department of Hepatobiliary Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Huiwen Luo
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Yaodong Wang
- Department of Urology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Xianfu Cai
- Department of Urology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Tiansheng Zhang
- Department of Urology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Yougang Liao
- Department of Urology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Decai Wang
- Department of Urology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
20
|
Dang B, Liang L, Li Z, Luo J, Zhong S. Bolstering CD8 + T Cells' Antitumor Immunity: A Promising Strategy to Improve the Response to Advanced Prostate Cancer Treatment. BIOLOGY 2025; 14:544. [PMID: 40427733 PMCID: PMC12108615 DOI: 10.3390/biology14050544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Revised: 05/07/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025]
Abstract
Prostate cancer is among the most frequently diagnosed and deadly cancers among men in the Western world. It is typically classified as an immune "cold" tumor due to its sparse immune cell presence and limited immunogenic response. Recent research has revealed the significant role of immune cells, especially CD8+ T cells, in both prostate cancer progression and treatment efficacy. This review integrates recent findings to provide a comprehensive overview of the current understanding of CD8+ T cell dynamics in prostate cancer and discusses emerging strategies to improve treatment outcomes. The ongoing exploration of new molecular targets and the development of innovative immunotherapeutic approaches hold promise for more effective management of prostate cancer, particularly in the context of advanced and resistant forms of the disease.
Collapse
Affiliation(s)
| | | | | | | | - Shangwei Zhong
- The Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang 421009, China; (B.D.); (L.L.); (Z.L.); (J.L.)
| |
Collapse
|
21
|
Kearns B, McKell A, Steveson I, Worley P, Barton B, Bennett J, Anderson D, Harris J, Christensen J, Barrott JJ. ARID1A and Its Impact Across the Hallmarks of Cancer. Int J Mol Sci 2025; 26:4644. [PMID: 40429787 PMCID: PMC12111594 DOI: 10.3390/ijms26104644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
ARID1A, a subunit of the SWI/SNF chromatin remodeling complex, has emerged as a pivotal tumor suppressor altered in a broad range of human malignancies. Its frequent inactivation across diverse cancer types has revealed pleiotropic roles that intersect multiple Hallmarks of Cancer. In this review, we integrate current knowledge on how ARID1A loss influences cellular processes including proliferative signaling, resistance to cell death, genomic instability, metabolic reprogramming, immune evasion, and more. We discuss the context-specific consequences of ARID1A deficiency, its cooperation with other oncogenic events, and its implications for therapeutic vulnerability-particularly in the realm of synthetic lethality and immune modulation. By mapping ARID1A's functional impact onto the established hallmarks framework, we highlight its centrality in cancer biology and underscore opportunities for biomarker-driven strategies and targeted interventions. Understanding ARID1A's multifaceted roles offers a compelling lens through which to explore chromatin dysregulation in cancer and guide translational advances.
Collapse
Affiliation(s)
- Bridger Kearns
- Department of Cell Biology & Physiology, Brigham Young University, Provo, UT 84602, USA; (B.K.); (A.M.); (I.S.); (P.W.); (B.B.); (J.B.); (D.A.); (J.H.); (J.C.)
- Simmons Center for Cancer Research, Brigham Young University, Provo, UT 84602, USA
| | - Andralyn McKell
- Department of Cell Biology & Physiology, Brigham Young University, Provo, UT 84602, USA; (B.K.); (A.M.); (I.S.); (P.W.); (B.B.); (J.B.); (D.A.); (J.H.); (J.C.)
| | - Isaac Steveson
- Department of Cell Biology & Physiology, Brigham Young University, Provo, UT 84602, USA; (B.K.); (A.M.); (I.S.); (P.W.); (B.B.); (J.B.); (D.A.); (J.H.); (J.C.)
| | - Peyton Worley
- Department of Cell Biology & Physiology, Brigham Young University, Provo, UT 84602, USA; (B.K.); (A.M.); (I.S.); (P.W.); (B.B.); (J.B.); (D.A.); (J.H.); (J.C.)
| | - Braeden Barton
- Department of Cell Biology & Physiology, Brigham Young University, Provo, UT 84602, USA; (B.K.); (A.M.); (I.S.); (P.W.); (B.B.); (J.B.); (D.A.); (J.H.); (J.C.)
| | - Jordan Bennett
- Department of Cell Biology & Physiology, Brigham Young University, Provo, UT 84602, USA; (B.K.); (A.M.); (I.S.); (P.W.); (B.B.); (J.B.); (D.A.); (J.H.); (J.C.)
| | - DeLaney Anderson
- Department of Cell Biology & Physiology, Brigham Young University, Provo, UT 84602, USA; (B.K.); (A.M.); (I.S.); (P.W.); (B.B.); (J.B.); (D.A.); (J.H.); (J.C.)
- Simmons Center for Cancer Research, Brigham Young University, Provo, UT 84602, USA
| | - Jacob Harris
- Department of Cell Biology & Physiology, Brigham Young University, Provo, UT 84602, USA; (B.K.); (A.M.); (I.S.); (P.W.); (B.B.); (J.B.); (D.A.); (J.H.); (J.C.)
| | - James Christensen
- Department of Cell Biology & Physiology, Brigham Young University, Provo, UT 84602, USA; (B.K.); (A.M.); (I.S.); (P.W.); (B.B.); (J.B.); (D.A.); (J.H.); (J.C.)
| | - Jared J. Barrott
- Department of Cell Biology & Physiology, Brigham Young University, Provo, UT 84602, USA; (B.K.); (A.M.); (I.S.); (P.W.); (B.B.); (J.B.); (D.A.); (J.H.); (J.C.)
- Simmons Center for Cancer Research, Brigham Young University, Provo, UT 84602, USA
| |
Collapse
|
22
|
Peng S, Hou X, Liu J, Huang F. Advances in polymer nanomaterials targeting cGAS-STING pathway for enhanced cancer immunotherapy. J Control Release 2025; 381:113560. [PMID: 40023225 DOI: 10.1016/j.jconrel.2025.02.056] [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: 09/13/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
Cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) pathway has been recognized as a promising target for cancer immunotherapy. Although various STING agonists have been developed, their clinical applications are still severely impeded by various issues, such as non-specific accumulation, adverse effects, rapid clearance, etc. In recent years, the emergence of nanomaterials has profoundly revolutionized STING agonists delivery, which promote tumor-targeted delivery, boost the immunotherapeutic effects and reduce systemic toxicity of STING agonists. In particular, polymer nanomaterials possess inherent advantages including controllable structure, tunable function and degradability. These properties afford them the capacity to serve as delivery vehicles for small-molecule STING agonists. Furthermore, the superior characteristics of polymer nanomaterials can enable their utilization as a novel STING agonist to stimulate anti-tumor immunity. In this review, the molecular mechanisms of cGAS-STING pathway activation are discussed. The recent development of small-molecules STING agonists is described. Then polymer nanomaterials are discussed as carriers for STING agonists in cancer immunotherapy, including polymersomes, polymer micelles, polymer capsules, and polymer nanogels. Additionally, polymer nanomaterials are identified as a novel class of STING agonists for efficient cancer immunotherapy, encompassing both polymer materials and polymer-STING agonists conjugates. The review also presents the combination of polymer-based cGAS-STING immunotherapy with chemotherapy, radiotherapy, phototherapy (both photodynamic and photothermal), chemodynamic therapy, and other therapeutic strategies. Furthermore, the discussion highlights recent advancements targeting the cGAS-STING pathway in clinically approved polymer nanomaterials and corresponding potent innovations. Finally, the potential challenges and perspectives of polymer nanomaterials for activating cGAS-STING pathway are outlined, emphasizing the critical scientific issue and hoping to offer guidance for their clinical translation.
Collapse
Affiliation(s)
- Shiyu Peng
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Xiaoxue Hou
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Jianfeng Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| | - Fan Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
23
|
Jiang Y, Liang X, Sun H, Yin P, Zhou J, Yu C. Immunomodulatory role of RNA modifications in sex hormone-dependent cancers. Front Immunol 2025; 16:1513037. [PMID: 40406121 PMCID: PMC12095187 DOI: 10.3389/fimmu.2025.1513037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Recent studies have identified that RNA epigenetic modifications, including m6A, m1A, m5C, etc, play pivotal roles in tumor progression. These modifications influence mRNA stability, RNA processing, translational efficiency, and decoding precision. However, comprehensive reviews detailing the connection between m6A RNA modifications and hormone-dependent cancers in both male and female populations remain scarce(breast cancer, ovarian cancer, and endometrial cancer, prostate cancer). In this article, we explore the cellular and molecular roles of various RNA modifications alongside the key elements of the tumor microenvironment. We examine how these RNA modifications influence the development of hormone-dependent cancers through their impact on immune mechanisms. By enhancing our understanding of the function of RNA modifications within the immune systems of four specific tumors, we offer fresh insights for their potential applications in diagnosis and treatment.
Collapse
Affiliation(s)
- Yujia Jiang
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiaolan Liang
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Hongyi Sun
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Ping Yin
- Department of Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Zhou
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chaoqin Yu
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
24
|
Qin F, Zheng H, Wu J, Liu Z, Zheng Y, Yang X, Chen J, Deng W, Luo Z, Tan J, Cai W, Jian B, Zeng Y, Qin X, Liao H. APOC1 expressed in macrophages promotes the pulmonary metastasis of colorectal cancer via CCL2/CCL5. Int Immunopharmacol 2025; 154:114611. [PMID: 40194454 DOI: 10.1016/j.intimp.2025.114611] [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: 03/16/2025] [Accepted: 03/31/2025] [Indexed: 04/09/2025]
Abstract
Metastasis is the main cause of death in colorectal cancer (CRC), and the lungs are common sites of metastasis. However, there is little effective target to intervene colorectal cancer pulmonary metastasis (CCPM), especially on its unique immune microenvironment. In this study, sixteen genes were identified as core CCPM-related differentially expressed genes (DEGs) between CRC and CCPM. Three genes including Apolipoprotein C1 (APOC1) were associated with prognosis, stage and metastasis of CRC. In immunohistochemistry, APOC1 was mainly expressed in macrophages, and expressed more in CCPM than CRC. Patients with synchronous CCPM, higher stage, poorer OS and CCPM-free interval tended to have higher expression. In experiments in vitro, knockdown of APOC1 in macrophages reduced the migration, invasion, and epithelial-mesenchymal transition of CRC cells. Knockdown of APOC1 in macrophages significantly decreased secretion of chemokines like CCL2 and CCL5. The pro-metastatic effect of macrophages expressing APOC1 was partially blocked by the antibodies of CCL2 and CCL5. Activation of STAT3 was a key process in APOC1's regulation of CCL2 and CCL5. In experiments in vivo, knockdown of APOC1 in macrophages reduced pulmonary metastasis. To conclude, APOC1 is one of core CCPM-related DEGs and associated with the metastasis and survival of CRC. Macrophages expressing APOC1 promote the CCPM by APOC1-STAT3-CCL2/CCL5 axis. APOC1 and macrophages expressing APOC1 play vital roles and may be potential therapeutic targets in CCPM.
Collapse
Affiliation(s)
- Fei Qin
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Haosheng Zheng
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Jiayan Wu
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Zui Liu
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Yuzhen Zheng
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Xingping Yang
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Junguo Chen
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Weihao Deng
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Ziyin Luo
- Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Jian Tan
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Weijie Cai
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Bozhu Jian
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Yushuai Zeng
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Xianyu Qin
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China.
| | - Hongying Liao
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China.
| |
Collapse
|
25
|
Gao Y, Zhang X, Xia S, Chen Q, Tong Q, Yu S, An R, Cheng C, Zou W, Liang L, Xie X, Song Z, Liu R, Zhang J. Spatial multi-omics reveals the potential involvement of SPP1 + fibroblasts in determining metabolic heterogeneity and promoting metastatic growth of colorectal cancer liver metastasis. Mol Ther 2025:S1525-0016(25)00374-0. [PMID: 40340245 DOI: 10.1016/j.ymthe.2025.05.004] [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: 12/20/2024] [Revised: 03/01/2025] [Accepted: 05/03/2025] [Indexed: 05/10/2025] Open
Abstract
This study investigates key microscopic regions involved in colorectal cancer liver metastasis (CRLM), focusing on the crucial role of cancer-associated fibroblasts (CAFs) in promoting tumor progression and providing molecular- and metabolism-level insights for its diagnosis and treatment using multi-omics. We followed 12 fresh surgical samples from 2 untreated CRLM patients. Among these, 4 samples were used for spatial transcriptomics (ST), 4 for spatial metabolomics, and 4 for single-cell RNA sequencing (scRNA-seq). Additionally, 92 frozen tissue samples from 40 patients were collected. Seven patients were used for immunofluorescence and RT-qPCR, while 33 patients were used for untargeted metabolomics. ST revealed that the spatial regions of CRLM consists of 7 major components, with fibroblast-dominated regions being the most prominent. These regions are characterized by diverse cell-cell interactions, and immunosuppressive and tumor growth-promoting environments. scRNA-seq identified that SPP1+ fibroblasts interact with CD44+ tumor cells, as confirmed through immunofluorescence. Spatial metabolomics revealed suberic acid and tetraethylene glycol as specific metabolic components of this structure, which was further validated by untargeted metabolomics. In conclusion, an SPP1+ fibroblast-rich spatial region with metabolic reprogramming capabilities and immunosuppressive properties was identified in CRLM, which potentially facilitates metastatic outgrowth through interactions with tumor cells.
Collapse
Affiliation(s)
- Yuzhen Gao
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China
| | - Xiuping Zhang
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing 100853, P.R. China
| | - Shenglong Xia
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Qing Chen
- Institute of Respiratory Diseases, Department of Basic Medicine, Xiamen Medical College, Xiamen 361023, Fujian, China; Organiod Platform of Medical Laboratory Science, Department of Basic Medicine, Xiamen Medical College, Xiamen 361023, Fujian, China
| | - Qingchao Tong
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Shaobo Yu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Rui An
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Cheng Cheng
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Wenbo Zou
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing 100853, P.R. China
| | - Leilei Liang
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou Zhejiang, China
| | - Xinyou Xie
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China.
| | - Rong Liu
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing 100853, P.R. China.
| | - Jun Zhang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou 310016, Zhejiang, China.
| |
Collapse
|
26
|
Yang X, Li L, Li R, Li X, Li S, Su C, Liao H. Arginine N-glycosylation of melittin enhances its bacteriostatic activity and antiproliferative therapeutic index. Org Biomol Chem 2025; 23:4471-4479. [PMID: 40223544 DOI: 10.1039/d5ob00398a] [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: 04/15/2025]
Abstract
Melittin is a natural antimicrobial peptide isolated from bee venom, and the non-specific cytotoxicity and hemolytic activity severely limit its clinical application. Glycosylation of proteins is very common in physiological and biochemical processes and can modulate the interaction of proteins with their target. In this study, eight glycosyl groups were used to modify the arginine of melittin at sites 22 and/or 24, and single and double arginine N-glycosylated peptides were designed and synthesized. Among the acquired 24 glycopeptides, MLT-1c, MLT-3c, MLT-1f, MLT-3f, MLT-1g, and MLT-3h were found to possess higher helicity, while MLT-3c, MLT-3f and MLT-3h showed dramatically reduced hemolytic activity, especially MLT-3c, whose HC50 value is 199.3 μM. MLT-1a, MLT-3a and MLT-2c exhibited improved inhibitory activity against Puzza streptococcus, and the MIC was 4 μg mL-1. MLT-1e and MLT-2g have the strongest tolerance to trypsase, and MLT-3c has the highest therapeutic index. In general, rhamnosyl-modified melittin MLT-3c could be a potent agent for antibacterial and antitumor therapy with high stability and low hemolytic side effects.
Collapse
Affiliation(s)
- Xiantao Yang
- School of Pharmacy, Chengdu Medical College, 783 Xindu Avenue, Xindu District, Chengdu 610500, China.
| | - Linji Li
- School of Pharmacy, Chengdu Medical College, 783 Xindu Avenue, Xindu District, Chengdu 610500, China.
| | - Rong Li
- Pidu area center, Chengdu Institute of Food Inspection, 456 Yong'an West Rd., Ande Street, Pidu District, Chengdu 611730, China
| | - Xiang Li
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Shuna Li
- Department of Otorhinolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Rd., Shanghai, 200092, China.
| | - Chunli Su
- School of Public Health, Chengdu Medical College, 783 Xindu Avenue, Xindu District, Chengdu 610500, China.
| | - Hongli Liao
- School of Pharmacy, Chengdu Medical College, 783 Xindu Avenue, Xindu District, Chengdu 610500, China.
| |
Collapse
|
27
|
Shalaby SM, Shawky SA, Ashour H, Sarhan W. The interplay between COX-2, chemotherapeutic drugs, and chemoresistance in colon cancer. Sci Rep 2025; 15:15837. [PMID: 40328989 PMCID: PMC12056169 DOI: 10.1038/s41598-025-98451-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
Chemoresistance and tumor relapse remain major clinical problems. Evidence indicates that COX2/PGE2/EP axis has a critical role in tumorogenesis and chemoresistance. This study assessed the relation of the COX-2 gene expression with chemoresistance in colon cancer (CC) patients. Also, it explored the effect of chemotherapy on COX-2 expression. The study included 24 patients with CC without chemotherapeutic treatment and 24 chemoresistant CC patients. Tumor and adjacent non-neoplastic colon tissue samples were collected and COX-2 mRNA expression was measured. Also, COX-2 and its related genes; TROP2 and DUSP4 expression were analysed in 5 flurouracil and Oxalliplatin treated Caco-2 and SW-620 cells. The results indicated significant upregulation of COX-2 expression in tissues of chemoresistant CC patients when compared with that in CC tissues without chemotherapy (p < 0.001). There was a relation between COX-2 expression with lymph nodes, metastases and staging in both groups. Concerning in-vitro experiments, there was a dose dependent significant increase of COX-2, TROP2 and DUSP4 mRNA and protein expression levels in 5flurouracil and Oxalliplatin treated cells. These findings demonstrated that overexpression of COX-2 in the chemoresistant CC patients. Both 5 flurouracil and Oxalliplatin induced COX-2 overexpression and in turn COX-2 upregulation may decrease the response of cancer to chemotherapy.
Collapse
Affiliation(s)
- Sally M Shalaby
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Salma A Shawky
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Hassan Ashour
- Surgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Walaa Sarhan
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
28
|
Adzavon YM, Culig Z, Sun Z. Interactions between androgen and IGF1 axes in prostate tumorigenesis. Nat Rev Urol 2025; 22:268-275. [PMID: 39375467 DOI: 10.1038/s41585-024-00942-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/09/2024]
Abstract
Androgen signalling through the androgen receptor (AR) is essential for prostate tumorigenesis. However, androgen signalling pathways also interact with other growth factor-mediated signalling pathways to regulate the prostatic cell cycle, differentiation, apoptosis and proliferation in the initiation and progression of prostate cancer. Insulin-like growth factor 1 (IGF1) is one of the most prominent growth factors in prostate tumorigenesis. Clinical and experimental evidence has demonstrated that IGF1 signalling supports both androgen-dependent and androgen-independent prostate tumorigenesis, suggesting that improved understanding of the interactions between the IGF1 and androgen axes might aid the development of new therapeutic strategies. Available data have shown a dynamic role of androgen-AR signalling in the activation of IGF1-signalling pathways by augmenting transcription of the IGF1 receptor in prostatic basal epithelial cells and by increasing IGF1 secretion through the suppression of IGF-binding protein 3 expression in prostatic stromal cells. In turn, IGF1 stimulates Wnt-β-catenin signalling in prostatic basal progenitors to promote prostatic oncogenic transformation and prostate cancer development. These findings highlight the cooperative, autocrine and paracrine interactions that underlie the oncogenic effects of androgens and IGF1 and open up new opportunities for therapeutic targeting.
Collapse
Affiliation(s)
- Yao Mawulikplimi Adzavon
- Department of Cell Biology, Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Oncology, Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Zoran Culig
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Zijie Sun
- Department of Cell Biology, Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
29
|
Guo S, Zhang L, Ren J, Lu Z, Ma X, Liu X, Jin H, Li J. The roles of enhancer, especially super-enhancer-driven genes in tumor metabolism and immunity. Int J Biol Macromol 2025; 308:142414. [PMID: 40132720 DOI: 10.1016/j.ijbiomac.2025.142414] [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: 10/16/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
Abnormal metabolism is a characteristic of malignant tumors. Numerous factors play roles in the regulation of tumor metabolism. As epigenetic regulators, enhancers, especially the super-enhancers (SEs), serve as platforms for transcription factors that regulate the expression of metabolism-related enzymes or transporters at the gene level. In this study, we review the effects of enhancer/ SE-driven genes on tumor metabolism and immunity. Enhancers/SEs play regulatory roles in glucose metabolism (glycolysis, gluconeogenesis, tricarboxylic acid (TCA) cycle, pyruvate, and pentose phosphate pathway, lipid metabolism (cholesterol, fatty acid, phosphatide, and sphingolipid), and amino acid metabolism (glutamine, tryptophan, arginine, and cystine). By regulating tumor metabolism, enhancers and SEs can reprogram tumor microenvironment, especially the status of various immune cells. Therefore, interfering enhancers/SEs that regulate the tumor metabolism is likely to enhance the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Songyue Guo
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Lu Zhang
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Jiao Ren
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Zhong Lu
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Xiaolin Ma
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Xinling Liu
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China.
| | - Hongchuan Jin
- Department of Medical Oncology, Cancer Center of Zhejiang University, Sir Run Run Shaw hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, China.
| | - Jiaqiu Li
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong, China; Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, Shandong, China.
| |
Collapse
|
30
|
Liang HQ, He QH, Wei QJ, Mo QZ, Yang GL, Wei FY, Wei LW, Li YJ, Qin M, Cheng JW. CTHRC1 expresses in cancer-associated fibroblasts and is associated with resistance to anti-androgen therapy in prostate cancer. Genes Genomics 2025; 47:541-557. [PMID: 40009323 DOI: 10.1007/s13258-025-01624-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND CTHRC1 overexpresses in prostate cancer and is associated with the proliferation, invasion and migration of prostate cancer cells. However, the roles and mechanisms of CTHRC1 expression in prostate cancer prognosis and treatment outcomes remain unknown. OBJECTIVE This study aimed to explore the expression and gene function of CTHRC1 in prostate cancer, investigate the prognostic value and potential effect in the treatment of prostate cancer. METHODS Bulk and single-cell RNA sequencing analyses were used to evaluate the expression of CTHRC1 in prostate cancer. All data used in the study were obtained from publicly available sources to ensure transparency. Study enrolled 1999 cases of prostate cancer and 531 normal controls. Single-cell RNA sequencing profile included 62,995 cells from seven prostate primary tumors. CTHRC1 expression and prognosis analyses were conducted with these samples and verified by immunohistochemical staining. CIBERSORT algorithm was used to assess the tumor immune infiltrating cells based on bulk mRNA sequencing profiles. Genomics of drug sensitivity in cancer (GDSC) database was used to predict IC50 to anti-androgen therapy (ADT) drugs of the samples. RESULTS CTHRC1 expressed in prostate cancer was higher than that in normal prostate tissue, and the expression increased with the progress of prostate cancer. CTHRC1 was the risk factor of progression-free interval (PFI). CTHRC1 was positively correlated with the infiltration of tumor-associated macrophages (TAMs). Myofibroblast-like cancer-associated fibroblasts (myCAFs) were the major CTHRC1 expressers in prostate cancer. TGF-β signaling activated in CTHRC1-positive myCAFs and was involved in TAMs polarization. Biological functions of myCAFs were enriched in hormone response and metabolism. CTHRC1 may regulate androgen receptor signaling through CCN2/CAV1/AR pathway. Moreover, ADT drug Bicalutamide and AZD3514 were less sensitive in the high CTHRC1 expression tumors. CONCLUSIONS As a potential molecular target of ADT resistance in prostate cancer, CTHRC1 provides a new promising molecular approach for the diagnosis and treatment of prostate cancer.
Collapse
Affiliation(s)
- Hai-Qi Liang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, China
| | - Qi-Huan He
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, China
| | - Qiu-Ju Wei
- Guangxi Medical University, Nanning, China
| | - Qi-Zhou Mo
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, China
| | - Guang-Lin Yang
- Department of Urology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fa-Ye Wei
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, China
| | - Li-Wei Wei
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, China
| | - Yu-Jian Li
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, China
| | - Min Qin
- Human Sperm Bank, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, China.
| | - Ji-Wen Cheng
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, China.
| |
Collapse
|
31
|
Li Y, Wang B, Zheng Y, Kang H, He A, Zhao L, Guo N, Liu H, Mardinoglu A, Mamun M, Gao Y, Chen X. The multifaceted role of post-translational modifications of LSD1 in cellular processes and disease pathogenesis. Genes Dis 2025; 12:101307. [PMID: 40028036 PMCID: PMC11870172 DOI: 10.1016/j.gendis.2024.101307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/31/2024] [Accepted: 03/13/2024] [Indexed: 03/05/2025] Open
Abstract
Post-translational modifications (PTMs) of proteins play a crucial role in living organisms, altering the properties and functions of proteins. There are over 450 known PTMs involved in various life activities. LSD1 (lysine-specific demethylase 1) is the first identified histone demethylase that can remove monomethylation or dimethylation modifications from histone H3 lysine K4 (H3K4) and histone H3 lysine K9 (H3K9). This ability of LSD1 allows it to inhibit or activate transcription. LSD1 has been found to abnormally express at the protein level in various tumors, making it relevant to multiple diseases. As a PTM enzyme, LSD1 itself undergoes various PTMs, including phosphorylation, acetylation, ubiquitination, methylation, SUMOylation, and S-nitrosylation, influencing its activity and function. Dysregulation of these PTMs has been implicated in a wide range of diseases, including cancer, metabolic disorders, neurological disorders, cardiovascular diseases, and bone diseases. Understanding the species of PTMs and functions regulated by various PTMs of LSD1 provides insights into its involvement in diverse physiological and pathological processes. In this review, we discuss the structural characteristics of LSD1 and amino acid residues that affect its enzyme activity. We also summarize the potential PTMs that occur on LSD1 and their involvement in cellular processes. Furthermore, we describe human diseases associated with abnormal expression of LSD1. This comprehensive analysis sheds light on the intricate interplay between PTMs and the functions of LSD1, highlighting their significance in health and diseases.
Collapse
Affiliation(s)
- Yinrui Li
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou, Henan 450001, China
- Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Bo Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou, Henan 450001, China
- Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yichao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou, Henan 450001, China
- Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Huiqin Kang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou, Henan 450001, China
- Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ang He
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou, Henan 450001, China
- Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Lijuan Zhao
- Henan Institute of Medical and Pharmaceutical Sciences, State Key Laboratory for Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ningjie Guo
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou, Henan 450001, China
- Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Hongmin Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou, Henan 450001, China
- Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm SE-100 44, Sweden
- Faculty of Dentistry, Oral & Craniofacial Sciences, Centre for Host-Microbiome Interactions, King's College London, London WC2R 2LS, UK
| | - M.A.A. Mamun
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou, Henan 450001, China
- Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ya Gao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality Control and Evaluation, Zhengzhou, Henan 450001, China
- Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xiaobing Chen
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer & Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan 450008, China
| |
Collapse
|
32
|
Xu J, Dong X, Peng Y, Yan J, Xu P, Li F, Zhang S, Chen L, Meng X, Wang K, Xing M, Li W, Huang DCS, Zhao Q, Chen B. ZMYND8 Reads H3K36me2 to Activate CEBPE Transcription and Suppress Multiple Myeloma Progression through the Inhibition of Adaptive UPR Pathways. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409219. [PMID: 40347515 PMCID: PMC12120720 DOI: 10.1002/advs.202409219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 02/10/2025] [Indexed: 05/14/2025]
Abstract
Multiple myeloma (MM) pathogenesis is closely associated with aberrant epigenetic regulation and resulting modifications, such as dimethylation of lysine 36 in histone H3 (H3K36me2). However, the recognition signature of H3K36me2 and its functional role in MM remain largely unknown. Here, the zinc-finger MYND-type-containing 8 (ZMYND8) is identified as a potential reader of the H3K36me2 mark that suppresses MM progression. ZMYND8 knockdown promotes the proliferation and invasion of MM cells. Combined transcriptomic and epigenomic analyses reveal that CCAAT/enhancer-binding protein epsilon (CEBPE) is a direct downstream target of ZMYND8. CEBPE modulates adaptive unfolded protein response (UPR) pathways through the transcriptional repression of ERN1, XBP1, and ATF6 to impair cell survival. Coimmunoprecipitation and chromatin immunoprecipitation assays show that ZMYND8 activates CEBPE expression in an H3K36me2-dependent manner and that its Pro-Trp-Trp-Pro domain is required for binding H3K36me2 modules, leading to CEBPE transcription. Low ZMYND8 expression is significantly correlated with adverse clinicopathological features and poor survival outcomes in MM patients. Furthermore, ZMYND8 upregulation increases the sensitivity of MM cells to carfilzomib. Taken together, these findings demonstrate that ZMYND8 epigenetically activates CEBPE transcription and suppresses MM cell growth by inhibiting the adaptive UPR, suggesting that ZMYND8 can be a novel therapeutic target for patients with MM.
Collapse
Affiliation(s)
- Jiaxuan Xu
- Department of HematologyNanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolThe State Key Laboratory of Pharmaceutical BiotechnologyChina‐Australia Institute of Translational MedicineSchool of Life SciencesNanjing UniversityDepartment of HematologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210023China
- School of Health Preservation and RehabilitationNanjing University of Chinese MedicineNanjing210023China
| | - Xiaoqing Dong
- Department of HematologyNanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolThe State Key Laboratory of Pharmaceutical BiotechnologyChina‐Australia Institute of Translational MedicineSchool of Life SciencesNanjing UniversityDepartment of HematologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210023China
| | - Yue Peng
- Department of HematologyNanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolThe State Key Laboratory of Pharmaceutical BiotechnologyChina‐Australia Institute of Translational MedicineSchool of Life SciencesNanjing UniversityDepartment of HematologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210023China
| | - Jie Yan
- Department of HematologyNanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolThe State Key Laboratory of Pharmaceutical BiotechnologyChina‐Australia Institute of Translational MedicineSchool of Life SciencesNanjing UniversityDepartment of HematologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210023China
| | - Peipei Xu
- Department of HematologyNanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolThe State Key Laboratory of Pharmaceutical BiotechnologyChina‐Australia Institute of Translational MedicineSchool of Life SciencesNanjing UniversityDepartment of HematologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210023China
| | - Feiyu Li
- Department of HematologyNanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolThe State Key Laboratory of Pharmaceutical BiotechnologyChina‐Australia Institute of Translational MedicineSchool of Life SciencesNanjing UniversityDepartment of HematologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210023China
| | - Suwen Zhang
- Department of HematologyNanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolThe State Key Laboratory of Pharmaceutical BiotechnologyChina‐Australia Institute of Translational MedicineSchool of Life SciencesNanjing UniversityDepartment of HematologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210023China
| | - Lanxin Chen
- Department of HematologyNanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolThe State Key Laboratory of Pharmaceutical BiotechnologyChina‐Australia Institute of Translational MedicineSchool of Life SciencesNanjing UniversityDepartment of HematologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210023China
| | - Xingjun Meng
- Department of HematologyNanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolThe State Key Laboratory of Pharmaceutical BiotechnologyChina‐Australia Institute of Translational MedicineSchool of Life SciencesNanjing UniversityDepartment of HematologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210023China
| | - Kangning Wang
- Institute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100005China
| | - Mengying Xing
- Department of HematologyNanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolThe State Key Laboratory of Pharmaceutical BiotechnologyChina‐Australia Institute of Translational MedicineSchool of Life SciencesNanjing UniversityDepartment of HematologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210023China
| | - Wenyang Li
- Department of HematologyNanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolThe State Key Laboratory of Pharmaceutical BiotechnologyChina‐Australia Institute of Translational MedicineSchool of Life SciencesNanjing UniversityDepartment of HematologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210023China
| | - David C. S. Huang
- The Walter and Eliza Hall Institute of Medical ResearchDepartment of Medical BiologyUniversity of MelbourneMelbourneVIC3052Australia
| | - Quan Zhao
- Department of HematologyNanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolThe State Key Laboratory of Pharmaceutical BiotechnologyChina‐Australia Institute of Translational MedicineSchool of Life SciencesNanjing UniversityDepartment of HematologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210023China
| | - Bing Chen
- Department of HematologyNanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolThe State Key Laboratory of Pharmaceutical BiotechnologyChina‐Australia Institute of Translational MedicineSchool of Life SciencesNanjing UniversityDepartment of HematologyNanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese MedicineNanjing210023China
| |
Collapse
|
33
|
Qiu Y, Wang Y, Liu J, Sun K, Liu B, Hou Q. Single-cell sequencing unveils the transcriptomic landscape of castration-resistant prostate cancer-associated fibroblasts and their association with prognosis and immunotherapy response. BMC Cancer 2025; 25:813. [PMID: 40307786 PMCID: PMC12044937 DOI: 10.1186/s12885-025-14212-x] [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: 10/04/2024] [Accepted: 04/23/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND The tumor microenvironment (TME) is increasingly acknowledged as a determinant in the malignant transformation and progression of castration-resistant prostate cancer (CRPC). Cancer-associated fibroblasts (CAFs), as a pivotal stromal cellular component in TME, are implicated in tumor progression and immune escape. However, the molecular characteristics and biological functions of CRPC-CAFs in prostate cancer necessitate further investigation. METHODS We ascertained the differential transcriptomic profiles between CRPC-CAFs and PCa-CAFs through single-cell RNA-sequencing (scRNA-seq). Bulk RNA-seq data were employed to assess the prognostic implications of CRPC-CAFs in PCa. In addition, we examined the impact of CRPC-CAFs on the efficacy of immunotherapy and the composition of the tumor immune milieu. Furthermore, a subcutaneous PCa model was applied to determine the potential of TGF-β signaling blockade to augment the response to immunotherapeutic interventions. RESULTS We observed a pronounced increase in the proportion of CAFs in CRPC compared to those in primary PCa. The functional pathways implicated in TGF-β signaling and ECM remodeling were remarkably upregulated in CRPC-CAFs. Moreover, gene regulatory network analysis uncovered substantial differences in the transcription factor activity profiles between CRPC-CAFs and PCa-CAFs. The elevated CRPC-CAFs abundance was associated with diminished recurrence-free survival and immunotherapy insensitivity. Substantially elevated infiltration of inhibitory immune cells and upregulated expression levels of immunosuppressive molecules were observed in patients with high CRPC-CAFs abundance. Importantly, administration of anti-TGF-β therapy remarkably potentiated the efficacy of anti-PD-1 immunotherapy through upregulating the anti-tumor immune response in the PCa model. CONCLUSION Our results highlighted the impact of CRPC-CAFs on clinical prognosis and immunosuppressive tumor milieu, indicating that CRPC-CAFs may function as a promising therapeutic target for CRPC.
Collapse
Affiliation(s)
- Yifeng Qiu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
- International Cancer Center, Shenzhen Key Laboratory, Hematology Institution of Shenzhen University, Shenzhen, China
| | - Yuhan Wang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Jiahe Liu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Kai Sun
- Department of Radiology, the Third People's Hospital of Longgang District, Shenzhen, China
- Shenzhen Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518116, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China.
| | - Qi Hou
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China.
- International Cancer Center, Shenzhen Key Laboratory, Hematology Institution of Shenzhen University, Shenzhen, China.
| |
Collapse
|
34
|
Cao C, Wu ZY, Liao W, Wei LJ, Liang HY, Yang X, Luo RZ, Liu LL. Clinicopathological characterization of Switch/Sucrose-non-fermentable (Swi/Snf) complex (ARID1A, SMARCA2, SMARCA4)-deficient endocervical adenocarcinoma. Cancer Cell Int 2025; 25:170. [PMID: 40301885 PMCID: PMC12042307 DOI: 10.1186/s12935-025-03794-y] [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: 11/06/2024] [Accepted: 04/17/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Subunits of the Switch/Sucrose-non-fermentable (Swi/Snf) complex, such as ARID1A, SMARCA4, SMARCA2, etc., have been implicated in the development of gynecologic cancers. However, their prevalence and clinical implications in endocervical adenocarcinoma (ECA) remain unclear. This study aimed to evaluate the expression of Swi/Snf complex subunits in ECA and characterize the clinicopathological and immune microenvironment features of Swi/Snf-deficient ECA. METHODS We evaluated 604 ECA using representative tissue microarrays, collected clinicopathologic data, reviewed histological features, and performed immunohistochemical staining for several Swi/Snf complex subunits, mismatch repair (MMR), immune cell markers, and immune checkpoint ligands proteins. RESULTS Among the 604 cases examined, five Swi/Snf subunit expression patterns were identified, including intact expression, deficient expression, 'checkerboard' expression, reduced expression, and heterogeneous expression. Deficiencies of ARID1A (3.97%, 24/604), SMARCA2 (2.32%,14/604), and SMARCA4 (1.49%, 9/604) were observed. Defining Swi/Snf deficiency as loss of any subunit, the overall deficiency rate was 5.96% (36/604). Swi/Snf-deficient ECA tended to advanced FIGO stage (III-IV, P = 0.041), larger tumor size (P < 0.001), deeper stromal invasion (≥ 1/3, P = 0.046), and higher lymph node metastasis rate (P = 0.037). Morphologically, Swi/Snf-deficient ECA displayed frequent poor differentiation (P = 0.001), medullary features (P < 0.001), high nuclear grade (P < 0.001), necrosis (P = 0.001), stromal tumor-infiltrating lymphocytes (sTILs, P < 0.001), peritumoral lymphocyte aggregation (P = 0.001), and tertiary lymphoid structures (TLS, P < 0.001). Immune subset analysis revealed significantly elevated densities of CD3⁺ T cells, CD8⁺ T cells, CD38⁺ plasma cells, CD56⁺ NK cells, CD68⁺ macrophages, and PD-1⁺ T cells in Swi/Snf-deficient ECA (P < 0.05). Swi/Snf-deficient ECA demonstrated higher PD-L1 combined positive score (CPS) positivity (P < 0.001), and was more frequently associated with mismatch repair deficiency (MMRD, P < 0.001). Survival analysis indicated shorter overall survival (median: 53 vs. 64.5 months, P = 0.0307) and disease-free survival (median: 52 vs. 60.5 months, P = 0.0228) in Swi/Snf-deficient ECA patients. CONCLUSIONS Swi/Snf complex deficiency is rare but significantly associated with NHPVA, aggressive pathological features, immunologically activated phenotypes, and MMRD. Swi/Snf status evaluation may inform novel therapeutic strategies for ECA patients.
Collapse
Affiliation(s)
- Chao Cao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651# Dong Feng Road East, Guangzhou, Guangdong, 510060, P.R. China
| | - Zi-Yun Wu
- Department of Urology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, P.R. China
| | - Wei Liao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Intensive Care Unit, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Li-Jun Wei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651# Dong Feng Road East, Guangzhou, Guangdong, 510060, P.R. China
| | - Hao-Yu Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651# Dong Feng Road East, Guangzhou, Guangdong, 510060, P.R. China
| | - Xia Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651# Dong Feng Road East, Guangzhou, Guangdong, 510060, P.R. China
| | - Rong-Zhen Luo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China.
- Department of Pathology, Sun Yat-sen University Cancer Center, 651# Dong Feng Road East, Guangzhou, Guangdong, 510060, P.R. China.
| | - Li-Li Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China.
- Department of Pathology, Sun Yat-sen University Cancer Center, 651# Dong Feng Road East, Guangzhou, Guangdong, 510060, P.R. China.
| |
Collapse
|
35
|
Wang Y, Wang Y, Bao L, Vale G, McDonald JG, Fang Y, Peng Y, Kumar A, Xing C, Brasó-Maristany F, Prat A, Arteaga CL, Wang Y, Luo W. ZMYND8 drives HER2 antibody resistance in breast cancer via lipid control of IL-27. Nat Commun 2025; 16:3908. [PMID: 40281007 PMCID: PMC12032076 DOI: 10.1038/s41467-025-59184-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Anti-HER2 antibodies are effective but often lead to resistance in patients with HER2+ breast cancer. Here, we report an epigenetic crosstalk with aberrant glycerophospholipid metabolism and inflammation as a key resistance mechanism of anti-HER2 therapies in HER2+ breast cancer. Histone reader ZMYND8 specifically confers resistance to cancer cells against trastuzumab and/or pertuzumab. Mechanistically, ZMYND8 enhances cPLA2α expression in resistant tumor cells through inducing c-Myc. cPLA2α inactivates phosphatidylcholine-specific phospholipase C to inhibit phosphatidylcholine breakdown into diacylglycerol, which diminishes protein kinase C activity leading to interleukin-27 secretion. Supplementation with interleukin-27 protein counteracts cPLA2α loss to reinforce trastuzumab resistance in HER2+ tumor cells and patient-derived organoids. Upregulation of ZMYND8, c-Myc, cPLA2α, and IL-27 is prevalent in HER2+ breast cancer patients following HER2-targeted therapies. Targeting c-Myc or cPLA2α effectively overcomes anti-HER2 therapy resistance in patient-derived xenografts. Collectively, this study uncovers a druggable signaling cascade that drives resistance to HER2-targeted therapies in HER2+ breast cancer.
Collapse
Affiliation(s)
- Yong Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Yanan Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Lei Bao
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Goncalo Vale
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey G McDonald
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Yisheng Fang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Yan Peng
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ashwani Kumar
- Eugene McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Cancer Institute and Blood Diseases, Hospital Clínic of Barcelona, Barcelona, Spain
- Reveal Genomics, S.L., Barcelona, Spain
| | - Aleix Prat
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Cancer Institute and Blood Diseases, Hospital Clínic of Barcelona, Barcelona, Spain
- Reveal Genomics, S.L., Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- Institute of Oncology-Hospital Quirónsalud, Barcelona, Spain
| | - Carlos L Arteaga
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Yingfei Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Weibo Luo
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA.
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
36
|
Campanelli G, Waxner N, Parkhomovsky N, Mak CK, Yin JH, Lin SJH, Vanderstichel R, Yang C, Levenson AS. Identification of metastasis-associated protein 1 (MTA1) as a new molecular marker for canine urothelial carcinoma. Front Vet Sci 2025; 12:1527167. [PMID: 40351767 PMCID: PMC12062746 DOI: 10.3389/fvets.2025.1527167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/01/2025] [Indexed: 05/14/2025] Open
Abstract
Background Although metastasis-associated protein 1 (MTA1) is known to play a role in cancer invasion and metastasis of various cancers, the clinical significance of its expression in canine urothelial carcinoma (UC) has not been explored. We sought to evaluate the expression of MTA1, cyclooxygenase 2 (COX2) and E-cadherin (E-cad) in association with clinicopathological parameters in clinical samples of canine UC. Methods We retrospectively analyzed UC tissues from 28 canine patients using immunohistochemistry for Ki67, CD31, MTA1, COX2, and E-cad staining. Statistical significance for marker staining intensities was evaluated by ANOVA or Student's t-test. The correlation between molecular markers in canine UC samples detected by IHC and clinicopathological features was calculated by the Wilcoxon (Mann-Whitney) and Kruskal-Wallis tests. Western blot analysis was performed for detection of EMT markers in canine cell lines. Results We show that MTA1 and COX2 are overexpressed in canine UC samples compared to normal canine bladder samples, whereas E-cad levels are higher in normal bladder. The results demonstrated that MTA1 expression correlated with aggressive clinicopathological features such as high tumor-grade, muscular/vascular invasion, and metastasis. The expression of MTA1 differed in tumors depending on their localization, with the highest being in the urethra adjoining the prostate. Unexpectedly, higher E-cad levels were detected in metastatic tumor cells compared to primary tumor cells. Conclusion These findings suggest that MTA1 may represent a key upstream effector tightly associated with COX2 and E-cad-mediated events in canine UC. Accordingly, MTA1 may be considered a feasible interceptive and therapeutic target for canine UC treatment.
Collapse
Affiliation(s)
- Gisella Campanelli
- Department of Veterinary Biomedical Sciences, Lewyt College of Veterinary Medicine, Long Island University, Brookville, NY, United States
| | - Noah Waxner
- Department of Veterinary Biomedical Sciences, Lewyt College of Veterinary Medicine, Long Island University, Brookville, NY, United States
| | - Nema Parkhomovsky
- College of Sciences, Long Island University, Brookville, NY, United States
| | - Chun Kuen Mak
- Department of Veterinary Clinical Sciences, Lewyt College of Veterinary Medicine, Long Island University, Brookville, NY, United States
| | - Ji-Hang Yin
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Susanne Je-Han Lin
- Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Raphael Vanderstichel
- Department of Veterinary Clinical Sciences, Lewyt College of Veterinary Medicine, Long Island University, Brookville, NY, United States
| | - Ching Yang
- Department of Veterinary Biomedical Sciences, Lewyt College of Veterinary Medicine, Long Island University, Brookville, NY, United States
| | - Anait S. Levenson
- Department of Veterinary Biomedical Sciences, Lewyt College of Veterinary Medicine, Long Island University, Brookville, NY, United States
| |
Collapse
|
37
|
Hao F, Zhang Y, Hou J, Zhao B. Chromatin remodeling and cancer: the critical influence of the SWI/SNF complex. Epigenetics Chromatin 2025; 18:22. [PMID: 40269969 PMCID: PMC12016160 DOI: 10.1186/s13072-025-00590-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/15/2025] [Indexed: 04/25/2025] Open
Abstract
The SWI/SNF complex was first identified in yeast and named after studies of mutants critical for the mating-type switch (SWI) and sucrose non-fermenting (SNF) pathways.The SWI/SNF complex plays a pivotal role in regulating gene expression by altering chromatin structure to promote or suppress the expression of specific genes, maintain stem cell pluripotency, and participate in various biological processes. Mutations in the SWI/SNF complex are highly prevalent in various human cancers, significantly impacting tumor suppressive or oncogenic functions and influencing tumor initiation and progression. This review focuses on the mechanisms by which ARID1A/ARID1B, PBRM1, SMARCB1, and SMARCA2/SMARCA4 contribute to cancer, the immunoregulatory roles of the SWI/SNF complex, its involvement in DNA repair pathways, synthetic lethality, and applications in precision oncology.
Collapse
Affiliation(s)
- Fengxiang Hao
- Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi Province, 030001, China
| | - Ying Zhang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi Province, 030001, China
| | - Jiayi Hou
- Department of Clinical Laboratory, Shanxi Provincial Academy of Traditional Chinese Medicine, Taiyuan, Shanxi Province, China
| | - Bin Zhao
- Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China.
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi Province, 030001, China.
| |
Collapse
|
38
|
Wang QC, Cao JY, Wang GY, Wang H, Peng H, Wang Q, Yin ZX, Jie-Zhou, Chang PA, Zhang GH, Yao WT, Wu JC, Pei CS. Circulating bile acid profiles characteristics and the potential predictive role in clear cell renal cell carcinoma progression. BMC Nephrol 2025; 26:206. [PMID: 40269745 PMCID: PMC12020223 DOI: 10.1186/s12882-025-04142-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 04/18/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND The incidence of clear cell renal cell carcinoma (ccRCC) has steadily increased over the past decade, and recent studies have linked bile acid (BA) metabolism to its development. However, the metabolic profile of BAs and their potential as biomarkers in ccRCC pathogenesis remain poorly characterized, making their evaluation crucial for advancing disease understanding and management. METHODS A total of 68 newly diagnosed ccRCC patients and 63 healthy controls were enrolled. Serum bile acid (BA) profiles were measured using Ultra Performance Liquid Chromatography-Tandem Mass Spectrometry (UPLC-MS/MS). The Orthogonal Projections to Latent Structures Discriminant Analysis (OPLS-DA) model analyzed differences in serum BA profiles between ccRCC patients and controls. Additionally, the relationship between BA profiles and tumor heterogeneity parameters was investigated. Receiver Operating Characteristic (ROC) analysis identified potential biomarkers for ccRCC pathogenesis. RESULTS The BA profile was altered in ccRCC patients and was not influenced by sex or age. Specifically, primary and secondary unconjugated BA fractions were significantly higher in the ccRCC population. Five BA metabolite candidates exhibited the most significant differences between ccRCC patients and controls. Deoxycholic acid (DCA) was associated with pathological pTNM stage classification and grade. Chenodeoxycholic acid (CDCA) and lithocholic acid (LCA), combined with testosterone, showed potential as biomarkers for the pathogenesis of ccRCC. CONCLUSION Alterations in the serum BA profile are observed in ccRCC. Deoxycholic acid (DCA) correlates with pathological pTNM stage classification and tumor grade. Additionally, CDCA combined with LCA show potential as biomarkers for ccRCC pathogenesis. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Qi-Chao Wang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Urology, Xuzhou Cancer Hospital, Affiliated Hospital of Jiangsu University, Xuzhou, China
| | - Jing-Yi Cao
- Department of Urology, Xuzhou Cancer Hospital, Affiliated Hospital of Jiangsu University, Xuzhou, China
| | - Guang-Yue Wang
- Department of Urology, Xuzhou Cancer Hospital, Affiliated Hospital of Jiangsu University, Xuzhou, China
| | - Heng Wang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hua Peng
- Department of Urology, Xuzhou Cancer Hospital, Affiliated Hospital of Jiangsu University, Xuzhou, China
| | - Qian Wang
- Department of Urology, Xuzhou Cancer Hospital, Affiliated Hospital of Jiangsu University, Xuzhou, China
| | - Zhi-Xiang Yin
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie-Zhou
- Department of Urology, Xuzhou Cancer Hospital, Affiliated Hospital of Jiangsu University, Xuzhou, China
| | - Ping-An Chang
- Department of Urology, Dongtai People Hospital, Yancheng, China
| | - Gui-Hua Zhang
- Department of Hematology, Xuzhou Cancer Hospital, Affiliated Hospital of Jiangsu University, Xuzhou, China
| | - Wen-Tao Yao
- Department of Urology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Jia-Cheng Wu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chang-Song Pei
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
39
|
Guo J, Li N, Liu Q, Hao Z, Zhu G, Wang X, Wang H, Pan Q, Xu B, Han Y, Zhang G, Lian Y, Zhang W, Gu Y, Lin N, Zeng X, Jin Z, Lan W, Jiang J, Gao D, Dong L, Yuan H, Liang C, Qin J. KMT2C deficiency drives transdifferentiation of double-negative prostate cancer and confer resistance to AR-targeted therapy. Cancer Cell 2025:S1535-6108(25)00139-4. [PMID: 40280125 DOI: 10.1016/j.ccell.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 01/25/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025]
Abstract
Double-negative prostate cancer (DNPC), characterized by an androgen receptor (AR)- and neuroendocrine-null phenotype, frequently emerges following androgen deprivation therapy (ADT). However, our understanding of the origins and regulatory mechanisms of DNPC remains limited. Here, we discover that tumors with KMT2C mutation or loss are highly susceptible to transitioning into DNPC following ADT. We clarify that DNPC primarily stems from luminal cell transdifferentiation rather than basal cell transformation. Antiandrogen treatment induces KMT2C binding at enhancers of a subset of AR-regulated genes, preserving the adenocarcinoma lineage. KMT2C maintains ASPP2 expression via enhancer-promoter communication post-AR inhibition, while its inactivation reduces ASPP2, triggering ΔNp63-dependent transdifferentiation. This DNPC transition maintains fatty acid (FA) synthesis through ΔNp63-mediated SREBP1c transactivation, fueling DNPC growth via HRAS palmitoylation and MAPK signaling activation. These findings highlight KMT2C as an epigenetic checkpoint against DNPC development and suggest the therapeutic potential of targeting fatty acid synthesis.
Collapse
Affiliation(s)
- Jiacheng Guo
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Ni Li
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China.
| | - Qiuli Liu
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Zongyao Hao
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230022, China
| | - Guanghui Zhu
- West China School of Public Health, West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610000, China
| | - Xuege Wang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Hanling Wang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Qiang Pan
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Beitao Xu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Ying Han
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Guoying Zhang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Yannan Lian
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Wei Zhang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Yongqiang Gu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Naiheng Lin
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xin Zeng
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zige Jin
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Weihua Lan
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jun Jiang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Dong Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liang Dong
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Huairui Yuan
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Chaozhao Liang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230022, China.
| | - Jun Qin
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
40
|
Lv Y, Mo X, Zhang R, Peng Y, Feng T, Zhang Y, Song G, Ge L, Liu Y, Yang G, Wang L. Prostate cancer exploits BRD9-driven metabolic reprogramming to shape the aggressive phenotype. Cell Death Dis 2025; 16:326. [PMID: 40263302 PMCID: PMC12015546 DOI: 10.1038/s41419-025-07561-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/24/2025] [Accepted: 03/17/2025] [Indexed: 04/24/2025]
Abstract
The aggressive phenotype of prostate cancer (PCa) requires adaptation to androgen deprivation (AD) to progress into castration-resistant PCa (CRPC), including adaptation to AD-induced oxidative stress. However, our understanding of the oncogenes that maintain the redox balance during CRPC progression is limited. Here, we identified Bromodomain-containing protein 9 (BRD9) as a metabolic checkpoint for reprogramming cell metabolism to support tumor growth and impart a castration-resistant phenotype under metabolic and oxidative stress. Following oxidation, BRD9 recruited the nuclear transcription factor-Y A-subunit (NFYA) to induce glycogen phosphorylase L (PYGL) expression, which directed glucose utilization through the pentose phosphate pathway, generating NADPH, and promoting clearance of reactive oxygen species (ROS), thus maintaining redox balance. By disturbing redox homeostasis, BRD9 inhibition exerted oxidative pressure on PCa cells, sensitizing them to radiotherapy. This work identified BRD9 as a novel component in antioxidant reprogramming and indicates BRD9 targeting as a promising treatment strategy for PCa therapy.
Collapse
Affiliation(s)
- Ye Lv
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Xinkai Mo
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ruojia Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yu Peng
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tingting Feng
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Yuang Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Guanhua Song
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Luna Ge
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Yu Liu
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Guiwen Yang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China.
| | - Lin Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
41
|
Zhu X, Yin G, Xu J, Tang X, Yu F. Dendrobium huoshanense polysaccharide inhibits NSCLC proliferation and immune evasion via FXR1-IL-35 axis signaling pathway. J Nat Med 2025:10.1007/s11418-025-01894-7. [PMID: 40259042 DOI: 10.1007/s11418-025-01894-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 03/10/2025] [Indexed: 04/23/2025]
Abstract
Dendrobium huoshanense has received special attention for its advantages in the treatment of lung cancer, but the underlying molecular mechanisms are not yet well understood. First, we obtained 8 active ingredients and 159 effective action targets of Dendrobium huoshanense using network pharmacology, and searching target interactions through STRING, constructing the PPI network and KEGG, GO and Hallmark enrichment analysis. Then, we combined target's enrichment analysis and GSEA enrichment analysis of IL-35, indicating the mechanism of cDHPs for non-small cell lung cancer (NSCLC) may be related to tight junction and NSCLC pathway. Further, FXR1 and ACTR3 were identified as core therapeutic targets, and high expression of FXR1 or ACTR3 was significantly associated with poor prognosis of patients. The analysis of single-cell data also indicated that the percentage of CD4-CTLA4-Treg cells may be increased by the expression of IL-35, resulting in a suppressive immune microenvironment. Next, In vivo experiment, we detected iTr35 by flow cytometry, detected IL-35 level by RT-PCR, Western blotting and ELISA, and detected NK cell activity to explore the immunomodulatory effects and anti-tumor mechanism of cDHPs. After cDHPs administration, the conversion of CD4+ T cells to iTr35 is inhibited, p35 and EBI3 in both protein and mRNA levels, the levels of IL-35 and IL-4 in serum decreased. The levels of IFN-γ, while the activity of NK cells in mice increased, enhancing the anti-tumor immune effect of the organism. Finally, analysis of sequencing data from the immunotherapy cohort of tumor-bearing mice obtained from the TISMO database shows that the combination of cDHPs and PD-1/PD-L1 antibodies improves effector and thus PD-1/PD-L1 antibody efficacy. These findings suggest that cDHPs inhibit NSCLC proliferation and immune escape via the FXR1-IL-35 axis signaling pathway.
Collapse
Affiliation(s)
- Xinying Zhu
- Translational Medicine Center, The Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, Anhui Province, China
| | - Guoquan Yin
- Clinical Laboratory, Yangzhou Blood Center in Jiangsu Province, Yangzhou, 225007, Jiangsu Province, China
| | - Jiaqian Xu
- Department of Medical Microbiology and Immunology, School of Preclinical Medicine, Wannan Medical College, Wuhu, 241001, Anhui Province, China
| | - Xiaolei Tang
- Translational Medicine Center, The Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, Anhui Province, China
| | - Fangliu Yu
- Department of Medical Microbiology and Immunology, School of Preclinical Medicine, Wannan Medical College, Wuhu, 241001, Anhui Province, China.
| |
Collapse
|
42
|
Pei X, Zheng F, Li Y, Li X, Lin Z, Han X, Tian Z, Cao K, Ren D, Li C. Disulfiram activation of prostaglandin E2 synthesis: a novel antifibrotic mechanism in pulmonary fibrosis. J Pharmacol Exp Ther 2025; 392:103588. [PMID: 40359874 DOI: 10.1016/j.jpet.2025.103588] [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: 02/06/2025] [Revised: 04/02/2025] [Accepted: 04/13/2025] [Indexed: 05/15/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by the pathological replacement of alveolar structures with thickened, inelastic fibrous tissue, which significantly hinders gas exchange in the lungs. Disulfiram (DSF), a Food and Drug Administration-approved drug for alcohol dependence, has shown potential in various diseases. This study investigates the effects of DSF on IPF and its mechanisms, focusing on the cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2) pathway. Utilizing primary diseased human lung fibroblast-IPF cells and A549 cells induced with transforming growth factor-beta 1 to model epithelial-mesenchymal transition (EMT), we employed a battery of in vitro assays to assess cellular viability, migratory capacity, and the expression of fibrosis-related genes and proteins. To further substantiate our in vitro findings, a bleomycin-induced mouse model of IPF was treated with DSF, and subjected to a comprehensive evaluation of pulmonary function, histological examination, hydroxyproline assay, and western blot analysis to quantify the extent of fibrosis. DSF reduced cell viability and migration in fibrotic cell models. It increased COX-2 and PGE2 levels, regulated EMT, and extracellular matrix collagen deposition. In vivo, DSF improved pulmonary function and reduced EMT and extracellular matrix accumulation in mice. The COX-2/PGE2 axis was identified as a critical mediator of DSF's effects. DSF exhibits antifibrotic properties in IPF by modulating the COX-2/PGE2 signaling pathway. This study provides a novel therapeutic strategy for IPF and highlights the potential of repurposing DSF for clinical use in this context. SIGNIFICANCE STATEMENT: Disulfiram shows promise in treating idiopathic pulmonary fibrosis by targeting the cyclooxygenase-2/prostaglandin E2 pathway, offering a new therapeutic strategy and highlighting its potential for repurposing in this context.
Collapse
Affiliation(s)
- Xiaolin Pei
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Fangxu Zheng
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Yin Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Xiaobo Li
- Beijing Tide Pharmaceutical Co, Ltd, Beijing, China; School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhoujun Lin
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Xiao Han
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Zhenhuan Tian
- Department of Thoracic surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Ke Cao
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Dunqiang Ren
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Chenggang Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.
| |
Collapse
|
43
|
You X, Li H, Li Q, Zhang Q, Cao Y, Fu W, Wang B. Astragaloside IV-PESV facilitates pyroptosis by enhancing palmitoylation of GSDMD protein mediated by ZDHHC1. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04122-x. [PMID: 40237800 DOI: 10.1007/s00210-025-04122-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/28/2025] [Indexed: 04/18/2025]
Abstract
Prostate cancer (PCa) is an epithelial malignancy affecting the prostate gland. Astragaloside IV combined with polypeptide extract from scorpion venom (PESV) has been reported to inhibit the growth of PCa. This study aimed to investigate the mechanisms by which this combination mitigates the progression of PCa. Bioinformatic analysis was utilized to investigate the correlation between zinc finger DHHC-type containing 1 (ZDHHC1) expression and PCa progression. The extent of pyroptosis in PCa cells was assessed by measuring cell viability, IL-1β and IL-18 secretion, LDH release, and HMGB1 content. PCa mouse models were constructed by subcutaneous injection of DU145 or PC-3 cells into nude mice, with subsequent monitoring of tumor weight and volume. ZDHHC1 expression was significantly lower in PCa patient tissues, which correlated with a poor prognosis. ZDHHC1 overexpression inhibited PC-3 and DU145 cell viability and increased IL-1β, IL-18, LDH, and HMGB1 levels in cell supernatants. Notably, the pyroptosis inhibitor LDC7559 partially reversed these effects. Co-IP assay demonstrated an interaction between ZDHHC1 and GSDMD. ZDHHC1 overexpression significantly enhanced GSDMD palmitoylation-mediated membrane translocation and pyroptosis; however, this effect was partially reversed by the palmitoylation inhibitor 2-BP. The combination of Astragaloside IV and PESV promoted GSDMD membrane translocation and pyroptosis in PCa cells, with ZDHHC1 knockdown partially reversing the effects of Astragaloside IV-PESV. Furthermore, treatment with Astragaloside IV-PESV significantly inhibited tumor tissue growth in tumor-bearing nude mouse models. Astragaloside IV-PESV enhances palmitoylation-mediated membrane translocation of GSDMD-N by upregulating ZDHHC1 expression, thereby facilitating pyroptosis in PCa cells and attenuating PCa progression.
Collapse
Affiliation(s)
- Xujun You
- Department of Andrology, Dongcheng District, Dongzhimen Hospital, Beijing University of Chinese Medicine, Hai Yun Cang on the 5 th, Beijing, 100700, China
- Department of Andrology, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518101, China
| | - Honghan Li
- The Seventh Clinical Medical College of Guangzhou, University of Traditional Chinese Medicine, Shenzhen, 518133, China
| | - Qixin Li
- Department of Andrology, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518101, China
| | - Qing Zhang
- Department of Andrology, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518101, China
| | - Yiguo Cao
- Department of Urology Surgery, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518101, China
| | - Wei Fu
- Department of Andrology, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518101, China.
| | - Bin Wang
- Department of Andrology, Dongcheng District, Dongzhimen Hospital, Beijing University of Chinese Medicine, Hai Yun Cang on the 5 th, Beijing, 100700, China.
| |
Collapse
|
44
|
Chen R, Chen T, Li X, Yu J, Lin M, Wen S, Zhang M, Chen J, Yi B, Zhong H, Li Z. SREBP2 as a central player in cancer progression: potential for targeted therapeutics. Front Pharmacol 2025; 16:1535691. [PMID: 40308757 PMCID: PMC12041066 DOI: 10.3389/fphar.2025.1535691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Recent studies have identified the reprogramming of lipid metabolism as a critical hallmark of malignancy. Enhanced cholesterol uptake and increased cholesterol biosynthesis significantly contribute to the rapid growth of tumors, with cholesterol also playing essential roles in cellular signaling pathways. Targeting cholesterol metabolism has emerged as a promising therapeutic strategy in oncology. The sterol regulatory element-binding protein-2 (SREBP2) serves as a primary transcriptional regulator of genes involved in cholesterol biosynthesis and is crucial for maintaining cholesterol homeostasis. Numerous studies have reported the upregulation of SREBP2 across various cancers, facilitating tumor progression. This review aims to provide a comprehensive overview of the structure, biological functions, and regulatory mechanisms of SREBP2. Furthermore, we summarize that SREBP2 plays a crucial role in various cancers and tumor microenvironment primarily by regulating cholesterol, as well as through several non-cholesterol pathways. We also particularly emphasize therapeutic agents targeting SREBP2 that are currently under investigation. This review seeks to enhance our understanding of SREBP2's involvement in cancer and provide theoretical references for cancer therapies that target SREBP2.
Collapse
Affiliation(s)
- Ruiqi Chen
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Tianyu Chen
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiang Li
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Junfeng Yu
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Min Lin
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Siqi Wen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Man Zhang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jinchi Chen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Bei Yi
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Huage Zhong
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, China
| | - Zhao Li
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
45
|
Bessot A, Gunter J, McGovern J, Bock N. Bone marrow adipocytes in cancer: Mechanisms, models, and therapeutic implications. Biomaterials 2025; 322:123341. [PMID: 40315628 DOI: 10.1016/j.biomaterials.2025.123341] [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: 11/21/2024] [Revised: 04/02/2025] [Accepted: 04/12/2025] [Indexed: 05/04/2025]
Abstract
Adipose tissue is the primary site of energy storage in the body and a key regulator of metabolism. However, different adipose depots exhibit distinct molecular and phenotypic characteristics that have yet to be fully unraveled. While initially considered inert, bone marrow adipocytes (BMAs) have been recognized as key regulators of bone homeostasis, and more recently bone pathologies, although many unknowns remain. In this review, we summarize the current knowledge on BMAs, focusing on their distinct characteristics, functional significance in bone physiology and metabolism, as well as their emerging role in cancer pathogenesis. We present and discuss the current methodologies for investigating BMA-cancer interactions, encompassing both in vitro 3D culture systems and in vivo models, and their limitations in accurately replicating the phenotypes and biological processes of the human species. We highlight the imperative for advancing towards humanized models to better mimic the complexities of human physiology and disease progression. Finally, therapeutic strategies targeting metabolism or BMA-secreted factors, such as anti-cholesterol drugs, hold considerable promise in cancer treatment. We present the synergistic avenue of combining conventional cancer therapies with agents targeting adipocyte signaling to amplify treatment efficacy. Developing preclinical models that more faithfully replicate human pathological and physiological processes will lead to more accurate mechanistic understanding of the role of BMAs in bone metastasis and lead to more relevant preclinical drug screening.
Collapse
Affiliation(s)
- Agathe Bessot
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia; Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia; Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Brisbane, QLD, 4000, Australia
| | - Jennifer Gunter
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia; Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia; Centre for Genomics and Personalised Health, QUT, Brisbane, QLD, 4102, Australia
| | - Jacqui McGovern
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia; Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia; Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Brisbane, QLD, 4000, Australia; Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies (CTET), QUT, Brisbane, QLD, 4000, Australia
| | - Nathalie Bock
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia; Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia; Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Brisbane, QLD, 4000, Australia; Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia; Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling, and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD, 4000, Australia.
| |
Collapse
|
46
|
Ji S, Cao L, Gao J, Du Y, Ye Z, Lou X, Liu F, Zhang Y, Xu J, Shi X, Wang H, Li P, Li Y, Chen H, Yang Z, Gao S, Zhang W, Huang D, Ni S, Wei M, Wang F, Wang Y, Ding T, Jing D, Fan G, Gong Z, Lu R, Qin Y, Chen J, Xu X, Wang P, Zhang B, Ding L, Robles AI, Rodriguez H, Chang DK, Hruban RH, Gao D, Gao D, Jin G, Zhou H, Wu J, Yu X. Proteogenomic characterization of non-functional pancreatic neuroendocrine tumors unravels clinically relevant subgroups. Cancer Cell 2025; 43:776-796.e14. [PMID: 40185092 DOI: 10.1016/j.ccell.2025.03.016] [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: 06/11/2024] [Revised: 01/27/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
The majority of neuroendocrine neoplasms in pancreas are non-functional pancreatic neuroendocrine tumors (NF-PanNETs), which exhibit a high occurrence of distant metastases with limited therapeutic options. Here, we perform a comprehensive molecular characterization of 108 NF-PanNETs through integrative analysis of genomic, transcriptomic, proteomic, and phosphoproteomic profiles. Proteogenomic analysis provides functional insights into the genomic driver alterations of NF-PanNETs, revealing a potential mediator of MEN1 alterations using Men1-conditional knockout mice. Machine-learning-based modeling uncovers a three-protein signature as an independent prognostic factor, which is validated by an independent external cohort. Proteomic and phosphoproteomic-based stratification identifies four subtypes with distinct molecular characteristics, immune microenvironments, and clinicopathological features. Drug screening using patient-derived tumor organoids identifies cyclin-dependent kinase (CDK) 5 and Calcium Voltage-Gated Channel Subunit Alpha1 D (CACNA1D) as ubiquitous and subtype-specific targets, respectively, with in vivo validation using xenograft models. Together, our proteogenomic analyses illustrate a comprehensive molecular landscape of NF-PanNETs, revealing biological insights and therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Lihua Cao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jing Gao
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yang Du
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xin Lou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Fen Liu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yehan Zhang
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Junfeng Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Xiaohan Shi
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Huan Wang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Penghao Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Yikai Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Hongxu Chen
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhicheng Yang
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China
| | - Suizhi Gao
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Wuhu Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Dan Huang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Shujuan Ni
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Miaoyan Wei
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Fei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Yan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Tian Ding
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Desheng Jing
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Guixiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Zhiyun Gong
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Renquan Lu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Jie Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NewYork, NY 10029, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Li Ding
- Department of Medicine, McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD 20850, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD 20850, USA
| | - David K Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - Ralph H Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Dong Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Daming Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China.
| | - Hu Zhou
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Shanghai Institute of Materia Medica-University of Ottawa Joint Research Center in Systems and Personalized Pharmacology, 555 Zuchongzhi Road, Shanghai 201203, China.
| | - Jianmin Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing 100142, China; Peking University International Cancer Institute, Peking University, Beijing 100191, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China.
| |
Collapse
|
47
|
Yang Y, Li Z, Yang Y, Xiao P, He Z, Zhang Z, Li Y, Shi L, Wang X, Tao Y, Fan J, Zhang F, Yang C, Yao F, Ji T, Zhang Y, Zhou B, Yu J, Guo A, Wei Z, Jiao W, Wu Y, Li Y, Wu D, Wu Y, Gao L, Hu Y, Pan J, Hu S, Yang X. The RBM39 degrader indisulam inhibits acute megakaryoblastic leukemia by altering the alternative splicing of ZMYND8. Cell Biosci 2025; 15:46. [PMID: 40223119 PMCID: PMC11995665 DOI: 10.1186/s13578-025-01380-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Acute megakaryoblastic leukemia (AMKL) is a rare hematological malignancy in adults but children. Alternative splicing (AS) has been shown to affect hematological cancer progression, making splicing factors promising targets. Our research aims to investigate the efficacy of the molecular glue degrader indisulam, which targets the splicing factor RNA binding motif protein 39 (RBM39) in AMKL models. RESULTS Public drug sensitivity data analysis revealed that AMKL cell lines exhibited the highest sensitivity to indisulam compared with other tumor types. Then we confirmed that RBM39 depletion by indisulam treatment induced AMKL cell cycle arrest and apoptosis. In AMKL mouse model, indisulam treatment significantly reduced the leukemic burden and prolonged the lifetime of AMKL mice. Mechanically, integration of transcriptomic and proteomic analyses revealed that indisulam-mediated RBM39 degradation resulted in AS of the transcription factor zinc finger MYND-type containing 8 (ZMYND8), an AMKL cell growth regulator. Finally, the effectiveness of indisulam depended on DDB1- and Cul4- Associated Factor 15 (DCAF15) expression because knockout of DCAF15 rescued the indisulam-induced RBM39 degradation and mis-splicing of ZMYND8. CONCLUSION Indisulam is a promising therapeutic candidate for AMKL and the RBM39-mediated ZMYND8 splicing plays an important role in promoting the development of AMKL.
Collapse
Affiliation(s)
- Ying Yang
- Department of Pediatrics, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Guiyang, 550001, China
| | - Zhiheng Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
- Department of Hematology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, 215003, China
| | - Yang Yang
- Institute of Pediatric Research, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Peifang Xiao
- Department of Hematology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, 215003, China
| | - Zhixu He
- Department of Pediatrics, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Guiyang, 550001, China
| | - Zimu Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Yizhen Li
- Department of Hematology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, 215003, China
| | - Lei Shi
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiaodong Wang
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Yanfang Tao
- Institute of Pediatric Research, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Junjie Fan
- Department of Hematology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Fenli Zhang
- Department of Pediatrics, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Guiyang, 550001, China
| | - Chunxia Yang
- Department of Pediatrics, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Guiyang, 550001, China
| | - Fahua Yao
- Department of Pediatrics, Guizhou Hospital, Shanghai Children's Medical Center, Guiyang, 550004, China
| | - Tongting Ji
- Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Yongping Zhang
- Department of Hematology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, 215003, China
| | - Bi Zhou
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Department of Pediatrics, Suzhou Hospital Affiliated to Anhui Medical University, Suzhou Municipal Hospital of Anhui Province, Suzhou, 234000, China
| | - Juanjuan Yu
- Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Ailian Guo
- Department of Hematology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, 215003, China
| | - Zhongling Wei
- Department of Hematology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, 215003, China
| | - Wanyan Jiao
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Department of Pediatric, Yancheng Third People's Hospital, Yancheng, 224000, China
| | - Yumeng Wu
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Department of Pediatric, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
| | - Yan Li
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Department of Pediatric, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Di Wu
- Institute of Pediatric Research, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Yijun Wu
- Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Li Gao
- Department of Hematology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, 215003, China
| | - Yixin Hu
- Department of Hematology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, 215003, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China.
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, 215003, China.
| | - Shaoyan Hu
- Department of Hematology, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou, 215003, China.
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, 215003, China.
| | - Xiaoyan Yang
- Department of Pediatrics, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Guiyang, 550001, China.
| |
Collapse
|
48
|
Kellen S, Makovec A, Miller CD, Nazari SS, Elliott A, Deacon A, John E, Vobugari N, Agarwal N, McKay RR, Barata PC, Ryan CJ, Sharifi N, Hwang J, Antonarakis ES. The Influence of the Germline HSD3B1 Adrenal-Permissive Allele (c.1100 C) on the Somatic Alteration Landscape, the Transcriptome, and Immune Cell Infiltration in Prostate Cancer. Cancers (Basel) 2025; 17:1270. [PMID: 40282446 PMCID: PMC12025685 DOI: 10.3390/cancers17081270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: The germline polymorphism in the HSD3B1 gene (c.1100 C) results in adrenal-permissive (CC) or adrenal-restrictive (AA) functions of the protein product by regulating the production of high-affinity ligands that activate androgen signaling. Prior studies have indicated that the CC genotype is associated with worse response to hormonal therapies in prostate cancer (PC) patients. Methods: To characterize the impact of germline HSD3B1 variants on somatic tumor features, we examined 6550 primary and metastatic PCs from the Caris Life Sciences database, in which the genomic and transcriptomic landscapes were acquired via paired whole-exome/whole-transcriptome sequencing. Results: The overall prevalence of the HSD3B1 AA genotype (restrictive-homozygous) was 48.8%, AC (permissive-heterozygous) was 32.8%, and CC (permissive-homozygous) was 14.9%. There was enrichment of the CC genotype in these PC patients as compared to prior reports that examined non-cancerous populations. However, the rates of the CC genotype varied between metastatic site and by race. Compared to the AA genotype, tumors harboring the CC genotype did not demonstrate increased AR alterations, nor higher expression of AR, FOXA1, HOXB13, or AR signaling signatures. We instead found significant changes in immune-associated hallmark pathways, immune cell fractions, and biomarkers that inform the use of immune therapies (TMB-high, MSI-high). Further, the CC and AA genotypes exhibited notable differences in the expression of immunoglobulins, MHC class I/II molecules, and cell surface targets. The differences in expression by HSD3B1 genotype were especially notable in lung and liver metastases. Conclusions: Our study indicates that in prostate cancers, HSD3B1 germline c.1100 allele status may not directly influence tumor-intrinsic genomics but is associated with novel functions beyond androgen signaling.
Collapse
Affiliation(s)
- Samuel Kellen
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; (S.K.); (A.M.); (A.D.); (E.J.)
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA;
| | - Allison Makovec
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; (S.K.); (A.M.); (A.D.); (E.J.)
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA;
| | - Carly D. Miller
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; (S.K.); (A.M.); (A.D.); (E.J.)
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA;
| | | | | | - Aiden Deacon
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; (S.K.); (A.M.); (A.D.); (E.J.)
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA;
| | - Emily John
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; (S.K.); (A.M.); (A.D.); (E.J.)
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA;
| | - Nikitha Vobugari
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA;
| | - Neeraj Agarwal
- Division of Hematology and Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake, UT 84112, USA;
| | - Rana R. McKay
- Department of Radiation Medicine and Applied Sciences, UC San Diego School of Medicine, La Jolla, CA 92093, USA;
| | - Pedro C. Barata
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Charles J. Ryan
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Nima Sharifi
- Desai Sethi Urology Institute and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Justin Hwang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; (S.K.); (A.M.); (A.D.); (E.J.)
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA;
| | - Emmanuel S. Antonarakis
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; (S.K.); (A.M.); (A.D.); (E.J.)
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA;
| |
Collapse
|
49
|
Karimpur-Zahmatkesh A, Khalaj-Kondori M. The perspective of targeting cancer cell metabolism: combination therapy approaches. Mol Biol Rep 2025; 52:375. [PMID: 40202553 DOI: 10.1007/s11033-025-10472-9] [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: 12/23/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025]
Abstract
Cancer cells are considered the most adaptable for their metabolic status, which supports growth, survival, rapid proliferation, invasiveness, and metastasis in a nutrient-deficient microenvironment. Since the discovery of altered glucose metabolism (aerobic glycolysis), which is generally known as a part of metabolic reprogramming and an innate trait of cancer cells, in 1930 via Dr. Otto Warburg, numerous studies have endeavored to recognize various aspects of cancer cell metabolism and find new methods for efficiently eradicating described cells by targeting their energy metabolism. In this way, the outcomes have mainly been promising. Accordingly, outlining the related results will indeed assist us in making a definitive path for developing targeted therapy strategies based on cancer cell-altered metabolism. The present study reviews the key features of cancer cell metabolism and treatment strategies based on them. It emphasizes the importance of targeting cancer cell dysregulated metabolic pathways that influence the cell energy supply and manage cancer cell growth and survival. This trial also introduces a multimodal therapeutic strategy hypothesis, a potential next-generation combination therapy approach, and suggests interdisciplinary research to recognize the complexities of cancer metabolism and exploit them for designing more efficacious cancer therapeutic strategies.
Collapse
Affiliation(s)
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
50
|
Xu X, Fei W, Wu M, He Y, Yang B, Lv C. Construction and validation of a nomogram for identifying the patients at risk for rapid progression of advanced hormone-sensitive prostate cancer. BMC Cancer 2025; 25:634. [PMID: 40200224 PMCID: PMC11980332 DOI: 10.1186/s12885-025-14035-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 03/28/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND This study aimed to evaluate the prognostic significance of lactate dehydrogenase (LDH) and fasting triglyceride-glucose (TyG) index in advanced hormone-sensitive prostate cancer (HSPC) patients, with the ultimate goal of developing and validating a nomogram for predicting castration-resistant prostate cancer (CRPC) free survival. MATERIALS AND METHODS The follow-up data of 207 CRPC patients who had androgen deprivation therapy as their initial and only treatment before progression were retrospectively reviewed. To assess prognostic variables, univariate and multivariate Cox regression analyses were performed. The concordance index (C-index), calibration curves, receiver operating characteristic (ROC) curves, and decision curve analyses (DCA) were utilized to construct and test a novel nomogram model. RESULTS TyG index, LDH, M stage and Gleason sum were determined to be independent prognostic markers and were combined to create a nomogram. This nomogram worked well in the tailored prediction of CRPC development at the sixth, twelve, eighteen, and twenty-fourth months. The C-indexes for the training and validation sets were 0.798 and 0.790, respectively. The ROC curves, calibration plots, and DCA all indicated good discrimination and prediction performance. Furthermore, the nomogram had a higher prognostic ability than the M stage and the Gleason sum. The nomogram-related risk score classified the patient population into two groups with significant progression differences. CONCLUSIONS The created nomogram could help identify patients at high risk for rapid progression of advanced HSPC, allowing for the formulation of tailored therapy regimens and follow-up methods in a timely manner.
Collapse
Affiliation(s)
- Xiaolong Xu
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Weiyu Fei
- Department of Emergency Intensive Care Unit, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Mingshuang Wu
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yi He
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| | - Bo Yang
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| | - Cuicui Lv
- Department of Endocrine, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|