1
|
Xu M, Xu C, Wang R, Tang Q, Zhou Q, Wu W, Wan X, Mo H, Pan J, Wang S. Treating human cancer by targeting EZH2. Genes Dis 2025; 12:101313. [PMID: 40028035 PMCID: PMC11870178 DOI: 10.1016/j.gendis.2024.101313] [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: 10/06/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 03/05/2025] Open
Abstract
Enhancer of zeste homolog 2 (EZH2), an epigenetic regulator that primarily inhibits downstream gene expression by tri-methylating histone H3, which is usually overexpressed in tumors and participates in many processes such as tumor occurrence and development, invasion, migration, drug resistance, and anti-tumor immunity as an oncogene, making it an important biomarker in cancer therapy. Collectively, several transcription factors and RNAs cooperate to facilitate the elevated expression of EZH2 in cancer. Although the significance of blocking EZH2 in cancer for inhibiting cancer progression is widely recognized, the clinical application of EZH2 inhibitors continues to encounter numerous challenges. In this review, drawing upon our comprehensive understanding of the factual underpinnings of EZH2's role in cancer, we aim to clarify the crucial importance of targeting EZH2 in cancer treatment. Furthermore, we summarize the current research landscape surrounding targeted EZH2 inhibitors and offer insights into potential future applications of these inhibitors.
Collapse
Affiliation(s)
- Mengfei Xu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Chunyan Xu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Rui Wang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Qing Tang
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Qichun Zhou
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Wanyin Wu
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Xinliang Wan
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Handan Mo
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Jun Pan
- Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Sumei Wang
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| |
Collapse
|
2
|
Liu Q, Yu M, Lin Z, Wu L, Xia P, Zhu M, Huang B, Wu W, Zhang R, Li K, Zhu L, Wang Q. COL1A1-positive endothelial cells promote gastric cancer progression via the ANGPTL4-SDC4 axis driven by endothelial-to-mesenchymal transition. Cancer Lett 2025; 623:217731. [PMID: 40254092 DOI: 10.1016/j.canlet.2025.217731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/19/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Gastric cancer (GC) is an aggressive and heterogeneous disease with poor survival outcomes. The progression of GC involves complex, multi-step processes. Endothelial cells (ECs) play a crucial role in tumor angiogenesis, proliferation, invasion, and metastasis, particularly through the process of endothelial-to-mesenchymal transition (EndoMT). However, the specific role and mechanisms of EndoMT in gastric cancer remain unclear. Based on 6 GC single-cell RNA-sequencing (scRNA-seq) cohorts (samples = 97), we established an EndoMT-related gene signature, termed EdMTS. Leveraging this gene signature, ssGSEA was applied to calculate sample scores across multiple bulk RNA-seq datasets, which include information on immunotherapy, metastasis, GC progression, and survival. Moreover, we applied the Monocle2 method to calculate cell pseudotime and used CellChat to analyze interactions between malignant and EC cells. We verified the molecular mechanism by multiple immunofluorescence and cell function experiments. Findings In this study, we established a single-cell atlas of ECs in GC and identified a subpopulation of COL1A1+ ECs that play a critical role in tumor progression and metastasis. These COL1A1+ ECs were significantly associated with worse clinical outcomes in GC patients. Further analysis revealed that COL1A1+ ECs originated from lymphatic ECs and underwent EndoMT through the upregulation of CEBPB, driving tumor invasiveness. Moreover, COL1A1+ ECs interacted with malignant cells via ANGPTL4-SDC4 axis, enhancing invasion and migration. These findings provide a deeper understanding of the role of COL1A1+ ECs in GC progression and highlight potential therapeutic targets for disrupting the EndoMT process in these cells to provide a benefit for GC patients.
Collapse
Affiliation(s)
- Quanzhong Liu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Miao Yu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Zihan Lin
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Lingxiang Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Peng Xia
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Mengyan Zhu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Bin Huang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Wei Wu
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Ruohan Zhang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Kening Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Lingjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
3
|
Zheng Z, Wang W, Feng M, Chen X, Ren F, Hou Y. The mechanism of EZH2/H3K27me3 downregulating CXCL10 to affect CD8 + T cell exhaustion to participate in the transformation from myelodysplastic syndrome to acute myeloid leukaemia. Br J Haematol 2025. [PMID: 40201935 DOI: 10.1111/bjh.20066] [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: 11/11/2024] [Accepted: 03/21/2025] [Indexed: 04/10/2025]
Abstract
Myelodysplastic syndromes (MDS) and acute myeloid leukaemia (AML) link to unfavourable prognoses. We explored the mechanism of enhancer of zeste homologue 2/histone H3 of lysine 27 (EZH2/H3K27me3) downregulating C-X-C motif chemokine 10 (CXCL10) to affect CD8+ T-cell exhaustion, participating in MDS-to-AML transformation. NHD13 mice were treated with GSK126 (EZH2 inhibitor) and CXCL10 neutralizing antibody, with transformation time, blood cell counts and CD8+ T cell determined. SKM-1 cells treated with short hairpin-EZH2, overexpressing-EZH2, GSK126 and CXCL10 were co-cultured with CD8+ T cells. EZH2, CXCL10, H3K27me3 and EZH2 levels and EZH2 enzyme activity were assessed. CD8+ T-cell cytotoxicity, exhaustion, apoptosis and SKM-1 cell malignant behaviours were evaluated. In vivo, EZH2 inhibition upregulated CXCL10, decelerating MDS to AML transformation and delaying CD8+ T-cell exhaustion. EZH2 inhibition elevated peripheral blood cells, alleviated splenomegaly, reduced CD8+ T cells, elevated CD8+ T cytotoxicity and abated CD8+ T-cell exhaustion in NHD13 mice. CXCL10 neutralizing antibody accelerated AML transformation by inhibiting CD8+ T-cell exhaustion via EZH2. In vitro, EZH2 overexpression facilitated CD8+ T-cell exhaustion and SKM-1 cell malignant behaviours. EZH2-mediated H3K27me3 curbed CXCL10 transcription and secretion. Collectively, EZH2/H3K27me3 downregulates CXCL10 to facilitate CD8+ T-cell exhaustion, accelerating transformation from MDS to AML.
Collapse
Affiliation(s)
- Zhuanzhen Zheng
- Department of Hemapathotology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Wenjing Wang
- Department of Hemapathotology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Mengjing Feng
- Department of Hemapathotology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiuhua Chen
- Department of Hemapathotology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Fanggang Ren
- Department of Hemapathotology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yanfei Hou
- Department of Hemapathotology, Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
4
|
Liao Q, Jin Z, Long H, Zhu B. Deciphering cancer complexity: perspective on hematopoietic remodeling-mediated immunosuppression. Oncogene 2025:10.1038/s41388-025-03361-9. [PMID: 40195467 DOI: 10.1038/s41388-025-03361-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 04/09/2025]
Affiliation(s)
- Qian Liao
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing, China
| | - Zheng Jin
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing, China
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
- Chongqing Key Laboratory of Immunotherapy, Chongqing, China.
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
- Chongqing Key Laboratory of Immunotherapy, Chongqing, China.
| |
Collapse
|
5
|
Zhang H, Pang Y, Yi L, Wang X, Wei P, Wang H, Lin S. Epigenetic regulators combined with tumour immunotherapy: current status and perspectives. Clin Epigenetics 2025; 17:51. [PMID: 40119465 PMCID: PMC11929245 DOI: 10.1186/s13148-025-01856-6] [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: 12/31/2024] [Accepted: 03/03/2025] [Indexed: 03/24/2025] Open
Abstract
Immunotherapy, particularly immune checkpoint inhibitor therapy, has demonstrated clinical benefits in solid tumours. Despite its satisfactory clinical efficacy, it still faces several issues, such as limited eligibility, low response rates and cytotoxicity. Cancer epigenetics implies that tumour cells exhibit unique phenotypes because of their unique characteristics, thus reprogramming of the epigenome holds promise for cancer therapy. Epigenetic regulation plays an important role in regulating gene expression during tumour development and maintenance. Epigenetic regulators induce cancer cell cycle arrest, apoptosis and differentiation of cancer cells, thereby exerting anti-tumour effects. Recent studies have revealed a significant correlation between epigenetic regulatory factors and immune checkpoint therapy. Epigenetics can modulate various aspects of the tumour immune microenvironment and immune response to enhance the sensitivity of immunotherapy, such as lowering the concentration required and mitigating cytotoxicity. This review primarily discusses DNA methyltransferase inhibitors, histone deacetylase inhibitors, enhancer of zeste homolog 2 inhibitors and lysine-specific demethylase 1 inhibitors, which are associated with transcriptional repression. This repression alters the expression of genes involved in the immune checkpoint, thereby enhancing the effectiveness of immunotherapy. We also discuss the potential and challenges of tumour immunotherapy and highlight its advantages, application challenges and clinical research on integrating epigenetic regulatory factors with tumour immunotherapy.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Gastroenterology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Yutong Pang
- Department of Gastroenterology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Ling Yi
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Xiaojue Wang
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Panjian Wei
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Haichao Wang
- Institute of Resources and Environment, Beijing Academy of Science and Technology, Beijing, 100089, China.
| | - Shuye Lin
- Department of Gastroenterology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
6
|
Ji Y, Xiao C, Fan T, Deng Z, Wang D, Cai W, Li J, Liao T, Li C, He J. The epigenetic hallmarks of immune cells in cancer. Mol Cancer 2025; 24:66. [PMID: 40038722 DOI: 10.1186/s12943-025-02255-4] [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: 11/17/2024] [Accepted: 01/30/2025] [Indexed: 03/06/2025] Open
Abstract
Targeting the dysregulation of epigenetic mechanisms in cancer has emerged as a promising therapeutic strategy. Although the significant rationale progress of epigenetic therapies in blocking cancer cells, how epigenetic regulation shapes tumor microenvironment (TME) and establishes antitumor immunity remains less understood. Recent study focus has been put on the epigenetic-mediated changes in the fate of immune cells, including the differentiation, expansion, recruitment, functionalization, and exhaustion of T cells, natural killer (NK) cells, tumor-associated macrophages (TAMs), dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), and B cells within the TME. Here, we review the latest molecular and clinical insights into how DNA modifications, histone modification, and epitranscriptome-related regulations shape immune cells of various cancers. We also discuss opportunities for leveraging epigenetic therapies to improve cancer immunotherapies. This review provides the epigenetic foundations of cancer immunity and proposes the future direction of combination therapies.
Collapse
Affiliation(s)
- Yu Ji
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Di Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenpeng Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jia Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tianle Liao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
7
|
Ma J, Zhang Y, Li J, Dang Y, Hu D. Regulation of histone H3K27 methylation in inflammation and cancer. MOLECULAR BIOMEDICINE 2025; 6:14. [PMID: 40042761 PMCID: PMC11882493 DOI: 10.1186/s43556-025-00254-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
Inflammation is a multifaceted defense mechanism of the immune system against infection. Chronic inflammation is intricately linked to all stages of tumorigenesis and is therefore associated with an elevated risk of developing serious cancers. Epigenetic mechanisms have the capacity to trigger inflammation as well as facilitate tumor development and transformation within an inflammatory context. They achieve this by dynamically modulating the expression of both pro-inflammatory and anti-inflammatory cytokines, which in turn sustains chronic inflammation. The aberrant epigenetic landscape reconfigures the transcriptional programs of inflammatory and oncogenic genes. This reconfiguration is pivotal in dictating the biological functions of both tumor cells and immune cells. Aberrant histone H3 lysine 27 site (H3K27) methylation has been shown to be involved in biological behaviors such as inflammation development, tumor progression, and immune response. The establishment and maintenance of this repressive epigenetic mark is dependent on the involvement of the responsible histone modifying enzymes enhancer of zeste homologue 2 (EZH2), jumonji domain containing 3 (JMJD3) and ubiquitously transcribed tetratricopeptide repeat gene X (UTX) as well as multiple cofactors. In addition, specific pharmacological agents have been shown to modulate H3K27 methylation levels, thereby modulating inflammation and carcinogenesis. This review comprehensively summarises the current characteristics and clinical significance of epigenetic regulation of H3K27 methylation in the context of inflammatory response and tumor progression.
Collapse
Affiliation(s)
- Jing Ma
- Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, No. 358 Datong Road, Pudong New Area, Shanghai, 200137, China
| | - Yalin Zhang
- Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, No. 358 Datong Road, Pudong New Area, Shanghai, 200137, China
| | - Jingyuan Li
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, (Shanghai University of Traditional Chinese Medicine), Shanghai, 200032, China
| | - Yanqi Dang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, (Shanghai University of Traditional Chinese Medicine), Shanghai, 200032, China.
| | - Dan Hu
- Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, No. 358 Datong Road, Pudong New Area, Shanghai, 200137, China.
| |
Collapse
|
8
|
Xiong X, Liu Y, Du Y, Lai X, Si C, Miao H. Unveiling the effects of GSK126 on osteosarcoma cells implications for apoptosis, autophagy, and cellular migration. Discov Oncol 2025; 16:245. [PMID: 40014170 PMCID: PMC11868014 DOI: 10.1007/s12672-025-02010-7] [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: 09/11/2024] [Accepted: 02/21/2025] [Indexed: 02/28/2025] Open
Abstract
Osteosarcoma, a malignant bone tumor, faces significant treatment challenges. Enhancer of zeste homolog 2 (EZH2) shows aberrant expression in various tumors including osteosarcoma, which is identified as a potential therapeutic target. The anti-cancer efficacy of EZH2 inhibitor GSK126 has attracted attention, yet its impact on osteosarcoma cells was not fully understood. The study investigated the effects of GSK126 on osteosarcoma cells, particularly in apoptosis, autophagy, and cell motility. Our findings revealed that GSK126 induced apoptosis and autophagy, evidenced by increased markers like cleaved caspase-3 and LC3-II, and decreased cellular migration, through downregulation of the Fuse Binding Protein 1 (FBP1)/C-Myc axis. These findings suggest GSK126 as a promising therapeutic against osteosarcoma, offering a dual action of promoting cell death and hindering migration.
Collapse
Affiliation(s)
- Xifeng Xiong
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Yulin Liu
- Guangdong Medical University, Zhanjiang, Guangdong, China
- Department of Pain, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Yanli Du
- Guangdong Medical University, Zhanjiang, Guangdong, China
- Department of Orthopedics, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Xudong Lai
- Department of Infectious Diseases, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Chunming Si
- Department of Orthopedics Center, Xinjiang Production and Construction Corps Hospital, Urumqi, Xinjiang, China.
| | - Haixiong Miao
- Department of Orthopedics, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China.
| |
Collapse
|
9
|
Bowen CM, Duzagac F, Martel-Martel A, Reyes-Uribe L, Zaheer M, Thompson J, Deng N, Sinha R, Mazumdar S, Taggart MW, Jain AK, Tosti E, Edelmann W, Sinha KM, Vilar E. Inhibition of histone methyltransferase EZH2 for immune interception of colorectal cancer in Lynch syndrome. JCI Insight 2025; 10:e177545. [PMID: 39946195 PMCID: PMC11949072 DOI: 10.1172/jci.insight.177545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/12/2025] [Indexed: 03/25/2025] Open
Abstract
Colorectal precancers in Lynch syndrome (LS) exhibit a distinct immune profile, presenting unique opportunities for developing immune-interception strategies to prevent carcinogenesis. Epigenetic modulation by EZH2 of immune-related genes is implicated in the carcinogenesis of different cancer types, including colorectal cancer. This study utilizes a mouse model of LS and ex vivo colonic organoids to assess the effects of the EZH2 inhibitor GSK503 on immune regulatory pathways, tumorigenesis, and epigenetic reprogramming. Our findings revealed that GSK503 significantly increased CD4+ and CD8+ T cells in both splenocytes and colonic mucosa of treated mice compared with controls. Additionally, a preventive dose of GSK503 over 9 weeks notably reduced adenoma multiplicity, demonstrating its efficacy as a preventive modality. Single-cell RNA-Seq and molecular analyses showed activation of immune and apoptotic markers, along with a reduction in H3K27 methylation levels in colonic crypts. ChIP sequencing further revealed decreased levels of H3K27me3 and H3K4me1, while levels of the active enhancer marks H3K4me3 and H3K27Ac increased in treated mice. Collectively, these findings indicate that EZH2 inhibition enhances immune responses through epigenetic reprogramming in the genome of LS mice, establishing a promising framework for the clinical development of EZH2 inhibitors as a cancer prevention strategy for LS carriers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nan Deng
- Department of Clinical Cancer Prevention
| | - Ria Sinha
- Department of Clinical Cancer Prevention
| | | | | | - Abhinav K. Jain
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Elena Tosti
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Winfried Edelmann
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | | |
Collapse
|
10
|
Zhu E, Liu Y, Xie S, Hou J, Yang X, Xu M, Yang F, Li Z, Zhu B, Zha H. IGF2-IGF1R signaling inhibition delays the growth of IGF2-high colorectal cancer by modulating MDSCs. Biochem Biophys Res Commun 2025; 746:151230. [PMID: 39756209 DOI: 10.1016/j.bbrc.2024.151230] [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: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025]
Abstract
Approximately 22 % of human colorectal cancers (CRC) are characterized by IGF2 overexpression, and the tumor-promoting role of IGF2 has been widely reported. Despite promising preclinical results, IGF2 signaling inhibition therapy has exhibited limited efficacy in treating unselected patients with CRC. Recent evidence suggests that IGF2-high CRC are more sensitive to IGF2 signaling blockade therapy in immune-deficient mice, suggesting that IGF2-high CRC can benefit from IGF2 signaling blockade therapy. However, T cells are absent in immunodeficient mice, and the effect of blocking IGF2 signaling on T cell-mediated antitumor immunity remains unknown. Herein, using an implanted mouse tumor model in immunocompetent hosts, we report that PQ401, an IGF2-IGF1R inhibitor, significantly inhibited the growth of IGF2-high CRC cells. PQ401 treatment increased the infiltration and function of tumor-infiltrating CD4+ and CD8+ T cells in a T cell-extrinsic manner. Our findings suggest that myeloid-derived suppressor cells (MDSCs) highly express the IGF2 receptor IGF1R. Moreover, PQ401 treatment inhibits the suppressive function and recruitment of MDSCs, thereby promoting the anti-tumor activity of T cells. These results provide a potential therapeutic regimen for patients with IGF2-high CRC.
Collapse
Affiliation(s)
- Enjian Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Ying Liu
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi Medical University, Zun Yi, Gui Zhou, China
| | - Shuanglong Xie
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China; School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Junlei Hou
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xuezhi Yang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China; Department of Radiology, 964th Hospital of Chinese People's Liberation Army, Changchun, 130000, China
| | - Minhao Xu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Fei Yang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Zhaoxia Li
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China.
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China; Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi Medical University, Zun Yi, Gui Zhou, China.
| | - Haoran Zha
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China; Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China.
| |
Collapse
|
11
|
Hou Y, Zak J, Shi Y, Pratumchai I, Dinner B, Wang W, Qin K, Weber EW, Teijaro JR, Wu P. Transient EZH2 Suppression by Tazemetostat during In Vitro Expansion Maintains T-Cell Stemness and Improves Adoptive T-Cell Therapy. Cancer Immunol Res 2025; 13:47-65. [PMID: 39365901 PMCID: PMC11717634 DOI: 10.1158/2326-6066.cir-24-0089] [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: 01/24/2024] [Revised: 06/13/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
The histone methyltransferase enhancer of zeste homolog 2 (EZH2) plays important roles in T-cell differentiation, proliferation, and function. Previous studies have demonstrated that genetic deletion of EZH2 in CD8+ or total T cells impairs their antiviral and antitumor activities, cytokine production, and ability to expand upon rechallenge. Contrary to the detrimental role of deleting T cell-intrinsic EZH2, in this study, we demonstrated that transient inhibition of EZH2 in T cells prior to the phenotypic onset of exhaustion with a clinically approved inhibitor, tazemetostat (Taz), delayed their dysfunctional progression and preserved T-cell stemness and polyfunctionality but had no negative impact on cell proliferation. Taz-induced T-cell epigenetic reprogramming increased the expression of the self-renewal T-cell transcription factor TCF1 by reducing H3K27 methylation at its promoter preferentially in rapidly dividing T cells. In a murine melanoma model, T cells depleted of EZH2 induced poor tumor control, whereas adoptively transferred T cells pretreated with Taz exhibited superior antitumor immunity, especially when used in combination with anti-PD-1 blockade. Collectively, these data highlight the potential of transient epigenetic reprogramming by EZH2 inhibition to enhance adoptive T-cell immunotherapy.
Collapse
Affiliation(s)
- Yingqin Hou
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
- Authors contributed equally
| | - Jaroslav Zak
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Authors contributed equally
| | - Yujie Shi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Isaraphorn Pratumchai
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Brandon Dinner
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Wenjian Wang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ke Qin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Evan W. Weber
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| | - John R. Teijaro
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Peng Wu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
12
|
Liu K, Li Y, Shen M, Xu W, Wu S, Yang X, Zhang B, Lin N. Epigenetic Regulation of Stromal and Immune Cells and Therapeutic Targets in the Tumor Microenvironment. Biomolecules 2025; 15:71. [PMID: 39858465 PMCID: PMC11764280 DOI: 10.3390/biom15010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/19/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
The tumor microenvironment (TME) plays a pivotal role in neoplastic initiation and progression. Epigenetic machinery, governing the expression of core oncogenes and tumor suppressor genes in transformed cells, significantly contributes to tumor development at both primary and distant sites. Recent studies have illuminated how epigenetic mechanisms integrate external cues and downstream signals, altering the phenotype of stromal cells and immune cells. This remolds the area surrounding tumor cells, ultimately fostering an immunosuppressive microenvironment. Therefore, correcting the TME by targeting the epigenetic modifications holds substantial promise for cancer treatment. This review synthesizes recent research that elucidates the impact of specific epigenetic regulations-ranging from DNA methylation to histone modifications and chromatin remodeling-on stromal and immune cells within the TME. Notably, we highlight their functional roles in either promoting or restricting tumor progression. We also discuss the potential applications of epigenetic agents for cancer treatment, envisaging their ability to normalize the ecosystem. This review aims to assist researchers in understanding the dynamic interplay between epigenetics and the TME, paving the way for better epigenetic therapy.
Collapse
Affiliation(s)
- Kang Liu
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Yue Li
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Minmin Shen
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Drug Clinical Trial Institution, Huzhou Central Hospital, Huzhou 313000, China
| | - Wei Xu
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Shanshan Wu
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Xinxin Yang
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Bo Zhang
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Nengming Lin
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (K.L.); (Y.L.); (M.S.); (W.X.); (S.W.); (X.Y.)
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Westlake University, Hangzhou 310024, China
| |
Collapse
|
13
|
Cao R, Ni J, Zhang X, Qi M, Zhang Z, Liu Z, Chen G. Recent Advances in enhancer of zeste homolog 2 Inhibitors: Structural insights and therapeutic applications. Bioorg Chem 2025; 154:108070. [PMID: 39709735 DOI: 10.1016/j.bioorg.2024.108070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/11/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024]
Abstract
Enhancer of Zeste Homolog 2 (EZH2) is overexpressed in many malignancies and plays a critical role in cancer progression. Therefore, it is considered a promising target for therapeutic intervention. Although several EZH2 inhibitors have entered clinical trials, only one has received FDA approval. In this review, we focus on the latest advancements in highly selective and potent dual-targeting EZH2 inhibitors, as well as proteolysis-targeted chimeras (PROTACs) and hydrophobic tagging (HYT) degraders. These novel compounds have been developed to address the existing gaps in the management of abnormal EZH2 expression. Notably, EZH2 inhibitors have shown great efficacy in antitumor therapy and have also demonstrated promising results in antiviral, anti-inflammatory, antisclerotic, bone protection, and nerve injury pain applications. The insights gained from this analysis could provide valuable guidance for future drug design and optimization of EZH2 inhibitors, potentially expediting the discovery of new inhibitors or degraders targeting EZH2.
Collapse
Affiliation(s)
- Ruolin Cao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jiayang Ni
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xiaoyu Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Minggang Qi
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zhen Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zhongbo Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
14
|
Kempkes RWM, Prinjha RK, de Winther MPJ, Neele AE. Novel insights into the dynamic function of PRC2 in innate immunity. Trends Immunol 2024; 45:1015-1030. [PMID: 39603889 DOI: 10.1016/j.it.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024]
Abstract
The polycomb repressive complex 2 (PRC2) is an established therapeutic target in cancer. PRC2 catalyzes methylation of histone H3 at lysine 27 (H3K27me3) and is known for maintaining eukaryote cell identity. Recent discoveries show that modulation of PRC2 not only impacts cell differentiation and tumor growth but also has immunomodulatory properties. Here, we integrate multiple immunological fields to understand PRC2 and its subunits in epigenetic canonical regulation and non-canonical mechanisms within innate immunity. We discuss how PRC2 regulates hematopoietic stem cell proliferation, myeloid cell differentiation, and shapes innate immune responses. The PRC2 catalytic domain EZH2 is upregulated in various human inflammatory diseases and its deletion or inhibition in experimental mouse models can reduce disease severity, emphasizing its importance in regulating inflammation.
Collapse
Affiliation(s)
- Rosalie W M Kempkes
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Disease, Amsterdam, the Netherlands
| | | | - Menno P J de Winther
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Disease, Amsterdam, the Netherlands.
| | - Annette E Neele
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, the Netherlands; Amsterdam Institute for Immunology and Infectious Disease, Amsterdam, the Netherlands.
| |
Collapse
|
15
|
Fischetti I, De Luca M, Jachetti E. Exploring the role of EZH2 modulation in shaping the tumor microenvironment. Epigenomics 2024; 16:1265-1268. [PMID: 39387445 PMCID: PMC11534139 DOI: 10.1080/17501911.2024.2410693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Affiliation(s)
- Irene Fischetti
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Michele De Luca
- Integrated Biology of Rare Tumors Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Elena Jachetti
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| |
Collapse
|
16
|
Huang Y, Tian Z, Bi J. Intracellular checkpoints for NK cell cancer immunotherapy. Front Med 2024; 18:763-777. [PMID: 39340588 DOI: 10.1007/s11684-024-1090-6] [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: 02/08/2024] [Accepted: 05/17/2024] [Indexed: 09/30/2024]
Abstract
Natural killer (NK) cells are key innate immune lymphocytes, which play important roles against tumors. However, tumor-infiltrating NK cells are always hypofunctional/exhaustive. On the one hand, this state is contributed by context-dependent interactions between inhibitory NK cell checkpoint receptors and their ligands, which usually vary in different tumor types and stages during tumor development. On the other hand, the inhibitory functions of intracellular checkpoint molecules of NK cells are more similar across different tumor types, representing common mechanisms limiting the potential of NK cell therapy. In this review, representative NK cell intracellular checkpoint molecules in different aspects of NK cell biology were reviewed, and therapeutic potentials were discussed by targeting these molecules to promote antitumor NK cell therapy.
Collapse
Affiliation(s)
- Yingying Huang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, 530021, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, 530021, China
- Collaborative Innovation Center of Regenerative Medicine and Medical BioResource Development and Application, Guangxi Medical University, Nanning, 530021, China
| | - Zhigang Tian
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Beijing, 100864, China
| | - Jiacheng Bi
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
17
|
Le M, Lu W, Tan X, Luo B, Yu T, Sun Y, Guo Z, Huang P, Zhu D, Wu Q, Ganesan A, Wen S. Design, Synthesis, and Biological Evaluation of Potent EZH2/LSD1 Dual Inhibitors for Prostate Cancer. J Med Chem 2024; 67:15586-15605. [PMID: 39196854 DOI: 10.1021/acs.jmedchem.4c01250] [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: 08/30/2024]
Abstract
As histone modification enzymes, EZH2 mediates H3K27 trimethylation (H3K27me3), whereas LSD1 removes methyl groups from H3K4me1/2 and H3K9me1/2. Synergistic anticancer effects of combining inhibitors of these two enzymes are observed in leukemia and prostate cancer. Thus, a series of EZH2/LSD1 dual inhibitors are designed and synthesized to evaluate their anticancer activity. After the structure-activity study, one of the best compounds, ML234, displayed excellent antiproliferative capacity against prostate cancer cell lines LNCAP, PC3, and 22RV1. Enzymatic assays ascertained that the anticancer potency of ML234 was mediated through coinhibition of EZH2 and LSD1. Moreover, the accumulation of H3K4me2 and H3K9me2 and the decrease of H3K27me3 induced by ML234 were verified by Western blot analysis. More importantly, the compound remarkably suppressed the tumor growth and enhanced the therapeutic efficacy of clinical drug enzalutamide in the 22RV1 xenograft mouse model, indicating that it may have potential as an anticancer agent in prostate cancer.
Collapse
Affiliation(s)
- Meiling Le
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Wenhua Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xiaozhuo Tan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Bingling Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Tiantian Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yameng Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zhirong Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Daqian Zhu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Qiang Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa 999078, Macau
| | - A Ganesan
- School of Chemistry, Pharmacy & Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - Shijun Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
18
|
Long M, Zhou Y, Guo D, Zhu Q, Liang H, Ji X, Chen N, Song H. Unzippable Siamese Nanoparticles for Programmed Two-Stage Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402456. [PMID: 38810924 DOI: 10.1002/adma.202402456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/13/2024] [Indexed: 05/31/2024]
Abstract
Epigenetic drugs (epi-drugs) can destruct cancer cells and initiate both innate and adaptive immunity, yet they have achieved very limited success in solid tumors so far, partly attributing to their concurrent induction of the myeloid-derived suppressor cell (MDSC) population. Here, dissociable Siamese nanoparticles (SIANPs) are developed for tumor cell-targeted delivery of epi-drug CM-272 and MDSC-targeted delivery of small molecule inhibitor Ibrutinib. The SIANPs are assembled via interparticle DNA annealing and detached via tumor microenvironment-triggered strand separation. Such binary regulation induces endogenous retrovirus expression and immunogenic cell death in tumor cells while restraining the immunosuppressive effects of MDSCs, and synergistically promotes dendritic cell maturation and CD8+ T cell activation for tumor inhibition. Significantly, immune microenvironment remodeling via SIANPs further overcomes tumor resistance to immune checkpoint blockade therapy. This study represents a two-pronged approach for orchestrating immune responses, and paves a new way for employing epi-drugs in cancer immunotherapy.
Collapse
Affiliation(s)
- Mei Long
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanfeng Zhou
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Daoxia Guo
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qingyun Zhu
- College of Chemistry and Materials Science, The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials, and Shanghai Frontiers Science Center of Biomimetic Catalysis, Shanghai Normal University, Shanghai, 200234, China
| | - Huan Liang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyuan Ji
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Nan Chen
- College of Chemistry and Materials Science, The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials, and Shanghai Frontiers Science Center of Biomimetic Catalysis, Shanghai Normal University, Shanghai, 200234, China
| | - Haiyun Song
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
19
|
Liu Q, Wu N, Hou P. PRPS2-mediated modulation of the antitumor immune response in lung cancer through CCL2-mediated tumor-associated macrophages and myeloid-derived suppressor cells. Thorac Cancer 2024; 15:1739-1748. [PMID: 38952044 PMCID: PMC11320087 DOI: 10.1111/1759-7714.15398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/08/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Phosphoribosyl pyrophosphate synthetase 2 (PRPS2) is known as an oncogene in many types of cancers, including lung cancer. However, its role in regulating tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC) remains unclear. Our study aimed to explore the involvement of PRPS2 in TAM and MDSC regulation. METHODS Stable Lewis lung cancer (LLC) cell lines were established using a lentivirus system. These LLC lines were then used to establish tumor model in mice. The levels of target genes were determined using qPCR, western blotting, and ELISA assays. The percentage of different immune cell types was analyzed using fluorescence-activated cell sorting. The chemotaxis ability of TAM and MDSC was evaluated using an in vitro transwell chemotaxis assay. RESULTS Notably, PRPS2 was found to regulate the chemotaxis of TAM and MDSC in tumor cells, as evidenced by the positive correlation of PRPS2 expression levels and abundance of TAM and MDSC populations. In addition, the expression of CCL2, mediated by PRPS2, was identified as a key factor in the chemotaxis of TAM and MDSC, as evidenced by a significant reduction in macrophages and MDSC numbers in the presence of the CCL2 antibody. Furthermore, in vivo experiments confirmed the involvement of PRPS2 in mediating CCL2 expression. PRPS2 was also found to regulate immune cell infiltration into tumors, whereas knockdown of CCL2 reversed the phenotype induced by PRPS2 overexpression. In tumor tissues from mice implanted with LLC-PRPS2-shCCL2 cells, a notable increase in CD4+ and CD8+ T cell percentages, alongside a marked decrease in TAMs, M-MDSC, and PMN-MDSC, was observed. CONCLUSION Taken together, PRPS2 plays a crucial role in modulating the antitumor immune response by reprogramming CCL2-mediated TAM and MDSC.
Collapse
Affiliation(s)
- Qing Liu
- Department of OncologyFujian Medical University Union HospitalFuzhouChina
- Fujian Key Laboratory of Translational Cancer MedicineFuzhouChina
| | - Ningzi Wu
- Fujian Medical UniversityFuzhouChina
| | - Peifeng Hou
- Department of OncologyFujian Medical University Union HospitalFuzhouChina
- Fujian Key Laboratory of Translational Cancer MedicineFuzhouChina
| |
Collapse
|
20
|
Luo Y, Lu J, Lei Z, Zhu H, Rao D, Wang T, Fu C, Zhang Z, Xia L, Huang W. Lysine methylation modifications in tumor immunomodulation and immunotherapy: regulatory mechanisms and perspectives. Biomark Res 2024; 12:74. [PMID: 39080807 PMCID: PMC11289998 DOI: 10.1186/s40364-024-00621-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/17/2024] [Indexed: 08/02/2024] Open
Abstract
Lysine methylation is a crucial post-translational modification (PTM) that significantly impacts gene expression regulation. This modification not only influences cancer development directly but also has significant implications for the immune system. Lysine methylation modulates immune cell functions and shapes the anti-tumor immune response, highlighting its dual role in both tumor progression and immune regulation. In this review, we provide a comprehensive overview of the intrinsic role of lysine methylation in the activation and function of immune cells, detailing how these modifications affect cellular processes and signaling pathways. We delve into the mechanisms by which lysine methylation contributes to tumor immune evasion, allowing cancer cells to escape immune surveillance and thrive. Furthermore, we discuss the therapeutic potential of targeting lysine methylation in cancer immunotherapy. Emerging strategies, such as immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T-cell (CAR-T) therapy, are being explored for their efficacy in modulating lysine methylation to enhance anti-tumor immune responses. By targeting these modifications, we can potentially improve the effectiveness of existing treatments and develop novel therapeutic approaches to combat cancer more effectively.
Collapse
Affiliation(s)
- Yiming Luo
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Junli Lu
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhen Lei
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - He Zhu
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Dean Rao
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Tiantian Wang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chenan Fu
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhiwei Zhang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030, Hubei, China
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Wenjie Huang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030, Hubei, China.
| |
Collapse
|
21
|
Liu W, Kuang T, Liu L, Deng W. The role of innate immune cells in the colorectal cancer tumor microenvironment and advances in anti-tumor therapy research. Front Immunol 2024; 15:1407449. [PMID: 39100676 PMCID: PMC11294098 DOI: 10.3389/fimmu.2024.1407449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/25/2024] [Indexed: 08/06/2024] Open
Abstract
Innate immune cells in the colorectal cancer microenvironment mainly include macrophages, neutrophils, natural killer cells, dendritic cells and bone marrow-derived suppressor cells. They play a pivotal role in tumor initiation and progression through the secretion of diverse cytokines, chemokines, and other factors that govern these processes. Colorectal cancer is a common malignancy of the gastrointestinal tract, and understanding the role of innate immune cells in the microenvironment of CRC may help to improve therapeutic approaches to CRC and increase the good prognosis. In this review, we comprehensively explore the pivotal role of innate immune cells in the initiation and progression of colorectal cancer (CRC), alongside an extensive evaluation of the current landscape of innate immune cell-based immunotherapies, thereby offering valuable insights for future research strategies and clinical trials.
Collapse
Affiliation(s)
| | | | | | - Wenhong Deng
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
22
|
Long L, Zhang H, Zhou Z, Duan L, Fan D, Wang R, Xu S, Qiao D, Zhu W. Pyrrole-containing hybrids as potential anticancer agents: An insight into current developments and structure-activity relationships. Eur J Med Chem 2024; 273:116470. [PMID: 38762915 DOI: 10.1016/j.ejmech.2024.116470] [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: 03/08/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/21/2024]
Abstract
Cancer poses a significant threat to human health. Therefore, it is urgent to develop potent anti-cancer drugs with excellent inhibitory activity and no toxic side effects. Pyrrole and its derivatives are privileged heterocyclic compounds with significant diverse pharmacological effects. These compounds can target various aspects of cancer cells and have been applied in clinical settings or are undergoing clinical trials. As a result, pyrrole has emerged as a promising drug scaffold and has been further probed to get novel entities for the treatment of cancer. This article reviews recent research progress on anti-cancer drugs containing pyrrole. It focuses on the mechanism of action, biological activity, and structure-activity relationships of pyrrole derivatives, aiming to assist in designing and synthesizing innovative pyrrole-based anti-cancer compounds.
Collapse
Affiliation(s)
- Li Long
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Han Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - ZhiHui Zhou
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Lei Duan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Dang Fan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Ran Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Shan Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China.
| | - Dan Qiao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China.
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China.
| |
Collapse
|
23
|
Guo B, Zheng Y, Fan Y, Yang Y, Wang Y, Qin L, An Y, Xu X, Zhang X, Sun G, Dou H, Shao C, Gong Y, Jiang B, Hu H. Enhanced Apc Min/+ adenoma formation after epithelial CUL4B deletion by recruitment of myeloid-derived suppressor cells. Neoplasia 2024; 53:101005. [PMID: 38761506 PMCID: PMC11127156 DOI: 10.1016/j.neo.2024.101005] [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: 12/04/2023] [Revised: 04/10/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
Colorectal cancer (CRC) stands as a prevalent malignancy globally. A pivotal event in CRC pathogenesis involves the loss-of-function mutation in the APC gene, leading to the formation of benign polyps. Despite the well-established role of APC, the contribution of CUL4B to CRC initiation in the pre-tumorous stage remains poorly understood. In this investigation, we generated a murine model by crossing ApcMin/+ mice with Cul4bΔIEC mice to achieve specific deletion of Cul4b in the gut epithelium against an ApcMin/+ background. By employing histological methods, RNA-sequencing (RNA-seq), and flow cytometry, we assessed alterations and characterized the immune microenvironment. Our results unveiled that CUL4B deficiency in gut epithelium expedited ApcMin/+ adenoma formation. Notably, CUL4B in adenomas restrained the accumulation of tumor-infiltrating myeloid-derived suppressor cells (MDSCs). In vivo inhibition of MDSCs significantly delayed the growth of CUL4B deleted ApcMin/+ adenomas. Furthermore, the addition of MDSCs to in vitro cultured ApcMin/+; Cul4bΔIEC adenoma organoids mitigated their alterations. Mechanistically, CUL4B directly interacted with the promoter of Csf3, the gene encoding granulocyte-colony stimulating factor (G-CSF) by coordinating with PRC2. Inhibiting CUL4B epigenetically activated the expression of G-CSF, promoting the recruitment of MDSCs. These findings offer novel insights into the tumor suppressor-like roles of CUL4B in regulating ApcMin/+ adenomas, suggesting a potential therapeutic strategy for CRC initiation and progression in the context of activated Wnt signaling.
Collapse
Affiliation(s)
- Beibei Guo
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yawen Zheng
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China; Department of Obstetrics & Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Yujia Fan
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yang Yang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yuxing Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Liping Qin
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yachun An
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiaoran Xu
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiyu Zhang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Gongping Sun
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Histoembryology, Shandong University Cheeloo Medical College, Shandong University School of Medicine, Jinan, China
| | - Hao Dou
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Yaoqin Gong
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Baichun Jiang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China.
| | - Huili Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, China.
| |
Collapse
|
24
|
Colon T, Kou Z, Choi BH, Tran F, Zheng E, Dai W. Enzyme-independent role of EZH2 in regulating cell cycle progression via the SKP2-KIP/CIP pathway. Sci Rep 2024; 14:13389. [PMID: 38862595 PMCID: PMC11166936 DOI: 10.1038/s41598-024-64338-4] [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: 09/19/2023] [Accepted: 06/07/2024] [Indexed: 06/13/2024] Open
Abstract
While EZH2 enzymatic activity is well-known, emerging evidence suggests that EZH2 can exert functions in a methyltransferase-independent manner. In this study, we have uncovered a novel mechanism by which EZH2 positively regulates the expression of SKP2, a critical protein involved in cell cycle progression. We demonstrate that depletion of EZH2 significantly reduces SKP2 protein levels in several cell types, while treatment with EPZ-6438, an EZH2 enzymatic inhibitor, has no effect on SKP2 protein levels. Consistently, EZH2 depletion leads to cell cycle arrest, accompanied by elevated expression of CIP/KIP family proteins, including p21, p27, and p57, whereas EPZ-6438 treatment does not modulate their levels. We also provide evidence that EZH2 knockdown, but not enzymatic inhibition, suppresses SKP2 mRNA expression, underscoring the transcriptional regulation of SKP2 by EZH2 in a methyltransferase-independent manner. Supporting this, analysis of the Cancer Genome Atlas database reveals a close association between EZH2 and SKP2 expression in human malignancies. Moreover, EZH2 depletion but not enzymatic inhibition positively regulates the expression of major epithelial-mesenchymal transition (EMT) regulators, such as ZEB1 and SNAIL1, in transformed cells. Our findings shed light on a novel mechanism by which EZH2 exerts regulatory effects on cell proliferation and differentiation through its methyltransferase-independent function, specifically by modulating SKP2 expression.
Collapse
Affiliation(s)
- Tania Colon
- Division of Environmental Medicine, Department of Medicine Grossman School of Medicine, New York University, 341 East 25th Street, New York, NY, 10010, USA
| | - Ziyue Kou
- Division of Environmental Medicine, Department of Medicine Grossman School of Medicine, New York University, 341 East 25th Street, New York, NY, 10010, USA
| | - Byeong Hyeok Choi
- Division of Environmental Medicine, Department of Medicine Grossman School of Medicine, New York University, 341 East 25th Street, New York, NY, 10010, USA
| | - Franklin Tran
- Division of Environmental Medicine, Department of Medicine Grossman School of Medicine, New York University, 341 East 25th Street, New York, NY, 10010, USA
| | - Edwin Zheng
- Division of Environmental Medicine, Department of Medicine Grossman School of Medicine, New York University, 341 East 25th Street, New York, NY, 10010, USA
| | - Wei Dai
- Division of Environmental Medicine, Department of Medicine Grossman School of Medicine, New York University, 341 East 25th Street, New York, NY, 10010, USA.
| |
Collapse
|
25
|
Chen Y, Zhu H, Luo Y, Tong S, Liu Y. EZH2: The roles in targeted therapy and mechanisms of resistance in breast cancer. Biomed Pharmacother 2024; 175:116624. [PMID: 38670045 DOI: 10.1016/j.biopha.2024.116624] [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: 01/28/2024] [Revised: 04/04/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Drug resistance presents a formidable challenge in the realm of breast cancer therapy. Accumulating evidence suggests that enhancer of zeste homolog 2 (EZH2), a component of the polycomb repressive complex 2 (PRC2), may serve as a key regulator in controlling drug resistance. EZH2 overexpression has been observed in breast cancer and many other malignancies, showing a strong correlation with poor outcomes. This review aims to summarize the mechanisms by which EZH2 regulates drug resistance, with a specific focus on breast cancer, in order to provide a comprehensive understanding of the underlying molecular processes. Additionally, we will discuss the current strategies and outcomes of targeting EZH2 using both single agents and combination therapies, with the goal of offering improved guidance for the clinical treatment of breast cancer patients who have developed drug resistance.
Collapse
Affiliation(s)
- Yun Chen
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Hongyan Zhu
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Yi Luo
- Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Biotheus Inc., Guangdong Province, Zhuhai 519080, PR China.
| | - Shuangmei Tong
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| |
Collapse
|
26
|
Feng M, Chai C, Hao X, Lai X, Luo Y, Zhang H, Tang W, Gao N, Pan G, Liu X, Wang Y, Xiong W, Wu Q, Wang J. Inherited KDM6A A649T facilitates tumor-immune escape and exacerbates colorectal signet-ring cell carcinoma outcomes. Oncogene 2024; 43:1757-1768. [PMID: 38622203 DOI: 10.1038/s41388-024-03029-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: 07/09/2023] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/17/2024]
Abstract
Childhood onset of colorectal signet-ring cell carcinoma (CR-SRCC) is extremely rare and featured as highly malignant with poor prognosis. Here we reported a CR-SRCC case of 11-year-old boy with a novel inherited X-linked KDM6AA694T mutation. The H3K27me3 demethylase KDM6A was frequently mutated in varieties of tumors and acts as a tumor suppressor. In vivo H3K27me3 demethylation assay demonstrated that KDM6AA694T had dampened H3K27me3 demethylase activity. Overexpression of KDM6AA694T in SRCC cell line KATO3 promoted cell proliferation, invasion and migration, which were further confirmed in vivo by constructing orthotopic tumor growth and lung metastasis model. Besides, expression of KDM6AA694T in immune cells suppresses inflammatory macrophage response and effector T cell response. In conclusion, we characterized a novel inherited KDM6AA694T mutant from a childhood-onset SRCC case and demonstrated that the mutant with impaired H3K27me3 demethylase activity could potentiate tumor malignancy and suppress antitumor immunity.
Collapse
Affiliation(s)
- Maoxiao Feng
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Chengwei Chai
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
- Department of Pediatric General Surgery, Guangdong Women and Children Hospital, Guangzhou, 511442, China.
| | - Xiaodong Hao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 253000, China
| | - Xiaojiang Lai
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yuanyuan Luo
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Hong Zhang
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wenzhu Tang
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Ningxin Gao
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Guihong Pan
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiaojie Liu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Wenjing Xiong
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Qiang Wu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Jun Wang
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China.
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
27
|
Xie H, Deng H, Yang X, Gao X, Yang S, Chen W, Wang Y, Yang N, Yong L, Hou X. Mesencephalic Astrocyte-derived Neurotrophic Factor Supports Hepatitis B Virus-induced Immunotolerance. Cell Mol Gastroenterol Hepatol 2024; 18:101360. [PMID: 38759839 PMCID: PMC11255368 DOI: 10.1016/j.jcmgh.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND & AIMS The immune tolerance induced by hepatitis B virus (HBV) is a major challenge for achieving effective viral clearance, and the mechanisms involved are not well-understood. One potential factor involved in modulating immune responses is mesencephalic astrocyte-derived neurotrophic factor (MANF), which has been reported to be increased in patients with chronic hepatitis B. In this study, our objective is to examine the role of MANF in regulating immune responses to HBV. METHODS We utilized a commonly used HBV-harboring mouse model, where mice were hydrodynamically injected with the pAAV/HBV1.2 plasmid. We assessed the HBV load by measuring the levels of various markers including hepatitis B surface antigen, hepatitis B envelope antigen, hepatitis B core antigen, HBV DNA, and HBV RNA. RESULTS Our study revealed that following HBV infection, both myeloid cells and hepatocytes exhibited increased expression of MANF. Moreover, we observed that mice with myeloid-specific MANF knockout (ManfMye-/-) displayed reduced HBV load and improved HBV-specific T cell responses. The decreased HBV-induced tolerance in ManfMye-/- mice was associated with reduced accumulation of myeloid-derived suppressor cells (MDSCs) in the liver. Restoring MDSC levels in ManfMye-/- mice through MDSC adoptive transfer reinstated HBV-induced tolerance. Mechanistically, we found that MANF promoted MDSC expansion by activating the IL-6/STAT3 pathway. Importantly, our study demonstrated the effectiveness of a combination therapy involving an hepatitis B surface antigen vaccine and nanoparticle-encapsulated MANF siRNA in effectively clearing HBV in HBV-carrier mice. CONCLUSION The current study reveals that MANF plays a previously unrecognized regulatory role in liver tolerance by expanding MDSCs in the liver through IL-6/STAT3 signaling, leading to MDSC-mediated CD8+ T cell exhaustion.
Collapse
Affiliation(s)
- Huiyuan Xie
- Department of Laboratory Medicine, the First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, P. R. China
| | - Haiyan Deng
- Health Science Center, Ningbo University, Ningbo, Zhejiang, P. R. China
| | - Xiaoping Yang
- Department of Hepatopancreatobiliary Surgery, the First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, P. R. China
| | - Xianxian Gao
- Health Science Center, Ningbo University, Ningbo, Zhejiang, P. R. China
| | - Shanru Yang
- Health Science Center, Ningbo University, Ningbo, Zhejiang, P. R. China
| | - Weiyi Chen
- Health Science Center, Ningbo University, Ningbo, Zhejiang, P. R. China
| | - Yixuan Wang
- Health Science Center, Ningbo University, Ningbo, Zhejiang, P. R. China
| | - Naibin Yang
- Department of Infection, the First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, P. R. China
| | - Liang Yong
- Laboratory of Stem Cell, the First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, P. R. China
| | - Xin Hou
- Health Science Center, Ningbo University, Ningbo, Zhejiang, P. R. China.
| |
Collapse
|
28
|
Fischetti I, Botti L, Sulsenti R, Cancila V, Enriquez C, Ferri R, Bregni M, Crivelli F, Tripodo C, Colombo MP, Jachetti E. Combined therapy targeting AR and EZH2 curbs castration-resistant prostate cancer enhancing anti-tumor T-cell response. Epigenomics 2024; 16:653-670. [PMID: 38530086 PMCID: PMC11160446 DOI: 10.2217/epi-2023-0374] [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: 10/25/2023] [Accepted: 03/07/2024] [Indexed: 03/27/2024] Open
Abstract
Aim: Castration-resistant prostate cancer (CRPC) eventually becomes resistant to androgen receptor pathway inhibitors like enzalutamide. Immunotherapy also fails in CRPC. We propose a new approach to simultaneously revert enzalutamide resistance and rewire anti-tumor immunity. Methods: We investigated in vitro and in subcutaneous and spontaneous mouse models the effects of combining enzalutamide and GSK-126, a drug inhibiting the epigenetic modulator EZH2. Results: Enzalutamide and GSK-126 synergized to reduce CRPC growth, also restraining tumor neuroendocrine differentiation. The anti-tumor activity was lost in immunodeficient mice. Indeed, the combination treatment awoke cytotoxic activity and IFN-γ production of tumor-specific CD8+ T lymphocytes. Conclusion: These results promote the combination of enzalutamide and GSK-126 in CRPC, also offering new avenues for immunotherapy in prostate cancer.
Collapse
Affiliation(s)
- Irene Fischetti
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Laura Botti
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Roberta Sulsenti
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Italy
| | - Claudia Enriquez
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Renata Ferri
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | | | | | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Italy
| | - Mario P. Colombo
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Elena Jachetti
- Molecular Immunology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| |
Collapse
|
29
|
Ibarrola J, Xiang RR, Sun Z, Lu Q, Hill MA, Jaffe IZ. Inhibition of the histone methyltransferase EZH2 induces vascular stiffness. Clin Sci (Lond) 2024; 138:251-268. [PMID: 38362910 PMCID: PMC11948989 DOI: 10.1042/cs20231478] [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: 11/15/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/17/2024]
Abstract
Vascular stiffness increases with aging, obesity and hypertension and predicts cardiovascular risk. The levels of histone H3-lysine-27 methylation (H3K27me) and the histone methyltransferase EZH2 both decrease in aging vessels, driving vascular stiffness. The impact of EZH2 inhibitors on vascular stiffness is unknown. We tested the hypothesis that the EZH2 inhibitor GSK126, currently in development for cancer treatment, increases vascular stiffness and explored underlying molecular mechanisms. Young (3 month) and middle-aged (12 month) male mice were treated with GSK126 for 1-2 months and primary human aortic smooth muscle cells (HASMCs) from young male and female donors were treated with GSK126 for 24-48 h. Stiffness was measured in vivo by pulse wave velocity and in vitro by atomic force microscopy (AFM) and vascular structure was quantified histologically. Extracellular matrix proteins were studied by qRT-PCR, immunoblotting, zymography and chromatin immunoprecipitation. GSK126 treatment decreased H3K27 methylation (H3K27me) and increased acetylation (H3K27ac) in mouse vessels and in HASMCs. In GSK126-treated mice, aortic stiffness increased without changes in vascular fibrosis. EZH2 inhibition enhanced elastin fiber degradation and matrix metalloprotease-2 (MMP2) expression. In HASMCs, GSK126 treatment increased synthetic phenotype markers and intrinsic HASMCs stiffness by AFM with altered cytoskeletal structure and increased nuclear actin staining. GSK126 also increased MMP2 protein expression, activity and enrichment of H3K27ac at the MMP2 promoter in HASMCs. GSK126 causes vascular stiffening, inducing MMP2 activity, elastin degradation, and modulation of SMC phenotype and cytoskeletal stiffness. These findings suggest that EZH2 inhibitors used to treat cancer could negatively impact the vasculature by enhancing stiffness and merits examination in human trials.
Collapse
Affiliation(s)
- Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Rachel R. Xiang
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Zhe Sun
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65203, USA
| | - Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Michael A. Hill
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65203, USA
| | - Iris Z. Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| |
Collapse
|
30
|
Lagarde CB, Kavalakatt J, Benz MC, Hawes ML, Arbogast CA, Cullen NM, McConnell EC, Rinderle C, Hebert KL, Khosla M, Belgodere JA, Hoang VT, Collins-Burow BM, Bunnell BA, Burow ME, Alahari SK. Obesity-associated epigenetic alterations and the obesity-breast cancer axis. Oncogene 2024; 43:763-775. [PMID: 38310162 DOI: 10.1038/s41388-024-02954-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024]
Abstract
Both breast cancer and obesity can regulate epigenetic changes or be regulated by epigenetic changes. Due to the well-established link between obesity and an increased risk of developing breast cancer, understanding how obesity-mediated epigenetic changes affect breast cancer pathogenesis is critical. Researchers have described how obesity and breast cancer modulate the epigenome individually and synergistically. In this review, the epigenetic alterations that occur in obesity, including DNA methylation, histone, and chromatin modification, accelerated epigenetic age, carcinogenesis, metastasis, and tumor microenvironment modulation, are discussed. Delineating the relationship between obesity and epigenetic regulation is vital to furthering our understanding of breast cancer pathogenesis.
Collapse
Affiliation(s)
- Courtney B Lagarde
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Joachim Kavalakatt
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Megan C Benz
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Mackenzie L Hawes
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Carter A Arbogast
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Nicole M Cullen
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Emily C McConnell
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Caroline Rinderle
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Katherine L Hebert
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Maninder Khosla
- Department of Biochemistry and Molecular Biology, LSU Health Science Center School of Medicine, New Orleans, LA, 70112, USA
| | - Jorge A Belgodere
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- Department of Biological and Agricultural Engineering, Louisiana State University and Agricultural Center, Baton Rouge, LA, 70803, USA
| | - Van T Hoang
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Bridgette M Collins-Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Bruce A Bunnell
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Matthew E Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, LSU Health Science Center School of Medicine, New Orleans, LA, 70112, USA.
- Stanley S. Scott Cancer Center, LSU Health Science Center School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
31
|
Li T, Han B, Wang L, Sun L, Cai Y, Yu M, Xiao W, Yang H. Activation of mucosal insulin receptor exacerbates intestinal inflammation by promoting tissue resident memory T cells differentiation through EZH2. J Transl Med 2024; 22:78. [PMID: 38243324 PMCID: PMC10797971 DOI: 10.1186/s12967-023-04789-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: 08/06/2023] [Accepted: 12/09/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Inflammatory Bowel Diseases (IBD), an autoimmune disease characterised by abnormal intestinal immunity, are related to vital morbidity around the world. However, therapeutic agents for IBD have not achieved desired benefit. Exploring new therapeutic targets for IBD, especially based on its abnormally intestinal immunity, could alleviate the flare-up and worsening of IBD. Tissue resident memory T cells (TRM) are core of multiple autoimmune diseases, including IBD. However, the mechanism of TRM differentiation remains to be investigated. METHODS The alterations in mRNA and lncRNA profile of intestinal intraepithelial lymphocytes (IELs), the largest component of intestinal TRM, were analyzed in DSS-induced chronic colitis. Based on it, we examined the function of rectal insulin instillation in a dextran sodium sulfate (DSS) induced chronic colitis. Furthermore, we investigated the downstream-target of the insulin pathway-EZH2 and the crucial role of EZH2 in intestinal tissue resident memory T cell differentiation by utilizing EZH2fl/flCD4cre mice. RESULTS Insulin receptor (INSR) expression was found to be significantly reduced. Activation of mucosal insulin pathway by rectal insulin instillation exacerbated colitis by disrupting IELs subgroups and up-regulating TNF-ɑ and IL-17 expression. Rectal insulin instillation promoted EZH2 expression and EZH2 inhibition alleviated chronic colitis. EZH2fl/flCD4cre mice restored the normal IEL subgroups and suppressed TNF-ɑ and IL-17 expression, exhibiting alleviated colitis. IELs from EZH2fl/flCD4cre mice exhibit significant changes in TRM related phenotype. CD4+TRM was significantly increased in chronic colitis and decreased in EZH2fl/flCD4cre mice. CONCLUSION Insulin receptor of intestinal mucosal T-cells could promote intestinal TRM differentiation via EZH2. Our discoveries suggest that therapies targeting colonic INSR and EZH2 could be potential treatment for IBD based on its regulatory effects on TRM. Insulin receptor inhibitors rather than insulin should be applied during colitis-active phase. In addition, EZH2 shows to be a downstream signal of the insulin pathway and EZH2 inhibitor could alleviating intestinal inflammation. However, the critical role of EZH2 in TRM differentiation restricts the anti-tumor effects of EZH2 inhibitor in vivo.
Collapse
Affiliation(s)
- Teming Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
- Department of General Surgery, Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Army Medical University, Shigatse, 857000, China
| | - Ben Han
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Liucan Wang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Lihua Sun
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yujiao Cai
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Min Yu
- Department of General Surgery, Chongqing General Hospital, Chongqing, 401147, China.
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
- Department of General Surgery, Chongqing General Hospital, Chongqing, 401147, China.
| |
Collapse
|
32
|
Yu X, Zhao H, Wang R, Chen Y, Ouyang X, Li W, Sun Y, Peng A. Cancer epigenetics: from laboratory studies and clinical trials to precision medicine. Cell Death Discov 2024; 10:28. [PMID: 38225241 PMCID: PMC10789753 DOI: 10.1038/s41420-024-01803-z] [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: 10/13/2023] [Revised: 12/23/2023] [Accepted: 01/04/2024] [Indexed: 01/17/2024] Open
Abstract
Epigenetic dysregulation is a common feature of a myriad of human diseases, particularly cancer. Defining the epigenetic defects associated with malignant tumors has become a focus of cancer research resulting in the gradual elucidation of cancer cell epigenetic regulation. In fact, most stages of tumor progression, including tumorigenesis, promotion, progression, and recurrence are accompanied by epigenetic alterations, some of which can be reversed by epigenetic drugs. The main objective of epigenetic therapy in the era of personalized precision medicine is to detect cancer biomarkers to improve risk assessment, diagnosis, and targeted treatment interventions. Rapid technological advancements streamlining the characterization of molecular epigenetic changes associated with cancers have propelled epigenetic drug research and development. This review summarizes the main mechanisms of epigenetic dysregulation and discusses past and present examples of epigenetic inhibitors in cancer diagnosis and treatment, with an emphasis on the development of epigenetic enzyme inhibitors or drugs. In the final part, the prospect of precise diagnosis and treatment is considered based on a better understanding of epigenetic abnormalities in cancer.
Collapse
Affiliation(s)
- Xinyang Yu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, (Zhuhai People's Hospital Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Hao Zhao
- Department of Spinal Surgery, Yichang Central People's Hospital Affiliated with China Three Gorges University, Yichang, Hubei, 443000, China
| | - Ruiqi Wang
- Department of Pharmacy, Zhuhai People's Hospital, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, Guangdong, 519000, China
| | - Yingyin Chen
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, (Zhuhai People's Hospital Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Xumei Ouyang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, (Zhuhai People's Hospital Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Wenting Li
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, (Zhuhai People's Hospital Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Yihao Sun
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, (Zhuhai People's Hospital Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China.
| | - Anghui Peng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, (Zhuhai People's Hospital Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China.
| |
Collapse
|
33
|
Hashemi M, Nazdari N, Gholamiyan G, Paskeh MDA, Jafari AM, Nemati F, Khodaei E, Abyari G, Behdadfar N, Raei B, Raesi R, Nabavi N, Hu P, Rashidi M, Taheriazam A, Entezari M. EZH2 as a potential therapeutic target for gastrointestinal cancers. Pathol Res Pract 2024; 253:154988. [PMID: 38118215 DOI: 10.1016/j.prp.2023.154988] [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: 08/15/2023] [Revised: 11/18/2023] [Accepted: 11/27/2023] [Indexed: 12/22/2023]
Abstract
Gastrointestinal (GI) cancers continue to be a major cause of mortality and morbidity globally. Understanding the molecular pathways associated with cancer progression and severity is essential for creating effective cancer treatments. In cancer research, there is a notable emphasis on Enhancer of zeste homolog 2 (EZH2), a key player in gene expression influenced by its irregular expression and capacity to attach to promoters and alter methylation status. This review explores the impact of EZH2 signaling on various GI cancers, such as colorectal, gastric, pancreatic, hepatocellular, esophageal, and cholangiocarcinoma. The primary function of EZH2 signaling is to facilitate the accelerated progression of cancer cells. Additionally, EZH2 has the capacity to modulate the reaction of GI cancers to chemotherapy and radiotherapy. Numerous pathways, including long non-coding RNAs and microRNAs, serve as upstream regulators of EZH2 in these types of cancer. EZH2's enzymatic activity enables it to attach to target gene promoters, resulting in methylation that modifies their expression. EZH2 could be considered as an independent prognostic factor, with increased expression correlating with a worse disease prognosis. Additionally, a range of gene therapies including small interfering RNA, and anti-tumor agents are being explored to target EZH2 for cancer treatment. This comprehensive review underscores the current insights into EZH2 signaling in gastrointestinal cancers and examines the prospect of therapies targeting EZH2 to enhance patient outcomes.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Naghmeh Nazdari
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ghazaleh Gholamiyan
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahshid Deldar Abad Paskeh
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Moghadas Jafari
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fateme Nemati
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Khodaei
- Department of Dermatology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghazal Abyari
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nazanin Behdadfar
- Young Researchers and Elite Club, Buinzahra Branch, Islamic Azad University, Buinzahra, Iran
| | - Behnaz Raei
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Rasoul Raesi
- Department of Health Services Management, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical-Surgical Nursing, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6 Vancouver, BC, Canada
| | - Peng Hu
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
34
|
Zhou Y, Xiao Y, Liu H, Chen Q, Zhu L, Zeng L, Liu X, Pan Y, Zhang J, Fu J, Shao C. Elevation of H3K27me3 level contributes to the radioresistance of nasopharyngeal carcinoma by inhibiting OAS1 expression. Am J Physiol Cell Physiol 2024; 326:C60-C73. [PMID: 38009194 DOI: 10.1152/ajpcell.00358.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 11/28/2023]
Abstract
Radiotherapy has long been a main treatment option for nasopharyngeal carcinoma (NPC). However, during clinical treatment, NPC is prone to developing radioresistance, resulting in treatment failure. This study aims to examine the role of histone methylation in the induction of radioresistance. It was found that the radioresistance of NPC cells was related to the increase of the level of histone H3 lysine 27 trimethylation (H3K27me3). Treatment of cells with histone methyltransferase inhibitor GSK126 increased the radiosensitivity of NPC cells by triggering Bcl2 apoptosis regulator/BCL2-associated X, apoptosis regulator (Bcl2/BAX) signaling pathway. Bioinformatics analysis indicated that the expression of 2'-5'-oligoadenylate synthetase 1 (OAS1) was reduced in the radioresistant cells but increased in the GSK126-treated cells. Chromatin immunoprecipitation assay confirmed that the decrease of OAS1 expression in radioresistant cells was mainly due to the enrichment of H3K27me3 in its promoter region. Furthermore, downregulation of OAS1 reduced apoptosis due to the inhibition of Bcl2/BAX pathway after irradiation, while OAS1 overexpression increased radiosensitivity. Our findings revealed for the first time that the increase of H3K27me3 level was associated with the decrease of OAS1 expression, leading to the inhibition of apoptosis and ultimately contributing to the radioresistance of NPC cells. Moreover, the histone methyltransferase inhibitor GSK126 could overcome the radioresistance and thus might be a potential therapeutic strategy for NPC.NEW & NOTEWORTHY Our findings revealed for the first time that the increase of H3K27me3 level was associated with the decrease of OAS1 expression, leading to the inhibition of apoptosis and ultimately contributing to the radioresistance of NPC cells. Moreover, we demonstrated that the histone methyltransferase inhibitor GSK126 could be a promising therapeutic strategy for NPC by overcoming radioresistance, providing valuable insights into the clinical treatment of NPC.
Collapse
Affiliation(s)
- Yuchuan Zhou
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yuqi Xiao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Hongxia Liu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Qianping Chen
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Lin Zhu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Liang Zeng
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Xinglong Liu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yan Pan
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Jianghong Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Jiamei Fu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
35
|
Chen Y, Xu J, Liu X, Guo L, Yi P, Cheng C. Potential therapies targeting nuclear metabolic regulation in cancer. MedComm (Beijing) 2023; 4:e421. [PMID: 38034101 PMCID: PMC10685089 DOI: 10.1002/mco2.421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/28/2023] [Accepted: 10/12/2023] [Indexed: 12/02/2023] Open
Abstract
The interplay between genetic alterations and metabolic dysregulation is increasingly recognized as a pivotal axis in cancer pathogenesis. Both elements are mutually reinforcing, thereby expediting the ontogeny and progression of malignant neoplasms. Intriguingly, recent findings have highlighted the translocation of metabolites and metabolic enzymes from the cytoplasm into the nuclear compartment, where they appear to be intimately associated with tumor cell proliferation. Despite these advancements, significant gaps persist in our understanding of their specific roles within the nuclear milieu, their modulatory effects on gene transcription and cellular proliferation, and the intricacies of their coordination with the genomic landscape. In this comprehensive review, we endeavor to elucidate the regulatory landscape of metabolic signaling within the nuclear domain, namely nuclear metabolic signaling involving metabolites and metabolic enzymes. We explore the roles and molecular mechanisms through which metabolic flux and enzymatic activity impact critical nuclear processes, including epigenetic modulation, DNA damage repair, and gene expression regulation. In conclusion, we underscore the paramount significance of nuclear metabolic signaling in cancer biology and enumerate potential therapeutic targets, associated pharmacological interventions, and implications for clinical applications. Importantly, these emergent findings not only augment our conceptual understanding of tumoral metabolism but also herald the potential for innovative therapeutic paradigms targeting the metabolism-genome transcriptional axis.
Collapse
Affiliation(s)
- Yanjie Chen
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Jie Xu
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaoyi Liu
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Linlin Guo
- Department of Microbiology and ImmunologyThe Indiana University School of MedicineIndianapolisIndianaUSA
| | - Ping Yi
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Chunming Cheng
- Department of Radiation OncologyJames Comprehensive Cancer Center and College of Medicine at The Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
36
|
Gao M, Li Y, Cao P, Liu H, Chen J, Kang S. Exploring the therapeutic potential of targeting polycomb repressive complex 2 in lung cancer. Front Oncol 2023; 13:1216289. [PMID: 37909018 PMCID: PMC10613995 DOI: 10.3389/fonc.2023.1216289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023] Open
Abstract
The pathogenesis of lung cancer (LC) is a multifaceted process that is influenced by a variety of factors. Alongside genetic mutations and environmental influences, there is increasing evidence that epigenetic mechanisms play a significant role in the development and progression of LC. The Polycomb repressive complex 2 (PRC2), composed of EZH1/2, SUZ12, and EED, is an epigenetic silencer that controls the expression of target genes and is crucial for cell identity in multicellular organisms. Abnormal expression of PRC2 has been shown to contribute to the progression of LC through several pathways. Although targeted inhibition of EZH2 has demonstrated potential in delaying the progression of LC and improving chemotherapy sensitivity, the effectiveness of enzymatic inhibitors of PRC2 in LC is limited, and a more comprehensive understanding of PRC2's role is necessary. This paper reviews the core subunits of PRC2 and their interactions, and outlines the mechanisms of aberrant PRC2 expression in cancer and its role in tumor immunity. We also summarize the important role of PRC2 in regulating biological behaviors such as epithelial mesenchymal transition, invasive metastasis, apoptosis, cell cycle regulation, autophagy, and PRC2-mediated resistance to LC chemotherapeutic agents in LC cells. Lastly, we explored the latest breakthroughs in the research and evaluation of medications that target PRC2, as well as the latest findings from clinical studies investigating the efficacy of these drugs in the treatment of various human cancers.
Collapse
Affiliation(s)
- Min Gao
- Department of Thoracic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Medical University, First Clinical Medical College, Hohhot, China
| | - Yongwen Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Peijun Cao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongyu Liu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Chen
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Shirong Kang
- Department of Thoracic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
37
|
Hegde M, Kumar A, Girisa S, Alqahtani MS, Abbas M, Goel A, Hui KM, Sethi G, Kunnumakkara AB. Exosomal noncoding RNA-mediated spatiotemporal regulation of lipid metabolism: Implications in immune evasion and chronic inflammation. Cytokine Growth Factor Rev 2023; 73:114-134. [PMID: 37419767 DOI: 10.1016/j.cytogfr.2023.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/06/2023] [Accepted: 06/06/2023] [Indexed: 07/09/2023]
Abstract
The hallmark of chronic inflammatory diseases is immune evasion. Successful immune evasion involves numerous mechanisms to suppress both adaptive and innate immune responses. Either direct contact between cells or paracrine signaling triggers these responses. Exosomes are critical drivers of these interactions and exhibit both immunogenic and immune evasion properties during the development and progression of various chronic inflammatory diseases. Exosomes carry diverse molecular cargo, including lipids, proteins, and RNAs that are crucial for immunomodulation. Moreover, recent studies have revealed that exosomes and their cargo-loaded molecules are extensively involved in lipid remodeling and metabolism during immune surveillance and disease. Many studies have also shown the involvement of lipids in controlling immune cell activities and their crucial upstream functions in regulating inflammasome activation, suggesting that any perturbation in lipid metabolism results in abnormal immune responses. Strikingly, the expanded immunometabolic reprogramming capacities of exosomes and their contents provided insights into the novel mechanisms behind the prophylaxis of inflammatory diseases. By summarizing the tremendous therapeutic potential of exosomes, this review emphasizes the role of exosome-derived noncoding RNAs in regulating immune responses through the modulation of lipid metabolism and their promising therapeutic applications.
Collapse
Affiliation(s)
- Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia; Computers and communications Department College of Engineering Delta University for Science and Technology, Gamasa 35712, Egypt
| | - Akul Goel
- California Institute of Technology (CalTech), Pasadena, CA, USA
| | - Kam Man Hui
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore 169610, Singapore
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
38
|
Li X, Yuan P, Yang H, Zong X, Yang C, Chen X, Li Y, Yan X, Wen Y, Zhu T, Zhang Q, Xue W, Dai J. Virus-Like Nanotherapeutic for Spatiotemporally Enhancing Antigen Presentation and Cross-Presentation toward Potential Personalized Immunotherapy. Adv Healthc Mater 2023; 12:e2300921. [PMID: 37531246 DOI: 10.1002/adhm.202300921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/13/2023] [Indexed: 08/04/2023]
Abstract
One of the major causes of immunotherapy resistance is the loss of major histocompatibility complex class I (MHC-I) molecules in tumor cells or the downregulation of the class I antigen presentation pathway. In this study, a novel virus-like nanotherapeutic (siRNA@HCM) is developed via encapsulating nanosized siRNA nanoparticles in a hybrid membrane comprising a personalized tumor cell membrane and a universal 293T membrane expressing the mutant vesicular stomatitis virus glycoprotein (mVSV-G). Upon intravenous administration, siRNA@HCM accumulates at the tumor site and provides two potent driving forces for antitumor immunity. First, mVSV-G induces the fusion of siRNA@HCM with tumor cell membranes and directly injects siRNAs into the cytoplasm, significantly improving tumor intrinsic MHC-I antigen presentation. Moreover, mVSV-G can promote the maturation of dendritic cells, thereby achieving highly efficient antigen cross-presentation. The results demonstrate that spatiotemporally enhancing tumor intrinsic antigen presentation and cross-presentation via siRNA@HCM can achieve satisfactory antitumor efficacy and excellent biocompatibility. Immune infiltration analysis shows that siRNA@HCM treatment turns cold tumors into hot tumors. In addition, it significantly promotes the therapeutic effect of programmed death-1 inhibitor. In summary, virus-like nanotherapeutics present a promising approach to enhance the antitumor immune response, with distinct advantages for potential personalized therapy and clinical applications.
Collapse
Affiliation(s)
- Xiaodi Li
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Pengfei Yuan
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Haiyuan Yang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Xiaoqing Zong
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Caiqi Yang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Xinjie Chen
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Yuchao Li
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Xiaodie Yan
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Yaoqi Wen
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Tianci Zhu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Qian Zhang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Jian Dai
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
39
|
Liu X, Tian F, Cui J, Gong L, Xiang L, Fan B, Liu S, Zhan J, Zhou Y, Jiang B, Wang M, Sun G, Gong Y, Zou Y. CUL4B functions as a tumor suppressor in KRAS-driven lung tumors by inhibiting the recruitment of myeloid-derived suppressor cells. Oncogene 2023; 42:3113-3126. [PMID: 37653114 DOI: 10.1038/s41388-023-02824-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/02/2023]
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. KRAS mutations are the most common oncogenic alterations found in lung cancer. Unfortunately, treating KRAS-mutant lung adenocarcinoma (ADC) remains a major oncotherapeutic challenge. Here, we used both autochthonous and transplantable KRAS-mutant tumor models to investigate the role of tumor-derived CUL4B in KRAS-driven lung cancers. We showed that knockout or knockdown of CUL4B promotes lung ADC growth and progression in both models. Mechanistically, CUL4B directly binds to the promoter of Cxcl2 and epigenetically represses its transcription. CUL4B deletion increases the expression of CXCL2, which binds to CXCR2 on myeloid-derived suppressor cells (MDSCs) and promotes their migration to the tumor microenvironment. Targeting of MDSCs significantly delayed the growth of CUL4B knockdown KRAS-mutant tumors. Collectively, our study provides mechanistic insights into the novel tumor suppressor-like functions of CUL4B in regulating KRAS-driven lung tumor development.
Collapse
Affiliation(s)
- Xiaochen Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Clinical Laboratory, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fei Tian
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jianfeng Cui
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Li Gong
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lu Xiang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Bowen Fan
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shuangteng Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jiafeng Zhan
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yadi Zhou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Baichun Jiang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Molin Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Gongping Sun
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yaoqin Gong
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
40
|
Lanzi C, Arrighetti N, Pasquali S, Cassinelli G. Targeting EZH2 in SMARCB1-deficient sarcomas: Advances and opportunities to potentiate the efficacy of EZH2 inhibitors. Biochem Pharmacol 2023; 215:115727. [PMID: 37541451 DOI: 10.1016/j.bcp.2023.115727] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 08/06/2023]
Abstract
Soft tissue sarcomas (STSs) are rare mesechymal malignancies characterized by distintive molecular, histological and clinical features. Many STSs are considered as predominatly epigenetic diseases due to underlying chromatin deregulation. Discovery of deregulated functional antagonism between the chromatin remodeling BRG1/BRM-associated (BAFs) and the histone modifying Polycomb repressor complexes (PRCs) has provided novel actionable targets. In epithelioid sarcoma (ES), extracranial, extrarenal malignant rhabdoid tumors (eMRTs) and synovial sarcoma (SS), the total or partial loss of the BAF core subunit SMARCB1, driven by different alterations, is associated with PRC2 deregulation and dependency on its enzymatic subunit, EZH2. In these SMARCB1-deficient STSs, aberrant EZH2 expression and/or activity emerged as a druggable vulnerability. Although preclinical investigation supported EZH2 targeting as a promising therapeutic option, clinical studies demonstrated a variable response to EZH2 inhibitors. Actually, whereas the clinical benefit recorded in ES patients prompted the FDA approval of the EZH2 inhibitor tazemetostat, the modest and sporadic responses observed in eMRT and SS patients highlighted the need to deepen mechanistic as well as pharmacological investigations to improve drug effectiveness. We summarize the current knowledge of different mechanisms driving SMARCB1 deficiency and EZH2 deregulation in ES, eMRT and SS along with preclinical and clinical studies of EZH2-targeting agents. Possible implication of the PRC2- and enzymatic-independent functions of EZH2 and of its homolog, EZH1, in the response to anti-EZH2 agents will be discussed together with combinatorial strategies under investigation to improve the efficacy of EZH2 targeting in these tumors.
Collapse
Affiliation(s)
- Cinzia Lanzi
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Noemi Arrighetti
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Sandro Pasquali
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Giuliana Cassinelli
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy.
| |
Collapse
|
41
|
Urbanova M, Cihova M, Buocikova V, Slopovsky J, Dubovan P, Pindak D, Tomas M, García-Bermejo L, Rodríguez-Garrote M, Earl J, Kohl Y, Kataki A, Dusinska M, Sainz B, Smolkova B, Gabelova A. Nanomedicine and epigenetics: New alliances to increase the odds in pancreatic cancer survival. Biomed Pharmacother 2023; 165:115179. [PMID: 37481927 DOI: 10.1016/j.biopha.2023.115179] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest cancers worldwide, primarily due to its robust desmoplastic stroma and immunosuppressive tumor microenvironment (TME), which facilitate tumor progression and metastasis. In addition, fibrous tissue leads to sparse vasculature, high interstitial fluid pressure, and hypoxia, thereby hindering effective systemic drug delivery and immune cell infiltration. Thus, remodeling the TME to enhance tumor perfusion, increase drug retention, and reverse immunosuppression has become a key therapeutic strategy. In recent years, targeting epigenetic pathways has emerged as a promising approach to overcome tumor immunosuppression and cancer progression. Moreover, the progress in nanotechnology has provided new opportunities for enhancing the efficacy of conventional and epigenetic drugs. Nano-based drug delivery systems (NDDSs) offer several advantages, including improved drug pharmacokinetics, enhanced tumor penetration, and reduced systemic toxicity. Smart NDDSs enable precise targeting of stromal components and augment the effectiveness of immunotherapy through multiple drug delivery options. This review offers an overview of the latest nano-based approaches developed to achieve superior therapeutic efficacy and overcome drug resistance. We specifically focus on the TME and epigenetic-targeted therapies in the context of PDAC, discussing the advantages and limitations of current strategies while highlighting promising new developments. By emphasizing the immense potential of NDDSs in improving therapeutic outcomes in PDAC, our review paves the way for future research in this rapidly evolving field.
Collapse
Affiliation(s)
- Maria Urbanova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Marina Cihova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Verona Buocikova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Jan Slopovsky
- 2nd Department of Oncology, National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia; Faculty of Medicine, Comenius University, Spitalska 24, 813 72 Bratislava, Slovakia
| | - Peter Dubovan
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia; Department of Surgical Oncology, National CancerInstitute in Bratislava, Klenova 1, 833 10 Bratislava, Slovakia; Faculty of Medicine, Slovak Medical University in Bratislava, Limbová12, 833 03 Bratislava
| | - Daniel Pindak
- Department of Surgical Oncology, National CancerInstitute in Bratislava, Klenova 1, 833 10 Bratislava, Slovakia; Faculty of Medicine, Slovak Medical University in Bratislava, Limbová12, 833 03 Bratislava
| | - Miroslav Tomas
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia; Department of Surgical Oncology, National CancerInstitute in Bratislava, Klenova 1, 833 10 Bratislava, Slovakia; Faculty of Medicine, Slovak Medical University in Bratislava, Limbová12, 833 03 Bratislava
| | - Laura García-Bermejo
- Biomarkers and Therapeutic Targets Group, Area4, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain
| | - Mercedes Rodríguez-Garrote
- Molecular Epidemiology and Predictive Tumor Markers Group, Area 3, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain; CIBERONC, Madrid, Spain
| | - Julie Earl
- Molecular Epidemiology and Predictive Tumor Markers Group, Area 3, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain; CIBERONC, Madrid, Spain
| | - Yvonne Kohl
- Department Bioprocessing & Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, 66280 Sulzbach, Germany
| | - Agapi Kataki
- 1st Department of Propaedeutic Surgery, National and Kapodistrian University of Athens, Vasilissis Sofias 114, 11527 Athens, Greece
| | - Maria Dusinska
- Health Effects Laboratory, Department of Environmental Chemistry, NILU-Norwegian Institute for Air Research, Instituttveien 18, 2002 Kjeller, Norway
| | - Bruno Sainz
- CIBERONC, Madrid, Spain; Instituto de Investigaciones Biomédicas"Alberto Sols" (IIBM), CSIC-UAM, 28029 Madrid, Spain; Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Area 3, Ramón y Cajal Health Research Institute (IRYCIS), 28034 Madrid, Spain
| | - Bozena Smolkova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Alena Gabelova
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 84505 Bratislava, Slovakia..
| |
Collapse
|
42
|
Uceda S, Echeverry-Alzate V, Reiriz-Rojas M, Martínez-Miguel E, Pérez-Curiel A, Gómez-Senent S, Beltrán-Velasco AI. Gut Microbial Metabolome and Dysbiosis in Neurodegenerative Diseases: Psychobiotics and Fecal Microbiota Transplantation as a Therapeutic Approach-A Comprehensive Narrative Review. Int J Mol Sci 2023; 24:13294. [PMID: 37686104 PMCID: PMC10487945 DOI: 10.3390/ijms241713294] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
The comprehensive narrative review conducted in this study delves into the mechanisms of communication and action at the molecular level in the human organism. The review addresses the complex mechanism involved in the microbiota-gut-brain axis as well as the implications of alterations in the microbial composition of patients with neurodegenerative diseases. The pathophysiology of neurodegenerative diseases with neuronal loss or death is analyzed, as well as the mechanisms of action of the main metabolites involved in the bidirectional communication through the microbiota-gut-brain axis. In addition, interventions targeting gut microbiota restructuring through fecal microbiota transplantation and the use of psychobiotics-pre- and pro-biotics-are evaluated as an opportunity to reduce the symptomatology associated with neurodegeneration in these pathologies. This review provides valuable information and facilitates a better understanding of the neurobiological mechanisms to be addressed in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sara Uceda
- BRABE Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Víctor Echeverry-Alzate
- BRABE Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Manuel Reiriz-Rojas
- BRABE Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Esther Martínez-Miguel
- Health Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Ana Pérez-Curiel
- Health Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | - Silvia Gómez-Senent
- Health Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
| | | |
Collapse
|
43
|
Xiao C, Fan T, Zheng Y, Tian H, Deng Z, Liu J, Li C, He J. H3K4 trimethylation regulates cancer immunity: a promising therapeutic target in combination with immunotherapy. J Immunother Cancer 2023; 11:e005693. [PMID: 37553181 PMCID: PMC10414074 DOI: 10.1136/jitc-2022-005693] [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] [Accepted: 05/03/2023] [Indexed: 08/10/2023] Open
Abstract
With the advances in cancer immunity regulation and immunotherapy, the effects of histone modifications on establishing antitumor immunological ability are constantly being uncovered. Developing combination therapies involving epigenetic drugs (epi-drugs) and immune checkpoint blockades or chimeric antigen receptor-T cell therapies are promising to improve the benefits of immunotherapy. Histone H3 lysine 4 trimethylation (H3K4me3) is a pivotal epigenetic modification in cancer immunity regulation, deeply involved in modulating tumor immunogenicity, reshaping tumor immune microenvironment, and regulating immune cell functions. However, how to integrate these theoretical foundations to create novel H3K4 trimethylation-based therapeutic strategies and optimize available therapies remains uncertain. In this review, we delineate the mechanisms by which H3K4me3 and its modifiers regulate antitumor immunity, and explore the therapeutic potential of the H3K4me3-related agents combined with immunotherapies. Understanding the role of H3K4me3 in cancer immunity will be instrumental in developing novel epigenetic therapies and advancing immunotherapy-based combination regimens.
Collapse
Affiliation(s)
- Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingjing Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Pickles OJ, Wanigasooriya K, Ptasinska A, Patel AJ, Robbins HL, Bryer C, Whalley CM, Tee L, Lal N, Pinna CM, Elzefzafy N, Taniere P, Beggs AD, Middleton GM. MHC Class II is Induced by IFNγ and Follows Three Distinct Patterns of Expression in Colorectal Cancer Organoids. CANCER RESEARCH COMMUNICATIONS 2023; 3:1501-1513. [PMID: 37565053 PMCID: PMC10411481 DOI: 10.1158/2767-9764.crc-23-0091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/17/2023] [Accepted: 07/05/2023] [Indexed: 08/12/2023]
Abstract
Tumor-specific MHC class II (tsMHC-II) expression impacts tumor microenvironmental immunity. tsMHC-II positive cancer cells may act as surrogate antigen-presenting cells and targets for CD4+ T cell-mediated lysis. In colorectal cancer, tsMHC-II negativity is common, in cell lines due to CIITA promoter methylation. To clarify mechanisms of tsMHC-II repression in colorectal cancer, we analyzed colorectal cancer organoids which are epigenetically faithful to tissue of origin. 15 primary colorectal cancer organoids were treated with IFNγ ± epigenetic modifiers: flow cytometry was used for tsMHC-II expression. qRT-PCR, total RNA sequencing, nanopore sequencing, bisulfite conversion/pyrosequencing, and Western blotting was used to quantitate CIITA, STAT1, IRF1, and JAK1 expression, mutations and promoter methylation and chromatin immunoprecipitation to quantitate H3K9ac, H3K9Me2, and EZH2 occupancy at CIITA. We define three types of response to IFNγ in colorectal cancer: strong, weak, and noninducibility. Delayed and restricted expression even with prolonged IFNγ exposure was due to IFNγ-mediated EZH2 occupancy at CIITA. tsMHC-II expression was enhanced by EZH2 and histone deacetylase inhibition in the weakly inducible organoids. Noninducibility is seen in three consensus molecular subtype 1 (CMS1) organoids due to JAK1 mutation. No organoid demonstrates CIITA promoter methylation. Providing IFNγ signaling is intact, most colorectal cancer organoids are class II inducible. Upregulation of tsMHC-II through targeted epigenetic therapy is seen in one of fifteen organoids. Our approach can serve as a blueprint for investigating the heterogeneity of specific epigenetic mechanisms of immune suppression across individual patients in other cancers and how these might be targeted to inform the conduct of future trials of epigenetic therapies as immune adjuvants more strategically in cancer. Significance Cancer cell expression of MHC class II significantly impacts tumor microenvironmental immunity. Previous studies investigating mechanisms of repression of IFNγ-inducible class II expression using cell lines demonstrate epigenetic silencing of IFN pathway genes as a frequent immune evasion strategy. Unlike cell lines, patient-derived organoids maintain epigenetic fidelity to tissue of origin. In the first such study, we analyze patterns, dynamics, and epigenetic control of IFNγ-induced class II expression in a series of colorectal cancer organoids.
Collapse
Affiliation(s)
- Oliver J. Pickles
- Institute of Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Edgbaston, Birmingham, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Kasun Wanigasooriya
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Anetta Ptasinska
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Akshay J. Patel
- Institute of Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Edgbaston, Birmingham, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Helen L. Robbins
- Institute of Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Claire Bryer
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Celina M. Whalley
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Louise Tee
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Neeraj Lal
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Claudia M.A. Pinna
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Nahla Elzefzafy
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Edgbaston, Birmingham, United Kingdom
- Cancer Biology Department, NCI, Cairo University, Cairo, Egypt
| | - Philippe Taniere
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Andrew D. Beggs
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Institute of Cancer and Genomic Science, College of Medical and Dental Science, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Gary M. Middleton
- Institute of Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Edgbaston, Birmingham, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
45
|
Cheng X, Wang H, Wang Z, Zhu B, Long H. Tumor-associated myeloid cells in cancer immunotherapy. J Hematol Oncol 2023; 16:71. [PMID: 37415162 PMCID: PMC10324139 DOI: 10.1186/s13045-023-01473-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023] Open
Abstract
Tumor-associated myeloid cells (TAMCs) are among the most important immune cell populations in the tumor microenvironment, and play a significant role on the efficacy of immune checkpoint blockade. Understanding the origin of TAMCs was found to be the essential to determining their functional heterogeneity and, developing cancer immunotherapy strategies. While myeloid-biased differentiation in the bone marrow has been traditionally considered as the primary source of TAMCs, the abnormal differentiation of splenic hematopoietic stem and progenitor cells, erythroid progenitor cells, and B precursor cells in the spleen, as well as embryo-derived TAMCs, have been depicted as important origins of TAMCs. This review article provides an overview of the literature with a focus on the recent research progress evaluating the heterogeneity of TAMCs origins. Moreover, this review summarizes the major therapeutic strategies targeting TAMCs with heterogeneous sources, shedding light on their implications for cancer antitumor immunotherapies.
Collapse
Affiliation(s)
- Xinyu Cheng
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China
| | - Huilan Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China
| | - Zhongyu Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China.
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China.
| |
Collapse
|
46
|
Deng P, Hu H. HSP90-Dependent Upregulation of EZH2 Promotes Hypoxia/Reoxygenation-Induced Pyroptosis by Inhibiting miR-22 in Endothelial Cells. J Inflamm Res 2023; 16:2615-2630. [PMID: 37360624 PMCID: PMC10289174 DOI: 10.2147/jir.s403531] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023] Open
Abstract
Objective Endothelial cell pyroptosis induced by hypoxia/reoxygenation (H/R) plays a key role in the pathogenesis of myocardial infarction (MI). However, the underlying mechanism is not clearly elucidated. Methods Human umbilical vein endothelial cells (HUVECs) exposed to H/R acted as in vitro model to investigate the mechanism of H/R-induced endothelial cell pyroptosis. CCK-8 assays were performed to investigate the viability of HUVECs. Calcein-AM/PI staining was carried out to quantify the death of HUVECs. The expression level of miR-22 was measured by RT-qPCR. The protein expression levels of zeste 2 polycomb repressive complex 2 subunit (EZH2), NLRP3, cleaved caspase-1 (c-caspase-1), GSDMD-N and heat shock protein 90 (HSP90) were measured by Western blot. Levels of IL-1β and IL-18 in culture medium were detected by ELISA. The intracellular localization of EZH2 was detected by immunofluorescence staining. Chromatin immunoprecipitation (ChIP) assay was used to detect the enrichment of EZH2 and H3K27me3 in the miR-22 promoter region. The binding between miR-22 and NLRP3 in HUVECs was confirmed by the dual luciferase assay. Reciprocal coimmunoprecipitation was conducted to detect the direct interaction between HSP90 and EZH2. Results H/R increased EZH2 expression, and the EZH2 siRNA could inhibit H/R-induced pyroptosis in HUVECs. H/R reduced miR-22 expression, which was reversed by EZH2 siRNA. Silencing of miR-22 by its inhibitor reversed EZH2 siRNA-induced pyroptosis inhibition in H/R-exposed HUVECs. Upregulation of miR-22 by its mimic suppressed EZH2 overexpression-enhanced pyroptosis in H/R-exposed HUVECs. ChIP assay confirmed that EZH2 bound to the miR-22 promoter region and repressed miR-22 expression through H3K27me3. Furthermore, luciferase reporter assay indicated that NLRP3 was a direct target of miR- 22 in HUVECs. Finally, HSP90 siRNA inhibited H/R-induced EZH2 expression, miR-22 downregulation, and pyroptosis in HUVECs. Conclusion H/R induces pyroptosis via the HSP90/EZH2/miR-22/NLRP3 signaling axis in endothelial cells.
Collapse
Affiliation(s)
- Paihe Deng
- Clinical Laboratory Medicine Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, People’s Republic of China
| | - Huimin Hu
- Clinical Laboratory Medicine Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, People’s Republic of China
| |
Collapse
|
47
|
Wang B, Liu Y, Liao Z, Wu H, Zhang B, Zhang L. EZH2 in hepatocellular carcinoma: progression, immunity, and potential targeting therapies. Exp Hematol Oncol 2023; 12:52. [PMID: 37268997 PMCID: PMC10236851 DOI: 10.1186/s40164-023-00405-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/15/2023] [Indexed: 06/04/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the leading cause of cancer-related death. The accumulation of genetic and epigenetic changes is closely related to the occurrence and development of HCC. Enhancer of zeste homolog 2 (EZH2, a histone methyltransferase) is suggested to be one of the principal factors that mediates oncogenesis by acting as a driver of epigenetic alternation. Recent studies show that EZH2 is widely involved in proliferation and metastasis of HCC cells. In this review, the functions of EZH2 in HCC progression, the role of EZH2 in tumor immunity and the application of EZH2-related inhibitors in HCC therapy are summarized.
Collapse
Affiliation(s)
- Bohan Wang
- Hepatic Surgery Center, Institute of Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yachong Liu
- Hepatic Surgery Center, Institute of Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhibin Liao
- Hepatic Surgery Center, Institute of Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haofeng Wu
- Hepatic Surgery Center, Institute of Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Institute of Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Lei Zhang
- Hepatic Surgery Center, Institute of Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Hepatobiliary Surgery, Shanxi Tongji Hospital, Tongji Medical College, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Shanxi Medical University, Huazhong University of Science and Technology, Taiyuan, 030032, China.
- Key Laboratory of Hepatobiliary and Pancreatic Diseases of Shanxi Province (Preparatory), Shanxi Tongji Hospital, Tongji Medical College, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Shanxi Medical University, Huazhong University of Science and Technology, Taiyuan, 030032, China.
| |
Collapse
|
48
|
Liu R, Wu J, Guo H, Yao W, Li S, Lu Y, Jia Y, Liang X, Tang J, Zhang H. Post-translational modifications of histones: Mechanisms, biological functions, and therapeutic targets. MedComm (Beijing) 2023; 4:e292. [PMID: 37220590 PMCID: PMC10200003 DOI: 10.1002/mco2.292] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023] Open
Abstract
Histones are DNA-binding basic proteins found in chromosomes. After the histone translation, its amino tail undergoes various modifications, such as methylation, acetylation, phosphorylation, ubiquitination, malonylation, propionylation, butyrylation, crotonylation, and lactylation, which together constitute the "histone code." The relationship between their combination and biological function can be used as an important epigenetic marker. Methylation and demethylation of the same histone residue, acetylation and deacetylation, phosphorylation and dephosphorylation, and even methylation and acetylation between different histone residues cooperate or antagonize with each other, forming a complex network. Histone-modifying enzymes, which cause numerous histone codes, have become a hot topic in the research on cancer therapeutic targets. Therefore, a thorough understanding of the role of histone post-translational modifications (PTMs) in cell life activities is very important for preventing and treating human diseases. In this review, several most thoroughly studied and newly discovered histone PTMs are introduced. Furthermore, we focus on the histone-modifying enzymes with carcinogenic potential, their abnormal modification sites in various tumors, and multiple essential molecular regulation mechanism. Finally, we summarize the missing areas of the current research and point out the direction of future research. We hope to provide a comprehensive understanding and promote further research in this field.
Collapse
Affiliation(s)
- Ruiqi Liu
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
| | - Jiajun Wu
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
- Otolaryngology & Head and Neck CenterCancer CenterDepartment of Head and Neck SurgeryZhejiang Provincial People's HospitalAffiliated People's Hospital, Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Haiwei Guo
- Otolaryngology & Head and Neck CenterCancer CenterDepartment of Head and Neck SurgeryZhejiang Provincial People's HospitalAffiliated People's Hospital, Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Weiping Yao
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
| | - Shuang Li
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentJinzhou Medical UniversityJinzhouLiaoningChina
| | - Yanwei Lu
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
| | - Yongshi Jia
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
| | - Xiaodong Liang
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
| | - Jianming Tang
- Department of Radiation OncologyThe First Hospital of Lanzhou UniversityLanzhou UniversityLanzhouGansuChina
| | - Haibo Zhang
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
| |
Collapse
|
49
|
Yang J, Xu J, Wang W, Zhang B, Yu X, Shi S. Epigenetic regulation in the tumor microenvironment: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2023; 8:210. [PMID: 37217462 PMCID: PMC10203321 DOI: 10.1038/s41392-023-01480-x] [Citation(s) in RCA: 120] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/17/2023] [Accepted: 04/28/2023] [Indexed: 05/24/2023] Open
Abstract
Over decades, researchers have focused on the epigenetic control of DNA-templated processes. Histone modification, DNA methylation, chromatin remodeling, RNA modification, and noncoding RNAs modulate many biological processes that are crucial to the development of cancers. Dysregulation of the epigenome drives aberrant transcriptional programs. A growing body of evidence suggests that the mechanisms of epigenetic modification are dysregulated in human cancers and might be excellent targets for tumor treatment. Epigenetics has also been shown to influence tumor immunogenicity and immune cells involved in antitumor responses. Thus, the development and application of epigenetic therapy and cancer immunotherapy and their combinations may have important implications for cancer treatment. Here, we present an up-to-date and thorough description of how epigenetic modifications in tumor cells influence immune cell responses in the tumor microenvironment (TME) and how epigenetics influence immune cells internally to modify the TME. Additionally, we highlight the therapeutic potential of targeting epigenetic regulators for cancer immunotherapy. Harnessing the complex interplay between epigenetics and cancer immunology to develop therapeutics that combine thereof is challenging but could yield significant benefits. The purpose of this review is to assist researchers in understanding how epigenetics impact immune responses in the TME, so that better cancer immunotherapies can be developed.
Collapse
Affiliation(s)
- Jing Yang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
50
|
Qiang N, Ao J, Nakamura M, Chiba T, Kusakabe Y, Kaneko T, Kurosugi A, Kogure T, Ma Y, Zhang J, Ogawa K, Kan M, Iwanaga T, Sakuma T, Kanayama K, Kanzaki H, Kojima R, Nakagawa R, Kondo T, Nakamoto S, Muroyama R, Kato J, Mimura N, Ma A, Jin J, Kato N. Alteration of the tumor microenvironment by pharmacological inhibition of EZH2 in hepatocellular carcinoma. Int Immunopharmacol 2023; 118:110068. [PMID: 37001386 DOI: 10.1016/j.intimp.2023.110068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/20/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023]
Abstract
Enhancer of zeste homolog 2 (EZH2), a core component of polycomb repressive component 2 is overexpressed in a variety of cancers and recognized as a therapeutic target molecule. However, EZH2 possesses immunomodulatory functions in the tumor microenvironment (TME). The impact of EZH2 on TME of hepatocellular carcinoma (HCC) using immunocompetent mouse model was evaluated in the present study. UNC1999, an EZH2 inhibitor, impaired growth of the murine HCC cells (H22 cells) and induced apoptosis in a dose-dependent manner. Although UNC1999 significantly inhibited the growth of H22 cells-derived and Hepa1-6 cells-derived tumors in nonobese diabetic/severe combined immunodeficiency mice, its antitumor effect was diminished in allogenic BALB/c and C57BL/6 mice. Flow cytometric analyses of TME cells in BALB/c mice demonstrated a significant decrease in the number of interferon‑γ+ CD8+ T cells and regulatory T cells and a significant increase in the number of myeloid-derived suppressor cells (MDSCs). Administration of Gr-1 neutralizing antibody concomitant with UNC1999 restored antitumor effect accompanied by an increase in the number of CD8+ T cells followed by a decrease in the number of MDSCs. Chemokine antibody array demonstrated an enhanced expression of chemokines responsible for MDSCs recruitment such as C5a, CCL8, and CCL9. In conclusion, the study results demonstrated that EZH2 inhibitor contributed to attenuation of tumor immunity caused by TME arrangement. Combination therapy with EZH2 inhibitors and agents that reduce MDSCs might represent a novel therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Na Qiang
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Junjie Ao
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masato Nakamura
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Tetsuhiro Chiba
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yuko Kusakabe
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tatsuya Kaneko
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akane Kurosugi
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tadayoshi Kogure
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yaojia Ma
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jiaqi Zhang
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Keita Ogawa
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Motoyasu Kan
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Terunao Iwanaga
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takafumi Sakuma
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kengo Kanayama
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroaki Kanzaki
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryuta Kojima
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryo Nakagawa
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takayuki Kondo
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shingo Nakamoto
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryosuke Muroyama
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jun Kato
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Naoya Mimura
- Department of Transfusion Medicine and Cell Therapy, Chiba University Hospital, Chiba, Japan
| | - Anqi Ma
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Naoya Kato
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|