1
|
Pan Y, Wang Y, Gou S. Proteolysis targeting chimera, molecular glue degrader and hydrophobic tag tethering degrader for targeted protein degradation: Mechanisms, strategies and application. Bioorg Chem 2025; 161:108491. [PMID: 40306190 DOI: 10.1016/j.bioorg.2025.108491] [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: 12/03/2024] [Revised: 02/13/2025] [Accepted: 04/16/2025] [Indexed: 05/02/2025]
Abstract
Targeted protein degradation (TPD) represents a revolutionary approach to drug discovery, offering a novel mechanism that outperforms traditional inhibitors. This approach employs small molecule drugs to induce the ubiquitination and subsequent degradation of target protein via the proteasome or lysosomal pathways. Key strategies within TPD include proteolysis targeting chimeras (PROTACs), hydrophobic tag tethering degraders (HyTTDs), and molecular glue degraders (MGDs). PROTACs have been undergone clinical evaluations, MGDs have been used in the clinic, and HyTTDs have shown significant progress in cancer treatment. Each of these strategies presents unique advantages and approaches to target protein degradation. This review summarizes five years of research on PROTACs, HyTTDs, and MGDs, highlighting their design principles, advantages, limitations, and future challenges to provide clear guidance and in-depth insights for advancing drug development.
Collapse
Affiliation(s)
- Yanchang Pan
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yuanjiang Wang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China
| | - Shaohua Gou
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China; Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, PR China.
| |
Collapse
|
2
|
Mitra A, Cutiongco MFA, Burla R, Zeng Y, Na Q, Kong M, Vinod B, Nai MH, Hübner B, Ludwig A, Lim CT, Shivashankar GV, Saggio I, Zhao W. Acute chromatin decompaction stiffens the nucleus as revealed by nanopillar-induced nuclear deformation in cells. Proc Natl Acad Sci U S A 2025; 122:e2416659122. [PMID: 40343993 DOI: 10.1073/pnas.2416659122] [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: 08/24/2024] [Accepted: 03/25/2025] [Indexed: 05/11/2025] Open
Abstract
Chromatin architecture is critical in determining nuclear mechanics. Most studies focus on the mechanical rigidity conferred by chromatin condensation from densely packed heterochromatin, but less is known on how transient chromatin decompaction impinge on nucleus stiffness. Here, we used an array of vertically aligned nanopillars to study nuclear deformability in situ after chromatin decompaction in cells. The nucleus significantly stiffened within 4 h of chromatin decompaction but softened at longer timescales. This acute stiffening of the nucleus was underpinned predominantly by an increase in nucleus volume and nuclear import, and partially by enhanced lamin protein recruitment to the periphery. The coupling between nucleus stiffening and acute chromatin decompaction was observed in low malignancy cancer cell lines (e.g. MCF7, PEO1, A549) but weakened in highly malignant counterparts (e.g. MDA-MB-231, HEYA8, HT1080) due to the capacity to efficiently compact heterochromatin into foci that sustains nucleus deformability required for confined migration. Our work signals how rapid chromatin remodeling is a physiologically relevant pathway to modulate nucleus mechanics and cell migration behavior.
Collapse
Affiliation(s)
- Aninda Mitra
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Marie F A Cutiongco
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Romina Burla
- Dipartimento di Biologia e Biotecnologie, Sapienza-Università di Roma, Roma 00185, Italy
| | - Yongpeng Zeng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Qin Na
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Mengya Kong
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Benjamin Vinod
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Mui Hoon Nai
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Barbara Hübner
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
- NTU Institute of Structural Biology, Nanyang Technological University, Singapore 636921, Singapore
| | - Alexander Ludwig
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
- NTU Institute of Structural Biology, Nanyang Technological University, Singapore 636921, Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
- Institute for Health Innovation and Technology, National University of Singapore, Singapore 117599, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - G V Shivashankar
- Department of Health Sciences and Technology, ETH Zürich, Zürich 8093, Switzerland
- Laboratory of Multiscale Bioimaging, Paul Scherrer Institut, Villigen, Aargau 5232, Switzerland
| | - Isabella Saggio
- Dipartimento di Biologia e Biotecnologie, Sapienza-Università di Roma, Roma 00185, Italy
| | - Wenting Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, Singapore 636921, Singapore
| |
Collapse
|
3
|
Kakimi K, Sugie T. Why combine and why neoadjuvant? Tumor immunological perspectives on chemoimmunotherapy in triple-negative breast cancer. Breast Cancer 2025:10.1007/s12282-025-01707-5. [PMID: 40327275 DOI: 10.1007/s12282-025-01707-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/18/2025] [Indexed: 05/07/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype characterized by limited targeted therapies and high recurrence rates. While immune checkpoint inhibitors (ICIs) have shown promise, their efficacy as monotherapy is limited. Clinically, ICIs demonstrate significant benefit primarily when combined with chemotherapy, particularly in the neoadjuvant setting for early-stage TNBC, which yields superior outcomes compared to adjuvant therapy. This review elucidates the tumor immunological principles underlying these observations. We discussed how the suppressive tumor microenvironment (TME), progressive T cell exhaustion, and associated epigenetic scarring constrain ICI monotherapy effectiveness. Crucially, we highlight the immunological advantages of the neoadjuvant approach: the presence of the primary tumor provides abundant antigens, and intact tumor-draining lymph nodes (TDLNs) act as critical sites for ICI-mediated priming and expansion of naïve and precursor exhausted T cells. This robust activation within TDLNs enhances systemic anti-tumor immunity and expands the T cell repertoire, a process less effectively achieved in the adjuvant setting after tumor resection. These mechanisms provide a strong rationale for the improved pathological complete response (pCR) rates and event-free survival observed with neoadjuvant chemoimmunotherapy, as demonstrated in trials like KEYNOTE-522. We further explore the implications for adjuvant therapy decisions based on treatment response, the challenges of ICI resistance, the need for predictive biomarkers, management of immune-related adverse events (irAEs), and future therapeutic directions. Understanding the dynamic interplay between chemotherapy, ICIs, T cells, and the TME, particularly the role of TDLNs in the neoadjuvant context, is essential for optimizing immunotherapy strategies and improving outcomes for patients with TNBC.
Collapse
Affiliation(s)
- Kazuhiro Kakimi
- Department of Immunology, Kindai University Faculty of Medicine, 377-2 Onohigashi, Osakasayama, Osaka, 589-8511, Japan.
- Chemotherapy Center, Kansai Medical University Kori Hospital, 8-45 Korihondori, Neyagawa, Osaka, 572-8551, Japan.
| | - Tomoharu Sugie
- Chemotherapy Center, Kansai Medical University Kori Hospital, 8-45 Korihondori, Neyagawa, Osaka, 572-8551, Japan.
| |
Collapse
|
4
|
Zebrowski K, June K, Thomas D, Djuric Z, Ballinger T, Kleer CG. Expression of EZH2 and Fatty Acid Synthase in Breast Tissues From Healthy Women With Breast Cancer Risk Factors. Appl Immunohistochem Mol Morphol 2025; 33:186-192. [PMID: 40181650 PMCID: PMC12055476 DOI: 10.1097/pai.0000000000001250] [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: 09/06/2024] [Accepted: 12/31/2024] [Indexed: 04/05/2025]
Abstract
Tissue-based biomarkers that identify women with increased breast cancer risk are needed for cancer prevention. Enhancer of zeste 2 (EZH2) and fatty acid synthase (FASN) are associated with breast cancer aggressiveness, but their expression in normal breast tissues and association with breast cancer risk factors are unclear. Further, there is a need to characterize healthy breast tissue cohorts for unbiased biomarker evaluation. In this study, we employed the Susan G. Komen healthy volunteer tissue bank to evaluate EZH2 and FASN expression and their relationship to breast cancer risk factors. Normal breast core biopsies from 40 healthy donors with low or high Gail scores (<11 or >20, respectively) and normal or obese body mass index (BMI, <25 kg/m 2 or >30 kg/m 2 , respectively) were stained for H&E, EZH2, and FASN and scored independently and blindly using the Allred method. We analyzed the associations between EZH2 and FASN with Gail score, BMI, menopausal status, hormone replacement therapy (HRT), and family history of breast cancer. None of the donors had BRCA1/2 mutations or developed breast cancer after 5 to 9 years. We found that premenopausal women had significantly higher expression of FASN and that EZH2 was higher with increasing Gail risk scores, compared with postmenopausal women. In postmenopausal women, increased EZH2 expression was associated with >5 years of HRT compared with <1 year or no HRT. No associations were found with BMI. This study provides validation of a healthy breast tissue cohort and initial characterization of EZH2 and FASN and their associations with breast cancer risk factors.
Collapse
Affiliation(s)
- Katelyn Zebrowski
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kaleb June
- Departments of Family Medicine and Nutritional Sciences, University of Michigan, Ann Arbor
| | - Dafydd Thomas
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Zora Djuric
- Departments of Family Medicine and Nutritional Sciences, University of Michigan, Ann Arbor
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Tarah Ballinger
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Celina G. Kleer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
5
|
Harada J, Kawashima K, Matsubara Y, Oshi M, Sasamoto M, Yamada A, Suganuma N, Fujii S. H3K27me3-mediated regulation of PD-L1 expression in triple-negative breast cancer (TNBC). Pathol Res Pract 2025; 269:155872. [PMID: 40023141 DOI: 10.1016/j.prp.2025.155872] [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: 10/18/2024] [Revised: 01/31/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
PURPOSE Enhancer of zeste homolog 2 (EZH2) is highly expressed in triple-negative breast cancer (TNBC) and induces massive histone modification via trimethylation at lysine 27 of histone H3 (H3K27me3). The expression level of programmed death ligand 1 (PD-L1) is crucial for determining the indications for immune checkpoint inhibitors in patients with TNBC. This study aimed to clarify the regulatory roles of EZH2 and H3K27me3 in the PD-L1 expression in TNBC cells. METHODS The change in the expression of PD-L1 at mRNA and protein levels was investigated by establishing an EZH2-knockdown MDA-MB-231 cell line using siRNA followed by RT-qPCR and western blotting analyses. Localization of the PD-L1 protein was assessed using immunofluorescence. Chromatin immunoprecipitation (ChIP) assays were performed to investigate the histone methylation status of PD-L1 promoter regions. The correlation among PD-L1, EZH2, and H3K27me3 protein expressions was explored in 57 patients with TNBC through immunohistochemistry. RESULTS Knockdown of EZH2 restored the PD-L1 expression and localization of PD-L1 protein in the cellular membrane. ChIP assay revealed that the knockdown of EZH2 diminished H3K27 trimethylation and enhanced H3K4 trimethylation in the promoter region of PD-L1. Immunohistochemical analysis of TNBC specimens reflected an inverse correlation between PD-L1 expression and H3K27me3 nuclear positivity; however, no correlation between H3K27me3 status and EZH2 expression was observed. CONCLUSIONS The downregulation of EZH2 can potentially enhance the efficacy of immune checkpoint inhibitors in patients with TNBC and may provide a new therapeutic strategy.
Collapse
Affiliation(s)
- Jotaro Harada
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Department of pathology, Yokohama City University Hospital, Yokohama, Japan
| | - Kei Kawashima
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuka Matsubara
- Department of Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masanori Oshi
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mahato Sasamoto
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Akimitsu Yamada
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Nobuyasu Suganuma
- Department of Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoshi Fujii
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Department of pathology, Yokohama City University Hospital, Yokohama, Japan.
| |
Collapse
|
6
|
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
|
7
|
Yin S, Brobbey C, Ball LE, Fu T, Sprague DJ, Gan W. BRD9 functions as a methylarginine reader to regulate AKT-EZH2 signaling. SCIENCE ADVANCES 2025; 11:eads6385. [PMID: 40279411 PMCID: PMC12024519 DOI: 10.1126/sciadv.ads6385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 03/20/2025] [Indexed: 04/27/2025]
Abstract
Recognition of methylarginine marks by effector proteins ("readers") is a critical link between arginine methylation and various cellular processes. Recently, we identified methylation of AKT1 at arginine-391 (R391), but the reader for this methylation has yet to be characterized. Here, we show that bromodomain-containing protein 9 (BRD9), a reader of acetylated lysine, unexpectedly recognizes methylated R391 of AKT1 through an aromatic cage in its bromodomain. Disrupting the methylarginine reader function of BRD9 suppresses AKT activation and tumorigenesis. RNA sequencing data show that BRD9 and AKT coregulate a hallmark transcriptional program in part through enhancer of zeste homolog 2 (EZH2)-mediated methylation of histone-3 lysine-27. We also find that inhibitors of BRD9 and EZH2 display synergistic effects on suppression of cell proliferation and tumor growth. Collectively, our study reveals a previously unknown function of BRD9 and a potential therapeutic strategy for cancer treatment by combining BRD9 and EZH2 inhibitors.
Collapse
Affiliation(s)
- Shasha Yin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Charles Brobbey
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Lauren E. Ball
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Tianmin Fu
- Department of Biological Chemistry and Pharmacology, Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel J. Sprague
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Wenjian Gan
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
8
|
Jeong Y, Jang H, Kim SB, Yu M, Kim RE, Choi WS, Jeon Y, Lim JY. Dual targeting of EZH2 and PD-L1 in Burkitt's lymphoma enhances immune activation and induces apoptotic pathway. Front Immunol 2025; 16:1578665. [PMID: 40308579 PMCID: PMC12040923 DOI: 10.3389/fimmu.2025.1578665] [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: 02/18/2025] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction Enhancer of zeste homolog 2 (EZH2) catalyzes H3K27me3, an epigenetic modification linked to gene silencing, and its overexpression contributes to the progression of hematological malignancies. This study compares the efficacy of a conventional EZH2 inhibitor with a PROTAC-based EZH2 degrader in human lymphoma cell lines. Furthermore, we investigate the anti-tumor effects of combining EZH2 degrader with anti-PD-1, an immune checkpoint inhibitor, focusing on immune cell interactions and underlying mechanisms. Methods The cytotoxic effects of the EZH2 degrader and EZH2 inhibitor were evaluated in Burkitt's, B-cell, cutaneous T-cell, and Hodgkin's lymphoma cell lines. Additionally, the combination therapy of the EZH2 degrader and anti-PD-1 was assessed both in vitro and in a hu-PBMC-CDX mouse model. Results We evaluated the effects of an EZH2 degrader on seven lymphoma cell lines and observed significant reductions in cell viability compared to EZH2 inhibitor, particularly in Burkitt's lymphoma cell lines. EZH2 degrader treatment reduced EZH2 and c-Myc expression, induced G2/M cell cycle arrest, and increased apoptosis markers, including cleaved caspase-3 and cleaved PARP. Furthermore, Burkitt's lymphoma is a PD-L1 negative tumor; however, treatment with the EZH2 degrader resulted in a slight increase in PD-L1 expression. Combining EZH2 degrader with anti-PD-1 significantly enhanced anti-tumor effects compared to monotherapy. In vivo studies using a humanized lymphoma mouse model demonstrated a synergistic anti-tumor effect of EZH2 degrader and anti-PD-1, which was attributed to apoptosis-related pathways. Discussion These findings aim to provide insights into the therapeutic potential of targeting EZH2 in combination with immune checkpoint inhibitors for improved treatment of lymphomas.
Collapse
Affiliation(s)
- Yurim Jeong
- Department of Biomedical Laboratory Science, Inje University, Gimhae, Republic of Korea
| | - Hyewon Jang
- Department of Biomedical Laboratory Science, Inje University, Gimhae, Republic of Korea
| | - Se Been Kim
- Department of Biomedical Laboratory Science, Inje University, Gimhae, Republic of Korea
| | - Minseo Yu
- Department of Biomedical Laboratory Science, Inje University, Gimhae, Republic of Korea
| | - Ra Eun Kim
- Department of Biomedical Laboratory Science, Inje University, Gimhae, Republic of Korea
| | - Wan-Su Choi
- Department of Biomedical Laboratory Science, Inje University, Gimhae, Republic of Korea
| | - Youngwoo Jeon
- Department of Hematology, Yeouido St. Mary Hospital, School of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung-Yeon Lim
- Department of Biomedical Laboratory Science, Inje University, Gimhae, Republic of Korea
| |
Collapse
|
9
|
Wan G, Li S, Tang Q, Qiu H, Zhang Q, Yu L. An updated patent review of EZH2 inhibitors (2024-present). Expert Opin Ther Pat 2025:1-14. [PMID: 40116819 DOI: 10.1080/13543776.2025.2483399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
INTRODUCTION EZH2 forms the PRC2 complex with SUZ12 and EED. As a crucial catalytic subunit of PRC2, EZH2 modifies histone H3K27 via its SET domain, resulting in chromatin condensation and suppressing the transcription of related target genes. EZH2 not only functions in PRC2-dependent transcriptional repression but can also activate gene expression in PRC2-independent circumstances or regulate the activity of downstream genes via its own activating mutations. On the basis of the critical role of EZH2 in cancer, the development of inhibitors targeting EZH2 provides a new strategy for cancer therapy. AREAS COVERED The purpose of this review is to summarize the molecular mechanisms of EZH2 inhibitors and emphasize the research progress on EZH2 inhibitors published in the patent literature in recent years. The literature and patent databases of PubMed, Web of Science, SCIFinder, WIPO, USPTO, EPO, and CNIPA were combined to search for more effective EZH2 inhibitors. EXPERT OPINION Recently, a wide range of structurally diverse EZH2 inhibitors, particularly EZH2 degraders, have been identified. These EZH2 modulators have demonstrated significant potential in treating various diseases, with cancer being a primary focus. The development of small molecules targeting EZH2 with distinct pharmacological effects is poised with numerous opportunities.
Collapse
Affiliation(s)
- Guoquan Wan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Siyan Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Qifan Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Huapei Qiu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Qiangsheng Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Luoting Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Goleij P, Heidari MM, Tabari MAK, Hadipour M, Rezaee A, Javan A, Sanaye PM, Larsen DS, Daglia M, Khan H. Polycomb repressive complex 2 (PRC2) pathway's role in cancer cell plasticity and drug resistance. Funct Integr Genomics 2025; 25:53. [PMID: 40048009 DOI: 10.1007/s10142-025-01563-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/17/2025] [Accepted: 02/23/2025] [Indexed: 05/13/2025]
Abstract
Polycomb Repressive Complex 2 (PRC2) is a central regulator of gene expression via the trimethylation of histone H3 on lysine 27. This epigenetic modification plays a crucial role in maintaining cell identity and controlling differentiation, while its dysregulation is closely linked to cancer progression. PRC2 silences tumor suppressor genes, promoting cell proliferation, metastasis, epithelial-mesenchymal transition, and cancer stem cell plasticity. Enhancement of zeste homolog 2 (EZH2) overexpression or gain-of-function mutations have been observed in several cancers, including lymphoma, breast, and prostate cancers, driving aggressive tumor behavior and drug resistance. In addition to EZH2, other PRC2 components, such as embryonic ectoderm development (EED) and suppressor of zeste 12, are essential for complex stability and function. EED, in particular, enhances EZH2 activity and has emerged as a therapeutic target. Inhibitors like MAK683 and EED226 disrupt EED's ability to maintain PRC2 activity, thereby reducing H3K27me3 levels and reactivating tumor suppressor genes. Valemetostat, a dual inhibitor of both EZH2 and EED, has shown promising results in aggressive cancers like diffuse large B-cell lymphoma and small-cell lung cancer, underlining the therapeutic potential of targeting multiple PRC2 components. PRC2's role extends beyond gene repression, as it contributes to metabolic reprogramming in tumors, regulating glycolysis and lipid synthesis to fuel cancer growth. Furthermore, PRC2 is implicated in chemoresistance, particularly by modulating DNA damage response and immune evasion. Tazemetostat, a selective EZH2 inhibitor, has demonstrated significant clinical efficacy in EZH2-mutant cancers, such as non-Hodgkin lymphomas and epithelioid sarcoma. However, the compensatory function of enhancer of zeste homolog 1 (EZH1) in some cancers requires dual inhibition strategies, as seen with agents like UNC1999 and Tulmimetostat, which target both EZH1 and EZH2. Given PRC2's multifaceted role in cancer biology, its inhibition represents a promising avenue for therapeutic intervention. The continued development of PRC2 inhibitors and exploration of their use in combination with standard chemotherapy or immunotherapy has great potential for improving patient outcomes in cancers driven by PRC2 dysregulation.
Collapse
Affiliation(s)
- Pouya Goleij
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, 6715847141, Iran.
- Immunology Board for Transplantation and Cell-Based Therapeutics (Immunotact), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Mohammad Mahdi Heidari
- Department of Pediatrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, 4815733971, Iran
| | - Mahboube Hadipour
- Department of Biochemistry, School of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, 7919693116, Iran
| | - Aryan Rezaee
- School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Alireza Javan
- School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Pantea Majma Sanaye
- School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, 4513956184, Iran
| | - Danaé S Larsen
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland, 1010, New Zealand
| | - Maria Daglia
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, 212013, China
| | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan.
- Department of Pharmacy, Korea University, Sejong, 20019, South Korea.
| |
Collapse
|
11
|
Liu D, Liu L, Zhang X, Zhao X, Li X, Che X, Wu G. Decoding driver and phenotypic genes in cancer: Unveiling the essence behind the phenomenon. Mol Aspects Med 2025; 103:101358. [PMID: 40037122 DOI: 10.1016/j.mam.2025.101358] [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: 10/27/2024] [Revised: 01/25/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Gray hair, widely regarded as a hallmark of aging. While gray hair is associated with aging, reversing this trait through gene targeting does not alter the fundamental biological processes of aging. Similarly, certain oncogenes (such as CXCR4, MMP-related genes, etc.) can serve as markers of tumor behavior, such as malignancy or prognosis, but targeting these genes alone may not lead to tumor regression. We pioneered the name of this class of genes as "phenotypic genes". Historically, cancer genetics research has focused on tumor driver genes, while genes influencing cancer phenotypes have been relatively overlooked. This review explores the critical distinction between driver genes and phenotypic genes in cancer, using the MAPK and PI3K/AKT/mTOR pathways as key examples. We also discuss current research techniques for identifying driver and phenotypic genes, such as whole-genome sequencing (WGS), RNA sequencing (RNA-seq), RNA interference (RNAi), CRISPR-Cas9, and other genomic screening methods, alongside the concept of synthetic lethality in driver genes. The development of these technologies will help develop personalized treatment strategies and precision medicine based on the characteristics of relevant genes. By addressing the gap in discussions on phenotypic genes, this review significantly contributes to clarifying the roles of driver and phenotypic genes, aiming at advancing the field of targeted cancer therapy.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Lei Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xiaoman Zhang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xinming Zhao
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xiaorui Li
- Department of Oncology, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, China.
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
12
|
Carbone FP, Ancona P, Volinia S, Terrazzan A, Bianchi N. Druggable Molecular Networks in BRCA1/BRCA2-Mutated Breast Cancer. BIOLOGY 2025; 14:253. [PMID: 40136510 PMCID: PMC11940086 DOI: 10.3390/biology14030253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025]
Abstract
Mutations in the tumor suppressor genes BRCA1 and BRCA2 are associated with the triple-negative breast cancer phenotype, particularly aggressive and hard-to-treat tumors lacking estrogen, progesterone, and human epidermal growth factor receptor 2. This research aimed to understand the metabolic and genetic links behind BRCA1 and BRCA2 mutations and investigate their relationship with effective therapies. Using the Cytoscape software, two networks were generated through a bibliographic analysis of articles retrieved from the PubMed-NCBI database. We identified 98 genes deregulated by BRCA mutations, and 24 were modulated by therapies. In particular, BIRC5, SIRT1, MYC, EZH2, and CSN2 are influenced by BRCA1, while BCL2, BAX, and BRIP1 are influenced by BRCA2 mutation. Moreover, the study evaluated the efficacy of several promising therapies, targeting only BRCA1/BRCA2-mutated cells. In this context, CDDO-Imidazolide was shown to increase ROS levels and induce DNA damage. Similarly, resveratrol decreased the expression of the anti-apoptotic gene BIRC5 while it increased SIRT1 both in vitro and in vivo. Other specific drugs were found to induce apoptosis selectively in BRCA-mutated cells or block cell growth when the mutation occurs, i.e., 3-deazaneplanocin A, genistein or daidzein, and PARP inhibitors. Finally, over-representation analysis on the genes highlights ferroptosis and proteoglycan pathways as potential drug targets for more effective treatments.
Collapse
Affiliation(s)
- Francesca Pia Carbone
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
| | - Pietro Ancona
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
| | - Stefano Volinia
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
- Genomics Core Facility, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
- Laboratory for Technologies of Advanced Therapies (LTTA), 44121 Ferrara, Italy
| | - Anna Terrazzan
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
- Genomics Core Facility, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
- Laboratory for Technologies of Advanced Therapies (LTTA), 44121 Ferrara, Italy
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
| |
Collapse
|
13
|
Colemon A, Prioleau T, Rouse C, Pendergast AM. ABL Kinases Modulate EZH2 Phosphorylation and Signaling in Metastatic Triple Negative Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638898. [PMID: 40027835 PMCID: PMC11870601 DOI: 10.1101/2025.02.18.638898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Triple-negative breast cancer (TNBC) remains a leading cause of cancer associated deaths in women owing to its highly metastatic potential and limited treatment options. Recent studies have shown that expression of proteins associated with epigenetic regulation of gene expression are associated with metastatic relapse, however targeting epigenetic regulatory proteins has not resulted in effective therapies for TNBC in the clinic. The ABL tyrosine kinases promote metastasis of breast cancer cells in mouse models. However, a role of ABL kinases in the regulation of epigenetic processes in solid tumor metastasis remains unexplored. Here we show that inactivation of ABL kinases in bone metastatic TNBC cells led to a significant enrichment in gene signatures associated with the PRC2 protein complex, revealing a functional link between ABL kinases and the PRC2 complex. ABL inactivation promotes EZH2-T487 phosphorylation through the regulation of a FAK-CDK1 signaling axis. We find that phosphorylated EZH2 T487 or a phosphomimic EZH2 T487D mutant exhibit increased binding to non-canonical binding partners of EZH2 including c-MYC and ZMYND8. Notably, we identify a therapeutic vulnerability in TNBC cells whereby combination treatment with ABL allosteric inhibitors and EZH2 inhibitors elicits a synergistic decrease in TNBC cell survival in vitro, and impairs TNBC metastasis, prolonging survival of tumor-bearing mice treated with the combination therapy. One Sentence Summary ABL Kinases indirectly impact EZH2 catalytic activity by blocking a signaling cascade that leads to changes in the phosphorylation, protein interactions, and function of the PRC2 catalytic component EZH2 in TNBC.
Collapse
|
14
|
Jager EC, van Hemel BM, Rutgers B, Zandee WT, Jansen L, Kruijff S, Links TP. Validation of the International Medullary Thyroid Cancer Grading System and Identification of EZH2 as a Prognostic and Potential Therapeutic Marker in Medullary Thyroid Cancer. Cancers (Basel) 2025; 17:737. [PMID: 40075585 PMCID: PMC11899120 DOI: 10.3390/cancers17050737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/06/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Medullary thyroid cancer (MTC) is a heterogeneous disease. While the International MTC Grading System (IMTCGS) provides baseline risk stratification, it lacks therapeutic relevance. In several cancers, EZH2 overexpression harbors an adverse prognosis, with several EZH2 inhibitors undergoing investigation. This study validated the IMTCGS and examined the prognostic value of EZH2 and other biomarkers. Methods: Clinical data were collected and MTC specimens were retrospectively reviewed and morphologically assessed. Immunohistochemistry (IHC) of Ki-67 allowed IMTCGS validation. IHC of EZH2, PD-L1 and PSMA was evaluated on a tissue microarray (TMA). Results: Of 64 MTCs, the median tumor size was 28 mm (IQR 15-40). Coagulative necrosis, ≥5 mitoses, and Ki-67 ≥ 5% was seen in nineteen (30%), three (5%) and seven (11%) cases. Median Ki-67 was 0.9% (IQR 0.4-2.1). Forty-three (67%) and twenty-one (33%) were classified as IMTCGS low- and high-risk, respectively. High-risk tumors were associated with lower distant metastasis-free survival (DMFS) (HR 5.651, p = 0.017), locoregional recurrence-free survival (LRFS) (HR 18.323, p < 0.001) and disease-specific survival (DSS) (HR 10.001, p = 0.002), but not with overall survival (OS) (HR 2.109, p = 0.146). EZH2 expression was identified in 39/46 (85%) cases on the TMA. An expression of ≥10% (9/46, 20%) was predictive for DMFS (HR 4.747, p = 0.030), LRFS (HR 4.242, 0.039), DSS (HR 19.736, p < 0.001) and OS (HR 8.386, p = 0.004). PD-L1 and PSMA had no prognostic value. Conclusions: This study validates the prognostic value of the IMTCGS and identifies EZH2 as a novel prognostic biomarker in MTC patients. The therapeutic potential of EZH2 warrants further investigation in larger cohorts.
Collapse
Affiliation(s)
- Eline C. Jager
- Department of Internal Medicine, Division of Endocrinology, University Medical Center Groningen, Hanzeplein 1, 9731 GZ Groningen, The Netherlands; (E.C.J.)
- Department of Surgery, Division of Surgical Oncology, University Medical Center Groningen, Hanzeplein 1, 9731 GZ Groningen, The Netherlands
| | - Bettien M. van Hemel
- Department of Pathology, University Medical Center Groningen, Hanzeplein 1, 9731 GZ Groningen, The Netherlands
| | - Bea Rutgers
- Department of Pathology, University Medical Center Groningen, Hanzeplein 1, 9731 GZ Groningen, The Netherlands
| | - Wouter T. Zandee
- Department of Internal Medicine, Division of Endocrinology, University Medical Center Groningen, Hanzeplein 1, 9731 GZ Groningen, The Netherlands; (E.C.J.)
| | - Liesbeth Jansen
- Department of Surgery, Division of Surgical Oncology, University Medical Center Groningen, Hanzeplein 1, 9731 GZ Groningen, The Netherlands
| | - Schelto Kruijff
- Department of Surgery, Division of Surgical Oncology, University Medical Center Groningen, Hanzeplein 1, 9731 GZ Groningen, The Netherlands
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Hanzeplein 1, 9731 GZ Groningen, The Netherlands
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Solnavägen 1, 171 77 Stockholm, Sweden
| | - Thera P. Links
- Department of Internal Medicine, Division of Endocrinology, University Medical Center Groningen, Hanzeplein 1, 9731 GZ Groningen, The Netherlands; (E.C.J.)
| |
Collapse
|
15
|
Yang Q, Zhou Z, Li L, Lu R, Hou G, Huang C, Huang J, Li H, Zhang Y, Li J, Zhang Y, Xu A, Chen R, Wang Y, Zhao X, Huang J, Wang Y, Zhao X, Yu J. The NEXT complex regulates H3K27me3 levels to affect cancer progression by degrading G4/U-rich lncRNAs. Nucleic Acids Res 2025; 53:gkaf107. [PMID: 39988317 PMCID: PMC11840553 DOI: 10.1093/nar/gkaf107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 01/13/2025] [Accepted: 02/05/2025] [Indexed: 02/25/2025] Open
Abstract
Polycomb repressive complex 2 (PRC2) is responsible for depositing H3K27me3 and plays essential roles in gene silencing during development and cancer. Meanwhile, the nuclear exosome targeting (NEXT) complex facilitates the degradation of numerous noncoding RNAs in the nucleoplasm. Here we find that the functional deficiency of the NEXT complex leads to an overall decrease in H3K27me3 levels. Specifically, ZCCHC8 depletion results in significant upregulation of nascent long noncoding RNAs (lncRNAs) containing G-quadruplex (G4) and U-Rich motifs (G4/U-Rich lncRNAs). The G4 motif binds to EZH2, blocking the chromatin recruitment of PRC2, while the U-Rich motif is specifically recognized by the NEXT complex for RNA exosome-mediated degradation. In tumor tissues with high ZCCHC8 expression in clear cell renal cell carcinoma (ccRCC) and lung adenocarcinoma (LUAD) patients, the NEXT complex excessively degrades nascent G4/U-Rich lncRNAs. Consequently, PRC2 core subunits are released and recruited to neighboring genomic loci, resulting in increased H3K27me3 levels and downregulation of adjacent genes, including tumor suppressors like SEMA5A and ARID1A. Notably, the EZH2 inhibitor Tazemetostat (EPZ-6438) exhibits greater sensitivity in cells with higher ZCCHC8 expression. Altogether, our findings demonstrate a novel mechanism that the NEXT complex regulates H3K27me3 levels by degrading nascent G4/U-Rich lncRNAs in cancer cells.
Collapse
Affiliation(s)
- Qianqian Yang
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Zihan Zhou
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Lian Li
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Runhui Lu
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Guofang Hou
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Caihu Huang
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Jiayi Huang
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Hongyan Li
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Yafan Zhang
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Junya Li
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Yixin Zhang
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Anan Xu
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Ran Chen
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Yanli Wang
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Xian Zhao
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Jian Huang
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Yiwei Wang
- Department of Urology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiaojing Zhao
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| | - Jianxiu Yu
- Department of Biochemistry and Molecular Cell Biology & Department of Thoracic Surgery Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai 200025, China
| |
Collapse
|
16
|
Liu R, Li Z, Chen R, Fang Z, Liu Z, Liu H. EZH2 serves as a viable therapeutic target for myeloma-induced osteolytic bone destruction. Nat Commun 2025; 16:1206. [PMID: 39885217 PMCID: PMC11782520 DOI: 10.1038/s41467-025-56506-5] [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: 04/11/2024] [Accepted: 01/21/2025] [Indexed: 02/01/2025] Open
Abstract
Myelomatous bone disease is a complication characterized by lytic bone lesions, reduced bone formation, bone pain, and increased fracture risk. Understanding these underlying mechanisms is crucial for developing effective therapeutic approaches. Here we show the role of enhancer of zeste homolog 2 (EZH2) in bone lesions induced by myeloma cells. Our research reveals that cytokines produced by myeloma-associated adipocytes activate the expression of EZH2 in myeloma cells. Furthermore, we find that EZH2 forms a transcriptional repression complex with transcription factor AP2α. This complex promotes trimethylation at lysine 27 of histone H3 (H3K27me3) in the promoter region of the tumor suppressor gene EMP1, resulting in transcriptional silencing. EMP1 silencing leads to increased myeloma cell proliferation and the concomitant secretion of osteolytic cytokines that contribute to bone destruction. Importantly, EZH2 inhibitors effectively treat myeloma-induced osteolytic lesions. Thus, targeting EZH2 represents a potential therapeutic strategy for preventing and managing myeloma bone disease.
Collapse
Affiliation(s)
- Rui Liu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Zongwei Li
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Rui Chen
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Zhihong Fang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
- Department of Hematology, Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, China
| | - Zhiqiang Liu
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| | - Huan Liu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China.
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China.
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
- Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, China.
| |
Collapse
|
17
|
Wang R, Dong X, Zhang X, Liao J, Cui W, Li W. Exploring viral mimicry combined with epigenetics and tumor immunity: new perspectives in cancer therapy. Int J Biol Sci 2025; 21:958-973. [PMID: 39897033 PMCID: PMC11781167 DOI: 10.7150/ijbs.103877] [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: 09/19/2024] [Accepted: 12/20/2024] [Indexed: 02/04/2025] Open
Abstract
Viral mimicry refers to an active antiviral response triggered by the activation of endogenous retroviruses (ERVs), usually manifested by the formation of double-stranded RNA (dsRNA) and activation of the cellular interferon response, which activates the immune system and produces anti-tumor effects. Epigenetic studies have shown that epigenetic modifications (e.g. DNA methylation, histone modifications, etc.) play a crucial role in tumorigenesis, progression, and treatment resistance. Particularly, alterations in DNA methylation may be closely associated with the suppression of ERVs expression, and treatment by demethylation may restore ERVs activity and thus strengthen the tumor immune response. Therefore, we propose that viral mimicry can induce immune responses in the tumor microenvironment by activating the expression of ERVs, and that epigenetic alterations may play a key regulatory role in this process. In this paper, we review the intersection of viral mimicry, epigenetics and tumor immunotherapy, and explore the possible interactions and synergistic effects among the three, aiming to provide a new theoretical basis and potential strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Ruirui Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University. Tongzipo Road 138, Changsha, Hunan, People's Republic of China
| | - Xin Dong
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiongjian Zhang
- Department of Radiology, The Third Xiangya Hospital of Central South University. Tongzipo Road 138, Changsha, Hunan, People's Republic of China
| | - Jinzhuang Liao
- Department of Radiology, The Third Xiangya Hospital of Central South University. Tongzipo Road 138, Changsha, Hunan, People's Republic of China
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Cui
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University. Tongzipo Road 138, Changsha, Hunan, People's Republic of China
| |
Collapse
|
18
|
Slachmuylders E, Laenen A, Vernemmen A, Keupers M, Nevelsteen I, Han SN, Neven P, Van Ongeval C, Wildiers H, Smeets A, Floris G. Expression patterns of H3K27me3 for differentiation of breast fibroadenomas and phyllodes tumors. APMIS 2025; 133:e13485. [PMID: 39454207 DOI: 10.1111/apm.13485] [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: 12/06/2023] [Accepted: 10/11/2024] [Indexed: 10/27/2024]
Abstract
Phyllodes tumors (PTs) are rare breast tumors showing overlapping features with fibroadenomas (FAs). Diagnosis on small biopsies is challenging. New diagnostic markers are needed. Here we evaluated immunohistochemical staining of histone 3 trimethyl-lysine-27 (H3K27me3) as a diagnostic and prognostic marker in a series of PTs. Surgically removed PTs at our institution (September 1990 and July 2022) and control FAs. Tissue micro-arrays (4 cores, 2 mm Ø) stained with H3K27me3, and scored with QuPath-derived H-score. Fisher exact test, Mann-Whitney U-test and chi-squared test used for group comparison. ROC analysis applied to define cutoffs. Cox proportional hazards models were used for assessing disease-free survival (DFS), overall survival (OS), and disease-specific survival (DSS) in PTs. We included 81 patients with PTs and 44 patients with FAs. QuPath-derived H-scores of stromal H3K27me3 were statically significantly lower in PTs than in FAs (p < 0.001). We identified exploratory cutoffs to discriminate FAs from benign and malignant PTs (AUC = 0.78 and 0.73, respectively). No associations between DFS, OS, or DSS and H3K27me3 expression were found. H3K27me3 expression differs between FAs and PTs, indicating potential as diagnostic marker, but it is not predictive for DFS, OS or DSS in PTs. Further validation is needed.
Collapse
Affiliation(s)
- E Slachmuylders
- Department of Imaging and Pathology, Laboratory of Translational Cell and Tissue Research, KU Leuven - University of Leuven, Leuven, Belgium
- Department of Pathology, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium
| | - A Laenen
- Interuniversity Centre for Biostatistics and Statistical Bioinformatics, KU Leuven - University of Leuven, Leuven, Belgium
| | - A Vernemmen
- Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - M Keupers
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - I Nevelsteen
- Department of Surgical Oncology, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven - University of Leuven, Leuven, Belgium
| | - S N Han
- Department of Oncology, KU Leuven - University of Leuven, Leuven, Belgium
- Department of Gynecology and Obstetrics, KU Leuven - University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - P Neven
- Department of Oncology, KU Leuven - University of Leuven, Leuven, Belgium
- Department of Gynecology and Obstetrics, KU Leuven - University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - C Van Ongeval
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - H Wildiers
- Department of Oncology, KU Leuven - University of Leuven, Leuven, Belgium
- Department of General Medical Oncology, KU Leuven - University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - A Smeets
- Department of Surgical Oncology, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven - University of Leuven, Leuven, Belgium
| | - G Floris
- Department of Imaging and Pathology, Laboratory of Translational Cell and Tissue Research, KU Leuven - University of Leuven, Leuven, Belgium
- Department of Pathology, University Hospitals Leuven, KU Leuven - University of Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Cao Y, Wang S, Ma J, Long M, Ma X, Yang X, Ji Y, Tang X, Liu J, Lin C, Yang Y, Du P. Mechanistic insights into SIRT7 and EZH2 regulation of cisplatin resistance in bladder cancer cells. Cell Death Dis 2024; 15:931. [PMID: 39719443 DOI: 10.1038/s41419-024-07321-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 12/26/2024]
Abstract
Cisplatin (CDDP) resistance has been established to significantly impact Bladder Cancer (BCa) therapy. On the other hand, the crucial regulatory involvement of SIRT7 and EZH2 in bladder cancer development is well known. Herein, the collaborative regulatory roles and underlying mechanisms of SIRT7 and EZH2 in CDDP resistance in bladder cancer were explored. Immunohistochemistry (IHC) and Western Blot (WB) analyses were used to assess the expression levels of SIRT7/EZH2 and RND3 in bladder cancer tissues, normal ureteral epithelial cells, and bladder cancer cell lines. Furthermore, the impact of various treatments on of UMUC3 cell proliferation and CDDP sensitivity was assessed using CCK-8 assays, plate cloning assays, and flow cytometry analysis. Additionally, the levels of H3K18ac and H3K27me3 at the promoter region of the RND3 gene, the binding abilities of SIRT7 and EZH2, and the succinylation level of the EZH2 protein were examined using ChIP-qPCR assays, CO-IP assays, and IP assays, respectively. Moreover, in vivo experiments were conducted using a bladder cancer mouse model created by subcutaneously injecting UMUC3 cells into Balb/c nude mice. According to the results, SIRT7 correlated with the sensitivity of bladder cancer cells to both the platinum-based chemotherapy and CDDP. Specifically, SIRT7 could bind to the RND3 promoter, downregulating H3K18ac and RND3, ultimately leading to an increased CDDP sensitivity in UMUC3 cells. Furthermore, EZH2 siRNA could decrease H3K27me3 levels in the RND3 promoter, upregulating RND3. Overall, in the promoter region of the RND3 gene, SIRT7 upregulated H3K27me3 and EZH2 downregulated H3K18ac, leading to a decline in RND3 expression and CDDP sensitivity in bladder cancer cells. Additionally, SIRT7 reduced the succinylation of the EZH2 protein resulting in an EZH2-mediated RND3 downregulation. Therefore, targeting SIRT7 and EZH2 could be a viable approach to enhancing CDDP efficacy in bladder cancer treatment.
Collapse
Affiliation(s)
- Yudong Cao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Shuo Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jinchao Ma
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Mengping Long
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiuli Ma
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiao Yang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yongpeng Ji
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xingxing Tang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jia Liu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Chen Lin
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yong Yang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Peng Du
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
20
|
Sarathi R, Sarumathy S, Durai Mavalavan VM. EVOLUTION OF METFORMIN IN BREAST CANCER THERAPY IN LAST TWO DECADES: A REVIEW. Exp Oncol 2024; 46:185-191. [PMID: 39704463 DOI: 10.15407/exp-oncology.2024.03.185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Indexed: 12/21/2024]
Abstract
Among women, breast cancer is one of the most prevalent cancers. The disease has a complex etiology, with multiple biological pathways contributing to its development. As insulin signaling has mitogenic effects, glucose is a necessary cellular metabolic substrate, and the growth and metastasis of breast cancer are closely related to cellular glucose metabolism. Anti-diabetic medications have drawn increased attention as a potential treatment for breast cancer. Metformin lowers cancer incidence and death rates in patients with type 2 diabetes, according to epidemiologic studies. Preclinical studies conducted in vivo and in vitro offer fascinating new insights into the cellular mechanisms underlying metformin oncostatic action. We present an overview of the mechanisms of anticancer effects of metformin and discuss its potential function as an adjuvant in the treatment of breast cancer.
Collapse
Affiliation(s)
- R Sarathi
- Department of Pharmacy Practice, SRM College of Pharmacy, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India
| | - S Sarumathy
- Department of Pharmacy Practice, SRM College of Pharmacy, Faculty of Medicine and Health Sciences, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India
| | - V M Durai Mavalavan
- Department of Medical Oncology, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India
| |
Collapse
|
21
|
Liapodimitri A, Tetens AR, Craig-Schwartz J, Lunsford K, Skalitzky KO, Koldobskiy MA. Progress Toward Epigenetic Targeted Therapies for Childhood Cancer. Cancers (Basel) 2024; 16:4149. [PMID: 39766049 PMCID: PMC11674401 DOI: 10.3390/cancers16244149] [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: 11/08/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Among the most significant discoveries from cancer genomics efforts has been the critical role of epigenetic dysregulation in cancer development and progression. Studies across diverse cancer types have revealed frequent mutations in genes encoding epigenetic regulators, alterations in DNA methylation and histone modifications, and a dramatic reorganization of chromatin structure. Epigenetic changes are especially relevant to pediatric cancers, which are often characterized by a low rate of genetic mutations. The inherent reversibility of epigenetic lesions has led to an intense interest in the development of epigenetic targeted therapies. Additionally, the recent appreciation of the interplay between the epigenome and immune regulation has sparked interest in combination therapies and synergistic immunotherapy approaches. Further, the recent appreciation of epigenetic variability as a driving force in cancer evolution has suggested new roles for epigenetic therapies in limiting plasticity and resistance. Here, we review recent progress and emerging directions in the development of epigenetic targeted therapeutics and their promise across the landscape of childhood cancers.
Collapse
Affiliation(s)
- Athanasia Liapodimitri
- Division of Pediatric Oncology, Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; (A.L.); (A.R.T.); (J.C.-S.); (K.L.); (K.O.S.)
| | - Ashley R. Tetens
- Division of Pediatric Oncology, Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; (A.L.); (A.R.T.); (J.C.-S.); (K.L.); (K.O.S.)
| | - Jordyn Craig-Schwartz
- Division of Pediatric Oncology, Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; (A.L.); (A.R.T.); (J.C.-S.); (K.L.); (K.O.S.)
| | - Kayleigh Lunsford
- Division of Pediatric Oncology, Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; (A.L.); (A.R.T.); (J.C.-S.); (K.L.); (K.O.S.)
| | - Kegan O. Skalitzky
- Division of Pediatric Oncology, Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; (A.L.); (A.R.T.); (J.C.-S.); (K.L.); (K.O.S.)
| | - Michael A. Koldobskiy
- Division of Pediatric Oncology, Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA; (A.L.); (A.R.T.); (J.C.-S.); (K.L.); (K.O.S.)
- Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
22
|
Wang MS, Sussman J, Xu JA, Patel R, Elghawy O, Rawla P. Pharmacological Advancements of PRC2 in Cancer Therapy: A Narrative Review. Life (Basel) 2024; 14:1645. [PMID: 39768352 PMCID: PMC11678550 DOI: 10.3390/life14121645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Polycomb repressive complex 2 (PRC2) is known to regulate gene expression and chromatin structure as it methylates H3K27, resulting in gene silencing. Studies have shown that PRC2 has dual functions in oncogenesis that allow it to function as both an oncogene and a tumor suppressor. Because of this, nuanced strategies are necessary to promote or inhibit PRC2 activity therapeutically. Given the therapeutic vulnerabilities and associated risks in oncological applications, a structured literature review on PRC2 was conducted to showcase similar cofactor competitor inhibitors of PRC2. Key inhibitors such as Tazemetostat, GSK126, Valemetostat, and UNC1999 have shown promise for clinical use within various studies. Tazemetostat and GSK126 are both highly selective for wild-type and lymphoma-associated EZH2 mutants. Valemetostat and UNC1999 have shown promise as orally bioavailable and SAM-competitive inhibitors of both EZH1 and EZH2, giving them greater efficacy against potential drug resistance. The development of other PRC2 inhibitors, particularly inhibitors targeting the EED or SUZ12 subunit, is also being explored with the development of drugs like EED 226. This review aims to bridge gaps in the current literature and provide a unified perspective on promising PRC2 inhibitors as therapeutic agents in the treatment of lymphomas and solid tumors.
Collapse
Affiliation(s)
- Michael S. Wang
- Hospital of the University of Pennsylvania, HUP 3400 Spruce St., Philadelphia, PA 19104, USA; (M.S.W.)
| | - Jonathan Sussman
- Hospital of the University of Pennsylvania, HUP 3400 Spruce St., Philadelphia, PA 19104, USA; (M.S.W.)
| | - Jessica A. Xu
- Hospital of the University of Pennsylvania, HUP 3400 Spruce St., Philadelphia, PA 19104, USA; (M.S.W.)
| | - Reema Patel
- University of Virginia School of Medicine, Charlottesville, VA 22903, USA;
| | - Omar Elghawy
- Hospital of the University of Pennsylvania, HUP 3400 Spruce St., Philadelphia, PA 19104, USA; (M.S.W.)
| | - Prashanth Rawla
- Parrish Healthcare, 951 North Washington Ave., Titusville, FL 32796, USA
| |
Collapse
|
23
|
Gilardini Montani MS, Benedetti R, Cirone M. Targeting EZH2 in Cancer: Mechanisms, Pathways, and Therapeutic Potential. Molecules 2024; 29:5817. [PMID: 39769907 PMCID: PMC11678268 DOI: 10.3390/molecules29245817] [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: 11/14/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Enhancer of zeste homolog 2 (EZH2) is a methyltransferase involved in cell cycle regulation, cell differentiation, and cell death and plays a role in modulating the immune response. Although it mainly functions by catalyzing the tri-methylation of H3 histone on K27 (H3K27), to inhibit the transcription of target genes, EZH2 can directly methylate several transcription factors or form complexes with them, regulating their functions. EZH2 expression/activity is often dysregulated in cancer, contributing to carcinogenesis and immune escape, thereby representing an important target in anti-cancer therapy. This review summarizes some of the mechanisms through which EZH2 regulates the expression and function of tumor suppressor genes and oncogenic molecules such as STAT3, mutant p53, and c-Myc and how it modulates the anti-cancer immune response. The influence of posttranslational modifications on EZH2 activity and stability and the possible strategies leading to its inhibition are also reviewed.
Collapse
Affiliation(s)
| | | | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| |
Collapse
|
24
|
Bakhtiari M, Jordan SC, Mumme HL, Sharma R, Shanmugam M, Bhasin SS, Bhasin M. ARMH1 is a novel marker associated with poor pediatric AML outcomes that affect the fatty acid synthesis and cell cycle pathways. Front Oncol 2024; 14:1445173. [PMID: 39703843 PMCID: PMC11655347 DOI: 10.3389/fonc.2024.1445173] [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: 06/06/2024] [Accepted: 11/04/2024] [Indexed: 12/21/2024] Open
Abstract
Introduction Despite remarkable progress in Pediatric Acute Myeloid Leukemia (pAML) treatments, the relapsed disease remains difficult to treat, making it pertinent to identify novel biomarkers of prognostic/therapeutic significance. Material and methods Bone marrow samples from 21 pAML patients were analyzed using single cell RNA sequencing, functional assays with ARMH1 knockdown and overexpression were performed in leukemia cell lines to evaluate impact on proliferation and migration, and chemotherapy sensitivity. Mitochondrial function was assessed via Seahorse assay, ARMH1 interacting proteins were studied using co-immunoprecipitation. Bulk RNA-seq was performed on ARMH1knockdown and over expressing cell lines to evaluate the pathways and networks impacted by ARMH1. Results Our data shows that ARMH1, a novel cancer-associated gene, is highly expressed in the malignant blast cells of multiple pediatric hematologic malignancies, including AML, T/B-ALL, and T/B-MPAL. Notably, ARMH1 expression is significantly elevated in blast cells of patients who relapsed or have a high-risk cytogenetic profile (MLL) compared to standard-risk (RUNX1, inv (16)). ARMH1 expression is also significantly correlated with the pediatric leukemia stem cell score of 6 genes (LSC6) associated with poor outcomes. Perturbation of ARMH1 (knockdown and overexpression) in leukemia cell lines significantly impacted cell proliferation and migration. The RNA-sequencing analysis on multiple ARMH1 knockdown and overexpressing cell lines established an association with mitochondrial fatty acid synthesis and cell cycle pathways.The investigation of the mitochondrial matrix shows that pharmacological inhibition of a key enzyme in fatty acid synthesis regulation, CPT1A, resulted in ARMH1 downregulation. ARMH1 knockdown also led to a significant reduction in CPT1A and ATP production as well as Oxygen Consumption Rate. Our data indicates that downregulating ARMH1 impacts cell proliferation by reducing key cell cycle regulators such as CDCA7 and EZH2. Further, we also established that ARMH1 is a key physical interactant of EZH2, associated with multiple cancers. Conclusion Our findings underscore further evaluation of ARMH1 as a potential candidate for targeted therapies and stratification of aggressive pAML to improve outcomes.
Collapse
Affiliation(s)
- Mojtaba Bakhtiari
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
| | - Sean C. Jordan
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
- Department of Biomedical Informatics, Emory University, Atlanta, GA, United States
| | - Hope L. Mumme
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
- Department of Biomedical Informatics, Emory University, Atlanta, GA, United States
| | - Richa Sharma
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, United States
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, United States
| | - Swati S. Bhasin
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
- Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - Manoj Bhasin
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
- Department of Biomedical Informatics, Emory University, Atlanta, GA, United States
- Department of Pediatrics, Emory University, Atlanta, GA, United States
| |
Collapse
|
25
|
Meng Q, Shen J, Ren Y, Liu Q, Wang R, Li Q, Jiang W, Wang Q, Zhang Y, Trinidad JC, Lu X, Wang T, Li Y, Yum C, Yi Y, Yang Y, Zhao D, Harris C, Kalantry S, Chen K, Yang R, Niu H, Cao Q. EZH2 directly methylates PARP1 and regulates its activity in cancer. SCIENCE ADVANCES 2024; 10:eadl2804. [PMID: 39602541 PMCID: PMC11601213 DOI: 10.1126/sciadv.adl2804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 10/24/2024] [Indexed: 11/29/2024]
Abstract
DNA repair dysregulation is a key driver of cancer development. Understanding the molecular mechanisms underlying DNA repair dysregulation in cancer cells is crucial for cancer development and therapies. Here, we report that enhancer of zeste homolog 2 (EZH2) directly methylates poly(adenosine diphosphate-ribose) polymerase-1 (PARP-1), an essential enzyme involved in DNA repair, and regulates its activity. Functionally, EZH2-catalyzed methylation represses PARP1 catalytic activity, down-regulates the recruitment of x-ray repair cross-complementing group-1 to DNA lesions and its associated DNA damage repair; on the other hand, it protects the cells from nicotinamide adenine dinucleotide overconsumption upon DNA damage formation. Meanwhile, EZH2-mediated methylation regulates PARP1 transcriptional and oncogenic activity, at least in part, through impairing PARP1-E2F1 interaction and E2F1 transcription factor activity. EZH2 and PARP1 inhibitors synergistically suppress prostate cancer growth. Collectively, our findings uncover an insight of EZH2 functions in fine-tuning PARP1 activity during DNA damage repair and cancer progression, which provides a rationale for combinational targeting EZH2 and PARP1 in cancer.
Collapse
Affiliation(s)
- Qingshu Meng
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Jiangchuan Shen
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405, USA
| | - Yanan Ren
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Qi Liu
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rui Wang
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Qiaqia Li
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Weihua Jiang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Quan Wang
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405, USA
| | - Yixiang Zhang
- Department of Chemistry, Biological Mass Spectrometry Facility, Indiana University, Bloomington, IN 47405, USA
- Research Technology Branch, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT 59840, USA
| | - Jonathan C. Trinidad
- Department of Chemistry, Biological Mass Spectrometry Facility, Indiana University, Bloomington, IN 47405, USA
| | - Xiaotong Lu
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Tingyou Wang
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yanqiang Li
- Basic and Translational Research Division, Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Chaehyun Yum
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yang Yi
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Yongyong Yang
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Dongyu Zhao
- Basic and Translational Research Division, Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Clair Harris
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sundeep Kalantry
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kaifu Chen
- Basic and Translational Research Division, Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Rendong Yang
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hengyao Niu
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405, USA
| | - Qi Cao
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
26
|
Sacco JL, Vaneman ZT, Self A, Sumner E, Kibinda S, Sankhe CS, Gomez EW. Chemomechanical regulation of EZH2 localization controls epithelial-mesenchymal transition. J Cell Sci 2024; 137:jcs262190. [PMID: 39450433 DOI: 10.1242/jcs.262190] [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: 04/08/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
The methyltransferase enhancer of zeste homolog 2 (EZH2) regulates gene expression, and aberrant EZH2 expression and signaling can drive fibrosis and cancer. However, it is not clear how chemical and mechanical signals are integrated to regulate EZH2 and gene expression. We show that culture of cells on stiff matrices in concert with transforming growth factor (TGF)-β1 promotes nuclear localization of EZH2 and an increase in the levels of the corresponding histone modification, H3K27me3, thereby regulating gene expression. EZH2 activity and expression are required for TGFβ1- and stiffness-induced increases in H3K27me3 levels as well as for morphological and gene expression changes associated with epithelial-mesenchymal transition (EMT). Inhibition of Rho associated kinase (ROCK) proteins or myosin II signaling attenuates TGFβ1-induced nuclear localization of EZH2 and decreases H3K27me3 levels in cells cultured on stiff substrata, suggesting that cellular contractility, in concert with a major cancer signaling regulator TGFβ1, modulates EZH2 subcellular localization. These findings provide a contractility-dependent mechanism by which matrix stiffness and TGFβ1 together mediate EZH2 signaling to promote EMT.
Collapse
Affiliation(s)
- Jessica L Sacco
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Zachary T Vaneman
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Ava Self
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Elix Sumner
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Stella Kibinda
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Chinmay S Sankhe
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Esther W Gomez
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
27
|
Ryu HJ, Lee C, Yoon SO. Epigenetic and Immune Profile Characteristics in Sinonasal Undifferentiated Carcinoma. Cancer Med 2024; 13:e70413. [PMID: 39565059 PMCID: PMC11577451 DOI: 10.1002/cam4.70413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/10/2024] [Accepted: 10/30/2024] [Indexed: 11/21/2024] Open
Abstract
INTRODUCTION Sinonasal undifferentiated carcinoma (SNUC) is a rare and highly aggressive malignancy originating in the nasal cavity and paranasal sinuses. Its pathogenesis and immune characteristics remain poorly understood. OBJECTIVES This study investigates the molecular aspects of SNUC, focusing on tumorigenesis and immunity. METHODS For this purpose, spatial transcriptome analysis was employed to compare the gene expression profiles of SNUC tumor cells with those of normal epithelial cells, as well as to compare tumor-infiltrating immune cells with immune cells from normal, tumor-free tissue areas. For validation, next-generation sequencing tests and clinical sample studies were conducted. RESULTS Spatial transcriptome analysis revealed notable upregulation of EZH2 and the histone family gene such as H3C2 (H3-clustered histone 2) in SNUC tumor cells. Additionally, gene set enrichment analysis identified significant activations in the histone deacetylase (HDAC) signaling pathway, histone acetyltransferase (HAT) pathway, polycomb repressive complex 2 (PRC2), and DNA methylation pathways. A notable decrease was observed in downregulated genes and pathways, including the mucin family of protein genes, the keratin protein gene, and the mucin glycosylation pathway. Next-generation sequencing did not reveal specific genetic mutations within these pathways, although mutations such as IDH2 R172S were noted. Clinical SNUC tissues confirmed increased immunoexpression of EZH2 and PRC2 markers. Analysis of tumor immunity revealed a characteristic immune cell signature, with a notable predominance of naïve B cells, macrophages, CD8 memory T cells, and Tregs in the SNUC microenvironment, alongside the increased expression of LAG3 in tumor-infiltrating immune cells. CONCLUSION Our study suggests epigenetic mechanisms, particularly via EZH2, play a crucial role in SNUC carcinogenesis. Furthermore, distinctive immune cell profiles in SNUC point to potential immune-related characteristics of this malignancy.
Collapse
Affiliation(s)
- Hyang Joo Ryu
- Department of PathologyYonsei University College of Medicine, Severance HospitalSeoulKorea
| | - Chung Lee
- Department of PathologyYonsei University College of Medicine, Severance HospitalSeoulKorea
| | - Sun Och Yoon
- Department of PathologyYonsei University College of Medicine, Severance HospitalSeoulKorea
| |
Collapse
|
28
|
Schade AE, Perurena N, Yang Y, Rodriguez CL, Krishnan A, Gardner A, Loi P, Xu Y, Nguyen VTM, Mastellone GM, Pilla NF, Watanabe M, Ota K, Davis RA, Mattioli K, Xiang D, Zoeller JJ, Lin JR, Morganti S, Garrido-Castro AC, Tolaney SM, Li Z, Barbie DA, Sorger PK, Helin K, Santagata S, Knott SRV, Cichowski K. AKT and EZH2 inhibitors kill TNBCs by hijacking mechanisms of involution. Nature 2024; 635:755-763. [PMID: 39385030 PMCID: PMC11578877 DOI: 10.1038/s41586-024-08031-6] [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: 06/03/2022] [Accepted: 09/06/2024] [Indexed: 10/11/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype and has the highest rate of recurrence1. The predominant standard of care for advanced TNBC is systemic chemotherapy with or without immunotherapy; however, responses are typically short lived1,2. Thus, there is an urgent need to develop more effective treatments. Components of the PI3K pathway represent plausible therapeutic targets; more than 70% of TNBCs have alterations in PIK3CA, AKT1 or PTEN3-6. However, in contrast to hormone-receptor-positive tumours, it is still unclear whether or how triple-negative disease will respond to PI3K pathway inhibitors7. Here we describe a promising AKT-inhibitor-based therapeutic combination for TNBC. Specifically, we show that AKT inhibitors synergize with agents that suppress the histone methyltransferase EZH2 and promote robust tumour regression in multiple TNBC models in vivo. AKT and EZH2 inhibitors exert these effects by first cooperatively driving basal-like TNBC cells into a more differentiated, luminal-like state, which cannot be effectively induced by either agent alone. Once TNBCs are differentiated, these agents kill them by hijacking signals that normally drive mammary gland involution. Using a machine learning approach, we developed a classifier that can be used to predict sensitivity. Together, these findings identify a promising therapeutic strategy for this highly aggressive tumour type and illustrate how deregulated epigenetic enzymes can insulate tumours from oncogenic vulnerabilities. These studies also reveal how developmental tissue-specific cell death pathways may be co-opted for therapeutic benefit.
Collapse
Affiliation(s)
- Amy E Schade
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Naiara Perurena
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Yoona Yang
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Carrie L Rodriguez
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Anjana Krishnan
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Alycia Gardner
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Patrick Loi
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Yilin Xu
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Van T M Nguyen
- Division of Cancer Biology, Institute of Cancer Research, London, UK
| | - G M Mastellone
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Natalie F Pilla
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Marina Watanabe
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Keiichi Ota
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rachel A Davis
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kaia Mattioli
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Dongxi Xiang
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jason J Zoeller
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Jia-Ren Lin
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stefania Morganti
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ana C Garrido-Castro
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sara M Tolaney
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Zhe Li
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - David A Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Peter K Sorger
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Kristian Helin
- Division of Cancer Biology, Institute of Cancer Research, London, UK
| | - Sandro Santagata
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Simon R V Knott
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Karen Cichowski
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Feng S, Marhon SA, Sokolowski DJ, D'Costa A, Soares F, Mehdipour P, Ishak C, Loo Yau H, Ettayebi I, Patel PS, Chen R, Liu J, Zuzarte PC, Ho KC, Ho B, Ning S, Huang A, Arrowsmith CH, Wilson MD, Simpson JT, De Carvalho DD. Inhibiting EZH2 targets atypical teratoid rhabdoid tumor by triggering viral mimicry via both RNA and DNA sensing pathways. Nat Commun 2024; 15:9321. [PMID: 39472584 PMCID: PMC11522499 DOI: 10.1038/s41467-024-53515-8] [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: 03/01/2023] [Accepted: 10/12/2024] [Indexed: 11/02/2024] Open
Abstract
Inactivating mutations in SMARCB1 confer an oncogenic dependency on EZH2 in atypical teratoid rhabdoid tumors (ATRTs), but the underlying mechanism has not been fully elucidated. We found that the sensitivity of ATRTs to EZH2 inhibition (EZH2i) is associated with the viral mimicry response. Unlike other epigenetic therapies targeting transcriptional repressors, EZH2i-induced viral mimicry is not triggered by cryptic transcription of endogenous retroelements, but rather mediated by increased expression of genes enriched for intronic inverted-repeat Alu (IR-Alu) elements. Interestingly, interferon-stimulated genes (ISGs) are highly enriched for dsRNA-forming intronic IR-Alu elements, suggesting a feedforward loop whereby these activated ISGs may reinforce dsRNA formation and viral mimicry. EZH2i also upregulates the expression of full-length LINE-1s, leading to genomic instability and cGAS/STING signaling in a process dependent on reverse transcriptase activity. Co-depletion of dsRNA sensing and cytoplasmic DNA sensing completely rescues the viral mimicry response to EZH2i in SMARCB1-deficient tumors.
Collapse
Affiliation(s)
- Shengrui Feng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- The First Affiliated Hospital of University of South China, Hengyang, Hunan, China.
| | - Sajid A Marhon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Dustin J Sokolowski
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
| | - Alister D'Costa
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Fraser Soares
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Parinaz Mehdipour
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Charles Ishak
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Helen Loo Yau
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Ilias Ettayebi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Parasvi S Patel
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Raymond Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Jiming Liu
- The Cardiac Development and Early Intervention Unit, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | | | - King Ching Ho
- Division of Hematology/Oncology, Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, ON, Canada
| | - Ben Ho
- Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Shiyao Ning
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Annie Huang
- The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
- Division of Hematology/Oncology, Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, ON, Canada
- Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Cheryl H Arrowsmith
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada
| | - Michael D Wilson
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
| | - Jared T Simpson
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Daniel D De Carvalho
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
30
|
Jeong SJ, Oh JH, Cho JY. ALYREF enhances breast cancer progression by regulating EZH2. Heliyon 2024; 10:e37749. [PMID: 39386827 PMCID: PMC11462240 DOI: 10.1016/j.heliyon.2024.e37749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/08/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Breast cancer is one of the most common malignant tumors in women worldwide. Similarly, Canine mammary tumors (CMTs) are mostly diagnosed as spontaneous diseases in female dogs. Many studies have suggested that CMTs serve as good models for human breast cancer. However, comparative approaches to histone modifications are still lacking. This study aimed to compare the canine mammary tumor Histone H3 lysine 4 trimethylation (H3K4me3) landscape with that in human breast cancer. Our H3K4me3 ChIP-seq data from CMTs revealed a significant enrichment of H3K4me3 in the ALYREF gene promoter in tumor tissues compared to normal tissues. Furthermore, our study and publicly available RNA-sequencing data revealed that ALYREF expression was elevated in malignant tissues and breast cancer cell lines, and its upregulation was associated with poor prognosis in humans. Depletion of ALYREF resulted in changes in cellular phenotypes, including increased proliferation and colony formation, as well as decreased apoptosis. ALYREF increased cell viability and anchorage-independent growth while decreasing apoptosis by regulating the mRNA expression and protein levels of enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2), which promotes hormone receptor-positive breast cancer and CMTs via epigenetic modifications. This suggests that ALYREF may function as a contributing factor to malignant transformation in both CMT and human breast cancer.
Collapse
Affiliation(s)
- Su-Jin Jeong
- Department of Biochemistry, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ji Hoon Oh
- Department of Biochemistry, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Je-Yoel Cho
- Department of Biochemistry, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
31
|
Jiang W, Liu L, Wang M, Li X, Zhou T, Hou X, Qiao L, Chen C, Zuo D, Liu J, Ren L. KLF15 suppresses stemness of pancreatic cancer by decreasing USP21-mediated Nanog stability. Cell Mol Life Sci 2024; 81:417. [PMID: 39367978 PMCID: PMC11455850 DOI: 10.1007/s00018-024-05442-6] [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] [Revised: 06/08/2024] [Accepted: 08/22/2024] [Indexed: 10/07/2024]
Abstract
The existence of cancer stem cells (CSCs) in pancreatic ductal adenocarcinoma (PDAC) is considered to be the key factor for metastasis and chemoresistance. Thus, novel therapeutic strategies for eradicating CSCs are urgently needed. Here we aimed to explore the role of KLF15 in stemness and the feasibility of using KLF15 to inhibit CSCs and improve chemotherapy sensitivity in PDAC. In this study, we report that KLF15 is negatively associated with poor survival and advanced pathological staging of PDAC. Moreover, tumorous KLF15 suppresses the stemness of PDAC by promoting the degradation of Nanog, and KLF15 directly interacts with Nanog, inhibiting interaction between Nanog with USP21. We also demonstrate that the KLF15/Nanog complex inhibit the stemness in vivo and in PDX cells. Tazemetostat suppresses stemness and sensitizes PDAC cells to gemcitabine by promoting KLF15 expression in PDAC. In summary, the findings of our study confirm the value of KLF15 level in diagnosis and prognosis of PDAC, it is the first time to explore the inhibition role of KLF15 in stemness of PDAC and the regulation mechanism of Nanog, contributing to provide a new therapeutic strategy that using Tazemetostat sensitizes PDAC cells to gemcitabine by promoting KLF15 expression for PDAC.
Collapse
Affiliation(s)
- Wenna Jiang
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Lin Liu
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Meng Wang
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Xueyang Li
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Tianxing Zhou
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Xupeng Hou
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Lu Qiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences of Tianjin Medical University, Tianjin, China
| | - Chong Chen
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Duo Zuo
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Jing Liu
- Department of Breast Oncoplastic Surgery, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Li Ren
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China.
| |
Collapse
|
32
|
Gonzalez ME, Brophy B, Eido A, Leonetti AE, Djomehri SI, Augimeri G, Carruthers NJ, Cavalcante RG, Giordano F, Andò S, Nesvizhskii AI, Fearon ER, Kleer CG. CCN6 Suppresses Metaplastic Breast Carcinoma by Antagonizing Wnt/β-Catenin Signaling to Inhibit EZH2-Driven EMT. Cancer Res 2024; 84:3235-3249. [PMID: 39024552 PMCID: PMC11444886 DOI: 10.1158/0008-5472.can-23-4054] [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: 12/21/2023] [Revised: 05/28/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Metaplastic breast carcinomas (mBrCA) are a highly aggressive subtype of triple-negative breast cancer with histologic evidence of epithelial-to-mesenchymal transition and aberrant differentiation. Inactivation of the tumor suppressor gene cellular communication network factor 6 (CCN6; also known as Wnt1-induced secreted protein 3) is a feature of mBrCAs, and mice with conditional inactivation of Ccn6 in mammary epithelium (Ccn6-KO) develop spindle mBrCAs with epithelial-to-mesenchymal transition. Elucidation of the precise mechanistic details of how CCN6 acts as a tumor suppressor in mBrCA could help identify improved treatment strategies. In this study, we showed that CCN6 interacts with the Wnt receptor FZD8 and coreceptor LRP6 on mBrCA cells to antagonize Wnt-induced activation of β-catenin/TCF-mediated transcription. The histone methyltransferase EZH2 was identified as a β-catenin/TCF transcriptional target in Ccn6-KO mBrCA cells. Inhibiting Wnt/β-catenin/TCF signaling in Ccn6-KO mBrCA cells led to reduced EZH2 expression, decreased histone H3 lysine 27 trimethylation, and deregulation of specific target genes. Pharmacologic inhibition of EZH2 reduced growth and metastasis of Ccn6-KO mBrCA mammary tumors in vivo. Low CCN6 is significantly associated with activated β-catenin and high EZH2 in human spindle mBrCAs compared with other subtypes. Collectively, these findings establish CCN6 as a key negative regulator of a β-catenin/TCF/EZH2 axis and highlight the inhibition of β-catenin or EZH2 as a potential therapeutic approach for patients with spindle mBrCAs. Significance: CCN6 deficiency drives metaplastic breast carcinoma growth and metastasis by increasing Wnt/β-catenin activation to upregulate EZH2, identifying EZH2 inhibition as a mechanistically guided treatment strategy for this deadly form of breast cancer.
Collapse
Affiliation(s)
- Maria E Gonzalez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Bryce Brophy
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Ahmad Eido
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Adele E Leonetti
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Sabra I Djomehri
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Giuseppina Augimeri
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | | | | | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Eric R Fearon
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan
| | - Celina G Kleer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
33
|
Sacco JL, Gomez EW. Epithelial-Mesenchymal Plasticity and Epigenetic Heterogeneity in Cancer. Cancers (Basel) 2024; 16:3289. [PMID: 39409910 PMCID: PMC11475326 DOI: 10.3390/cancers16193289] [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: 08/06/2024] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
The tumor microenvironment comprises various cell types and experiences dynamic alterations in physical and mechanical properties as cancer progresses. Intratumoral heterogeneity is associated with poor prognosis and poses therapeutic challenges, and recent studies have begun to identify the cellular mechanisms that contribute to phenotypic diversity within tumors. This review will describe epithelial-mesenchymal (E/M) plasticity and its contribution to phenotypic heterogeneity in tumors as well as how epigenetic factors, such as histone modifications, histone modifying enzymes, DNA methylation, and chromatin remodeling, regulate and maintain E/M phenotypes. This review will also report how mechanical properties vary across tumors and regulate epigenetic modifications and E/M plasticity. Finally, it highlights how intratumoral heterogeneity impacts therapeutic efficacy and provides potential therapeutic targets to improve cancer treatments.
Collapse
Affiliation(s)
- Jessica L. Sacco
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA;
| | - Esther W. Gomez
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA 16802, USA;
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
34
|
Shao M, Tian M, Chen K, Jiang H, Zhang S, Li Z, Shen Y, Chen F, Shen B, Cao C, Gu N. Leveraging Random Effects in Cistrome-Wide Association Studies for Decoding the Genetic Determinants of Prostate Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400815. [PMID: 39099406 PMCID: PMC11423091 DOI: 10.1002/advs.202400815] [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: 01/23/2024] [Revised: 07/09/2024] [Indexed: 08/06/2024]
Abstract
Cistrome-wide association studies (CWAS) are pivotal for identifying genetic determinants of diseases by correlating genetically regulated cistrome states with phenotypes. Traditional CWAS typically develops a model based on cistrome and genotype data to associate predicted cistrome states with phenotypes. The random effect cistrome-wide association study (RECWAS), reevaluates the necessity of cistrome state prediction in CWAS. RECWAS utilizes either a linear model or marginal effect for initial feature selection, followed by kernel-based feature aggregation for association testing is introduced. Through simulations and analysis of prostate cancer data, a thorough evaluation of CWAS and RECWAS is conducted. The results suggest that RECWAS offers improved power compared to traditional CWAS, identifying additional genomic regions associated with prostate cancer. CWAS identified 102 significant regions, while RECWAS found 50 additional significant regions compared to CWAS, many of which are validated. Validation encompassed a range of biological evidence, including risk signals from the GWAS catalog, susceptibility genes from the DisGeNET database, and enhancer-domain scores. RECWAS consistently demonstrated improved performance over traditional CWAS in identifying genomic regions associated with prostate cancer. These findings demonstrate the benefits of incorporating kernel methods into CWAS and provide new insights for genetic discovery in complex diseases.
Collapse
Affiliation(s)
- Mengting Shao
- Key Laboratory for Bio‐Electromagnetic Environment and Advanced Medical TheranosticsSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjing211166P. R. China
| | - Min Tian
- Key Laboratory for Bio‐Electromagnetic Environment and Advanced Medical TheranosticsSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjing211166P. R. China
| | - Kaiyang Chen
- Key Laboratory for Bio‐Electromagnetic Environment and Advanced Medical TheranosticsSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjing211166P. R. China
| | - Hangjin Jiang
- Center for Data ScienceZhejiang UniversityHangzhou310058P. R. China
| | - Shuting Zhang
- Key Laboratory for Bio‐Electromagnetic Environment and Advanced Medical TheranosticsSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjing211166P. R. China
| | - Zhenghui Li
- Key Laboratory for Bio‐Electromagnetic Environment and Advanced Medical TheranosticsSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjing211166P. R. China
| | - Yan Shen
- Key Laboratory for Bio‐Electromagnetic Environment and Advanced Medical TheranosticsSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjing211166P. R. China
| | - Feng Chen
- Key Laboratory for Bio‐Electromagnetic Environment and Advanced Medical TheranosticsSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjing211166P. R. China
| | - Baixin Shen
- Department of UrologyThe Second Affiliated Hospital of Nanjing Medical UniversityNanjing210011P. R. China
| | - Chen Cao
- Key Laboratory for Bio‐Electromagnetic Environment and Advanced Medical TheranosticsSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjing211166P. R. China
- Department of UrologyThe Second Affiliated Hospital of Nanjing Medical UniversityNanjing210011P. R. China
| | - Ning Gu
- Key Laboratory for Bio‐Electromagnetic Environment and Advanced Medical TheranosticsSchool of Biomedical Engineering and InformaticsNanjing Medical UniversityNanjing211166P. R. China
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering MedicineInstitute of Clinical MedicineNanjing Drum Tower HospitalMedical SchoolNanjing UniversityNanjing210093P. R. China
| |
Collapse
|
35
|
Akhlaghipour I, Moghbeli M. MicroRNA-98 as a novel diagnostic marker and therapeutic target in cancer patients. Discov Oncol 2024; 15:385. [PMID: 39210158 PMCID: PMC11362465 DOI: 10.1007/s12672-024-01270-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
The progress of cancer treatment methods in the last decade has significantly reduced mortality rate among these patients. Nevertheless, cancer is still recognized as one of the main causes of human deaths. One of the main reasons for the high death rate in cancer patients is the late diagnosis in the advanced tumor stages. Therefore, it is necessary to investigate the molecular biology of tumor progressions in order to introduce early diagnostic markers. MicroRNAs (miRNAs) have an important role in regulating cellular processes associated with tumor progression. Due to the high stability of miRNAs in body fluids, they are widely used as non-invasive markers in the early tumor diagnosis. Since, deregulation of miR-98 has been reported in a wide range of cancers, we investigated the molecular mechanisms of miR-98 during tumor progression. It has been reported that miR-98 mainly inhibits the tumor growth by the modulation of transcription factors and signaling pathways. Therefore, miR-98 can be introduced as a tumor marker and therapeutic target among cancer patients.
Collapse
Affiliation(s)
- Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
36
|
Gallardo A, López-Onieva L, Belmonte-Reche E, Fernández-Rengel I, Serrano-Prados A, Molina A, Sánchez-Pozo A, Landeira D. EZH2 represses mesenchymal genes and upholds the epithelial state of breast carcinoma cells. Cell Death Dis 2024; 15:609. [PMID: 39174513 PMCID: PMC11341823 DOI: 10.1038/s41419-024-07011-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
Emerging studies support that the polycomb repressive complex 2 (PRC2) regulates phenotypic changes of carcinoma cells by modulating their shifts among metastable states within the epithelial and mesenchymal spectrum. This new role of PRC2 in cancer has been recently proposed to stem from the ability of its catalytic subunit EZH2 to bind and modulate the transcription of mesenchymal genes during epithelial-mesenchymal transition (EMT) in lung cancer cells. Here, we asked whether this mechanism is conserved in other types of carcinomas. By combining TGF-β-mediated reversible induction of epithelial to mesenchymal transition and inhibition of EZH2 methyltransferase activity, we demonstrate that EZH2 represses a large set of mesenchymal genes and favours the residence of breast cancer cells towards the more epithelial spectrum during EMT. In agreement, analysis of human patient samples supports that EZH2 is required to efficiently repress mesenchymal genes in breast cancer tumours. Our results indicate that PRC2 operates through similar mechanisms in breast and lung cancer cells. We propose that PRC2-mediated direct transcriptional modulation of the mesenchymal gene expression programme is a conserved molecular mechanism underlying cell dissemination across human carcinomas.
Collapse
Affiliation(s)
- Amador Gallardo
- Centre for Genomics and Oncological Research (GENYO), Avenida de la Ilustración 114, 18016, Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Hospital Virgen de las Nieves, Granada, Spain
| | - Lourdes López-Onieva
- Centre for Genomics and Oncological Research (GENYO), Avenida de la Ilustración 114, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Hospital Virgen de las Nieves, Granada, Spain
- Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Efres Belmonte-Reche
- Centre for Genomics and Oncological Research (GENYO), Avenida de la Ilustración 114, 18016, Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Hospital Virgen de las Nieves, Granada, Spain
| | - Iván Fernández-Rengel
- Centre for Genomics and Oncological Research (GENYO), Avenida de la Ilustración 114, 18016, Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Hospital Virgen de las Nieves, Granada, Spain
| | - Andrea Serrano-Prados
- Centre for Genomics and Oncological Research (GENYO), Avenida de la Ilustración 114, 18016, Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Hospital Virgen de las Nieves, Granada, Spain
| | - Aldara Molina
- Centre for Genomics and Oncological Research (GENYO), Avenida de la Ilustración 114, 18016, Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Hospital Virgen de las Nieves, Granada, Spain
| | - Antonio Sánchez-Pozo
- Centre for Genomics and Oncological Research (GENYO), Avenida de la Ilustración 114, 18016, Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Hospital Virgen de las Nieves, Granada, Spain
| | - David Landeira
- Centre for Genomics and Oncological Research (GENYO), Avenida de la Ilustración 114, 18016, Granada, Spain.
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain.
- Instituto de Investigación Biosanitaria ibs.GRANADA, Hospital Virgen de las Nieves, Granada, Spain.
| |
Collapse
|
37
|
Kiri S, Ryba T. Cancer, metastasis, and the epigenome. Mol Cancer 2024; 23:154. [PMID: 39095874 PMCID: PMC11295362 DOI: 10.1186/s12943-024-02069-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Cancer is the second leading cause of death worldwide and disease burden is expected to increase globally throughout the next several decades, with the majority of cancer-related deaths occurring in metastatic disease. Cancers exhibit known hallmarks that endow them with increased survival and proliferative capacities, frequently as a result of de-stabilizing mutations. However, the genomic features that resolve metastatic clones from primary tumors are not yet well-characterized, as no mutational landscape has been identified as predictive of metastasis. Further, many cancers exhibit no known mutation signature. This suggests a larger role for non-mutational genome re-organization in promoting cancer evolution and dissemination. In this review, we highlight current critical needs for understanding cell state transitions and clonal selection advantages for metastatic cancer cells. We examine links between epigenetic states, genome structure, and misregulation of tumor suppressors and oncogenes, and discuss how recent technologies for understanding domain-scale regulation have been leveraged for a more complete picture of oncogenic and metastatic potential.
Collapse
Affiliation(s)
- Saurav Kiri
- College of Medicine, University of Central Florida, 6850 Lake Nona Blvd., Orlando, 32827, Florida, USA.
| | - Tyrone Ryba
- Department of Natural Sciences, New College of Florida, 5800 Bay Shore Rd., Sarasota, 34243, Florida, USA.
| |
Collapse
|
38
|
Huang Z, Tang Y, Zhang J, Huang J, Cheng R, Guo Y, Kleer CG, Wang Y, Xue L. Hypoxia makes EZH2 inhibitor not easy-advances of crosstalk between HIF and EZH2. LIFE METABOLISM 2024; 3:loae017. [PMID: 38911968 PMCID: PMC11192520 DOI: 10.1093/lifemeta/loae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/28/2024] [Accepted: 05/03/2024] [Indexed: 06/25/2024]
Abstract
Histone methylation plays a crucial role in tumorigenesis. Enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase that regulates chromatin structure and gene expression. EZH2 inhibitors (EZH2is) have been shown to be effective in treating hematologic malignancies, while their effectiveness in solid tumors remains limited. One of the major challenges in the treatment of solid tumors is their hypoxic tumor microenvironment. Hypoxia-inducible factor 1-alpha (HIF-1α) is a key hypoxia responder that interacts with EZH2 to promote tumor progression. Here we discuss the implications of the relationship between EZH2 and hypoxia for expanding the application of EZH2is in solid tumors.
Collapse
Affiliation(s)
- Zhanya Huang
- Cancer Center of Peking University Third Hospital, Beijing 100191, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Yuanjun Tang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Jianlin Zhang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Jiaqi Huang
- Cancer Center of Peking University Third Hospital, Beijing 100191, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Rui Cheng
- Cancer Center of Peking University Third Hospital, Beijing 100191, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Yunyun Guo
- Cancer Center of Peking University Third Hospital, Beijing 100191, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Celina G Kleer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Yuqing Wang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Lixiang Xue
- Cancer Center of Peking University Third Hospital, Beijing 100191, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
39
|
Jiang L, Huang L, Jiang W. H3K27me3-mediated epigenetic regulation in pluripotency maintenance and lineage differentiation. CELL INSIGHT 2024; 3:100180. [PMID: 39072246 PMCID: PMC11278802 DOI: 10.1016/j.cellin.2024.100180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024]
Abstract
Cell fate determination is an intricate process which is orchestrated by multiple regulatory layers including signal pathways, transcriptional factors, epigenetic modifications, and metabolic rewiring. Among the sophisticated epigenetic modulations, the repressive mark H3K27me3, deposited by PRC2 (polycomb repressive complex 2) and removed by demethylase KDM6, plays a pivotal role in mediating the cellular identity transition through its dynamic and precise alterations. Herein, we overview and discuss how H3K27me3 and its modifiers regulate pluripotency maintenance and early lineage differentiation. We primarily highlight the following four aspects: 1) the two subcomplexes PRC2.1 and PRC2.2 and the distribution of genomic H3K27 methylation; 2) PRC2 as a critical regulator in pluripotency maintenance and exit; 3) the emerging role of the eraser KDM6 in early differentiation; 4) newly identified additional factors influencing H3K27me3. We present a comprehensive insight into the molecular principles of the dynamic regulation of H3K27me3, as well as how this epigenetic mark participates in pluripotent stem cell-centered cell fate determination.
Collapse
Affiliation(s)
- Liwen Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Linfeng Huang
- Wang-Cai Biochemistry Lab, Division of Natural and Applied Sciences, Duke Kunshan University, Kunshan, Jiangsu, China
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| |
Collapse
|
40
|
Chida K, Kanazawa H, Kinoshita H, Roy AM, Hakamada K, Takabe K. The role of lidocaine in cancer progression and patient survival. Pharmacol Ther 2024; 259:108654. [PMID: 38701900 PMCID: PMC11162934 DOI: 10.1016/j.pharmthera.2024.108654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/17/2024] [Accepted: 04/30/2024] [Indexed: 05/05/2024]
Abstract
Since its development in 1943, lidocaine has been one of the most commonly used local anesthesia agents for surgical procedures. Lidocaine alters neuronal signal transmission by prolonging the inactivation of fast voltage-gated sodium channels in the cell membrane of neurons, which are responsible for action potential propagation. Recently, it has attracted attention due to emerging evidence suggesting its potential antitumor properties, particularly in the in vitro setting. Further, local administration of lidocaine around the tumor immediately prior to surgical removal has been shown to improve overall survival in breast cancer patients. However, the exact mechanisms driving these antitumor effects remain largely unclear. In this article, we will review the existing literature on the mechanism of lidocaine as a local anesthetic, its effects on the cancer cells and the tumor microenvironment, involved pathways, and cancer progression. Additionally, we will explore recent reports highlighting its impact on clinical outcomes in cancer patients. Taken together, there remains significant ambiguity surrounding lidocaine's functions and roles in cancer biology, particularly in perioperative setting.
Collapse
Affiliation(s)
- Kohei Chida
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan.
| | - Hirofumi Kanazawa
- The University of Texas Health Science Center at Tyler School of Medicine, TX, USA.
| | - Hirotaka Kinoshita
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan.
| | - Arya Mariam Roy
- Department of Hematology and Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Kenichi Hakamada
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan.
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan; Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY 14263, USA; Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan; Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan; Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
41
|
Xu H, Zhang J, Zhuang J, Chen Y, Chen L, Wang J, Cao R, Liu F, Wang K, Zhang X, Wang L, Chen G. 2,2- dimethylbenzopyran derivatives containing pyridone structural fragments as selective dual-targeting inhibitors of HIF-1α and EZH2 for the treatment of lung cancer. Bioorg Chem 2024; 147:107419. [PMID: 38703440 DOI: 10.1016/j.bioorg.2024.107419] [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: 01/16/2024] [Revised: 04/16/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024]
Abstract
We formerly reported that EZH2 inhibitors sensitized HIF-1 inhibitor-resistant cells and inhibited HIF-1α to promote SUZ12 transcription, leading to enhanced EZH2 enzyme activity and elevated H3K27me3 levels, and conversely, inhibition of EZH2 promoted HIF-1α transcription. HIF-1α and EZH2 interacted to form a negative feedback loop that reinforced each other's activity. In this paper, a series of 2,2- dimethylbenzopyran derivatives containing pyridone structural fragments were designed and synthesized with DYB-03, a HIF-1α inhibitor previously reported by our group, and Tazemetostat, an EZH2 inhibitor approved by FDA, as lead compounds. Among these compounds, D-01 had significant inhibitory activities on HIF-1α and EZH2. In vitro experiments showed that D-01 significantly inhibited the migration of A549 cells, clone, invasion and angiogenesis. Moreover, D-01 had good pharmacokinetic profiles. All the results about compound D-01 could lay a foundation for the research and development of HIF-1α and EZH2 dual-targeting compounds.
Collapse
Affiliation(s)
- Huashen Xu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jie Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China; Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, PR China
| | - Junning Zhuang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yuanguang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Lu Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jianmin Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China; Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, PR China
| | - 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
| | - Fuqin Liu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Kaibo Wang
- 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
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China; Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, 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
|
42
|
Vlodavsky I, Hilwi M, Kayal Y, Soboh S, Ilan N. Impact of heparanase-2 (Hpa2) on cancer and inflammation: Advances and paradigms. FASEB J 2024; 38:e23670. [PMID: 38747803 DOI: 10.1096/fj.202400286r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/09/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
HPSE2, the gene-encoding heparanase 2 (Hpa2), is mutated in urofacial syndrome (UFS), a rare autosomal recessive congenital disease attributed to peripheral neuropathy. Hpa2 lacks intrinsic heparan sulfate (HS)-degrading activity, the hallmark of heparanase (Hpa1), yet it exhibits a high affinity toward HS, thereby inhibiting Hpa1 enzymatic activity. Hpa2 regulates selected genes that promote normal differentiation, tissue homeostasis, and endoplasmic reticulum (ER) stress, resulting in antitumor, antiangiogenic, and anti-inflammatory effects. Importantly, stress conditions induce the expression of Hpa2, thus establishing a feedback loop, where Hpa2 enhances ER stress which, in turn, induces Hpa2 expression. In most cases, cancer patients who retain high levels of Hpa2 survive longer than patients bearing Hpa2-low tumors. Experimentally, overexpression of Hpa2 attenuates the growth of tumor xenografts, whereas Hpa2 gene silencing results in aggressive tumors. Studies applying conditional Hpa2 knockout (cHpa2-KO) mice revealed an essential involvement of Hpa2 contributed by the host in protecting against cancer and inflammation. This was best reflected by the distorted morphology of the Hpa2-null pancreas, including massive infiltration of immune cells, acinar to adipocyte trans-differentiation, and acinar to ductal metaplasia. Moreover, orthotopic inoculation of pancreatic ductal adenocarcinoma (PDAC) cells into the pancreas of Hpa2-null vs. wild-type mice yielded tumors that were by far more aggressive. Likewise, intravenous inoculation of cancer cells into cHpa2-KO mice resulted in a dramatically increased lung colonization reflecting the involvement of Hpa2 in restricting the formation of a premetastatic niche. Elucidating Hpa2 structure-activity-relationships is expected to support the development of Hpa2-based therapies against cancer and inflammation.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Maram Hilwi
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Yasmin Kayal
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Soaad Soboh
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Neta Ilan
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
43
|
Herbein G. Cellular Transformation by Human Cytomegalovirus. Cancers (Basel) 2024; 16:1970. [PMID: 38893091 PMCID: PMC11171319 DOI: 10.3390/cancers16111970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Epstein-Barr virus (EBV), Kaposi sarcoma human virus (KSHV), human papillomavirus (HPV), hepatitis B and C viruses (HBV, HCV), human T-lymphotropic virus-1 (HTLV-1), and Merkel cell polyomavirus (MCPyV) are the seven human oncoviruses reported so far. While traditionally viewed as a benign virus causing mild symptoms in healthy individuals, human cytomegalovirus (HCMV) has been recently implicated in the pathogenesis of various cancers, spanning a wide range of tissue types and malignancies. This perspective article defines the biological criteria that characterize the oncogenic role of HCMV and based on new findings underlines a critical role for HCMV in cellular transformation and modeling the tumor microenvironment as already reported for the other human oncoviruses.
Collapse
Affiliation(s)
- Georges Herbein
- Department Pathogens & Inflammation-EPILAB EA4266, University of Franche-Comté (UFC), 25000 Besançon, France;
- Department of Virology, CHU Besançon, 25000 Besançon, France
| |
Collapse
|
44
|
Wang H, Wang L, Zhang S, Liu Q, Gao F. EZH2 G553C significantly increases the risk of brain metastasis from lung cancer due to salt bridge instability. Cancer Cell Int 2024; 24:175. [PMID: 38764053 PMCID: PMC11103815 DOI: 10.1186/s12935-024-03362-w] [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: 10/21/2023] [Accepted: 05/07/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND The incidence and mortality of lung cancer is the highest in China and the world. Brain is the most common distant metastasis site of lung cancer. Its transfer mechanism and predictive biomarkers are still unclear. EZH2 participates in the catalysis of transcriptional inhibition complex, mediates chromatin compactness, leads to the silencing of its downstream target genes, participates in the silencing of multiple tumor suppressor genes, and is related to cell proliferation, apoptosis and cycle regulation. In physiology, EZH2 has high activity in stem cells or progenitor cells, inhibits genes related to cell cycle arrest and promotes self-renewal. To detect the expression and mutation of EZH2 gene in patients with brain metastasis of lung cancer, and provide further theoretical basis for exploring the pathogenesis of brain metastasis of lung cancer and finding reliable biomarkers to predict brain metastasis of lung cancer. METHODS This study investigated susceptible genes for brain metastasis of lung cancer. The second-generation sequencing technology was applied to screen the differential genes of paired samples (brain metastasis tissues, lung cancer tissues and adjacent tissues) of lung cancer patients with brain metastasi. RESULTS It revealed that there was a significant difference in the G553C genotype of EZH2 between lung cancer brain metastasis tissues and lung cancer tissues (p = 0.045). The risk of lung cancer brain metastasis in G allele carriers was 2.124 times higher than that in C allele carriers. Immunohistochemistry showed that compared with lung cancer patients and lung cancer patients with brain metastasis, the expression level of EZH2 in lung cancer tissues of lung cancer patients was significantly higher than that in adjacent lung tissues (p < 0.0001), and higher than that in brain metastasis tissues (p = 0.0309). RNA in situ immunohybridization showed that EZH2 mRNA expression was gradually high in lung cancer adjacent tissues, lung cancer tissues and lung cancer brain metastasis tissues. CONCLUSIONS EZH2 G553C polymorphism contributes to the prediction of brain metastasis of lung cancer, in which G allele carriers are more prone to brain metastasis.
Collapse
Affiliation(s)
- Hanjun Wang
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, People's Republic of China
- Department of Pathology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ling Wang
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Sheng Zhang
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Qicai Liu
- Department of Reproductive Medicine Centre, 1st Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China.
| | - Feng Gao
- Department of Pathology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, People's Republic of China.
| |
Collapse
|
45
|
Kong L, Jin X. Dysregulation of deubiquitination in breast cancer. Gene 2024; 902:148175. [PMID: 38242375 DOI: 10.1016/j.gene.2024.148175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/04/2023] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
Breast cancer (BC) is a highly frequent malignant tumor that poses a serious threat to women's health and has different molecular subtypes, histological subtypes, and biological features, which act by activating oncogenic factors and suppressing cancer inhibitors. The ubiquitin-proteasome system (UPS) is the main process contributing to protein degradation, and deubiquitinases (DUBs) are reverse enzymes that counteract this process. There is growing evidence that dysregulation of DUBs is involved in the occurrence of BC. Herein, we review recent research findings in BC-associated DUBs, describe their nature, classification, and functions, and discuss the potential mechanisms of DUB-related dysregulation in BC. Furthermore, we present the successful treatment of malignant cancer with DUB inhibitors, as well as analyzing the status of targeting aberrant DUBs in BC.
Collapse
Affiliation(s)
- Lili Kong
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo 315211, Zhejiang, China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo 315211, Zhejiang, China.
| |
Collapse
|
46
|
Luo Y, Li L, Hu Q, Zhang Z, Liu F, Peng Y, Zou Y, Chen L. Iron overload increases the sensitivity of endometriosis stromal cells to ferroptosis via a PRC2-independent function of EZH2. Int J Biochem Cell Biol 2024; 169:106553. [PMID: 38417568 DOI: 10.1016/j.biocel.2024.106553] [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: 12/18/2023] [Revised: 02/05/2024] [Accepted: 02/21/2024] [Indexed: 03/01/2024]
Abstract
Given the high concentration of iron in the micro-environment of ovarian endometriosis, it is plausible to hypothesize that ectopic endometrial cells may be more susceptible to undergoing ferroptosis. Manipulation of ferroptosis has been explored as a potential therapeutic strategy to treat related diseases. In this study, we examined the impact on ectopic endometrial stromal cells (EESCs) of iron overload and an inducer of ferroptosis. We found that the iron concentration in the ovarian endometriosis was much higher than control samples. Treatment of cultured EESCs with ferric ammonium citrate (FAC) increase the sensitivity to undergo ferroptosis. By analyzing the RNA-seq results, it was discovered that zeste 2 polycomb repressive complex 2 subunit (EZH2) was significantly downregulated in ferroptosis induced EESCs. Moreover, overexpression of EZH2 effectively prevented the induction of ferroptosis. In addition, the activity or expression of EZH2 is directly and specifically inhibited by the methyltransferase inhibitor GSK343, which raises the sensitivity of stromal cells to ferroptosis. Taken together, our findings revealed that EZH2 act as a suppressor in the induced cell ferroptosis through a PRC2-independent methyltransferase mechanism. Therefore, blocking EZH2 expression and inducing ferroptosis may be effective treatment approaches for ovarian endometriosis.
Collapse
Affiliation(s)
- Yong Luo
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China
| | - Liping Li
- Prenatal Diagnosis Center, Jiangxi Key Laboratory of Birth Defect Prevention and Control, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China
| | - Qiwen Hu
- School of Medicine, Nanchang University, Nanchang, China
| | - Ziyu Zhang
- Department of pathology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China
| | - Faying Liu
- Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China; Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China
| | - Yongbao Peng
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China
| | - Yang Zou
- Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China; Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China
| | - Lina Chen
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
47
|
Kaur P, Shankar E, Gupta S. EZH2-mediated development of therapeutic resistance in cancer. Cancer Lett 2024; 586:216706. [PMID: 38331087 DOI: 10.1016/j.canlet.2024.216706] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/29/2024] [Accepted: 02/03/2024] [Indexed: 02/10/2024]
Abstract
Enhancer of zeste homolog 2 (EZH2) regulates gene expression and plays a definite role in cell proliferation, apoptosis, and senescence. Overexpression of EZH2 has been found in various human malignancies, including prostate, breast, and ovarian cancers, and is associated with increased metastasis and poor prognosis. EZH2 catalyzes trimethylation of lysine 27 of histone H3 (H3K27me3) as a canonical role in a PRC2-dependent manner. This mechanism silences various tumor suppressor genes through EZH2-mediated histone lysine methyltransferase activity. As a non-canonical role, EZH2 partners with other signaling molecules to undergo post-translational modification to orchestrate its function as a co-activator playing a critical role in cancer progression. Dysregulation of EZH2 has also been associated with therapeutic resistance in cancer cells. Given the role of EZH2 in promoting carcinogenesis and therapy resistance, both canonical and non-canonical EZH2 inhibitors have been used to combat multiple cancer types. Moreover, combining EZH2 inhibitors with other therapeutic modalities have shown to enhance the therapeutic efficacy and overcome potential resistance mechanisms in these cancerous cells. Therefore, targeting EZH2 through canonical and non-canonical modes appears to be a promising therapeutic strategy to enhance efficacy and overcome resistance in multiple cancers.
Collapse
Affiliation(s)
- Parminder Kaur
- Department of Urology, Case Western Reserve University, Cleveland, OH, 44016, USA; The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44016, USA.
| | - Eswar Shankar
- Department of Urology, Case Western Reserve University, Cleveland, OH, 44016, USA; Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA.
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, Cleveland, OH, 44016, USA; The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44016, USA; Department of Pharmacology, Case Western Reserve University, Cleveland, OH, 44016, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH, 44016, USA; Department of Nutrition, Case Western Reserve University, Cleveland, OH, 44016, USA; Division of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
48
|
Ozgun G, Yaras T, Akman B, Özden-Yılmaz G, Landman N, Karakülah G, van Lohuizen M, Senturk S, Erkek-Ozhan S. Retinoids and EZH2 inhibitors cooperate to orchestrate anti-oncogenic effects on bladder cancer cells. Cancer Gene Ther 2024; 31:537-551. [PMID: 38233533 DOI: 10.1038/s41417-024-00725-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 01/19/2024]
Abstract
The highly mutated nature of bladder cancers harboring mutations in chromatin regulatory genes opposing Polycomb-mediated repression highlights the importance of targeting EZH2 in bladder cancer. Furthermore, the critical role of the retinoic acid signaling pathway in the development and homeostasis of the urothelium, and the anti-oncogenic effects of retinoids are well established. Therefore, our aim is to simultaneously target EZH2 and retinoic acid signaling in bladder cancer to potentiate the therapeutic response. Here we report that this coordinated targeting strategy stimulates an anti-oncogenic profile, as reflected by inducing a synergistic reduction in cell viability that was associated with increased apoptosis and cell cycle arrest in a cooperative and orchestrated manner. This study characterized anti-oncogenic transcriptional reprogramming centered on the transcriptional regulator CHOP by stimulating the endoplasmic reticulum stress response. We further portrayed a molecular mechanism whereby EZH2 maintains H3K27me3-mediated repression of a subset of genes involved in unfolded protein responses, reflecting the molecular mechanism underlying this co-targeting strategy. These findings highlight the importance of co-targeting the EZH2 and retinoic acid pathway in bladder cancers and encourage the design of novel treatments employing retinoids coupled with EZH2 inhibitors in bladder carcinoma.
Collapse
Affiliation(s)
- Gizem Ozgun
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Tutku Yaras
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Burcu Akman
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Gülden Özden-Yılmaz
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Nick Landman
- Division of Molecular Genetics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Maarten van Lohuizen
- Division of Molecular Genetics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Serif Senturk
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | | |
Collapse
|
49
|
Zhang M, Wu K, Zhang W, Lin X, Cao Q, Zhang L, Chen K. The therapeutic potential of targeting the CHD protein family in cancer. Pharmacol Ther 2024; 256:108610. [PMID: 38367868 PMCID: PMC10942663 DOI: 10.1016/j.pharmthera.2024.108610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/06/2024] [Accepted: 02/02/2024] [Indexed: 02/19/2024]
Abstract
Accumulating evidence indicates that epigenetic events undergo deregulation in various cancer types, playing crucial roles in tumor development. Among the epigenetic factors involved in the epigenetic remodeling of chromatin, the chromodomain helicase DNA-binding protein (CHD) family frequently exhibits gain- or loss-of-function mutations in distinct cancer types. Therefore, targeting CHD remodelers holds the potential for antitumor treatment. In this review, we discuss epigenetic regulations of cancer development. We emphasize proteins in the CHD family, delving deeply into the intricate mechanisms governing their functions. Additionally, we provide an overview of current therapeutic strategies targeting CHD family members in preclinical trials. We further discuss the promising approaches that have demonstrated early signs of success in cancer treatment.
Collapse
Affiliation(s)
- Min Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Kaiyuan Wu
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Weijie Zhang
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Orthopaedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xia Lin
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Qi Cao
- Department of Urology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lili Zhang
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA; Prostate Cancer Program, Dana-Farber and Harvard Cancer Center, Harvard University, Boston, MA 02115, USA
| | - Kaifu Chen
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA; Prostate Cancer Program, Dana-Farber and Harvard Cancer Center, Harvard University, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
50
|
Ding Y, Zhou G, Hu W. Epigenetic regulation of TGF-β pathway and its role in radiation response. Int J Radiat Biol 2024; 100:834-848. [PMID: 38506660 DOI: 10.1080/09553002.2024.2327395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 02/27/2024] [Indexed: 03/21/2024]
Abstract
PURPOSE Transforming growth factor (TGF-β) plays a dual role in tumor progression as well as a pivotal role in radiation response. TGF-β-related epigenetic regulations, including DNA methylation, histone modifications (including methylation, acetylation, phosphorylation, ubiquitination), chromatin remodeling and non-coding RNA regulation, have been found to affect the occurrence and development of tumors as well as their radiation response in multiple dimensions. Due to the significance of radiotherapy in tumor treatment and the essential roles of TGF-β signaling in radiation response, it is important to better understand the role of epigenetic regulation mechanisms mediated by TGF-β signaling pathways in radiation-induced targeted and non-targeted effects. CONCLUSIONS By revealing the epigenetic mechanism related to TGF-β-mediated radiation response, summarizing the existing relevant adjuvant strategies for radiotherapy based on TGF-β signaling, and discovering potential therapeutic targets, we hope to provide a new perspective for improving clinical treatment.
Collapse
Affiliation(s)
- Yunan Ding
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Wentao Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| |
Collapse
|