1
|
Zhang BW, Huang T, Yang YF, Li MY, Shao GB. Lysine methyltransferase SETD7 in cancer: functions, molecular mechanisms and therapeutic implications. Mol Biol Rep 2025; 52:389. [PMID: 40232640 DOI: 10.1007/s11033-025-10494-3] [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: 12/04/2024] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Since its discovery as a histone methyltransferase, SETD7 has been implicated in many signaling pathways and carcinogenesis. SETD7 catalyzes the methylation of histone H3 and non-histone proteins, regulating their translation, stability and activity. SETD7 is frequently abnormally expressed and has a significant influence on cell proliferation, invasion, autophagy and immune response. As cancer is a complex disease, an outstanding concept in cancer biology is the "hallmarks of cancer". In this review, we focus on the involvement of SETD7 in the hallmarks of cancer, describing its functions and underlying mechanisms in detail. Additionally, we discuss non-coding RNAs and chemical inhibitors targeting SETD7, highlighting the potential and importance of SETD7 in cancer therapy.
Collapse
Affiliation(s)
- Bo-Wen Zhang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Ting Huang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Yi-Fan Yang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Ming-Yang Li
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Gen-Bao Shao
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
- Present Address: Jiangsu University, No.301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu province, China.
| |
Collapse
|
2
|
Zheng Z, Lin Y, Guo H, Liu Z, Jie X, Wang G, Zhou G. CHAF1B promotes the progression of lung squamous-cell carcinoma by inhibiting SETD7 expression. Front Med 2025; 19:318-328. [PMID: 39862337 DOI: 10.1007/s11684-024-1122-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 01/27/2025]
Abstract
The p60 subunit of the chromatin assembly factor-1 complex, that is, chromatin assembly factor-1 subunit B (CHAF1B), is a histone H3/H4 chaperone crucial for the transcriptional regulation of cell differentiation and self-renewal. CHAF1B is overexpressed in several cancers and may represent a potential target for cancer therapy. However, its expression and clinical significance in lung squamous-cell carcinoma (LUSC) remain unclear. In this study, we performed weighted gene correlation network analysis to analyze the Gene Expression Omnibus GSE68793 LUSC dataset and identified CHAF1B as one of the most important driver gene candidates. Immunohistochemical analysis of 126 LUSC tumor samples and 80 adjacent normal lung tissues showed the marked upregulation of CHAF1B in tumor tissues and the negative association of its expression level with patient survival outcomes. Silencing of CHAF1B suppressed LUSC proliferation in vitro and LUSC tumor growth in vivo. Furthermore, bulk RNA sequencing of CHAF1B knockdown cells indicated SET domain containing 7 (SETD7) as a significant CHAF1B target gene. In addition, CHAF1B competitively binds to the SETD7 promoter region and represses its transcription. Altogether, these results imply that CHAF1B plays a vital role in LUSC tumorigenesis and may represent a potential molecular target for this deadly disease.
Collapse
Affiliation(s)
- Zhuo Zheng
- Guizhou University Medical College, Guiyang, 550025, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yongfang Lin
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hua Guo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zheng Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiaoliang Jie
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Guizhen Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Guangbiao Zhou
- Guizhou University Medical College, Guiyang, 550025, China.
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
3
|
Li L, Zhu J, Chen Y, Li H, Han Y, Zhang L, Wang B. Interaction Between YTH Domain-Containing Family Protein 2 and SET Domain-Containing Lysine Methyltransferase 7 Suppresses Autophagy in Osteoarthritis Chondrocytes, Exacerbating Cartilage Damage. J Gene Med 2025; 27:e70005. [PMID: 39789715 DOI: 10.1002/jgm.70005] [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: 06/04/2024] [Revised: 10/31/2024] [Accepted: 11/18/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Osteoarthritis (OA) is characterized by progressive cartilage degeneration mediated by various molecular pathways, including inflammatory and autophagic processes. SET domain-containing lysine methyltransferase 7 (SETD7), a methyltransferase, has been implicated in OA pathology. This study investigates the expression pattern of SETD7 in OA and its role in promoting interleukin-1 beta (IL-1β)-induced chondrocyte injury through modulation of autophagy and inflammation. METHODS The expression of SETD7 in cartilage tissues from OA patients and healthy controls was quantified using quantitative reverse transcription PCR and Western blot analysis. Small interfering RNA targeting SETD7 (si-SETD7) was transfected into human articular chondrocytes (HACs) treated with IL-1β to examine its impact on cellular viability, apoptosis, inflammatory responses, and autophagy. Functional assays including Cell Counting Kit-8, flow cytometry, enzyme-linked immunosorbent assay, and commercial kits were employed to assess biochemical changes. Interaction between YTH N6-methyladenosine RNA binding protein 2 (YTHDF2) and SETD7 was explored using RNA immunoprecipitation and co-immunoprecipitation assays. RESULTS SETD7 was overexpressed in OA cartilage compared with controls and increased further upon IL-1β treatment. Knockdown of SETD7 in IL-1β-treated HACs improved cellular viability, decreased apoptosis, and reversed the adverse effects on lactate dehydrogenase release and inflammatory markers (tumor necrosis factor-alpha and interleukin-6) while enhancing antioxidant enzymes (catalase, malondialdehyde, and superoxide dismutase). Additionally, autophagy was restored, as evidenced by changes in the levels of autophagy related 5, Beclin1, and sequestosome 1. Interfering with autophagy using chloroquine negated the protective effects of SETD7 knockdown. Furthermore, YTHDF2 was found to stabilize SETD7 mRNA, influencing its expression and enhancing IL-1β-induced chondrocyte injury. CONCLUSION SETD7 plays a critical role in the pathogenesis of OA by modulating chondrocyte survival, apoptosis, inflammation, and autophagy. The interaction between YTHDF2 and SETD7 exacerbates chondrocyte injury under inflammatory conditions, highlighting potential therapeutic targets for OA treatment. The YTHDF2/SETD7 axis offers a novel insight into the molecular mechanisms governing cartilage degeneration in OA.
Collapse
Affiliation(s)
- Lexiang Li
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| | - Jun Zhu
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| | - Yi Chen
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| | - Haobo Li
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| | - Yaguang Han
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| | - Lei Zhang
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| | - Bo Wang
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| |
Collapse
|
4
|
Chen J, Wang T, Zhang D, Wang H, Huang Z, Yang Z, Li J, Hu T, Wang X, Li X. KDM5D histone demethylase mediates p38α inactivation via its enzymatic activity to inhibit cancer progression. Proc Natl Acad Sci U S A 2024; 121:e2402022121. [PMID: 39636854 DOI: 10.1073/pnas.2402022121] [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: 03/07/2024] [Accepted: 10/16/2024] [Indexed: 12/07/2024] Open
Abstract
The p38 MAP kinase (MAPK) signaling pathway plays pivotal roles in various cellular processes. Phosphorylation serves as a canonical way to regulate p38α activation through a phosphorylation cascade. Thus, understanding the mechanism governing p38α phosphorylation is important. The present study demonstrated that p38α undergoes methylation at K165, which promote its phosphorylation in tumor cells. Inhibition of p38α methylation impairs p38α phosphorylation, repressing tumor progression in vitro and in vivo. Mechanistically, KDM5D is a demethylase that interacts with p38α, mediating demethylation at K165 and inhibiting p38α phosphorylation. Moreover, KDM5D is expressed at low levels in non-small cell lung cancer (NSCLC), and high KDM5D expression is positively correlated with cancer survival. KDM5D markedly inhibits cell proliferation and migration via inactivating p38α, thereby slowing cancer progression in xenograft models. In summary, these findings highlight KDM5D as a demethylase of p38α at K165, elucidating a unique role for lysine demethylation in integrating cytoplasmic kinase-signaling cascades. The present results revealed the critical role of KDM5D in suppressing tumor progression, suggesting that KDM5D can serve as a potential drug target for combating hyperactive p38α-driven lung cancer.
Collapse
Affiliation(s)
- Jingying Chen
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng 475004, China
- Institute of Translational Medicine, Henan University, Kaifeng 475004, China
| | - Ting Wang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng 475004, China
| | - Dongzhe Zhang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng 475004, China
| | - Huiling Wang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng 475004, China
| | - Zhiang Huang
- The First Affiliated Hospital, Henan University, Kaifeng 475004, China
| | - Zhongxin Yang
- The First Affiliated Hospital, Henan University, Kaifeng 475004, China
| | - Jizhuo Li
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng 475004, China
| | - Tianyi Hu
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng 475004, China
| | - Xin Wang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng 475004, China
| | - Xia Li
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng 475004, China
- Institute of Translational Medicine, Henan University, Kaifeng 475004, China
| |
Collapse
|
5
|
Gadewal N, Patidar D, Natu A, Gupta S, Bastikar V. In silico screening of phytochemicals against chromatin modifier, SETD7 for remodeling of the immunosuppressive tumor microenvironment in renal cancer. Mol Divers 2024:10.1007/s11030-024-11038-w. [PMID: 39602041 DOI: 10.1007/s11030-024-11038-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: 06/04/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024]
Abstract
The tumor microenvironment and immune evasion function in a complex cellular network profoundly challenge the clinical outcome of promising therapies. Our recently published study reported that the subset of genes upregulated in ccRCC due to H3K4me1 and DNA demethylation potentially leads to an immunosuppressive environment. Thus, modulating H3K4me1 chromatin modifier SETD7 with a natural inhibitor in combination with immunotherapy might improve the immune landscape for a better therapeutic outcome. The present study was conducted via virtual screening and MD simulation using compounds from the literature, IMPPAT, and SuperNatural database. The phytochemical IMPHY002979 showed better binding affinity and lower energy than the reported R-PFI-2 and cyproheptadine inhibitors. The phytochemicals interact with the SET domain through H-bonding, as confirmed by MD simulation and molecular interaction analysis. Further, the compound was assessed using ADME parameters and free energy estimation, showing better pharmacokinetic properties. Therefore, the non-accessibility of the histone methyltransferase activity domain of SET7 with IMPHY002979 can downregulate H3K4me1 and, thereby, the expression of genes potentially responsible for immunosuppressive TME. Thus, patient stratification based on molecular markers for immunotherapy and combining epigenetic modulators with therapeutic drugs will improve the efficacy of immunotherapy in ccRCC.
Collapse
Affiliation(s)
- Nikhil Gadewal
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Cancer Research Institute, Kharghar, Navi Mumbai, MH, 410210, India
- Center for Computational Biology & Translational Research, Amity Institute of Biotechnology, Amity University, Mumbai, MH, India
| | - Diya Patidar
- Center for Computational Biology & Translational Research, Amity Institute of Biotechnology, Amity University, Mumbai, MH, India
| | - Abhiram Natu
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Cancer Research Institute, Kharghar, Navi Mumbai, MH, 410210, India
- Training School Complex, Homi Bhabha National Institute, Anushakti Nagar, Mumbai, MH, 400094, India
| | - Sanjay Gupta
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Cancer Research Institute, Kharghar, Navi Mumbai, MH, 410210, India.
- Training School Complex, Homi Bhabha National Institute, Anushakti Nagar, Mumbai, MH, 400094, India.
| | - Virupaksha Bastikar
- Center for Computational Biology & Translational Research, Amity Institute of Biotechnology, Amity University, Mumbai, MH, India.
| |
Collapse
|
6
|
Wang Z, Townley SL, Zhang S, Liu M, Li M, Labaf M, Patalano S, Venkataramani K, Siegfried KR, Macoska JA, Han D, Gao S, Risbridger GP, Taylor RA, Lawrence MG, He HH, Selth LA, Cai C. FOXA2 rewires AP-1 for transcriptional reprogramming and lineage plasticity in prostate cancer. Nat Commun 2024; 15:4914. [PMID: 38851846 PMCID: PMC11162502 DOI: 10.1038/s41467-024-49234-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 05/29/2024] [Indexed: 06/10/2024] Open
Abstract
FOXA family proteins act as pioneer factors by remodeling compact chromatin structures. FOXA1 is crucial for the chromatin binding of the androgen receptor (AR) in both normal prostate epithelial cells and the luminal subtype of prostate cancer (PCa). Recent studies have highlighted the emergence of FOXA2 as an adaptive response to AR signaling inhibition treatments. However, the role of the FOXA1 to FOXA2 transition in regulating cancer lineage plasticity remains unclear. Our study demonstrates that FOXA2 binds to distinct classes of developmental enhancers in multiple AR-independent PCa subtypes, with its binding depending on LSD1. Moreover, we reveal that FOXA2 collaborates with JUN at chromatin and promotes transcriptional reprogramming of AP-1 in lineage-plastic cancer cells, thereby facilitating cell state transitions to multiple lineages. Overall, our findings underscore the pivotal role of FOXA2 as a pan-plasticity driver that rewires AP-1 to induce the differential transcriptional reprogramming necessary for cancer cell lineage plasticity.
Collapse
Affiliation(s)
- Zifeng Wang
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, 02125, USA
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA
- Yale Stem Cell Center, Department of Cell Biology and Department of Genetics, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Scott L Townley
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Bedford Park, SA, 5042, Australia
- Freemasons Centre for Male Health and Wellbeing, Flinders University, Bedford Park, SA, 5042, Australia
| | - Songqi Zhang
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, 02125, USA
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Mingyu Liu
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, 02125, USA
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Muqing Li
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, 02125, USA
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Maryam Labaf
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, 02125, USA
- Department of Mathematics, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Susan Patalano
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, 02125, USA
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Kavita Venkataramani
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Kellee R Siegfried
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Jill A Macoska
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, 02125, USA
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Dong Han
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, 02125, USA
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA
| | - Shuai Gao
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, 10595, USA
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York, 10595, USA
| | - Gail P Risbridger
- Melbourne Urological Research Alliance, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, VIC, 3800, Australia
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, 3010, Australia
- Cabrini Institute, Cabrini Health, Malvern, VIC, 3144, Australia
| | - Renea A Taylor
- Melbourne Urological Research Alliance, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, 3010, Australia
- Cabrini Institute, Cabrini Health, Malvern, VIC, 3144, Australia
- Department of Physiology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, VIC, 3800, Australia
| | - Mitchell G Lawrence
- Melbourne Urological Research Alliance, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Cancer Program, Monash University, Melbourne, VIC, 3800, Australia
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, 3010, Australia
- Cabrini Institute, Cabrini Health, Malvern, VIC, 3144, Australia
| | - Housheng Hansen He
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G1L7, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G1L7, Canada
| | - Luke A Selth
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Bedford Park, SA, 5042, Australia
- Freemasons Centre for Male Health and Wellbeing, Flinders University, Bedford Park, SA, 5042, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, 5000, Australia
| | - Changmeng Cai
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA, 02125, USA.
- Department of Biology, University of Massachusetts Boston, Boston, MA, 02125, USA.
| |
Collapse
|
7
|
Zhang J, Duan B, Li F, Jing X, Li R, Cai S, Cao L, Jiang Q, Zhou J, Zhou J, Qin Y, Wang X, Tong D, Huang C. SETD7 Promotes Cell Proliferation and Migration via Methylation-mediated TAF7 in Clear Cell Renal Cell Carcinoma. Int J Biol Sci 2024; 20:3008-3027. [PMID: 38904013 PMCID: PMC11186372 DOI: 10.7150/ijbs.93201] [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: 12/13/2023] [Accepted: 05/09/2024] [Indexed: 06/22/2024] Open
Abstract
SET domain containing 7(SETD7), a member of histone methyltransferases, is abnormally expressed in multiple tumor types. However, the biological function and underlying molecular mechanism of SETD7 in clear cell renal cell carcinoma (ccRCC) remain unclear. Here, we explored the biological effects of SETD7-TAF7-CCNA2 axis on proliferation and metastasis in ccRCC. We identified both SETD7 and TAF7 were up-regulated and significantly promoted the proliferation and migration of ccRCC cells. Concurrently, there was a significant positive correlation between the expression of SETD7 and TAF7, and the two were colocalized in the nucleus. Mechanistically, SETD7 methylates TAF7 at K5 and K300 sites, resulting in the deubiquitination and stabilization of TAF7. Furthermore, re-expression of TAF7 could partially restore SETD7 knockdown inhibited ccRCC cells proliferation and migration. In addition, TAF7 transcriptionally activated to drive the expression of cyclin A2 (CCNA2). And more importantly, the methylation of TAF7 at K5 and K300 sites exhibited higher transcriptional activity of CCNA2, which promotes formation and progression of ccRCC. Our findings reveal a unique mechanism that SETD7 mediated TAF7 methylation in regulating transcriptional activation of CCNA2 in ccRCC progression and provide a basis for developing effective therapeutic strategies by targeting members of SETD7-TAF7-CCNA2 axis.
Collapse
Affiliation(s)
- Jinyuan Zhang
- Institute of Genetics and Development Biology, Translational Medcine Institute, Xi'an Jiaotong University, Xi'an 710301, China
| | - Baojun Duan
- Institute of Genetics and Development Biology, Translational Medcine Institute, Xi'an Jiaotong University, Xi'an 710301, China
- Department of Medical Oncology of Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Fang Li
- Institute of Genetics and Development Biology, Translational Medcine Institute, Xi'an Jiaotong University, Xi'an 710301, China
| | - Xintao Jing
- Institute of Genetics and Development Biology, Translational Medcine Institute, Xi'an Jiaotong University, Xi'an 710301, China
| | - Rufeng Li
- Institute of Genetics and Development Biology, Translational Medcine Institute, Xi'an Jiaotong University, Xi'an 710301, China
| | - Shuang Cai
- Institute of Genetics and Development Biology, Translational Medcine Institute, Xi'an Jiaotong University, Xi'an 710301, China
| | - Li Cao
- Institute of Genetics and Development Biology, Translational Medcine Institute, Xi'an Jiaotong University, Xi'an 710301, China
| | - Qiuyu Jiang
- Institute of Genetics and Development Biology, Translational Medcine Institute, Xi'an Jiaotong University, Xi'an 710301, China
| | - Jing Zhou
- Institute of Genetics and Development Biology, Translational Medcine Institute, Xi'an Jiaotong University, Xi'an 710301, China
| | - Jiancheng Zhou
- Department of Urology of Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Yannan Qin
- Institute of Genetics and Development Biology, Translational Medcine Institute, Xi'an Jiaotong University, Xi'an 710301, China
| | - Xiaofei Wang
- Institute of Genetics and Development Biology, Translational Medcine Institute, Xi'an Jiaotong University, Xi'an 710301, China
| | - Dongdong Tong
- Institute of Genetics and Development Biology, Translational Medcine Institute, Xi'an Jiaotong University, Xi'an 710301, China
| | - Chen Huang
- Institute of Genetics and Development Biology, Translational Medcine Institute, Xi'an Jiaotong University, Xi'an 710301, China
| |
Collapse
|
8
|
Liu N, Wang A, Xue M, Zhu X, Liu Y, Chen M. FOXA1 and FOXA2: the regulatory mechanisms and therapeutic implications in cancer. Cell Death Discov 2024; 10:172. [PMID: 38605023 PMCID: PMC11009302 DOI: 10.1038/s41420-024-01936-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
FOXA1 (Forkhead Box A1) and FOXA2 (Forkhead Box A2) serve as pioneering transcription factors that build gene expression capacity and play a central role in biological processes, including organogenesis and differentiation, glycolipid metabolism, proliferation, migration and invasion, and drug resistance. Notably, FOXA1 and FOXA2 may exert antagonistic, synergistic, or complementary effects in the aforementioned biological processes. This article focuses on the molecular mechanisms and clinical relevance of FOXA1 and FOXA2 in steroid hormone-induced malignancies and highlights potential strategies for targeting FOXA1 and FOXA2 for cancer therapy. Furthermore, the article describes the prospect of targeting upstream regulators of FOXA1/FOXA2 to regulate its expression for cancer therapy because of the drug untargetability of FOXA1/FOXA2.
Collapse
Affiliation(s)
- Na Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| | - Anran Wang
- Department of Radiotherapy and Oncology, Gusu School, Nanjing Medical University, The First People's Hospital of Kunshan, Suzhou, 215300, Jiangsu Province, China
| | - Mengen Xue
- Department of Radiotherapy and Oncology, Gusu School, Nanjing Medical University, The First People's Hospital of Kunshan, Suzhou, 215300, Jiangsu Province, China
| | - Xiaoren Zhu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Yang Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Minbin Chen
- Department of Radiotherapy and Oncology, Gusu School, Nanjing Medical University, The First People's Hospital of Kunshan, Suzhou, 215300, Jiangsu Province, China.
| |
Collapse
|
9
|
Wang Z, Petricca J, Liu M, Zhang S, Chen S, Li M, Besschetnova A, Patalano S, Venkataramani K, Siegfried KR, Macoska JA, Han D, Gao S, Vedadi M, Arrowsmith CH, He HH, Cai C. SETD7 functions as a transcription repressor in prostate cancer via methylating FOXA1. Proc Natl Acad Sci U S A 2023; 120:e2220472120. [PMID: 37549269 PMCID: PMC10438836 DOI: 10.1073/pnas.2220472120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 06/29/2023] [Indexed: 08/09/2023] Open
Abstract
Dysregulation of histone lysine methyltransferases and demethylases is one of the major mechanisms driving the epigenetic reprogramming of transcriptional networks in castration-resistant prostate cancer (CRPC). In addition to their canonical histone targets, some of these factors can modify critical transcription factors, further impacting oncogenic transcription programs. Our recent report demonstrated that LSD1 can demethylate the lysine 270 of FOXA1 in prostate cancer (PCa) cells, leading to the stabilization of FOXA1 chromatin binding. This process enhances the activities of the androgen receptor and other transcription factors that rely on FOXA1 as a pioneer factor. However, the identity of the methyltransferase responsible for FOXA1 methylation and negative regulation of the FOXA1-LSD1 oncogenic axis remains unknown. SETD7 was initially identified as a transcriptional activator through its methylation of histone 3 lysine 4, but its function as a methyltransferase on nonhistone substrates remains poorly understood, particularly in the context of PCa progression. In this study, we reveal that SETD7 primarily acts as a transcriptional repressor in CRPC cells by functioning as the major methyltransferase targeting FOXA1-K270. This methylation disrupts FOXA1-mediated transcription. Consistent with its molecular function, we found that SETD7 confers tumor suppressor activity in PCa cells. Moreover, loss of SETD7 expression is significantly associated with PCa progression and tumor aggressiveness. Overall, our study provides mechanistic insights into the tumor-suppressive and transcriptional repression activities of SETD7 in mediating PCa progression and therapy resistance.
Collapse
Affiliation(s)
- Zifeng Wang
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA02125
- Department of Biology, University of Massachusetts Boston, Boston, MA02125
| | - Jessica Petricca
- Department of Medical Biophysics, University of Toronto, Toronto, ONM5G1L7, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ONM5G1L7, Canada
| | - Mingyu Liu
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA02125
- Department of Biology, University of Massachusetts Boston, Boston, MA02125
| | - Songqi Zhang
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA02125
- Department of Biology, University of Massachusetts Boston, Boston, MA02125
| | - Sujun Chen
- Department of Medical Biophysics, University of Toronto, Toronto, ONM5G1L7, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ONM5G1L7, Canada
- West China School of Public Health, West China Fourth Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan610041, China
| | - Muqing Li
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA02125
- Department of Biology, University of Massachusetts Boston, Boston, MA02125
| | - Anna Besschetnova
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA02125
- Department of Biology, University of Massachusetts Boston, Boston, MA02125
| | - Susan Patalano
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA02125
- Department of Biology, University of Massachusetts Boston, Boston, MA02125
| | | | | | - Jill A. Macoska
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA02125
- Department of Biology, University of Massachusetts Boston, Boston, MA02125
| | - Dong Han
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA02125
- Department of Biology, University of Massachusetts Boston, Boston, MA02125
| | - Shuai Gao
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY10595
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY10595
| | - Masoud Vedadi
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ONM5S 1A8, Canada
- Structural Genomics Consortium, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - Cheryl H. Arrowsmith
- Department of Medical Biophysics, University of Toronto, Toronto, ONM5G1L7, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ONM5G1L7, Canada
- Structural Genomics Consortium, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - Housheng Hansen He
- Department of Medical Biophysics, University of Toronto, Toronto, ONM5G1L7, Canada
- Princess Margaret Cancer Center, University Health Network, Toronto, ONM5G1L7, Canada
| | - Changmeng Cai
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, MA02125
- Department of Biology, University of Massachusetts Boston, Boston, MA02125
| |
Collapse
|