DOI QR코드

DOI QR Code

Tumor antigen PRAME is a potential therapeutic target of p53 activation in melanoma cells

  • Yong-Kyu Lee (Department of Integrative Bioscience and Biotechnology, Sejong University) ;
  • Hyeon Ho Heo (Department of Integrative Bioscience and Biotechnology, Sejong University) ;
  • Nackhyoung Kim (Department of Integrative Bioscience and Biotechnology, Sejong University) ;
  • Ui-Hyun Park (Department of Integrative Bioscience and Biotechnology, Sejong University) ;
  • Hyesook Youn (Department of Integrative Bioscience and Biotechnology, Sejong University) ;
  • Eun-Yi Moon (Department of Integrative Bioscience and Biotechnology, Sejong University) ;
  • Eun-Joo Kim (Department of Molecular Biology, Dankook University) ;
  • Soo-Jong Um (Department of Integrative Bioscience and Biotechnology, Sejong University)
  • 투고 : 2023.12.26
  • 심사 : 2024.04.23
  • 발행 : 2024.06.30

초록

Upregulation of PRAME (preferentially expressed antigen of melanoma) has been implicated in the progression of a variety of cancers, including melanoma. The tumor suppressor p53 is a transcriptional regulator that mediates cell cycle arrest and apoptosis in response to stress signals. Here, we report that PRAME is a novel repressive target of p53. This was supported by analysis of melanoma cell lines carrying wild-type p53 and human melanoma databases. mRNA expression of PRAME was downregulated by p53 overexpression and activation using DNA-damaging agents, but upregulated by p53 depletion. We identified a p53-responsive element (p53RE) in the promoter region of PRAME. Luciferase and ChIP assays showed that p53 represses the transcriptional activity of the PRAME promoter and is recruited to the p53RE together with HDAC1 upon etoposide treatment. The functional significance of p53 activation-mediated PRAME downregulation was demonstrated by measuring colony formation and p27 expression in melanoma cells. These data suggest that p53 activation, which leads to PRAME downregulation, could be a therapeutic strategy in melanoma cells.

키워드

과제정보

This study was supported by the Basic Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science and ICT (2021R1A4A5033289), and also supported by Korea Basic Science Institute (National research Facilities and Equipment Center) grant funded by the Ministry of Education (2023R1A6C101A045).

참고문헌

  1. Ikeda H, Lethe B, Lehmann F et al (1997) Characterization of an antigen that is recognized on a melanoma showing partial HLA Loss by CTL expressing an NK inhibitory receptor. Immunity 2, 199-208 
  2. Xu Y, Zou R, Wang J, Wang ZW and Zhu X (2020) The role of the cancer testis antigen PRAME in tumorigenesis and immunotherapy in human cancer. Cell Prolif 53, e12770 
  3. Kern CH, Yang M and Liu WS (2021) The PRAME family of cancer testis antigens is essential for germline development and gametogenesis. Biol Reprod 105, 290-304 
  4. Oberthuer A, Hero B, Spitz R, Berthold F and Fischer M (2004) The tumor-associated antigen PRAME is universally expressed in high-stage neuroblastoma and associated with poor outcome. Clin Cancer Res 10, 4307-4313 
  5. Epping MT, Hart AA, Glas AM, Krijgsman O and Bernards R (2008) PRAME expression and clinical outcome of breast cancer. Br J Cancer 99, 398-403 
  6. Cazzato G, Colagrande A, Ingravallo G et al (2022) PRAME immuno-expression in cutaneous sebaceous carcinoma: a single institutional experience. J Clin Med 11, 6936 
  7. Cazzato G, Cascardi E, Colagrande A et al (2022) PRAME immunoexpression in 275 cutaneous melanocytic lesions: a double institutional experience. Diagnostics (Basel) 12, 2197
  8. Cascardi E, Cazzato G, Ingravallo G et al (2023) PReferentially Expressed Antigen in MElanoma (PRAME): preliminary communication on a translational tool able to early detect oral malignant melanoma (OMM). J Cancer 14, 628-633 
  9. Yan H, Zhao RM, Wang ZJ, Zhao FR and Wang SL (2015) Knockdown of PRAME enhances adriamycin-induced apoptosis in chronic myeloid leukemia cells. Eur Rev Med Pharmacol Sci 19, 4827-4834 
  10. Zhu H, Wang J, Yin J et al (2018) Downregulation of PRAME suppresses proliferation and promotes apoptosis in hepatocellular carcinoma through the activation of p53 mediated pathway. Cell Physiol Biochem 45, 1121-1135 
  11. Chen X, Jiang M, Zhou S et al (2023) PRAME promotes cervical cancer proliferation and migration via Wnt/β-catenin pathway regulation. Cancers (Basel) 15, 1801 
  12. Bose M (2023) Preferentially expressed antigen in melanoma is a multifaceted cancer testis antigen with diverse roles as a biomarker and therapeutic target. Int J Transl Med 3, 334-359 
  13. Cassalia F, Danese A, Tudurachi I et al (2024) PRAME updated: diagnostic, prognostic, and therapeutic role in skin cancer. Int J Mol Sci 25, 1582 
  14. Qian J, Zhu ZH, Lin J et al (2011) Hypomethylation of PRAME promoter is associated with poor prognosis in myelodysplastic syndrome. Br J Haematol 154, 153-155 
  15. Zhang W, Barger CJ, Eng KH et al (2016) PRAME expression and promoter hypomethylation in epithelial ovarian cancer. Oncotarget 7, 45352-45369 
  16. Lee YK, Park UH, Kim EJ, Hwang JT, Jeong JC and Um SJ (2017) Tumor antigen PRAME is up-regulated by MZF1 in cooperation with DNA hypomethylation in melanoma cells. Cancer Lett 403, 144-151 
  17. Dai C and Gu W (2010) p53 post-translational modification: deregulated in tumorigenesis. Mol Med 16, 528-536 
  18. Lee JY and Gu W (2013) SIRT1: regulator of p53 deacetylation. Genes Cancer 4, 112-117 
  19. Riley T, Sontag E, Chen P and Levine A (2008) Transcriptional control of human p53-regulated genes. Nat Rev Mol Cell Biol 9, 402-412 
  20. Ceribelli M, Alcalay M, Vigano MA and Mantovani R (2006) Repression of new p53 targets revealed by ChIP on chip experiments. Cell Cycle 5, 1102-1110 
  21. Rinn JL and Huarte M (2011) To repress or not to repress: this is the guardian's question. Trends Cell Biol 21, 344-353 
  22. Luca M, Lenzi R, Leejackson D, Gutman M, Fidler I and Bareli M (1993) p53 mutations are infrequent and do not correlate with the metastatic potential of human-melanoma cells. Int J Oncol 3, 19-22 
  23. Weiss J, Heine M, Arden KC et al (1995) Mutation and expression of TP53 in malignant melanomas. Recent Results Cancer Res 139, 137-154 
  24. Houben R, Hesbacher S, Schmid CP et al (2011) High-level expression of wild-type p53 in melanoma cells is frequently associated with inactivity in p53 reporter gene assays. PLoS One 6, e22096 
  25. Avery-Kiejda KA, Bowden NA, Croft AJ et al (2011) p53 in human melanoma fails to regulate target genes associated with apoptosis and the cell cycle and may contribute to proliferation. BMC Cancer 11, 203 
  26. Pfister NT and Prives C (2017) Transcriptional regulation by wild-type and cancer-related mutant forms of p53. Cold Spring Harb Perspect Med 7, a026054 
  27. Yeudall WA, Vaughan CA, Miyazaki H et al (2012) Gain-of-function mutant p53 upregulates CXC chemokines and enhances cell migration. Carcinogenesis. 33, 442-451 
  28. Yan W, Liu G, Scoumanne A and Chen X (2008) Suppression of inhibitor of differentiation 2, a target of mutant p53, is required for gain-of-function mutations. Cancer Res 68, 6789-6796 
  29. Zalcenstein A, Stambolsky P, Weisz L et al (2003) Mutant p53 gain of function: repression of CD95 (Fas/APO-1) gene expression by tumor-associated p53 mutants. Oncogene 22, 5667-5676