DOI QR코드

DOI QR Code

YPEL3 expression induces cellular senescence via the Hippo signaling pathway in human breast cancer cells

  • Yeonju Kwon (College of Pharmacy and Center for Metareceptome Research, Chung-Ang University) ;
  • Hyein Lee (College of Pharmacy and Center for Metareceptome Research, Chung-Ang University) ;
  • Hyemin Park (College of Pharmacy and Center for Metareceptome Research, Chung-Ang University) ;
  • Boyoung Lee (College of Pharmacy and Center for Metareceptome Research, Chung-Ang University) ;
  • Tae‑Uk Kwon (College of Pharmacy and Center for Metareceptome Research, Chung-Ang University) ;
  • Yeo‑Jung Kwon (College of Pharmacy and Center for Metareceptome Research, Chung-Ang University) ;
  • Young‑Jin Chun (College of Pharmacy and Center for Metareceptome Research, Chung-Ang University)
  • Received : 2023.06.28
  • Accepted : 2023.08.14
  • Published : 2023.10.15

Abstract

The Hippo pathway is a signaling pathway that controls organ size in animals by regulating cell proliferation and apoptosis. Yes-associated protein 1 (YAP1), an oncogene associated with the development and progression of breast cancer, is downregulated by the Hippo pathway and is associated with the development and progression of breast cancer. Yippee-like 3 (YPEL3) is a target gene of the tumor suppressor protein p53, and its activation has been shown to inhibit cell growth, induce cellular senescence, and suppress tumor cell metastasis. In this study, we found that YAP1 inhibits the expression of YPEL3 expression in breast cancer cells. Furthermore, a decrease in lamin B1, a marker protein of cellular senescence, coupled with the activation of senescence-associated β-galactosidase indicated that upregulating YPEL3 levels through YAP1 downregulation can induce cellular senescence. Additionally, elevated YPEL3 levels resulted in higher levels of oxygen consumption rate in mitochondria, thus promoting apoptosis. This suggests that YPEL3 plays a crucial role in regulating oxidative stress and cell apoptosis in breast cancer cells. Therefore, the interaction between YAP1 and YPEL3 represents a novel mechanism of cellular senescence mediated by the Hippo signaling pathway. Collectively, our fndings suggest that the Hippo signaling pathway plays an important role in regulating cellular senescence, which could have implications for the development of new therapeutic strategies for diseases such as cancer.

Keywords

Acknowledgement

This research was supported by the National Research Foundation of Korea (NRF) funded by the Korean government (MSIP) (Grant no. 2021R1A2C201239513, 2022R1A5A600076012) and the Chung-Ang University Graduate Research Scholarship in 2021. The funding agency had no role in the study design, data collection or analysis, the decision to publish, or the preparation of the manuscript.

References

  1. Han Y (2019) Analysis of the role of the Hippo pathway in cancer. J Transl Med 17:116. https://doi.org/10.1186/s12967-019-1869-4 
  2. Johnson R, Halder G (2014) The two faces of Hippo: targeting the Hippo pathway for regenerative medicine and cancer treatment. Nat Rev Drug Discov 13:63-79. https://doi.org/10.1038/nrd4161 
  3. Zhang J, Ji JY, Yu M et al (2009) YAP-dependent induction of amphiregulin identifes a non-cell-autonomous component of the Hippo pathway. Nat Cell Biol 11:1444-1450. https://doi.org/10.1038/ncb1993 
  4. Tuttle R, Simon M, Hitch DC et al (2011) Senescence-associated gene YPEL3 is downregulated in human colon tumors. Ann Surg Oncol 18:1791-1796. https://doi.org/10.1245/s10434-011-1558-x 
  5. Kelley KD, Miller KR, Todd A, Kelley AR, Tuttle R, Berberich SJ (2010) YPEL3, a p53-regulated gene that induces cellular senescence. Cancer Res 70:3566-3575. https://doi.org/10.1158/0008-5472.CAN-09-3219 
  6. Kong X, Li Y, Zhang X (2018) Increased expression of the YPEL3 gene in human colonic adenocarcinoma tissue and the efects on proliferation, migration, and invasion of colonic adenocarcinoma cells in vitro via the Wnt/β-catenin signaling pathway. Med Sci Monit 24:4767-4775. https://doi.org/10.12659/MSM.908173 
  7. Xie Q, Chen J, Feng H et al (2013) YAP/TEAD-mediated transcription controls cellular senescence. Cancer Res 73:3615-3624. https://doi.org/10.1158/0008-5472.CAN-12-3793 
  8. Lee S, Lee J (2019) Cellular senescence: a promising strategy for cancer therapy. BMB Rep 52:35-41. https://doi.org/10.5483/BMBRep.2019.52.1.294 
  9. Kumari R, Jat P (2021) Mechanisms of cellular senescence: cell cycle arrest and senescence-associated secretory phenotype. Front Cell Dev Biol 9:645593. https://doi.org/10.3389/fcell.2021.645593 
  10. He L, Chen Y, Feng J, Sun W, Li S, Ou M, Tang L (2017) Cellular senescence regulated by SWI/SNF complex subunits through p53/p21 and p16/pRB pathway. Int J Biochem Cell Biol 90:29-37. https://doi.org/10.1016/j.biocel.2017.07.007 
  11. Zeng S, Shen WH, Liu L (2018) Senescence and cancer. Cancer Transl Med 4:70-74. https://doi.org/10.4103/ctm.ctm_22_18 
  12. Kim H, Chang J, Shao L et al (2017) DNA damage and senescence in osteoprogenitors expressing Osx1 may cause their decrease with age. Aging Cell 16:693-703. https://doi.org/10.1111/acel.12597 
  13. Wiley CD, Velarde MC, Lecot P et al (2016) Mitochondrial dysfunction induces senescence with a distinct secretory phenotype. Cell Metab 23:303-314. https://doi.org/10.1016/j.cmet.2015.11.011 
  14. Vergnes L, Peterfy M, Bergo MO, Young SG, Reue K (2004) Lamin B1 is required for mouse development and nuclear integrity. Proc Natl Acad Sci USA 101:10428-10433. https://doi.org/10.1073/pnas.0401424101 
  15. Freund A, Laberge R, Demaria M, Campisi J (2012) Lamin B1 loss is a senescence-associated biomarker. Mol Biol Cell 23:2066-2075. https://doi.org/10.1091/mbc.e11-10-0884 
  16. Campisi J, Kim SH, Lim CS, Rubio M (2001) Cellular senescence, cancer, and aging: the telomere connection. Exp Gerontol 36:1619-1637. https://doi.org/10.1016/S0531-5565(01)00160-7 
  17. Wang C, Zhu X, Feng W et al (2015) Verteporfn inhibits YAP function through up-regulating 14-3-3σ sequestering YAP in the cytoplasm. Am J Cancer Res 6:27-37 
  18. Guo Q, Quan M, Xu L et al (2022) Enhanced nuclear localization of YAP1-2 contributes to EGF-induced EMT in NSCLC. J Cell Mol Med 26:1013-1023. https://doi.org/10.1111/jcmm.17150 
  19. Tuttle R, Miller KR, Maiorano JN, Termuhlen PM, Gao Y, Berberich SJ (2012) Novel senescence associated gene, YPEL3, is repressed by estrogen in ER+ mammary tumor cells and required for tamoxifen-induced cellular senescence. Int J Cancer 130:2291-2299. https://doi.org/10.1002/ijc.26239 
  20. Wang AS, Ong PF, Chojnowski A, Clavel C, Dreesen O (2017) Loss of lamin B1 is a biomarker to quantify cellular senescence in photoaged skin. Sci Rep 7:15678. https://doi.org/10.1038/s41598-017-15901-9 
  21. Kurz DJ, Decary S, Hong Y, Erusalimsky JD (2000) Senescence-associated (β)-galactosidase refects an increase in lysosomal mass during replicative aging of human endothelial cells. J Cell Sci 113:3613-3622. https://doi.org/10.1242/jcs.113.20.3613 
  22. Olga M, Veronique B, Antoine R, Xavier D-S, Gerardo F (2009) Mitochondrial dysfunction contributes to oncogene-induced senescence. Mol Cell Biol 29:4495-4507. https://doi.org/10.1128/MCB.01868-08 
  23. Wang D, Liu Y, Zhang R et al (2016) Apoptotic transition of senescent cells accompanied with mitochondrial hyper-function. Oncotarget 7:28286-28300. https://doi.org/10.18632/oncotarget.8536 
  24. Pan D (2010) The Hippo signaling pathway in development and cancer. Dev Cell 19:491-505. https://doi.org/10.1016/j.devcel.2010.09.011 
  25. Guo L, Chen Y, Luo J, Zheng J, Shao G (2019) YAP1 overexpression is associated with poor prognosis of breast cancer patients and induces breast cancer cell growth by inhibiting PTEN. FEBS Open Bio 9:437-445. https://doi.org/10.1002/2211-5463.12597 
  26. Chen J, You H, Li Y, Xu Y, He Q, Harris RC (2018) EGF receptor-dependent YAP activation is important for renal recovery from AKI. J Am Soc Nephrol 29:2372-2385. https://doi.org/10.1681/ASN.2017121272 
  27. Ando T, Arang N, Wang Z et al (2021) EGFR regulates the hippo pathway by promoting the tyrosine phosphorylation of MOB1. Commun Biol 4:1237. https://doi.org/10.1038/s42003-021-02744-4 
  28. Gire V, Dulic V (2015) Senescence from G2 arrest, revisited. Cell Cycle 14:297-304. https://doi.org/10.1080/15384101.2014.1000134 
  29. Kim S, Mehta HH, Wan J et al (2018) Mitochondrial peptides modulate mitochondrial function during cellular senescence. Aging 10:1239-1256. https://doi.org/10.18632/aging.101463 
  30. Ma S, Tang T, Probst G et al (2022) Transcriptional repression of estrogen receptor alpha by YAP reveals the Hippo pathway as therapeutic target for ER+ breast cancer. Nat Commun 13:1061. https://doi.org/10.1038/s41467-022-28691-0