• 검색 결과가 없습니다.

Expression of estrogen receptorin B-RafV600E-expressing HOSE

Our group published thatnormal thyrocyte infected with B-RafV600E lentivrus increased in mRNA level of thyroid stimulating hormone(TSHR)(Kim et al., 2014).Accordingly, we assumedthatB-RafV600E-expressing HOSEwould showupregulation of estrogen receptor level. Therefore, we analyzedestrogen receptor alpha and beta expression by real-time PCR and immunoblotting (Fig. 5A and 5B). Unlike our expectation, B-RafV600E-expressing HOSEdid not show the upregulation estrogen receptor α and β.

18 A.

Fig5.Level of estrogen receptor α and β in B-RafV600E-expressing HOSE.

(A) Measurementof mRNA. At 12 days after infection, we performed Real-time PCR. (B) Measurement of protein. On the same day, we performed immunoblotting.

19

6. Estrogen regulates translocation of p-Erk1/2 in B-RafV600E-expressing HOSE.

We found that estrogen inhibits B-RafV600E induced senescence in HOSE. But, we wondered that how estrogen inhibitedOIS.Estrogen regulated translocation of p-Erk1/2 (Chen et al., 2005).For these reasons, we analyzed localization of p-Erk1/2 by using confocal microscopy and counted by using image of immunocytochemistry (Fig. 6A). p-Erk1/2 is mainly accumulated in cytosol in B-RafV600E-expressing HOSE. But, p-Erk1/2 located in nuclear in estrogen treated B-RafV600E-expressing HOSE. Next, we focused on which mechanism is involved in translocation of p-Erk1/2. It was established that PEA15 regulated location of Erk1/2 (Lee et al., 2015). Therefore, we measured level of PEA15 and p-PEA15S104 (Fig. 6B) (Chen et al., 2008; Lee et al., 2015). We observed that p-PEA15S104 is increased in estrogen treated B-RafV600E-expressing HOSE. These results suggested that estrogen regulates translocation of p-Erk1/2 via phosphorylation of PEA15.

20 A.

21 B.

Fig6. Estrogen mediated translocation of p- Erk1/2.

(A) Localization of p-Erk1/2. We performed confocal microscopy 19 days after infection and 16 days after treatment of E2and counted by using image J. (B) Level of protein.

Immunoblotting of PEA15, p-PEA15S104 and α-Tubulin is performed19 days after infection and 16 days after treatment of E2.

22

IV.Discussion

If residue 600 amino acid val of the B-Raf is optionally substituted with glu, B-Raf always has the active form in cell signaling pathway which causes a more than cell growth and can be oncogene (Michaloglou et al., 2008). B-RafV600E has been reported up to 14% in serous carcinoma (SOC) (Grisham et al., 2013) and up to 71% in serous borderline tumor (SBT) (Malpica and Wong, 2016). For patients receiving the post menopause in women and ovarian cancer resection may increase the risk of ovarian cancer by implementing the estrogen replacement therapy (Risch, 1996; Riman et al., 2002; Morch et al., 2009).

In this study, we observed whether estrogen regulates B-RafV600E-induced senescence in human primary ovarian epithelial cells (HOSE). Thus, we overexpressed B-Raf protein by using lentivirus system. B-RafV600E-expressing HOSE was decreased in cell growth and increased in SA-B-GAL and expression of p21 but not expression of p16. Next, to identify estrogen effect, 40nM 17-b-estradiol (E2) was treated in normal HOSE and B-RafV600E-expressing HOSE every 2 day. B-RafV600E-B-RafV600E-expressing HOSE treated E2 increased in cell growth and level of cyclin D1 and decreased in SA-β-gal compared with B-RafV600E-expressing HOSE. Other hand, no significant difference in normal HOSE and normal HOSE treated E2 better than B-Rafv600e-expressing HOSE. After that, we observed estrogen receptor α and β whether overexpression of B-Raf affects to expression of estrogen receptors.

However, we couldn’t found the difference.To know how estrogen inhibits B-RafV600E-induced senescence and how cell growth was reversed, we focused on relation between estrogen and phosphorylation of Erk1/2. Estrogen can translocate phosphorylation of Erk1/2 into nucleus (Chen et al., 2005). As you know, Erk1/2 must translocate in nucleus to proliferate cells .So, we carried out localization of p-Erk1/2 through Immunofluorescence and confocal microscopy. We proved that p-Erk1/2 was accumulated in cytosol in RafV600E-expressing HOSE. Other hand, p-Erk1/2 was translocated in nucleus in B-RafV600E-expressing HOSE treated E2. It was reported that pea-15 regulates p-Erk1/2(Krueger et al., 2005; Lee et al., 2015). In our experiment, to identify whether PEA15

23

was involved in translocation of Erk1/2, we observed level of protein including pea-15, p-pea15S104 and p-Erk1/2. As a result, phosphorylation of pea-15 increased in B-RafV600E-expressing HOSE treated E2.

In further study, we will count amount of p-Erk1/2 in nucleus. And then, we will prove interaction between p-Erk1/2 and PEA15 by performing immunoprecipitation.

24

REFERENCES

1. Aronica SM, Kraus WL, Katzenellenbogen BS: Estrogen action via the cAMP signaling pathway: stimulation of adenylate cyclase and cAMP-regulated gene transcription. Proc Natl Acad Sci U S A 91: 8517-8521, 1994

2. Chen JR, Plotkin LI, Aguirre JI, Han L, Jilka RL, Kousteni S, Bellido T, Manolagas SC:

Transient versus sustained phosphorylation and nuclear accumulation of ERKs underlie anti-versus pro-apoptotic effects of estrogens. J Biol Chem 280: 4632-4638, 2005

3. Cho KR, Shih Ie M: Ovarian cancer. Annu Rev Pathol 4: 287-313, 2009

4. Ciucci A, Zannoni GF, Buttarelli M, Lisi L, Travaglia D, Martinelli E, Scambia G, Gallo D: Multiple direct and indirect mechanisms drive estrogen-induced tumor growth in high grade serous ovarian cancers. Oncotarget 7: 8155-8171, 2016 5. Courtois-Cox S, Jones SL, Cichowski K: Many roads lead to oncogene-induced

senescence. Oncogene 27: 2801-2809, 2008

6. Cui J, Shen Y, Li R: Estrogen synthesis and signaling pathways during aging: from periphery to brain. Trends Mol Med 19: 197-209, 2013

7. Gottsch ML, Navarro VM, Zhao Z, Glidewell-Kenney C, Weiss J, Jameson JL, Clifton DK, Levine JE, Steiner RA: Regulation of Kiss1 and dynorphin gene expression in the murine brain by classical and nonclassical estrogen receptor pathways. J Neurosci 29: 9390-9395, 2009

8. Grisham RN, Iyer G, Garg K, DeLair D, Hyman DM, Zhou Q, Iasonos A, Berger MF, Dao F, Spriggs DR, Levine DA, Aghajanian C, Solit DB: BRAF mutation is associated with early stage disease and improved outcome in patients with low-grade serous ovarian cancer. Cancer 119: 548-554, 2013

9. Haas MJ, Raheja P, Jaimungal S, Sheikh-Ali M, Mooradian AD: Estrogen-dependent inhibition of dextrose-induced endoplasmic reticulum stress and superoxide generation in endothelial cells. Free Radic Biol Med 52: 2161-2167, 2012

10. Kim YH, Choi YW, Han JH, Lee J, Soh EY, Park SH, Kim JH, Park TJ: TSH signaling overcomes B-RafV600E-induced senescence in papillary thyroid carcinogenesis through regulation of DUSP6. Neoplasia 16: 1107-1120, 2014

11. Klotz DM, Hewitt SC, Ciana P, Raviscioni M, Lindzey JK, Foley J, Maggi A,

25

DiAugustine RP, Korach KS: Requirement of estrogen receptor-alpha in insulin-like growth factor-1 (1)-induced uterine responses and in vivo evidence for IGF-1/estrogen receptor cross-talk. J Biol Chem 277: 8531-8537, 2002

12. Krueger J, Chou FL, Glading A, Schaefer E, Ginsberg MH: Phosphorylation of phosphoprotein enriched in astrocytes (PEA-15) regulates extracellular signal-regulated kinase-dependent transcription and cell proliferation. Mol Biol Cell 16:

3552-3561, 2005

13. Lee YY, Kim HS, Lim IK: Downregulation of PEA-15 reverses G1 arrest, and nuclear and chromatin changes of senescence phenotype via pErk1/2 translocation to nuclei. Cell Signal 27: 1102-1109, 2015

14. Malpica A, Wong KK: The molecular pathology of ovarian serous borderline tumors. Ann Oncol 27 Suppl 1: i16-i19, 2016

15. Martin MB, Franke TF, Stoica GE, Chambon P, Katzenellenbogen BS, Stoica BA, McLemore MS, Olivo SE, Stoica A: A role for Akt in mediating the estrogenic functions of epidermal growth factor and insulin-like growth factor I.

Endocrinology 141: 4503-4511, 2000

16. Meng L, Quezada M, Levine P, Han Y, McDaniel K, Zhou T, Lin E, Glaser S, Meng F, Francis H, Alpini G: Functional role of cellular senescence in biliary injury. Am J Pathol 185: 602-609, 2015

17. Micevych P, Dominguez R: Membrane estradiol signaling in the brain. Front Neuroendocrinol 30: 315-327, 2009

18. Michaloglou C, Vredeveld LC, Mooi WJ, Peeper DS: BRAF(E600) in benign and malignant human tumours. Oncogene 27: 877-895, 2008

19. Mooi WJ, Peeper DS: Oncogene-induced cell senescence--halting on the road to cancer. N Engl J Med 355: 1037-1046, 2006

20. Morch LS, Lokkegaard E, Andreasen AH, Kruger-Kjaer S, Lidegaard O: Hormone therapy and ovarian cancer. JAMA 302: 298-305, 2009

21. Ng A, Barker N: Ovary and fimbrial stem cells: biology, niche and cancer origins.

Nat Rev Mol Cell Biol 16: 625-638, 2015

22. Prossnitz ER, Arterburn JB, Smith HO, Oprea TI, Sklar LA, Hathaway HJ: Estrogen signaling through the transmembrane G protein-coupled receptor GPR30. Annu Rev Physiol 70: 165-190, 2008

26

23. Richardson TE, Yu AE, Wen Y, Yang SH, Simpkins JW: Estrogen prevents oxidative damage to the mitochondria in Friedreich's ataxia skin fibroblasts. PLoS One 7:

e34600, 2012

24. Riman T, Dickman PW, Nilsson S, Correia N, Nordlinder H, Magnusson CM, Weiderpass E, Persson IR: Hormone replacement therapy and the risk of invasive epithelial ovarian cancer in Swedish women. J Natl Cancer Inst 94: 497-504, 2002 25. Risch HA: Estrogen replacement therapy and risk of epithelial ovarian cancer.

Gynecol Oncol 63: 254-257, 1996

27

관련 문서