#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

SPDEF Inhibits Prostate Carcinogenesis by Disrupting a Positive Feedback Loop in Regulation of the Foxm1 Oncogene


Development of prostate cancer is a multistep process that involves the loss of tumor suppressor functions and activation of oncogenes. SPDEF transcription factor is expressed in normal prostate epithelium and its expression changes during prostate carcinogenesis (PCa). Since the role of SPDEF in PCa remains controversial, we generated transgenic mice with loss- and gain-of-function of SPDEF to demonstrate that SPDEF functions as a tumor suppressor in PCa. In animal models, the loss of SPDEF promoted PCa and increased the levels of Foxm1, a well-known oncogenic protein. Overexpression of SPDEF in prostate epithelium decreased PCa and reduced Foxm1 levels. Proliferation defects in SPDEF-containing tumor cells were corrected by re-expression of Foxm1, providing direct evidence that SPDEF inhibits tumor cell proliferation through Foxm1. We further showed that SPDEF directly bound to Foxm1 promoter and prevented its auto-regulatory activation. In prostate cancer patients, the low SPDEF and high Foxm1 were found in most aggressive prostate tumors that were associated with poor prognosis. The combined two-gene signature of low SPDEF and high Foxm1 was a strong predictor of survival in prostate cancer patients. The present study identified novel molecular mechanism of prostate cancer progression, providing a crosstalk between SPDEF tumor suppressor and Foxm1 oncogene.


Vyšlo v časopise: SPDEF Inhibits Prostate Carcinogenesis by Disrupting a Positive Feedback Loop in Regulation of the Foxm1 Oncogene. PLoS Genet 10(9): e32767. doi:10.1371/journal.pgen.1004656
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004656

Souhrn

Development of prostate cancer is a multistep process that involves the loss of tumor suppressor functions and activation of oncogenes. SPDEF transcription factor is expressed in normal prostate epithelium and its expression changes during prostate carcinogenesis (PCa). Since the role of SPDEF in PCa remains controversial, we generated transgenic mice with loss- and gain-of-function of SPDEF to demonstrate that SPDEF functions as a tumor suppressor in PCa. In animal models, the loss of SPDEF promoted PCa and increased the levels of Foxm1, a well-known oncogenic protein. Overexpression of SPDEF in prostate epithelium decreased PCa and reduced Foxm1 levels. Proliferation defects in SPDEF-containing tumor cells were corrected by re-expression of Foxm1, providing direct evidence that SPDEF inhibits tumor cell proliferation through Foxm1. We further showed that SPDEF directly bound to Foxm1 promoter and prevented its auto-regulatory activation. In prostate cancer patients, the low SPDEF and high Foxm1 were found in most aggressive prostate tumors that were associated with poor prognosis. The combined two-gene signature of low SPDEF and high Foxm1 was a strong predictor of survival in prostate cancer patients. The present study identified novel molecular mechanism of prostate cancer progression, providing a crosstalk between SPDEF tumor suppressor and Foxm1 oncogene.


Zdroje

1. HanahanD, WeinbergRA (2011) Hallmarks of cancer: the next generation. Cell 144: 646–674.

2. BergesRR, VukanovicJ, EpsteinJI, CarMichelM, CisekL, et al. (1995) Implication of cell kinetic changes during the progression of human prostatic cancer. Clin Cancer Res 1: 473–480.

3. UzgareAR, KaplanPJ, GreenbergNM (2003) Differential expression and/or activation of P38MAPK, erk1/2, and jnk during the initiation and progression of prostate cancer. Prostate 55: 128–139.

4. GaoH, OuyangX, Banach-PetroskyWA, GeraldWL, ShenMM, et al. (2006) Combinatorial activities of Akt and B-Raf/Erk signaling in a mouse model of androgen-independent prostate cancer. Proc Natl Acad Sci U S A 103: 14477–14482.

5. CairnsP, OkamiK, HalachmiS, HalachmiN, EstellerM, et al. (1997) Frequent inactivation of PTEN/MMAC1 in primary prostate cancer. Cancer Res 57: 4997–5000.

6. GreenbergNM, DeMayoF, FinegoldMJ, MedinaD, TilleyWD, et al. (1995) Prostate cancer in a transgenic mouse. Proc Natl Acad Sci U S A 92: 3439–3443.

7. KaplanPJ, MohanS, CohenP, FosterBA, GreenbergNM (1999) The insulin-like growth factor axis and prostate cancer: lessons from the transgenic adenocarcinoma of mouse prostate (TRAMP) model. Cancer Res 59: 2203–2209.

8. KalinTV, WangIC, AckersonTJ, MajorML, DetrisacCJ, et al. (2006) Increased levels of the FoxM1 transcription factor accelerate development and progression of prostate carcinomas in both TRAMP and LADY transgenic mice. Cancer Res 66: 1712–1720.

9. WangIC, MelitonL, TretiakovaM, CostaRH, KalinichenkoVV, et al. (2008) Transgenic expression of the forkhead box M1 transcription factor induces formation of lung tumors. Oncogene 27: 4137–4149.

10. CaiY, BalliD, UstiyanV, FulfordL, HillerA, et al. (2013) Foxm1 expression in prostate epithelial cells is essential for prostate carcinogenesis. J Biol Chem 288: 22527–22541.

11. WangIC, MelitonL, RenX, ZhangY, BalliD, et al. (2009) Deletion of Forkhead Box M1 transcription factor from respiratory epithelial cells inhibits pulmonary tumorigenesis. PLoS ONE 4: e6609.

12. WeiGH, BadisG, BergerMF, KiviojaT, PalinK, et al. (2010) Genome-wide analysis of ETS-family DNA-binding in vitro and in vivo. EMBO J 29: 2147–2160.

13. ParkKS, KorfhagenTR, BrunoMD, KitzmillerJA, WanH, et al. (2007) SPDEF regulates goblet cell hyperplasia in the airway epithelium. J Clin Invest 117: 978–988.

14. GregorieffA, StangeDE, KujalaP, BegthelH, van den BornM, et al. (2009) The ets-domain transcription factor Spdef promotes maturation of goblet and paneth cells in the intestinal epithelium. Gastroenterology 137: 1333–1345 e1331–1333.

15. RodabaughKJ, Mhawech-FaucegliaP, GrothJ, LeleS, SoodAK (2007) Prostate-derived Ets factor is overexpressed in serous epithelial ovarian tumors. Int J Gynecol Pathol 26: 10–15.

16. HorstD, GuX, BhasinM, YangQ, VerziM, et al. (2010) Requirement of the epithelium-specific Ets transcription factor Spdef for mucous gland cell function in the gastric antrum. J Biol Chem 285: 35047–35055.

17. CarverBS, TranJ, ChenZ, Carracedo-PerezA, AlimontiA, et al. (2009) ETS rearrangements and prostate cancer initiation. Nature 457: E1 discussion E2–3.

18. SteffanJJ, KoulS, MeachamRB, KoulHK (2012) The transcription factor SPDEF suppresses prostate tumor metastasis. J Biol Chem 287: 29968–29978.

19. SchaeferJS, SabherwalY, ShiHY, SriramanV, RichardsJ, et al. (2010) Transcriptional regulation of p21/CIP1 cell cycle inhibitor by PDEF controls cell proliferation and mammary tumor progression. J Biol Chem 285: 11258–11269.

20. NoahTK, LoYH, PriceA, ChenG, KingE, et al. (2013) SPDEF functions as a colorectal tumor suppressor by inhibiting beta-catenin activity. Gastroenterology 144: 1012–1023 e1016.

21. OettgenP, FingerE, SunZ, AkbaraliY, ThamrongsakU, et al. (2000) PDEF, a novel prostate epithelium-specific ets transcription factor, interacts with the androgen receptor and activates prostate-specific antigen gene expression. J Biol Chem 275: 1216–1225.

22. ChenH, NandiAK, LiX, BieberichCJ (2002) NKX-3.1 interacts with prostate-derived Ets factor and regulates the activity of the PSA promoter. Cancer Res 62: 338–340.

23. JohnsonTR, KoulS, KumarB, KhandrikaL, VeneziaS, et al. (2010) Loss of PDEF, a prostate-derived Ets factor is associated with aggressive phenotype of prostate cancer: regulation of MMP 9 by PDEF. Mol Cancer 9: 148.

24. FeldmanRJ, SementchenkoVI, GayedM, FraigMM, WatsonDK (2003) Pdef expression in human breast cancer is correlated with invasive potential and altered gene expression. Cancer Res 63: 4626–4631.

25. GhadersohiA, OdunsiK, ZhangS, AzrakRG, BundyBN, et al. (2008) Prostate-derived Ets transcription factor as a favorable prognostic marker in ovarian cancer patients. Int J Cancer 123: 1376–1384.

26. GunawardaneRN, SgroiDC, WrobelCN, KohE, DaleyGQ, et al. (2005) Novel role for PDEF in epithelial cell migration and invasion. Cancer Res 65: 11572–11580.

27. SoodAK, SaxenaR, GrothJ, DesoukiMM, CheewakriangkraiC, et al. (2007) Expression characteristics of prostate-derived Ets factor support a role in breast and prostate cancer progression. Hum Pathol 38: 1628–1638.

28. GuX, ZerbiniLF, OtuHH, BhasinM, YangQ, et al. (2007) Reduced PDEF expression increases invasion and expression of mesenchymal genes in prostate cancer cells. Cancer Res 67: 4219–4226.

29. KalinTV, UstiyanV, KalinichenkoVV (2011) Multiple faces of FoxM1 transcription factor: Lessons from transgenic mouse models. Cell Cycle 10: 396–405.

30. TaylorBS, SchultzN, HieronymusH, GopalanA, XiaoY, et al. (2010) Integrative genomic profiling of human prostate cancer. Cancer Cell 18: 11–22.

31. SbonerA, DemichelisF, CalzaS, PawitanY, SetlurSR, et al. (2010) Molecular sampling of prostate cancer: a dilemma for predicting disease progression. BMC Med Genomics 3: 8.

32. WangZA, MitrofanovaA, BergrenSK, Abate-ShenC, CardiffRD, et al. (2013) Lineage analysis of basal epithelial cells reveals their unexpected plasticity and supports a cell-of-origin model for prostate cancer heterogeneity. Nat Cell Biol 15: 274–283.

33. BuchwalterG, HickeyMM, CromerA, SelforsLM, GunawardaneRN, et al. (2013) PDEF promotes luminal differentiation and acts as a survival factor for ER-positive breast cancer cells. Cancer Cell 23: 753–767.

34. TurnerDP, FindlayVJ, MoussaO, SemenchenkoVI, WatsonPM, et al. (2011) Mechanisms and functional consequences of PDEF protein expression loss during prostate cancer progression. Prostate 71: 1723–1735.

35. GhadersohiA, SharmaS, ZhangS, AzrakRG, WildingGE, et al. (2011) Prostate-derived Ets transcription factor (PDEF) is a potential prognostic marker in patients with prostate cancer. Prostate 71: 1178–1188.

36. HallerAC, TanW, Payne-OndracekR, UnderwoodW, TianL, et al. (2014) High SPDEF may identify patients who will have a prolonged response to androgen deprivation therapy. Prostate 74: 509–519.

37. SoodAK, WangJ, Mhawech-FaucegliaP, JanaB, LiangP, et al. (2009) Sam-pointed domain containing Ets transcription factor in luminal breast cancer pathogenesis. Cancer Epidemiol Biomarkers Prev 18: 1899–1903.

38. de VisserKE, EichtenA, CoussensLM (2006) Paradoxical roles of the immune system during cancer development. Nat Rev Cancer 6: 24–37.

39. BalliD, RenX, ChouFS, CrossE, ZhangY, et al. (2011) Foxm1 transcription factor is required for macrophage migration during lung inflammation and tumor formation. Oncogene 31: 3875–3888.

40. HalasiM, GartelAL (2009) A novel mode of FoxM1 regulation: positive auto-regulatory loop. Cell Cycle 8: 1966–1967.

41. ChenG, KorfhagenTR, XuY, KitzmillerJ, WertSE, et al. (2009) SPDEF is required for mouse pulmonary goblet cell differentiation and regulates a network of genes associated with mucus production. J Clin Invest 119: 2914–2924.

42. BalliD, UstiyanV, ZhangY, WangIC, MasinoAJ, et al. (2013) Foxm1 transcription factor is required for lung fibrosis and epithelial-to-mesenchymal transition. EMBO J 32: 231–244.

43. WatsonPA, Ellwood-YenK, KingJC, WongvipatJ, LebeauMM, et al. (2005) Context-dependent hormone-refractory progression revealed through characterization of a novel murine prostate cancer cell line. Cancer Res 65: 11565–11571.

44. BeltekiG, HaighJ, KabacsN, HaighK, SisonK, et al. (2005) Conditional and inducible transgene expression in mice through the combinatorial use of Cre-mediated recombination and tetracycline induction. Nucleic Acids Res 33: e51.

45. UstiyanV, WangIC, RenX, ZhangY, SnyderJ, et al. (2009) Forkhead box M1 transcriptional factor is required for smooth muscle cells during embryonic development of blood vessels and esophagus. Dev Biol 336: 266–279.

46. RenX, ShahTA, UstiyanV, ZhangY, ShinnJ, et al. (2013) FOXM1 promotes allergen-induced goblet cell metaplasia and pulmonary inflammation. Mol Cell Biol 33: 371–386.

47. BarrettT, TroupDB, WilhiteSE, LedouxP, RudnevD, et al. (2009) NCBI GEO: archive for high-throughput functional genomic data. Nucleic Acids Res 37: D885–890.

48. MantelN (1966) Evaluation of survival data and two new rank order statistics arising in its consideration. Cancer Chemother Rep 50: 163–170.

49. GentlemanRC, CareyVJ, BatesDM, BolstadB, DettlingM, et al. (2004) Bioconductor: open software development for computational biology and bioinformatics. Genome Biol 5: R80.

50. MajorML, LepeR, CostaRH (2004) Forkhead Box M1B (FoxM1B) Transcriptional Activity Requires Binding of Cdk/Cyclin Complexes for Phosphorylation-Dependent Recruitment of p300/CBP Co-activators. Mol Cell Biol 24: 2649–2661.

51. KalinTV, WangIC, MelitonL, ZhangY, WertSE, et al. (2008) Forkhead Box m1 transcription factor is required for perinatal lung function. Proc Natl Acad Sci U S A 105: 19330–19335.

52. BalliD, ZhangY, SnyderJ, KalinichenkoVV, KalinTV (2011) Endothelial cell-specific deletion of transcription factor FoxM1 increases urethane-induced lung carcinogenesis. Cancer Res 71: 40–50.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 9
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#