Vol 89, No 10 (2018)
Research paper
Published online: 2018-10-31

open access

Page views 1782
Article views/downloads 1203
Get Citation

Connect on Social Media

Connect on Social Media

The role of the βKlotho gene in uterine endometrial cancer

Katarzyna Monika Wójcik-Krowiranda1, Sylwia Szczepaniec1, Andrzej Bieńkiewicz1
Pubmed: 30393845
Ginekol Pol 2018;89(10):563-567.

Abstract

Objectives: Endometrial cancer is the most common cancer of the female genital organs in developed countries, accounting for approximately 50% of all gynecological cancers. The Klotho gene was discovered in 1997 as an anti-aging gene that, when overexpressed, may extend the lifespan, but when disrupted, may be a factor responsible for premature aging syndrome. The aim of the study is to assess the relationship between the clinical and pathological features of endometrial cancer and βKlotho gene expression.
Material and methods: The expression of βKlotho gene was studied in 138 cases of endometrioid endometrial carcinoma specimens using Real Time PCR reaction in RNA isolated tissue samples by commercial tests. The expression profile was correlated with the clinicopathological characteristics of endometrial carcinoma. The chi-square independence test and Fisher’s test for four-field tables were used to assess the statistical significance of the observed relationships.
Results: Significant relationships were found between βKlotho gene expression and FIGO clinical stage, the degree of histological differentiation and the presence of metastases in the lymph nodes. Higher levels of gene expression correlate with lower degrees of clinical staging according to FIGO, the presence of highly-differentiated endometrial cancer (G1) and the absence of lymph node metastases.
Conclusions: The βKlotho gene expression might be involved in endometrioid endometrial cancer tumorgenesis. The
βKlotho may in future be used as an useful indicator for endometrial cancer, although further studies are needed.

Article available in PDF format

View PDF Download PDF file

References

  1. Brooks AN, Kilgour E, Smith PD. Molecular pathways: fibroblast growth factor signaling: a new therapeutic opportunity in cancer. Clin Cancer Res. 2012; 18(7): 1855–1862.
  2. Wang Y, Sun Z. Current understanding of klotho. Ageing Res Rev. 2009; 8(1): 43–51.
  3. Kuro-o M, Matsumura Y, Aizawa H, et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 1997; 390(6655): 45–51.
  4. Kuro-o M. Klotho and aging. Biochim Biophys Acta. 2009; 1790(10): 1049–1058.
  5. Kuro-o M. Klotho and βKlotho. Adv Exp Med Biol. 2012; 728: 25–40.
  6. Matsumura Y, Aizawa H, Shiraki-Iida T, et al. Identification of the human klotho gene and its two transcripts encoding membrane and secreted klotho protein. Biochem Biophys Res Commun. 1998; 242(3): 626–630.
  7. Li SA, Watanabe M, Yamada H, et al. Immunohistochemical localization of Klotho protein in brain, kidney, and reproductive organs of mice. Cell Struct Funct. 2004; 29(4): 91–99.
  8. Razzaque MS. The role of Klotho in energy metabolism. Nat Rev Endocrinol. 2012; 8(10): 579–587.
  9. Ito S, Kinoshita S, Shiraishi N, et al. Molecular cloning and expression analyses of mouse betaklotho, which encodes a novel Klotho family protein. Mech Dev. 2000; 98(1-2): 115–119.
  10. Ito S, Fujimori T, Furuya A, et al. Impaired negative feedback suppression of bile acid synthesis in mice lacking βKlotho. Journal of Clinical Investigation. 2005; 115(8): 2202–2208.
  11. Ogawa Y, Kurosu H, Yamamoto M, et al. BetaKlotho is required for metabolic activity of fibroblast growth factor 21. Proc Natl Acad Sci U S A. 2007; 104(18): 7432–7437.
  12. Kharitonenkov A, Dunbar JD, Bina HA, et al. FGF-21/FGF-21 receptor interaction and activation is determined by betaKlotho. J Cell Physiol. 2008; 215(1): 1–7.
  13. Ito S, Kinoshita S, Shiraishi N, et al. Molecular cloning and expression analyses of mouse betaklotho, which encodes a novel Klotho family protein. Mech Dev. 2000; 98(1-2): 115–119.
  14. Szymczyk A, Krzywański R, Forma E. Struktura i funkcje białka βKlotho. Folia Medica Lodziensia. 2013; 40: 99–132.
  15. http://www.nextprot.org.
  16. Suzuki H, Watkins DN, Jair KW, et al. Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat Genet. 2004; 36(4): 417–422.
  17. Nojima M, Suzuki H, Toyota M, et al. Frequent epigenetic inactivation of SFRP genes and constitutive activation of Wnt signaling in gastric cancer. Oncogene. 2007; 26(32): 4699–4713.
  18. Urakami S, Shiina H, Enokida H, et al. Combination analysis of hypermethylated Wnt-antagonist family genes as a novel epigenetic biomarker panel for bladder cancer detection. Clin Cancer Res. 2006; 12(7 Pt 1): 2109–2211.
  19. Szymczyk A, Forma E, Krześlak A, et al. Genetic polymorphism of Klotho gene. Folia Medica Lodziensia. 2012; 39: 189–205.
  20. Poh W, Wong W, Ong H, et al. Klotho-beta overexpression as a novel target for suppressing proliferation and fibroblast growth factor receptor-4 signaling in hepatocellular carcinoma. Mol Cancer. 2012; 23: 11–14.
  21. Ye X, Guo Yu, Zhang Qi, et al. βKlotho suppresses tumor growth in hepatocellular carcinoma by regulating Akt/GSK-3β/cyclin D1 signaling pathway. PLoS One. 2013; 8(1): e55615.
  22. Knights V, Cook SJ. De-regulated FGF receptors as therapeutic targets in cancer. Pharmacol Ther. 2010; 125(1): 105–117.
  23. Liu Z, Qi S, Zhao X, et al. Metformin inhibits 17β-estradiol-induced epithelial-to-mesenchymal transition via βKlotho-related ERK1/2 signaling and AMPKα signaling in endometrial adenocarcinoma cells. Oncotarget. 2016; 7(16).