open access

Vol 68, No 6 (2017)
Original paper
Submitted: 2017-02-03
Accepted: 2017-03-07
Published online: 2017-09-18
Get Citation

The significance of TRIP11 and T3 signalling pathway in renal cancer progression and survival of patients

Piotr Popławski, Agnieszka Piekiełko-Witkowska, Alicja Nauman
·
Pubmed: 29022645
·
Endokrynol Pol 2017;68(6):631-641.

open access

Vol 68, No 6 (2017)
Original Paper
Submitted: 2017-02-03
Accepted: 2017-03-07
Published online: 2017-09-18

Abstract

Introduction: TRIP11 is a multifunctional protein localizing either to Golgi apparatus, acting as a golgin, or in the nucleus, acting as coactivator of transcription mediated by thyroid hormone receptor (THR) and hypoxia induced factor (HIF). Triiodothyronine (T3) regulates nuclear localization of TRIP11 by inducing its phosphorylation. The exact mechanism of this regulation unknown. The expressions of THR and HIF are disturbed in various cancers, including renal cell cancer (RCC). In this study we aimed to analyze: 1) the mechanism of T3-dependent subcellular localization of TRIP11; 2) the significance of TRIP11 and T3 signaling pathway in RCC progression. Material and methods: TRIP11 subcellular localization was analyzed using immunocytochemistry in RCC-derived cell line treated with T3, T3-agarose and PI3K inhibitor, wortmannin. The expressions of TRIP11 and genes involved in T3 signaling and hypoxia were investigated using qPRC in 36 pairs of RCC tumor-control samples, followed by validation/survival analysis in an independent cohort of >450 renal cancer patients. Results: Wortmannin disrupted T3-dependent nuclear transport of TRIP11. T3-agarose did not change TRIP11 localization, precluding extracellular T3-mediated mechanism. The expressions of TRIP11, HIF-1β, THRA, THRB, FURIN, VEGFA, and GLUT1 were disturbed in renal cancer. Expressions of TRIP11 and HIF-1β correlated with tumor grades. Decreased expressions of TRIP11, THRA, and THRB correlated with poor survival of RCC patients. Conclusions: 1) T3 induces nuclear TRIP11 localization via PI3K-dependent mechanism; 2) disturbed expression of T3 signaling pathway genes correlates with RCC progression. The specific mechanisms by which altered T3 signaling may contribute to RCC progression require further investigation.

Abstract

Introduction: TRIP11 is a multifunctional protein localizing either to Golgi apparatus, acting as a golgin, or in the nucleus, acting as coactivator of transcription mediated by thyroid hormone receptor (THR) and hypoxia induced factor (HIF). Triiodothyronine (T3) regulates nuclear localization of TRIP11 by inducing its phosphorylation. The exact mechanism of this regulation unknown. The expressions of THR and HIF are disturbed in various cancers, including renal cell cancer (RCC). In this study we aimed to analyze: 1) the mechanism of T3-dependent subcellular localization of TRIP11; 2) the significance of TRIP11 and T3 signaling pathway in RCC progression. Material and methods: TRIP11 subcellular localization was analyzed using immunocytochemistry in RCC-derived cell line treated with T3, T3-agarose and PI3K inhibitor, wortmannin. The expressions of TRIP11 and genes involved in T3 signaling and hypoxia were investigated using qPRC in 36 pairs of RCC tumor-control samples, followed by validation/survival analysis in an independent cohort of >450 renal cancer patients. Results: Wortmannin disrupted T3-dependent nuclear transport of TRIP11. T3-agarose did not change TRIP11 localization, precluding extracellular T3-mediated mechanism. The expressions of TRIP11, HIF-1β, THRA, THRB, FURIN, VEGFA, and GLUT1 were disturbed in renal cancer. Expressions of TRIP11 and HIF-1β correlated with tumor grades. Decreased expressions of TRIP11, THRA, and THRB correlated with poor survival of RCC patients. Conclusions: 1) T3 induces nuclear TRIP11 localization via PI3K-dependent mechanism; 2) disturbed expression of T3 signaling pathway genes correlates with RCC progression. The specific mechanisms by which altered T3 signaling may contribute to RCC progression require further investigation.
Get Citation

Keywords

RCC, thyroid hormones, hypoxia, TRIP11

About this article
Title

The significance of TRIP11 and T3 signalling pathway in renal cancer progression and survival of patients

Journal

Endokrynologia Polska

Issue

Vol 68, No 6 (2017)

Article type

Original paper

Pages

631-641

Published online

2017-09-18

Page views

1572

Article views/downloads

1550

DOI

10.5603/EP.a2017.0052

Pubmed

29022645

Bibliographic record

Endokrynol Pol 2017;68(6):631-641.

Keywords

RCC
thyroid hormones
hypoxia
TRIP11

Authors

Piotr Popławski
Agnieszka Piekiełko-Witkowska
Alicja Nauman

References (38)
  1. Szymański Ł, Matak D, Bartnik E, et al. Thyroid Hormones as Renal Cell Cancer Regulators. J Signal Transduct. 2016; 2016: 1362407.
  2. Wojcicka A, Piekielko-Witkowska A, Kedzierska H, et al. Epigenetic regulation of thyroid hormone receptor beta in renal cancer. PLoS One. 2014; 9(5): e97624.
  3. Master A, Wójcicka A, Piekiełko-Witkowska A, et al. Untranslated regions of thyroid hormone receptor beta 1 mRNA are impaired in human clear cell renal cell carcinoma. Biochim Biophys Acta. 2010; 1802(11): 995–1005.
  4. Puzianowska-Kuznicka M, Nauman A, Madej A, et al. Expression of thyroid hormone receptors is disturbed in human renal clear cell carcinoma. Cancer Lett. 2000; 155(2): 145–152.
  5. Ríos RM, Sanchís A, Tassin AM, et al. GMAP-210 recruits gamma-tubulin complexes to cis-Golgi membranes and is required for Golgi ribbon formation. Cell. 2004; 118(3): 323–335.
  6. Chang KH, Chen Y, Chen TT, et al. A thyroid hormone receptor coactivator negatively regulated by the retinoblastoma protein. Proc Natl Acad Sci U S A. 1997; 94(17): 9040–9045.
  7. Beischlag TV, Taylor RT, Rose DW, et al. Recruitment of thyroid hormone receptor/retinoblastoma-interacting protein 230 by the aryl hydrocarbon receptor nuclear translocator is required for the transcriptional response to both dioxin and hypoxia. J Biol Chem. 2004; 279(52): 54620–54628.
  8. Labrecque MP, Takhar MK, Jagdeo JM, et al. A TRIP230-retinoblastoma protein complex regulates hypoxia-inducible factor-1α-mediated transcription and cancer cell invasion. PLoS One. 2014; 9(6): e99214.
  9. Rankin EB, Giaccia AJ. Hypoxic control of metastasis. Science. 2016; 352(6282): 175–180.
  10. Perra A, Plateroti M, Columbano A. T3/TRs axis in hepatocellular carcinoma: new concepts for an old pair. Endocr Relat Cancer. 2016; 23(8): R353–R369.
  11. Moeller LC, Cao X, Dumitrescu AM, et al. Thyroid hormone mediated changes in gene expression can be initiated by cytosolic action of the thyroid hormone receptor beta through the phosphatidylinositol 3-kinase pathway. Nucl Recept Signal. 2006; 4: e020.
  12. Zimna A, Kurpisz M. Hypoxia-Inducible Factor-1 in Physiological and Pathophysiological Angiogenesis: Applications and Therapies. Biomed Res Int. 2015; 2015: 549412.
  13. Kim WGu, Zhao Li, Kim DW, et al. Inhibition of tumorigenesis by the thyroid hormone receptor β in xenograft models. Thyroid. 2014; 24(2): 260–269.
  14. Zhang L, Cooper-Kuhn CM, Nannmark U, et al. Stimulatory effects of thyroid hormone on brain angiogenesis in vivo and in vitro. J Cereb Blood Flow Metab. 2010; 30(2): 323–335.
  15. Moeller LC, Führer D. Thyroid hormone, thyroid hormone receptors, and cancer: a clinical perspective. Endocr Relat Cancer. 2013; 20(2): R19–R29.
  16. Chen Y, Chen PL, Chen CF, et al. Thyroid hormone, T3-dependent phosphorylation and translocation of Trip230 from the Golgi complex to the nucleus. Proc Natl Acad Sci U S A. 1999; 96(8): 4443–4448.
  17. Boguslawska J, Kedzierska H, Poplawski P, et al. Expression of Genes Involved in Cellular Adhesion and Extracellular Matrix Remodeling Correlates with Poor Survival of Patients with Renal Cancer. J Urol. 2016; 195(6): 1892–1902.
  18. Aguirre-Gamboa R, Gomez-Rueda H, Martínez-Ledesma E, et al. SurvExpress: an online biomarker validation tool and database for cancer gene expression data using survival analysis. PLoS One. 2013; 8(9): e74250.
  19. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. 2013; 499(7456): 43–49.
  20. Moretto FC, De Sibio MT, Luvizon AC, et al. Triiodothyronine (T3) induces HIF1A and TGFA expression in MCF7 cells by activating PI3K. Life Sci. 2016; 154: 52–57.
  21. Verga Falzacappa C, Petrucci E, Patriarca V, et al. Thyroid hormone receptor TRbeta1 mediates Akt activation by T3 in pancreatic beta cells. J Mol Endocrinol. 2007; 38(1-2): 221–233.
  22. Davis PJ, Davis FB, Lin HY, et al. Translational implications of nongenomic actions of thyroid hormone initiated at its integrin receptor. Am J Physiol Endocrinol Metab. 2009; 297(6): E1238–E1246.
  23. McMahon S, Grondin F, McDonald PP, et al. Hypoxia-enhanced expression of the proprotein convertase furin is mediated by hypoxia-inducible factor-1: impact on the bioactivation of proproteins. J Biol Chem. 2005; 280(8): 6561–6569.
  24. Chen RN, Huang YH, Lin YC, et al. Thyroid hormone promotes cell invasion through activation of furin expression in human hepatoma cell lines. Endocrinology. 2008; 149(8): 3817–3831.
  25. Thomas G. Furin at the cutting edge: from protein traffic to embryogenesis and disease. Nat Rev Mol Cell Biol. 2002; 3(10): 753–766.
  26. Barron CC, Bilan PJ, Tsakiridis T, et al. Facilitative glucose transporters: Implications for cancer detection, prognosis and treatment. Metabolism. 2016; 65(2): 124–139.
  27. Qian CN, Tan MH, Yang JP, et al. Revisiting tumor angiogenesis: vessel co-option, vessel remodeling, and cancer cell-derived vasculature formation. Chin J Cancer. 2016; 35: 10.
  28. Stacker SA, Achen MG. The VEGF signaling pathway in cancer: the road ahead. Chin J Cancer. 2013; 32(6): 297–302.
  29. Qian CN, Huang D, Wondergem B, et al. Complexity of tumor vasculature in clear cell renal cell carcinoma. Cancer. 2009; 115(10 Suppl): 2282–2289.
  30. Ferrara N. Vascular endothelial growth factor. Arterioscler Thromb Vasc Biol. 2009; 29(6): 789–791.
  31. Baldewijns MM, Thijssen VL, Van den Eynden GG, et al. High-grade clear cell renal cell carcinoma has a higher angiogenic activity than low-grade renal cell carcinoma based on histomorphological quantification and qRT-PCR mRNA expression profile. Br J Cancer. 2007; 96(12): 1888–1895.
  32. Rioux-Leclercq N, Fergelot P, Zerrouki S, et al. Plasma level and tissue expression of vascular endothelial growth factor in renal cell carcinoma: a prospective study of 50 cases. Hum Pathol. 2007; 38(10): 1489–1495.
  33. Smits P, Bolton AD, Funari V, et al. Lethal skeletal dysplasia in mice and humans lacking the golgin GMAP-210. N Engl J Med. 2010; 362(3): 206–216.
  34. Roboti P, Sato K, Lowe M. The golgin GMAP-210 is required for efficient membrane trafficking in the early secretory pathway. J Cell Sci. 2015; 128(8): 1595–1606.
  35. Hellewell AL, Adams JC. Insider trading: Extracellular matrix proteins and their non-canonical intracellular roles. Bioessays. 2016; 38(1): 77–88.
  36. Jacob A, Prekeris R. The regulation of MMP targeting to invadopodia during cancer metastasis. Front Cell Dev Biol. 2015; 3: 4.
  37. Martínez-Iglesias O, Garcia-Silva S, Tenbaum SP, et al. Thyroid hormone receptor beta1 acts as a potent suppressor of tumor invasiveness and metastasis. Cancer Res. 2009; 69(2): 501–509.
  38. Bassett JH, Williams GR. Role of Thyroid Hormones in Skeletal Development and Bone Maintenance. Endocr Rev. 2016; 37: 135–87.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Via MedicaWydawcą jest  VM Media Group sp. z o.o., Grupa Via Medica, ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl