207
Views
2
CrossRef citations to date
0
Altmetric
ORIGINAL RESEARCH

EPAS1/HIF-2α Acts as an Unanticipated Tumor-Suppressive Role in Papillary Thyroid Carcinoma

ORCID Icon, &
Pages 2165-2174 | Received 23 Feb 2023, Accepted 25 May 2023, Published online: 31 May 2023

References

  • Megwalu UC, Moon PK. Thyroid cancer incidence and mortality trends in the United States: 2000–2018. Thyroid. 2022;32(5):560–570. doi:10.1089/thy.2021.0662
  • Guo K, Qian K, Shi Y, et al. Clinical and molecular characterizations of papillary thyroid cancer in children and young adults: a multicenter retrospective study. Thyroid. 2021;31(11):1693–1706. doi:10.1089/thy.2021.0003
  • Ghossein R, Livolsi VA. Papillary thyroid carcinoma tall cell variant. Thyroid. 2008;18:1179–1181. doi:10.1089/thy.2008.0164
  • Cuomo F, Giani C, Cobellis G. The role of the kinase inhibitors in thyroid cancers. Pharmaceutics. 2022;14(5):1040. doi:10.3390/pharmaceutics14051040
  • Bhandari V, Hoey C, Liu LY, et al. Molecular landmarks of tumor hypoxia across cancer types. Nat Genet. 2019;51(2):308–318. doi:10.1038/s41588-018-0318-2
  • Wu Q, You L, Nepovimova E, et al. Hypoxia-inducible factors: master regulators of hypoxic tumor immune escape. J Hematol Oncol. 2022;15(1):77. doi:10.1186/s13045-022-01292-6
  • Galardo MN, Gorga A, Merlo JP, et al. Participation of HIFs in the regulation of Sertoli cell lactate production. Biochimie. 2017;132:9–18. doi:10.1016/j.biochi.2016.10.006
  • Gonzalez FJ, Xie C, Jiang C. The role of hypoxia-inducible factors in metabolic diseases. Nat Rev Endocrinol. 2018;15(1):21–32. doi:10.1038/s41574-018-0096-z
  • Schodel J, Grampp S, Maher ER, et al. Hypoxia, hypoxia-inducible transcription factors, and renal cancer. Eur Urol. 2016;69(4):646–657. doi:10.1016/j.eururo.2015.08.007
  • Garcia Garcia CJ, Huang Y, Fuentes NR, et al. Stromal HIF2 regulates immune suppression in the pancreatic cancer microenvironment. Gastroenterology. 2022;162(7):2018–2031. doi:10.1053/j.gastro.2022.02.024
  • Islam F, Gopalan V, Law S, Lam AK, Pillai S. Molecular deregulation of EPAS1 in the pathogenesis of esophageal squamous cell carcinoma. Front Oncol. 2020;10:1534. doi:10.3389/fonc.2020.01534
  • Hoefflin R, Harlander S, Schafer S, et al. HIF-1alpha and HIF-2alpha differently regulate tumour development and inflammation of clear cell renal cell carcinoma in mice. Nat Commun. 2020;11(1):4111. doi:10.1038/s41467-020-17873-3
  • Nakazawa MS, Eisinger-Mathason TS, Sadri N, et al. Epigenetic re-expression of HIF-2alpha suppresses soft tissue sarcoma growth. Nat Commun. 2016;7:10539. doi:10.1038/ncomms10539
  • Acker T, Diez-Juan A, Aragones J, et al. Genetic evidence for a tumor suppressor role of HIF-2alpha. Cancer Cell. 2005;8(2):131–141. doi:10.1016/j.ccr.2005.07.003
  • Mazumdar J, Hickey MM, Pant DK, et al. HIF-2alpha deletion promotes Kras-driven lung tumor development. Proc Natl Acad Sci USA. 2010;107(32):14182–14187. doi:10.1073/pnas.1001296107
  • Westerlund I, Shi Y, Toskas K, et al. Combined epigenetic and differentiation-based treatment inhibits neuroblastoma tumor growth and links HIF2alpha to tumor suppression. Proc Natl Acad Sci USA. 2017;114(30):E6137–E6146. doi:10.1073/pnas.1700655114
  • Xu Q, Gao S, Miao J. The relationship between autophagy-related genes and the staging and prognosis of thyroid cancer: a bioinformatics analysis. Gland Surg. 2021;10(8):2511–2527. doi:10.21037/gs-21-480
  • Wicks EE, Semenza GL. Hypoxia-inducible factors: cancer progression and clinical translation. J Clin Invest. 2022;132(11). doi:10.1172/JCI159839
  • Wigerup C, Pahlman S, Bexell D. Therapeutic targeting of hypoxia and hypoxia-inducible factors in cancer. Pharmacol Ther. 2016;164:152–169. doi:10.1016/j.pharmthera.2016.04.009
  • Cowman SJ, Koh MY. Revisiting the HIF switch in the tumor and its immune microenvironment. Trends Cancer. 2022;8(1):28–42. doi:10.1016/j.trecan.2021.10.004
  • Luan Y, Gao C, Miao Y, Li Y, Wang Z, Qiu X. Clinicopathological and prognostic significance of HIF-1alpha and HIF-2alpha expression in small cell lung cancer. Pathol Res Pract. 2013;209(3):184–189. doi:10.1016/j.prp.2012.10.017
  • Chen W, Hill H, Christie A, et al. Targeting renal cell carcinoma with a HIF-2 antagonist. Nature. 2016;539(7627):112–117. doi:10.1038/nature19796
  • Martinez-Saez O, Gajate Borau P, Alonso-Gordoa T, Molina-Cerrillo J, Grande E. Targeting HIF-2 alpha in clear cell renal cell carcinoma: a promising therapeutic strategy. Crit Rev Oncol Hematol. 2017;111:117–123. doi:10.1016/j.critrevonc.2017.01.013
  • Yang Y, Chen W, Mai W, Gao Y. HIF-2alpha regulates proliferation, invasion, and metastasis of hepatocellular carcinoma cells via VEGF/Notch1 signaling axis after insufficient radiofrequency ablation. Front Oncol. 2022;12:998295. doi:10.3389/fonc.2022.998295
  • Singhal R, Mitta SR, Das NK, et al. HIF-2alpha activation potentiates oxidative cell death in colorectal cancers by increasing cellular iron. J Clin Invest. 2021;131(12):1.
  • Liu YM, Ying SP, Huang YR, et al. Expression of HIF-1alpha and HIF-2alpha correlates to biological and clinical significance in papillary thyroid carcinoma. World J Surg Oncol. 2016;14(1):30. doi:10.1186/s12957-016-0785-9
  • Rawluszko-Wieczorek AA, Horbacka K, Krokowicz P, Misztal M, Jagodzinski PP. Prognostic potential of DNA methylation and transcript levels of HIF1A and EPAS1 in colorectal cancer. Mol Cancer Res. 2014;12(8):1112–1127. doi:10.1158/1541-7786.MCR-14-0054
  • Xiong Y, Liu L, Xia Y, et al. Tumor infiltrating mast cells determine oncogenic HIF-2alpha-conferred immune evasion in clear cell renal cell carcinoma. Cancer Immunol Immunother. 2019;68(5):731–741. doi:10.1007/s00262-019-02314-y
  • Guo M, Chen Z, Li Y, et al. Tumor mutation burden predicts relapse in papillary thyroid carcinoma with changes in genes and immune microenvironment. Front Endocrinol. 2021;12:674616. doi:10.3389/fendo.2021.674616
  • Shao QP, Wei C, Yang J, Zhang WZ. miR-3609 decelerates the clearance of sorafenib in hepatocellular carcinoma cells by targeting EPAS-1 and reducing the activation of the pregnane X receptor pathway. Onco Targets Ther. 2020;13:7213–7227. doi:10.2147/OTT.S246471