DOI QR코드

DOI QR Code

The role of hypoxia on the acquisition of epithelial-mesenchymal transition and cancer stemness: a possible link to epigenetic regulation

  • Yeo, Chang Dong (Division of Pulmonology, Department of Internal Medicine, The Cancer Research Institute, College of Medicine, The Catholic University of Korea) ;
  • Kang, Nahyeon (Division of Pulmonology, Department of Internal Medicine, The Cancer Research Institute, College of Medicine, The Catholic University of Korea) ;
  • Choi, Su Yeon (Division of Pulmonology, Department of Internal Medicine, The Cancer Research Institute, College of Medicine, The Catholic University of Korea) ;
  • Kim, Bit Na (Division of Pulmonology, Department of Internal Medicine, The Cancer Research Institute, College of Medicine, The Catholic University of Korea) ;
  • Park, Chan Kwon (Division of Pulmonology, Department of Internal Medicine, The Cancer Research Institute, College of Medicine, The Catholic University of Korea) ;
  • Kim, Jin Woo (Division of Pulmonology, Department of Internal Medicine, The Cancer Research Institute, College of Medicine, The Catholic University of Korea) ;
  • Kim, Young Kyoon (Division of Pulmonology, Department of Internal Medicine, The Cancer Research Institute, College of Medicine, The Catholic University of Korea) ;
  • Kim, Seung Joon (Division of Pulmonology, Department of Internal Medicine, The Cancer Research Institute, College of Medicine, The Catholic University of Korea)
  • Received : 2016.09.29
  • Accepted : 2017.06.15
  • Published : 2017.07.01

Abstract

A hypoxic microenvironment leads to cancer progression and increases the metastatic potential of cancer cells within tumors via epithelial-mesenchymal transition (EMT) and cancer stemness acquisition. The hypoxic response pathway can occur under oxygen tensions of < 40 mmHg through hypoxia-inducible factors (HIFs), which are considered key mediators in the adaptation to hypoxia. Previous studies have shown that cellular responses to hypoxia are required for EMT and cancer stemness maintenance through $HIF-1{\alpha}$ and $HIF-2{\alpha}$. The principal transcription factors of EMT include Twist, Snail, Slug, Sip1 (Smad interacting protein 1), and ZEB1 (zinc finger E-box-binding homeobox 1). HIFs bind to hypoxia response elements within the promoter region of these genes and also target cancer stem cell-associated genes and mediate transcriptional responses to hypoxia during stem cell differentiation. Acquisition of stemness characteristics in epithelial cells can be induced by activation of the EMT process. The mechanism of these phenotypic changes includes epigenetic alterations, such as DNA methylation, histone modification, chromatin remodeling, and microRNAs. Increased expression of EMT and pluripotent genes also play a role through demethylation of their promoters. In this review, we summarize the role of hypoxia on the acquisition of EMT and cancer stemness and the possible association with epigenetic regulation, as well as their therapeutic applications.

Keywords

Acknowledgement

Supported by : National Research Foundation of Korea (NRF)

References

  1. Foster JG, Wong SC, Sharp TV. The hypoxic tumor microenvironment: driving the tumorigenesis of non-smallcell lung cancer. Future Oncol 2014;10:2659-2674. https://doi.org/10.2217/fon.14.201
  2. Hockel M, Vaupel P. Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst 2001;93:266-276. https://doi.org/10.1093/jnci/93.4.266
  3. Gao T, Li JZ, Lu Y, et al. The mechanism between epithelial mesenchymal transition in breast cancer and hypoxia microenvironment. Biomed Pharmacother 2016;80:393-405. https://doi.org/10.1016/j.biopha.2016.02.044
  4. Li M, Wang YX, Luo Y, et al. Hypoxia inducible factor-$1{\alpha}$-dependent epithelial to mesenchymal transition under hypoxic conditions in prostate cancer cells. Oncol Rep 2016;36:521-527. https://doi.org/10.3892/or.2016.4766
  5. Ye LY, Chen W, Bai XL, et al. Hypoxia-induced epithelialto-mesenchymal transition in hepatocellular carcinoma induces an immunosuppressive tumor microenvironment to promote metastasis. Cancer Res 2016;76:818-830. https://doi.org/10.1158/0008-5472.CAN-15-0977
  6. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelialmesenchymal transitions in development and disease. Cell 2009;139:871-890. https://doi.org/10.1016/j.cell.2009.11.007
  7. Li L, Li W. Epithelial-mesenchymal transition in human cancer: comprehensive reprogramming of metabolism, epigenetics, and differentiation. Pharmacol Ther 2015;150:33-46. https://doi.org/10.1016/j.pharmthera.2015.01.004
  8. Garg M. Targeting microRNAs in epithelial-to-mesenchymal transition-induced cancer stem cells: therapeutic approaches in cancer. Expert Opin Ther Targets 2015;19:285-297. https://doi.org/10.1517/14728222.2014.975794
  9. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature 2001;414:105-111. https://doi.org/10.1038/35102167
  10. Medema JP. Cancer stem cells: the challenges ahead. Nat Cell Biol 2013;15:338-344. https://doi.org/10.1038/ncb2717
  11. Mimeault M, Batra SK. Hypoxia-inducing factors as master regulators of stemness properties and altered metabolism of cancer- and metastasis-initiating cells. J Cell Mol Med 2013;17:30-54. https://doi.org/10.1111/jcmm.12004
  12. Shukla S, Meeran SM. Epigenetics of cancer stem cells: pathways and therapeutics. Biochim Biophys Acta 2014;1840:3494-3502. https://doi.org/10.1016/j.bbagen.2014.09.017
  13. Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet 2012;13:484-492. https://doi.org/10.1038/nrg3230
  14. Vaupel P, Hockel M, Mayer A. Detection and characterization of tumor hypoxia using pO2 histography. Antioxid Redox Signal 2007;9:1221-1235. https://doi.org/10.1089/ars.2007.1628
  15. Bertout JA, Patel SA, Simon MC. The impact of O2 availability on human cancer. Nat Rev Cancer 2008;8:967-975. https://doi.org/10.1038/nrc2540
  16. Myszczyszyn A, Czarnecka AM, Matak D, et al. The role of hypoxia and cancer stem cells in renal cell carcinoma pathogenesis. Stem Cell Rev 2015;11:919-943. https://doi.org/10.1007/s12015-015-9611-y
  17. Yeh KA, Biade S, Lanciano RM, et al. Polarographic needle electrode measurements of oxygen in rat prostate carcinomas: accuracy and reproducibility. Int J Radiat Oncol Biol Phys 1995;33:111-118. https://doi.org/10.1016/0360-3016(95)00036-X
  18. Vaupel P, Mayer A. Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metastasis Rev 2007;26:225-239. https://doi.org/10.1007/s10555-007-9055-1
  19. Koch CJ, Evans SM. Optimizing hypoxia detection and treatment strategies. Semin Nucl Med 2015;45:163-176. https://doi.org/10.1053/j.semnuclmed.2014.10.004
  20. Shimoda LA, Semenza GL. HIF and the lung: role of hypoxia-inducible factors in pulmonary development and disease. Am J Respir Crit Care Med 2011;183:152-156. https://doi.org/10.1164/rccm.201009-1393PP
  21. Sharp FR, Bernaudin M. HIF1 and oxygen sensing in the brain. Nat Rev Neurosci 2004;5:437-448. https://doi.org/10.1038/nrn1408
  22. Harris AL. Hypoxia: a key regulatory factor in tumour growth. Nat Rev Cancer 2002;2:38-47. https://doi.org/10.1038/nrc704
  23. Gordan JD, Simon MC. Hypoxia-inducible factors: central regulators of the tumor phenotype. Curr Opin Genet Dev 2007;17:71-77. https://doi.org/10.1016/j.gde.2006.12.006
  24. Zhao J, Du F, Luo Y, Shen G, Zheng F, Xu B. The emerging role of hypoxia-inducible factor-2 involved in chemo/radioresistance in solid tumors. Cancer Treat Rev 2015;41:623-633. https://doi.org/10.1016/j.ctrv.2015.05.004
  25. Zhang J, Tian XJ, Xing J. Signal transduction pathways of EMT induced by TGF-beta, SHH, and WNT and their crosstalks. J Clin Med 2016;5:41. https://doi.org/10.3390/jcm5040041.
  26. Jiang J, Tang YL, Liang XH. EMT: a new vision of hypoxia promoting cancer progression. Cancer Biol Ther 2011;11:714-723. https://doi.org/10.4161/cbt.11.8.15274
  27. Imai T, Horiuchi A, Wang C, et al. Hypoxia attenuates the expression of E-cadherin via up-regulation of SNAIL in ovarian carcinoma cells. Am J Pathol 2003;163:1437-1447. https://doi.org/10.1016/S0002-9440(10)63501-8
  28. Hung JJ, Yang MH, Hsu HS, Hsu WH, Liu JS, Wu KJ. Prognostic significance of hypoxia-inducible factor-1alpha, TWIST1 and Snail expression in resectable nonsmall cell lung cancer. Thorax 2009;64:1082-1089. https://doi.org/10.1136/thx.2009.115691
  29. Nishi H, Nakada T, Hokamura M, et al. Hypoxia-inducible factor-1 transactivates transforming growth factorbeta3 in trophoblast. Endocrinology 2004;145:4113-4118. https://doi.org/10.1210/en.2003-1639
  30. Diecke S, Jung SM, Lee J, Ju JH. Recent technological updates and clinical applications of induced pluripotent stem cells. Korean J Intern Med 2014;29:547-557. https://doi.org/10.3904/kjim.2014.29.5.547
  31. Kim N, Cho SG. Clinical applications of mesenchymal stem cells. Korean J Intern Med 2013;28:387-402. https://doi.org/10.3904/kjim.2013.28.4.387
  32. Clarke MF, Dick JE, Dirks PB, et al. Cancer stem cells: perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res 2006;66:9339-9344. https://doi.org/10.1158/0008-5472.CAN-06-3126
  33. Hamburger AW, Salmon SE. Primary bioassay of human tumor stem cells. Science 1977;197:461-463. https://doi.org/10.1126/science.560061
  34. Sourisseau T, Hassan KA, Wistuba I, et al. Lung cancer stem cell: fancy conceptual model of tumor biology or cornerstone of a forthcoming therapeutic breakthrough? J Thorac Oncol 2014;9:7-17. https://doi.org/10.1097/JTO.0000000000000028
  35. Giangreco A, Arwert EN, Rosewell IR, Snyder J, Watt FM, Stripp BR. Stem cells are dispensable for lung homeostasis but restore airways after injury. Proc Natl Acad Sci U S A 2009;106:9286-9291. https://doi.org/10.1073/pnas.0900668106
  36. Akunuru S, James Zhai Q, Zheng Y. Non-small cell lung cancer stem/progenitor cells are enriched in multiple distinct phenotypic subpopulations and exhibit plasticity. Cell Death Dis 2012;3:e352. https://doi.org/10.1038/cddis.2012.93
  37. Yang CH, Wang HL, Lin YS, et al. Identification of CD24 as a cancer stem cell marker in human nasopharyngeal carcinoma. PLoS One 2014;9:e99412. https://doi.org/10.1371/journal.pone.0099412
  38. Li Z, Bao S, Wu Q, et al. Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer Cell 2009;15:501-513. https://doi.org/10.1016/j.ccr.2009.03.018
  39. McCord AM, Jamal M, Shankavaram UT, Lang FF, Camphausen K, Tofilon PJ. Physiologic oxygen concentration enhances the stem-like properties of CD133+ human glioblastoma cells in vitro. Mol Cancer Res 2009;7:489-497. https://doi.org/10.1158/1541-7786.MCR-08-0360
  40. Koukourakis MI, Kakouratos C, Kalamida D, et al. Hypoxia-inducible proteins $HIF1{\alpha}$ and lactate dehydrogenase LDH5, key markers of anaerobic metabolism, relate with stem cell markers and poor post-radiotherapy outcome in bladder cancer. Int J Radiat Biol 2016;92:353-363. https://doi.org/10.3109/09553002.2016.1162921
  41. Seidel S, Garvalov BK, Wirta V, et al. A hypoxic niche regulates glioblastoma stem cells through hypoxia inducible factor 2 alpha. Brain 2010;133:983-995. https://doi.org/10.1093/brain/awq042
  42. Wang Z, Sun J, Feng Y, Tian X, Wang B, Zhou Y. Oncogenic roles and drug target of CXCR4/CXCL12 axis in lung cancer and cancer stem cell. Tumour Biol 2016;37:8515-8528. https://doi.org/10.1007/s13277-016-5016-z
  43. Cho BS, Kim HJ, Konopleva M. Targeting the CXCL12/CXCR4 axis in acute myeloid leukemia: from bench to bedside. Korean J Intern Med 2017;32:248-257. https://doi.org/10.3904/kjim.2016.244
  44. Nian WQ, Chen FL, Ao XJ, Chen ZT. CXCR4 positive cells from Lewis lung carcinoma cell line have cancer metastatic stem cell characteristics. Mol Cell Biochem 2011;355:241-248. https://doi.org/10.1007/s11010-011-0860-z
  45. Romain B, Hachet-Haas M, Rohr S, et al. Hypoxia differentially regulated CXCR4 and CXCR7 signaling in colon cancer. Mol Cancer 2014;13:58. https://doi.org/10.1186/1476-4598-13-58
  46. Liang Z, Brooks J, Willard M, et al. CXCR4/CXCL12 axis promotes VEGF-mediated tumor angiogenesis through Akt signaling pathway. Biochem Biophys Res Commun 2007;359:716-722. https://doi.org/10.1016/j.bbrc.2007.05.182
  47. Fabregat I, Malfettone A, Soukupova J. New insights into the crossroads between EMT and stemness in the context of cancer. J Clin Med 2016;5:37. https://doi.org/10.3390/jcm5030037.
  48. Mani SA, Guo W, Liao MJ, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008;133:704-715. https://doi.org/10.1016/j.cell.2008.03.027
  49. Morel AP, Lievre M, Thomas C, Hinkal G, Ansieau S, Puisieux A. Generation of breast cancer stem cells through epithelial-mesenchymal transition. PLoS One 2008;3:e2888. https://doi.org/10.1371/journal.pone.0002888
  50. Rhim AD, Mirek ET, Aiello NM, et al. EMT and dissemination precede pancreatic tumor formation. Cell 2012;148:349-361. https://doi.org/10.1016/j.cell.2011.11.025
  51. Kurrey NK, Jalgaonkar SP, Joglekar AV, et al. Snail and slug mediate radioresistance and chemoresistance by antagonizing p53-mediated apoptosis and acquiring a stem-like phenotype in ovarian cancer cells. Stem Cells 2009;27:2059-2068. https://doi.org/10.1002/stem.154
  52. Wellner U, Schubert J, Burk UC, et al. The EMT-activator ZEB1 promotes tumorigenicity by repressing stemnessinhibiting microRNAs. Nat Cell Biol 2009;11:1487-1495. https://doi.org/10.1038/ncb1998
  53. Mima K, Okabe H, Ishimoto T, et al. CD44s regulates the TGF-beta-mediated mesenchymal phenotype and is associated with poor prognosis in patients with hepatocellular carcinoma. Cancer Res 2012;72:3414-3423. https://doi.org/10.1158/0008-5472.CAN-12-0299
  54. Nomura A, Banerjee S, Chugh R, et al. CD133 initiates tumors, induces epithelial-mesenchymal transition and increases metastasis in pancreatic cancer. Oncotarget 2015;6:8313-8322.
  55. Huangyang P, Shang Y. Epigenetic regulation of epithelial to mesenchymal transition. Curr Cancer Drug Targets 2013;13:973-985. https://doi.org/10.2174/15680096113136660103
  56. Hennessy BT, Gonzalez-Angulo AM, Stemke-Hale K, et al. Characterization of a naturally occurring breast cancer subset enriched in epithelial-to-mesenchymal transition and stem cell characteristics. Cancer Res 2009;69:4116-4124.
  57. Chen Y, Wang K, Qian CN, Leach R. DNA methylation is associated with transcription of snail and slug genes. Biochem Biophys Res Commun 2013;430:1083-1090. https://doi.org/10.1016/j.bbrc.2012.12.034
  58. Deneberg S, Guardiola P, Lennartsson A, et al. Prognostic DNA methylation patterns in cytogenetically normal acute myeloid leukemia are predefined by stem cell chromatin marks. Blood 2011;118:5573-5582. https://doi.org/10.1182/blood-2011-01-332353
  59. Chang CJ, Chao CH, Xia W, et al. p53 regulates epithelial-mesenchymal transition and stem cell properties through modulating miRNAs. Nat Cell Biol 2011;13:317-323. https://doi.org/10.1038/ncb2173
  60. Taube JH, Malouf GG, Lu E, et al. Epigenetic silencing of microRNA-203 is required for EMT and cancer stem cell properties. Sci Rep 2013;3:2687. https://doi.org/10.1038/srep02687
  61. Simonsson S, Gurdon J. DNA demethylation is necessary for the epigenetic reprogramming of somatic cell nuclei. Nat Cell Biol 2004;6:984-990. https://doi.org/10.1038/ncb1176
  62. El Helou R, Wicinski J, Guille A, et al. Brief reports: a distinct DNA methylation signature defines breast cancer stem cells and predicts cancer outcome. Stem Cells 2014;32:3031-3036. https://doi.org/10.1002/stem.1792
  63. Yamazaki Y, Fujita TC, Low EW, Alarcon VB, Yanagimachi R, Marikawa Y. Gradual DNA demethylation of the Oct4 promoter in cloned mouse embryos. Mol Reprod Dev 2006;73:180-188. https://doi.org/10.1002/mrd.20411
  64. Liu CC, Lin JH, Hsu TW, et al. IL-6 enriched lung cancer stem-like cell population by inhibition of cell cycle regulators via DNMT1 upregulation. Int J Cancer 2015;136:547-559.
  65. Suzuki M, Mohamed S, Nakajima T, et al. Aberrant methylation of CXCL12 in non-small cell lung cancer is associated with an unfavorable prognosis. Int J Oncol 2008;33:113-119.
  66. Zhi Y, Chen J, Zhang S, Chang X, Ma J, Dai D. Down-regulation of CXCL12 by DNA hypermethylation and its involvement in gastric cancer metastatic progression. Dig Dis Sci 2012;57:650-659. https://doi.org/10.1007/s10620-011-1922-5
  67. Fridrichova I, Smolkova B, Kajabova V, et al. CXCL12 and ADAM23 hypermethylation are associated with advanced breast cancers. Transl Res 2015;165:717-730. https://doi.org/10.1016/j.trsl.2014.12.006
  68. Kubarek L, Jagodzinski PP. Epigenetic up-regulation of CXCR4 and CXCL12 expression by 17 beta-estradiol and tamoxifen is associated with formation of DNA methyltransferase 3B4 splice variant in Ishikawa endometrial adenocarcinoma cells. FEBS Lett 2007;581:1441-1448. https://doi.org/10.1016/j.febslet.2007.02.070
  69. Przybylski M, Kozlowska A, Pietkiewicz PP, Lutkowska A, Lianeri M, Jagodzinski PP. Increased CXCR4 expression in AsPC1 pancreatic carcinoma cells with RNA interference-mediated knockdown of DNMT1 and DNMT3B. Biomed Pharmacother 2010;64:254-258. https://doi.org/10.1016/j.biopha.2009.06.008
  70. Koga C, Kobayashi S, Nagano H, et al. Reprogramming using microRNA-302 improves drug sensitivity in hepatocellular carcinoma cells. Ann Surg Oncol 2014;21 Suppl 4:S591-S600. https://doi.org/10.1245/s10434-014-3705-7
  71. Liu C, Kelnar K, Liu B, et al. The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med 2011;17:211-215. https://doi.org/10.1038/nm.2284
  72. Shimono Y, Zabala M, Cho RW, et al. Downregulation of miRNA-200c links breast cancer stem cells with normal stem cells. Cell 2009;138:592-603. https://doi.org/10.1016/j.cell.2009.07.011
  73. Gwak JM, Kim HJ, Kim EJ, et al. MicroRNA-9 is associated with epithelial-mesenchymal transition, breast cancer stem cell phenotype, and tumor progression in breast cancer. Breast Cancer Res Treat 2014;147:39-49. https://doi.org/10.1007/s10549-014-3069-5
  74. Yu T, Liu K, Wu Y, et al. MicroRNA-9 inhibits the proliferation of oral squamous cell carcinoma cells by suppressing expression of CXCR4 via the Wnt/beta-catenin signaling pathway. Oncogene 2014;33:5017-5027. https://doi.org/10.1038/onc.2013.448
  75. Yin P, Peng R, Peng H, et al. MiR-451 suppresses cell proliferation and metastasis in A549 lung cancer cells. Mol Biotechnol 2015;57:1-11. https://doi.org/10.1007/s12033-014-9796-3
  76. Sarvi S, Mackinnon AC, Avlonitis N, et al. CD133+ cancer stem-like cells in small cell lung cancer are highly tumorigenic and chemoresistant but sensitive to a novel neuropeptide antagonist. Cancer Res 2014;74:1554-1565. https://doi.org/10.1158/0008-5472.CAN-13-1541
  77. Chen Q, Lipkina G, Song Q, Kramer RH. Promoter methylation regulates cadherin switching in squamous cell carcinoma. Biochem Biophys Res Commun 2004;315:850-856. https://doi.org/10.1016/j.bbrc.2004.01.143
  78. Peter ME. Let-7 and miR-200 microRNAs: guardians against pluripotency and cancer progression. Cell Cycle 2009;8:843-852. https://doi.org/10.4161/cc.8.6.7907
  79. Chen Y, Ramjiawan RR, Reiberger T, et al. CXCR4 inhibition in tumor microenvironment facilitates anti- programmed death receptor-1 immunotherapy in sorafenib-treated hepatocellular carcinoma in mice. Hepatology 2015;61:1591-1602. https://doi.org/10.1002/hep.27665

Cited by

  1. Cancer stem cells (CSCs): metabolic strategies for their identification and eradication vol.475, pp.9, 2018, https://doi.org/10.1042/bcj20170164
  2. Chemoresistance and the Self-Maintaining Tumor Microenvironment vol.10, pp.12, 2017, https://doi.org/10.3390/cancers10120471
  3. Hypoxia Can Induce Migration of Glioblastoma Cells Through a Methylation-Dependent Control of ODZ1 Gene Expression vol.9, pp.None, 2017, https://doi.org/10.3389/fonc.2019.01036
  4. Poor outcome in hypoxic endometrial carcinoma is related to vascular density vol.120, pp.11, 2017, https://doi.org/10.1038/s41416-019-0461-2
  5. Low Numbers of Vascular Vessels Correlate to Progression in Hormone-Naïve Prostate Carcinomas Undergoing Radical Prostatectomy vol.11, pp.9, 2017, https://doi.org/10.3390/cancers11091356
  6. Red Blood Cell-Membrane-Coated Poly(Lactic-co-glycolic Acid) Nanoparticles for Enhanced Chemo- and Hypoxia-Activated Therapy vol.2, pp.9, 2017, https://doi.org/10.1021/acsabm.9b00584
  7. ERO1α promotes hypoxic tumor progression and is associated with poor prognosis in pancreatic cancer vol.10, pp.57, 2017, https://doi.org/10.18632/oncotarget.27235
  8. Hypoxia-induced cancer stemness acquisition is associated with CXCR4 activation by its aberrant promoter demethylation vol.19, pp.None, 2019, https://doi.org/10.1186/s12885-019-5360-7
  9. Circular RNA circNRIP1 Sponges microRNA-138-5p to Maintain Hypoxia-Induced Resistance to 5-Fluorouracil Through HIF-1α-Dependent Glucose Metabolism in Gastric Carcinoma vol.12, pp.None, 2017, https://doi.org/10.2147/cmar.s246272
  10. Novel Therapeutic Strategies for Ovarian Cancer Stem Cells vol.10, pp.None, 2020, https://doi.org/10.3389/fonc.2020.00319
  11. Hypoxia-Induced Epithelial-Mesenchymal Transition in Cancers: HIF-1α and Beyond vol.10, pp.None, 2020, https://doi.org/10.3389/fonc.2020.00486
  12. Colorectal Cancer Stem Cells in the Progression to Liver Metastasis vol.10, pp.None, 2020, https://doi.org/10.3389/fonc.2020.01511
  13. Genome-wide analysis of the hypoxia-related DNA methylation-driven genes in lung adenocarcinoma progression vol.40, pp.2, 2017, https://doi.org/10.1042/bsr20194200
  14. Hypoxia modulates stem cell properties and induces EMT through N‐glycosylation of EpCAM in breast cancer cells vol.235, pp.4, 2017, https://doi.org/10.1002/jcp.29252
  15. The Redox Theory of Development vol.32, pp.10, 2020, https://doi.org/10.1089/ars.2019.7976
  16. The Role of Epithelial-to-Mesenchymal Transition in Cutaneous Squamous Cell Carcinoma : Epithelial-to-Mesenchymal Transition in Cutaneous SCC vol.21, pp.6, 2017, https://doi.org/10.1007/s11864-020-00735-x
  17. Defining lung cancer stem cells exosomal payload of miRNAs in clinical perspective vol.12, pp.6, 2017, https://doi.org/10.4252/wjsc.v12.i6.406
  18. Cancer-Associated Fibroblasts: Versatile Players in the Tumor Microenvironment vol.12, pp.9, 2017, https://doi.org/10.3390/cancers12092652
  19. TGF-β induced EMT and stemness characteristics are associated with epigenetic regulation in lung cancer vol.10, pp.None, 2020, https://doi.org/10.1038/s41598-020-67325-7
  20. Hypoxia-Mediated Decrease of Ovarian Cancer Cells Reaction to Treatment: Significance for Chemo- and Immunotherapies vol.21, pp.24, 2020, https://doi.org/10.3390/ijms21249492
  21. MODERN CONCEPTS ON THE ROLE OF HYPOXIA IN THE DEVELOPMENT OF TUMOR RADIORESISTANCE vol.19, pp.6, 2017, https://doi.org/10.21294/1814-4861-2020-19-6-141-147
  22. Cisplatin prevents breast cancer metastasis through blocking early EMT and retards cancer growth together with paclitaxel vol.11, pp.5, 2017, https://doi.org/10.7150/thno.46460
  23. The CXCL12 Crossroads in Cancer Stem Cells and Their Niche vol.13, pp.3, 2017, https://doi.org/10.3390/cancers13030469
  24. HIF-Prolyl Hydroxylase Domain Proteins (PHDs) in Cancer-Potential Targets for Anti-Tumor Therapy? vol.13, pp.5, 2017, https://doi.org/10.3390/cancers13050988
  25. Hypoxia-Induced Cancer Cell Responses Driving Radioresistance of Hypoxic Tumors: Approaches to Targeting and Radiosensitizing vol.13, pp.5, 2017, https://doi.org/10.3390/cancers13051102
  26. hCINAP is potentially a direct target gene of HIF-1 and is required for hypoxia-induced EMT and apoptosis in cervical cancer cells vol.99, pp.2, 2017, https://doi.org/10.1139/bcb-2020-0090
  27. The Cancer Stem Cell Niche in Ovarian Cancer and Its Impact on Immune Surveillance vol.22, pp.8, 2021, https://doi.org/10.3390/ijms22084091
  28. Cancer stem cell characteristics and their potential as therapeutic targets vol.38, pp.7, 2017, https://doi.org/10.1007/s12032-021-01524-8
  29. Hypoxia in Lung Cancer Management: A Translational Approach vol.13, pp.14, 2017, https://doi.org/10.3390/cancers13143421
  30. The epithelial-mesenchymal transition regulators Twist, Slug, and Snail are associated with aggressive tumour features and poor outcome in prostate cancer patients vol.7, pp.3, 2017, https://doi.org/10.1002/cjp2.202
  31. Pluripotency inducing Yamanaka factors: role in stemness and chemoresistance of liver cancer vol.21, pp.8, 2021, https://doi.org/10.1080/14737140.2021.1915137
  32. Prolonged hypoxia switched on cancer stem cell-like plasticity in HepG2 tumourspheres cultured in serum-free media vol.57, pp.9, 2021, https://doi.org/10.1007/s11626-021-00625-y
  33. Vimentin Is at the Heart of Epithelial Mesenchymal Transition (EMT) Mediated Metastasis vol.13, pp.19, 2017, https://doi.org/10.3390/cancers13194985