DOI QR코드

DOI QR Code

Emerging role of RUNX3 in the regulation of tumor microenvironment

  • Manandhar, Sarala (Laboratory of Vascular Homeostasis Regulation, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University) ;
  • Lee, You Mie (Laboratory of Vascular Homeostasis Regulation, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University)
  • Received : 2018.01.20
  • Published : 2018.04.30

Abstract

A number of genes have been therapeutically targeted to relieve cancer, but cancer relapse is still a growing issue. The concept that the surrounding tumor environment is critical for the progression of cancer may foster an answer to the issue of cancer malignancy. Runt domain transcription factors (RUNX1, 2, and 3) are evolutionarily conserved and have been intensively studied for their roles in normal development and pathological conditions. During tumor growth, a hypoxic microenvironment and infiltration of the tumor by immune cells are common phenomena. In this review, we briefly introduce the consequences of hypoxia and immune cell infiltration into the tumor microenvironment with a focus on RUNX3 as a critical regulator. Furthermore, based on our current knowledge of the functional role of RUNX3 in hypoxia and immune cell maintenance, a probable therapeutic intervention is suggested for the effective management of tumor growth and malignancy.

Keywords

References

  1. Klemm F and Joyce JA (2015) Microenvironmental regulation of therapeutic response in cancer. Cell Biol 25, 198-213
  2. Balkwill FR, Capasso M and Hagemann T (2012) The tumor microenvironment at a glance. J Cell Sci 125, 5591-5596 https://doi.org/10.1242/jcs.116392
  3. Hockel M and Vaupel P (2001) Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. J Natl Cancer Inst 93, 266-276 https://doi.org/10.1093/jnci/93.4.266
  4. Ito Y (1999) Molecular basis of tissue-specific gene expression mediated by the runt domain transcription factor PEBP2/CBF. Genes Cells 4, 685-696 https://doi.org/10.1046/j.1365-2443.1999.00298.x
  5. Ito Y (2008) RUNX genes in development and cancer: regulation of viral gene expression and the discovery of RUNX family genes. Adv Cancer Res 99, 33-76
  6. Kundu M, Compton S, Garrett-Beal L et al (2005) Runx1 deficiency predisposes mice to T-lymphoblastic lymphoma. Blood 106, 3621-3624 https://doi.org/10.1182/blood-2005-04-1447
  7. Ito Y (2004) Oncogenic potential of the RUNX gene family: 'overview'. Oncogene 23, 4198-4208 https://doi.org/10.1038/sj.onc.1207755
  8. Leong DT, Lim J, Goh X et al (2010) Cancer-related ectopic expression of the bone-related transcription factor RUNX2 in non-osseous metastatic tumor cells is linked to cell proliferation and motility. Cancer Res 12, R89 https://doi.org/10.1186/bcr2762
  9. Woolf E, Xiao C, Fainaru O et al (2003) Runx3 and Runx1 are required for CD8 T cell development during thymopoiesis. Proc Natl Acad Sci U S A 100, 7731-7736 https://doi.org/10.1073/pnas.1232420100
  10. Levanon D, Bettoun D, Harris-Cerruti C et al (2002) The Runx3 transcription factor regulates development and survival of TrkC dorsal root ganglia neurons. EMBO J 21, 3454-3463 https://doi.org/10.1093/emboj/cdf370
  11. Bae SC and Choi JK (2004) Tumor suppressor activity of RUNX3. Oncogene 23, 4336-4340 https://doi.org/10.1038/sj.onc.1207286
  12. Chen F, Bai J, Li W et al (2013) RUNX3 suppresses migration, invasion and angiogenesis of human renal cell carcinoma. PLoS One 8, e56241 https://doi.org/10.1371/journal.pone.0056241
  13. Chen F, Wang M, Bai J et al (2014) Role of RUNX3 in suppressing metastasis and angiogenesis of human prostate cancer. PLoS One 9, e86917 https://doi.org/10.1371/journal.pone.0086917
  14. Chuang LS and Ito Y (2010) RUNX3 is multifunctional in carcinogenesis of multiple solid tumors. Oncogene 29, 2605-2615 https://doi.org/10.1038/onc.2010.88
  15. Kim HJ, Park J, Lee SK, Kim KR, Park KK and Chung WY (2015) Loss of RUNX3 expression promotes cancerassociated bone destruction by regulating CCL5, CCL19 and CXCL11 in non-small cell lung cancer. J Pathol 237, 520-531 https://doi.org/10.1002/path.4597
  16. Li QL, Ito K, Sakakura C et al (2002) Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell 109, 113-124 https://doi.org/10.1016/S0092-8674(02)00690-6
  17. Kudo Y, Tsunematsu T and Takata T (2011) Oncogenic role of RUNX3 in head and neck cancer. J Cell Biochem 112, 387-393 https://doi.org/10.1002/jcb.22967
  18. Lee CW, Chuang LS, Kimura S et al (2011) RUNX3 functions as an oncogene in ovarian cancer. Gynecol Oncol 122, 410-417 https://doi.org/10.1016/j.ygyno.2011.04.044
  19. Park J, Kim HJ, Kim KR et al (2017) Loss of RUNX3 expression inhibits bone invasion of oral squamous cell carcinoma. Oncotarget 8, 9079-9092
  20. Inoue K, Ozaki S, Shiga T et al (2002) Runx3 controls the axonal projection of proprioceptive dorsal root ganglion neurons. Nat Neurosci 5, 946-954 https://doi.org/10.1038/nn925
  21. Lau QC, Raja E, Salto-Tellez M et al (2006) RUNX3 is frequently inactivated by dual mechanisms of protein mislocalization and promoter hypermethylation in breast cancer. Cancer Res 66, 6512-6520 https://doi.org/10.1158/0008-5472.CAN-06-0369
  22. Ito K, Liu Q, Salto-Tellez M et al (2005) RUNX3, a novel tumor suppressor, is frequently inactivated in gastric cancer by protein mislocalization. Cancer Res 65, 7743-7750 https://doi.org/10.1158/0008-5472.CAN-05-0743
  23. Li QL, Kim HR, Kim WJ et al (2004) Transcriptional silencing of the RUNX3 gene by CpG hypermethylation is associated with lung cancer. Biochem Biophys Res Commun 314, 223-228 https://doi.org/10.1016/j.bbrc.2003.12.079
  24. Kim WJ, Kim EJ, Jeong P et al (2005) RUNX3 inactivation by point mutations and aberrant DNA methylation in bladder tumors. Cancer Res 65, 9347-9354 https://doi.org/10.1158/0008-5472.CAN-05-1647
  25. Zhang X, Wang L, Zeng X, Fujita T and Liu W (2017) Runx3 inhibits melanoma cell migration through regulation of cell shape change. Cell Biol International 41, 1048-1055 https://doi.org/10.1002/cbin.10824
  26. Cameron ER and Neil JC (2004) The Runx genes: lineage-specific oncogenes and tumor suppressors. Oncogene 23, 4308-4314 https://doi.org/10.1038/sj.onc.1207130
  27. Salto-Tellez M, Peh BK, Ito K et al (2006) RUNX3 protein is overexpressed in human basal cell carcinomas. Oncogene 25, 7646-7649 https://doi.org/10.1038/sj.onc.1209739
  28. Tsunematsu T, Kudo Y, Iizuka S et al (2009) RUNX3 has an oncogenic role in head and neck cancer. PLoS One 4, e5892 https://doi.org/10.1371/journal.pone.0005892
  29. Bledsoe KL, McGee-Lawrence ME, Camilleri ET et al (2014) RUNX3 facilitates growth of Ewing sarcoma cells. J Cell Physiol 229, 2049-2056 https://doi.org/10.1002/jcp.24663
  30. Hanahan D and Weinberg RA (2000) The hallmarks of cancer. Cell 100, 57-70 https://doi.org/10.1016/S0092-8674(00)81683-9
  31. Hanahan D and Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144, 646-674 https://doi.org/10.1016/j.cell.2011.02.013
  32. Quail DF and Joyce JA (2013) Microenvironmental regulation of tumor progression and metastasis. Nat Med 19, 1423-1437 https://doi.org/10.1038/nm.3394
  33. Hanahan D and Coussens LM (2012) Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell 21, 309-322 https://doi.org/10.1016/j.ccr.2012.02.022
  34. Weber CE and Kuo PC (2012) The tumor microenvironment. Surg Oncol 21, 172-177 https://doi.org/10.1016/j.suronc.2011.09.001
  35. Blagosklonny MV (2004) Antiangiogenic therapy and tumor progression. Cancer Cell 5, 13-17 https://doi.org/10.1016/S1535-6108(03)00336-2
  36. Semenza GL (2000) Hypoxia, clonal selection, and the role of HIF-1 in tumor progression. Crit Rev Biochem Mol Biol 35, 71-103 https://doi.org/10.1080/10409230091169186
  37. Qian BZ and Pollard JW (2010) Macrophage diversity enhances tumor progression and metastasis. Cell 141, 39-51 https://doi.org/10.1016/j.cell.2010.03.014
  38. Condeelis J and Pollard JW (2006) Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell 124, 263-266 https://doi.org/10.1016/j.cell.2006.01.007
  39. Murdoch C, Giannoudis A and Lewis CE (2004) Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood 104, 2224-2234 https://doi.org/10.1182/blood-2004-03-1109
  40. Grivennikov SI, Greten FR and Karin M (2010) Immunity, inflammation, and cancer. Cell 140, 883-899 https://doi.org/10.1016/j.cell.2010.01.025
  41. Chuang LS, Ito K and Ito Y (2013) RUNX family: Regulation and diversification of roles through interacting proteins. Int J Cancer 132, 1260-1271 https://doi.org/10.1002/ijc.27964
  42. Ito Y, Bae SC and Chuang LS (2015) The RUNX family: developmental regulators in cancer. Nat Rev Cancer 15, 81-95 https://doi.org/10.1038/nrc3877
  43. Lotem J, Levanon D, Negreanu V et al (2015) Runx3 at the interface of immunity, inflammation and cancer. Biochim Biophys Acta 1855, 131-143
  44. Afaloniati H, Karagiannis GS, Hardas A, Poutahidis T and Angelopoulou K (2017) Inflammation-driven colon neoplasmatogenesis in uPA-deficient mice is associated with an increased expression of Runx transcriptional regulators. Exp Cell Res 361, 257-264 https://doi.org/10.1016/j.yexcr.2017.10.025
  45. Le XF, Groner Y, Kornblau SM et al (1999) Regulation of AML2/CBFA3 in hematopoietic cells through the retinoic acid receptor alpha-dependent signaling pathway. J Biol Chem 274, 21651-21658 https://doi.org/10.1074/jbc.274.31.21651
  46. Kalev-Zylinska ML, Horsfield JA, Flores MV et al (2003) Runx3 is required for hematopoietic development in zebrafish. Dev Dyn 228, 323-336 https://doi.org/10.1002/dvdy.10388
  47. Facciabene A, Motz GT and Coukos G (2012) T-regulatory cells: key players in tumor immune escape and angiogenesis. Cancer Res 72, 2162-2171 https://doi.org/10.1158/0008-5472.CAN-11-3687
  48. Hossain DM, Panda AK, Chakrabarty S et al (2015) MEK inhibition prevents tumour-shed transforming growth factor-beta-induced T-regulatory cell augmentation in tumour milieu. Immunology 144, 561-573 https://doi.org/10.1111/imm.12397
  49. Dinesh RK, Skaggs BJ, La Cava A, Hahn BH and Singh RP (2010) CD8+ Tregs in lupus, autoimmunity, and beyond. Autoimm Rev 9, 560-568 https://doi.org/10.1016/j.autrev.2010.03.006
  50. Chakraborty S, Panda AK, Bose S et al (2017) Transcriptional regulation of FOXP3 requires integrated activation of both promoter and CNS regions in tumor-induced CD8(+) Treg cells. Sci Rep 7, 1628 https://doi.org/10.1038/s41598-017-01788-z
  51. Egawa T, Tillman RE, Naoe Y, Taniuchi I and Littman DR (2007) The role of the Runx transcription factors in thymocyte differentiation and in homeostasis of naive T cells. J Exp Med 204, 1945-1957 https://doi.org/10.1084/jem.20070133
  52. Cruz-Guilloty F, Pipkin ME, Djuretic IM et al (2009) Runx3 and T-box proteins cooperate to establish the transcriptional program of effector CTLs. J Exp Med 206, 51-59 https://doi.org/10.1084/jem.20081242
  53. Li Y, Ji X, Su Z et al (2014) Downregulation of Runx3 is closely related to the decreased Th1-associated factors in patients with gastric carcinoma. Tumour Biol 35, 12235-12244 https://doi.org/10.1007/s13277-014-2532-6
  54. Massague J (2008) TGFbeta in Cancer. Cell 134, 215-230 https://doi.org/10.1016/j.cell.2008.07.001
  55. Krishnan V, Chong YL, Tan TZ et al (2018) TGF-beta promotes genomic instability after loss of RUNX3. Cancer Res 78, 88-102 https://doi.org/10.1158/0008-5472.CAN-17-1178
  56. Voon DC, Hor YT and Ito Y (2015) The RUNX complex: reaching beyond haematopoiesis into immunity. Immunology 146, 523-536 https://doi.org/10.1111/imm.12535
  57. Mei PJ, Bai J, Liu H et al (2011) RUNX3 expression is lost in glioma and its restoration causes drastic suppression of tumor invasion and migration. J Cancer Res Clin Oncol 137, 1823-1830 https://doi.org/10.1007/s00432-011-1063-4
  58. Kim BR, Kang MH, Kim JL et al (2016) RUNX3 inhibits the metastasis and angiogenesis of colorectal cancer. Oncol Rep 36, 2601-2608 https://doi.org/10.3892/or.2016.5086
  59. Cheng HC, Liu YP, Shan YS et al (2013) Loss of RUNX3 increases osteopontin expression and promotes cell migration in gastric cancer. Carcinogenesis 34, 2452-2459 https://doi.org/10.1093/carcin/bgt218
  60. Sanchez-Martin L, Estecha A, Samaniego R, Sanchez-Ramon S, Vega MA and Sanchez-Mateos P (2011) The chemokine CXCL12 regulates monocyte-macrophage differentiation and RUNX3 expression. Blood 117, 88-97 https://doi.org/10.1182/blood-2009-12-258186
  61. Horikawa N, Abiko K, Matsumura N et al (2017) Expression of Vascular Endothelial Growth Factor in Ovarian Cancer Inhibits Tumor Immunity through the Accumulation of Myeloid-Derived Suppressor Cells. Clin Cancer Res 23, 587-599 https://doi.org/10.1158/1078-0432.CCR-16-0387
  62. Oussa NA, Dahmani A, Gomis M, Richaud M, Andreev E and Navab-Daneshmand AR (2016) VEGF Requires the Receptor NRP-1 To Inhibit Lipopolysaccharide-Dependent Dendritic Cell Maturation. J Immunol 197, 3927-3935 https://doi.org/10.4049/jimmunol.1601116
  63. Peng Z, Wei D, Wang L et al (2006) RUNX3 inhibits the expression of vascular endothelial growth factor and reduces the angiogenesis, growth, and metastasis of human gastric cancer. Clin Cancer Res 12, 6386-6394 https://doi.org/10.1158/1078-0432.CCR-05-2359
  64. Gordan JD, Thompson CB and Simon MC (2007) HIF and c-Myc: sibling rivals for control of cancer cell metabolism and proliferation. Cancer Cell 12, 108-113 https://doi.org/10.1016/j.ccr.2007.07.006
  65. Simon MC and Keith B (2008) The role of oxygen availability in embryonic development and stem cell function. Nat Rev Mol Cell Biol 9, 285-296 https://doi.org/10.1038/nrm2354
  66. Lin Q and Yun Z (2010) Impact of the hypoxic tumor microenvironment on the regulation of cancer stem cell characteristics. Cancer Biol Ther 9, 949-956 https://doi.org/10.4161/cbt.9.12.12347
  67. Lee SH, Kim J, Kim WH and Lee YM (2009) Hypoxic silencing of tumor suppressor RUNX3 by histone modification in gastric cancer cells. Oncogene 28, 184-194 https://doi.org/10.1038/onc.2008.377
  68. Chen H, Yan Y, Davidson TL, Shinkai Y and Costa M (2006) Hypoxic stress induces dimethylated histone H3 lysine 9 through histone methyltransferase G9a in mammalian cells. Cancer Res 66, 9009-9016 https://doi.org/10.1158/0008-5472.CAN-06-0101
  69. Chen MW, Hua KT, Kao HJ et al (2010) H3K9 histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule Ep-CAM. Cancer Res 70, 7830-7840 https://doi.org/10.1158/0008-5472.CAN-10-0833
  70. Liu S, Ye D, Guo W et al (2015) G9a is essential for EMT-mediated metastasis and maintenance of cancer stem cell-like characters in head and neck squamous cell carcinoma. Oncotarget 6, 6887-6901
  71. Dong C, Wu Y, Yao J et al (2012) G9a interacts with Snail and is critical for Snail-mediated E-cadherin repression in human breast cancer. J Clin Invest 122, 1469-1486 https://doi.org/10.1172/JCI57349
  72. Yuan Y, Tang AJ, Castoreno AB et al (2013) Gossypol and an HMT G9a inhibitor act in synergy to induce cell death in pancreatic cancer cells. Cell Death Dis 4, e690 https://doi.org/10.1038/cddis.2013.191
  73. Kim MS, Kwon HJ, Lee YM et al (2001) Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat Med 7, 437-443 https://doi.org/10.1038/86507
  74. Iwatani K, Fujimoto T and Ito T (2010) Cyclin D1 blocks the anti-proliferative function of RUNX3 by interfering with RUNX3-p300 interaction. Biochem Biophys Res Commun 400, 426-431 https://doi.org/10.1016/j.bbrc.2010.08.094
  75. Lee YS, Lee JW, Jang JW et al (2013) Runx3 inactivation is a crucial early event in the development of lung adenocarcinoma. Cancer Cell 24, 603-616 https://doi.org/10.1016/j.ccr.2013.10.003
  76. Huang C, Ida H, Ito K, Zhang H and Ito Y (2007) Contribution of reactivated RUNX3 to inhibition of gastric cancer cell growth following suberoylanilide hydroxamic acid (vorinostat) treatment. Biochem Pharmacol 73, 990-1000 https://doi.org/10.1016/j.bcp.2006.12.013
  77. Shio S, Kodama Y, Ida H et al (2011) Loss of RUNX3 expression by histone deacetylation is associated with biliary tract carcinogenesis. Cancer Sci 102, 776-783 https://doi.org/10.1111/j.1349-7006.2011.01848.x
  78. He L and Hannon GJ (2004) MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genetics 5, 522-531 https://doi.org/10.1038/nrg1379
  79. Kulshreshtha R, Davuluri RV, Calin GA and Ivan M (2008) A microRNA component of the hypoxic response. Cell Death Differ 15, 667-671 https://doi.org/10.1038/sj.cdd.4402310
  80. Nicoloso MS, Spizzo R, Shimizu M, Rossi S and Calin GA (2009) MicroRNAs--the micro steering wheel of tumour metastases. Nat Rev Cancer 9, 293-302 https://doi.org/10.1038/nrc2619
  81. Wentz-Hunter KK and Potashkin JA (2011) The Role of miRNAs as Key Regulators in the Neoplastic Microenvironment. Mol Biol Int 2011, 839872
  82. Paudel D, Zhou W, Ouyang Y et al (2016) MicroRNA-130b functions as a tumor suppressor by regulating RUNX3 in epithelial ovarian cancer. Gene 586, 48-55 https://doi.org/10.1016/j.gene.2016.04.001
  83. Lee SH, Jung YD, Choi YS and Lee YM (2015) Targeting of RUNX3 by miR-130a and miR-495 cooperatively increases cell proliferation and tumor angiogenesis in gastric cancer cells. Oncotarget 6, 33269-33278
  84. Pande S, Ali SA, Dowdy C et al (2009) Subnuclear targeting of the Runx3 tumor suppressor and its epigenetic association with mitotic chromosomes. J Cell Phys 218, 473-479 https://doi.org/10.1002/jcp.21630
  85. Kumar A, Singhal M, Chopra C et al (2016) Threonine 209 phosphorylation on RUNX3 by Pak1 is a molecular switch for its dualistic functions. Oncogene 35, 4857-4865 https://doi.org/10.1038/onc.2016.18
  86. Zhu X, Xu JJ, Hu SS et al (2014) Pim-1 acts as an oncogene in human salivary gland adenoid cystic carcinoma. J Exp Clin Cancer Res 33, 114 https://doi.org/10.1186/s13046-014-0114-5
  87. Tsang YH, Lamb A, Romero-Gallo J et al (2010) Helicobacter pylori CagA targets gastric tumor suppressor RUNX3 for proteasome- mediated degradation. Oncogene 29, 5643-5650 https://doi.org/10.1038/onc.2010.304
  88. Kumar V and Gabrilovich DI (2014) Hypoxia-inducible factors in regulation of immune responses in tumour microenvironment. Immunology 143, 512-519 https://doi.org/10.1111/imm.12380
  89. Lee SH, Bae SC, Kim KW and Lee YM (2014) RUNX3 inhibits hypoxia-inducible factor-1alpha protein stability by interacting with prolyl hydroxylases in gastric cancer cells. Oncogene 33, 1458-1467 https://doi.org/10.1038/onc.2013.76
  90. Zhou WN, Du YF, Bai J et al (2017) RUNX3 plays a tumor suppressor role by inhibiting cell migration, invasion and angiogenesis in oral squamous cell carcinoma. Oncol Rep 38, 2378-2386 https://doi.org/10.3892/or.2017.5857
  91. Bauer O, Hantisteanu S, Lotem J and Groner Y (2014) Carcinogen-induced skin tumor development requires leukocytic expression of the transcription factor Runx3. Cancer Pre Res (Phila) 7, 913-926 https://doi.org/10.1158/1940-6207.CAPR-14-0098-T
  92. Brenner O, Levanon D, Negreanu V et al (2004) Loss of Runx3 function in leukocytes is associated with spontaneously developed colitis and gastric mucosal hyperplasia. Proc Nat Acad Sci U S A 101, 16016-16021 https://doi.org/10.1073/pnas.0407180101
  93. Fainaru O, Woolf E, Lotem J et al (2004) Runx3 regulates mouse TGF-beta-mediated dendritic cell function and its absence results in airway inflammation. EMBO J 23, 969-979 https://doi.org/10.1038/sj.emboj.7600085
  94. Naoe Y, Setoguchi R, Akiyama K et al (2007) Repression of interleukin-4 in T helper type 1 cells by Runx/Cbf beta binding to the Il4 silencer. J Exp Med 204, 1749-1755 https://doi.org/10.1084/jem.20062456
  95. Fan L, Wang X, Fan L et al (2016) MicroRNA-145 influences the balance of Th1/Th2 via regulating RUNX3 in asthma patients. Exp Lung Res 42, 417-424 https://doi.org/10.1080/01902148.2016.1256452
  96. Mantovani A, Allavena P, Sica A and Balkwill F (2008) Cancer-related inflammation. Nature 454, 436-444 https://doi.org/10.1038/nature07205