DOI QR코드

DOI QR Code

Zinc finger protein 143 expression is closely related to tumor malignancy via regulating cell motility in breast cancer

  • Paek, A Rome (Translational Research Branch, Division of Translational Science, Research Institute, National Cancer Center) ;
  • Mun, Ji Young (Department of Biomedical Laboratory Science (Seongnam campus) Eulji University) ;
  • Hong, Kyeong-Man (Omics Core Laboratory, Research Institute, National Cancer Center) ;
  • Lee, Jongkeun (Clinical Genomics Analysis Branch, Research Institute, National Cancer Center) ;
  • Hong, Dong Wan (Clinical Genomics Analysis Branch, Research Institute, National Cancer Center) ;
  • You, Hye Jin (Translational Research Branch, Division of Translational Science, Research Institute, National Cancer Center)
  • Received : 2017.09.05
  • Accepted : 2017.10.09
  • Published : 2017.12.31

Abstract

We previously reported the involvement of zinc-finger protein 143 (ZNF143) on cancer cell motility in colon cancer cells. Here, ZNF143 was further characterized in breast cancer. Immunohistochemistry was used to determine the expression of ZNF143 in normal tissues and in tissues from metastatic breast cancer at various stages. Notably, ZNF143 was selectively expressed in duct and gland epithelium of normal breast tissues, which decreased when the tissue became malignant. To determine the molecular mechanism how ZNF143 affects breast cancer progression, it was knocked down by infecting benign breast cancer cells with short-hairpin (sh) RNA-lentiviral particles against ZNF143 (MCF7 sh-ZNF143). MCF7 sh-ZNF143 cells showed different cell-cell contacts and actin filament (F-actin) structures when compared with MCF7 sh-Control cells. In migration and invasion assays, ZNF143 knockdown induced increased cellular motility in breast carcinoma cells. This was reduced by the recovery of ZNF143 expression. Taken together, these results suggest that ZNF143 expression contributes to breast cancer progression.

Keywords

References

  1. Li CI, Uribe DJ and Daling JR (2005) Clinical characteristics of different histologic types of breast cancer. Br J Cancer 93, 1046-1052 https://doi.org/10.1038/sj.bjc.6602787
  2. Pestalozzi BC, Zahrieh D, Mallon E et al (2008) Distinct clinical and prognostic features of infiltrating lobular carcinoma of the breast: combined results of 15 International Breast Cancer Study Group clinical trials. J Clin Oncol 26, 3006-3014 https://doi.org/10.1200/JCO.2007.14.9336
  3. Perou CM, Sorlie T, Eisen MB et al (2000) Molecular portraits of human breast tumours. Nature 406, 747-752 https://doi.org/10.1038/35021093
  4. Gatza ML, Silva GO, Parker JS, Fan C and Perou CM (2014) An integrated genomics approach identifies drivers of proliferation in luminal-subtype human breast cancer. Nat Genet 46, 1051-1059 https://doi.org/10.1038/ng.3073
  5. Cancer Genome Atlas N (2012) Comprehensive molecular portraits of human breast tumours. Nature 490, 61-70 https://doi.org/10.1038/nature11412
  6. Ciriello G, Gatza ML, Beck AH et al (2015) Comprehensive Molecular Portraits of Invasive Lobular Breast Cancer. Cell 163, 506-519 https://doi.org/10.1016/j.cell.2015.09.033
  7. Heng YJ, Lester SC, Tse GM et al (2017) The molecular basis of breast cancer pathological phenotypes. J Pathol 241, 375-391 https://doi.org/10.1002/path.4847
  8. Lamouille S, Xu J and Derynck R (2014) Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 15, 178-196 https://doi.org/10.1038/nrm3758
  9. Kalluri R and Weinberg RA (2009) The basics of epithelial-mesenchymal transition. J Clin Invest 119, 1420-1428 https://doi.org/10.1172/JCI39104
  10. Nieto MA and Cano A (2012) The epithelial-mesenchymal transition under control: global programs to regulate epithelial plasticity. Semin Cancer Biol 22, 361-368 https://doi.org/10.1016/j.semcancer.2012.05.003
  11. Huang RY, Guilford P and Thiery JP (2012) Early events in cell adhesion and polarity during epithelial-mesenchymal transition. J Cell Sci 125, 4417-4422 https://doi.org/10.1242/jcs.099697
  12. Myslinski E, Krol A and Carbon P (1998) ZNF76 and ZNF143 are two human homologs of the transcriptional activator Staf. J Biol Chem 273, 21998-22006 https://doi.org/10.1074/jbc.273.34.21998
  13. Bailey SD, Zhang X, Desai K et al (2015) ZNF143 provides sequence specificity to secure chromatin interactions at gene promoters. Nat Commun 2, 6186
  14. Halbig KM, Lekven AC and Kunkel GR (2012) The transcriptional activator ZNF143 is essential for normal development in zebrafish. BMC Mol Biol 13, 3 https://doi.org/10.1186/1471-2199-13-3
  15. Myslinski E, Gerard MA, Krol A and Carbon P (2006) A genome scale location analysis of human Staf/ZNF143- binding sites suggests a widespread role for human Staf/ZNF143 in mammalian promoters. J Biol Chem 281, 39953-39962 https://doi.org/10.1074/jbc.M608507200
  16. Ngondo-Mbongo RP, Myslinski E, Aster JC and Carbon P (2013) Modulation of gene expression via overlapping binding sites exerted by ZNF143, Notch1 and THAP11. Nucleic Acids Res 41, 4000-4014 https://doi.org/10.1093/nar/gkt088
  17. Vinckevicius A, Parker JB and Chakravarti D (2015) Genomic Determinants of THAP11/ZNF143/HCFC1 Complex Recruitment to Chromatin. Mol Cell Biol 35, 4135-4146 https://doi.org/10.1128/MCB.00477-15
  18. Ishiguchi H, Izumi H, Torigoe T et al (2004) ZNF143 activates gene expression in response to DNA damage and binds to cisplatin-modified DNA. Int J Cancer 111, 900-909 https://doi.org/10.1002/ijc.20358
  19. Kawatsu Y, Kitada S, Uramoto H et al (2014) The combination of strong expression of ZNF143 and high MIB-1 labelling index independently predicts shorter disease-specific survival in lung adenocarcinoma. Br J Cancer 110, 2583-2592 https://doi.org/10.1038/bjc.2014.202
  20. Myslinski E, Gerard MA, Krol A and Carbon P (2007) Transcription of the human cell cycle regulated BUB1B gene requires hStaf/ZNF143. Nucleic Acids Res 35, 3453-3464 https://doi.org/10.1093/nar/gkm239
  21. Paek AR, Lee CH and You HJ (2014) A role of zinc-finger protein 143 for cancer cell migration and invasion through ZEB1 and E-cadherin in colon cancer cells. Mol Carcinog 53 Suppl 1, E161-168 https://doi.org/10.1002/mc.22083
  22. Lu W, Chen Z, Zhang H, Wang Y, Luo Y and Huang P (2012) ZNF143 transcription factor mediates cell survival through upregulation of the GPX1 activity in the mitochondrial respiratory dysfunction. Cell Death Dis 3, e422 https://doi.org/10.1038/cddis.2012.156
  23. Izumi H, Yasuniwa Y, Akiyama M et al (2011) Forced Expression of ZNF143 Restrains Cancer Cell Growth. Cancers (Basel) 3, 3909-3920 https://doi.org/10.3390/cancers3043909
  24. Wei S, Wang L, Zhang L et al (2016) ZNF143 enhances metastasis of gastric cancer by promoting the process of EMT through PI3K/AKT signaling pathway. Tumour Biol 77, 12813-12821
  25. Ngondo RP and Carbon P (2014) Transcription factor abundance controlled by an auto-regulatory mechanism involving a transcription start site switch. Nucleic Acids Res 42, 2171-2184 https://doi.org/10.1093/nar/gkt1136
  26. Chambers I, Frampton J, Goldfarb P, Affara N, McBain W and Harrison PR (1986) The structure of the mouse glutathione peroxidase gene: the selenocysteine in the active site is encoded by the 'termination' codon, TGA. EMBO J 5, 1221-1227
  27. Gyorffy B, Lanczky A, Eklund AC et al (2010) An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients. Breast Cancer Res Treat 123, 725-731 https://doi.org/10.1007/s10549-009-0674-9
  28. Neve RM, Chin K, Fridlyand J et al (2006) A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 10, 515-527 https://doi.org/10.1016/j.ccr.2006.10.008
  29. Paek AR, Kim SH, Kim SS, Kim KT and You HJ (2010) IGF-1 induces expression of zinc-finger protein 143 in colon cancer cells through phosphatidylinositide 3-kinase and reactive oxygen species. Exp Mol Med 42, 696-702 https://doi.org/10.3858/emm.2010.42.10.068
  30. Choi JA, Jung YS, Kim JY, Kim HM and Lim IK (2016) Inhibition of breast cancer invasion by TIS21/BTG2/Pc3-Akt1-Sp1-Nox4 pathway targeting actin nucleators, mDia genes. Oncogene 35, 83-93 https://doi.org/10.1038/onc.2015.64
  31. Blanchoin L, Boujemaa-Paterski R, Sykes C and Plastino J (2014) Actin dynamics, architecture, and mechanics in cell motility. Physiol Rev 94, 235-263 https://doi.org/10.1152/physrev.00018.2013
  32. Li J, Yang B, Zhou Q et al (2013) Autophagy promotes hepatocellular carcinoma cell invasion through activation of epithelial-mesenchymal transition. Carcinogenesis 34, 1343-1351 https://doi.org/10.1093/carcin/bgt063
  33. Freije JM, Diez-Itza I, Balbin M et al (1994) Molecular cloning and expression of collagenase-3, a novel human matrix metalloproteinase produced by breast carcinomas. J Biol Chem 269, 16766-16773
  34. Pivetta E, Scapolan M, Pecolo M et al (2011) MMP-13 stimulates osteoclast differentiation and activation in tumour breast bone metastases. Breast Cancer Res 13, R105 https://doi.org/10.1186/bcr3047
  35. Alexandrova AY (2014) Plasticity of tumor cell migration: acquisition of new properties or return to the past? Biochemistry (Mosc) 79, 947-963 https://doi.org/10.1134/S0006297914090107
  36. Chhabra ES and Higgs HN (2007) The many faces of actin: matching assembly factors with cellular structures. Nat Cell Biol 9, 1110-1121 https://doi.org/10.1038/ncb1007-1110
  37. Lee JJ, Kim M and Kim HP (2016) Epigenetic regulation of long noncoding RNA UCA1 by SATB1 in breast cancer. BMB Rep 49, 578-583 https://doi.org/10.5483/BMBRep.2016.49.10.156
  38. Lee CS, Ghim J, Song P, Suh P-G and Ryu SH (2016) Loss of phospholipase D2 impairs VEGF-induced angiogenesis. BMB Rep 49, 191-196 https://doi.org/10.5483/BMBRep.2016.49.3.219
  39. Li B and Dewey CN (2011) RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12, 323 https://doi.org/10.1186/1471-2105-12-323