Biopolym. Cell. 2015; 31(6):429-435.
Genomics, Transcriptomics and Proteomics
Heterogeneity of premetastatic niches gene expression in breast cancer cells
1Tashireva L. A., 2, 3Denisov E. V., 1, 3Savelieva O. E., 2, 3Geraschenko T. S., 1, 3, 4Zavyalova M. V., 1, 4Perelmuter V. M.
  1. Dep of Pathological Anatomy and Cytology
    Tomsk Cancer Research Institute
    5, Kooperativny Str, Tomsk, Russian Federation, 634050
  2. Laboratory of Molecular Oncology and Immunology
    Tomsk Cancer Research Institute
    5, Kooperativny Str, Tomsk, Russian Federation, 634050
  3. Tomsk State University
    36, Lenina ave, Tomsk, Russian Federation, 634050
  4. Siberian State Medical University
    2, Moskovsky tr., Tomsk, Russian Federation, 634050

Abstract

Aim. To investigate the expression of the genes TGFB1, TNF, CSF1, CSF2, VEGFA and HIF1A in the patients with invasive breast carcinoma of no special type considering the intratumoral morphological heterogeneity. Methods. The technology of laser capture microdissection PALM was used to isolate five types of morphological tumor structures from three patients with invasive carcinoma of no special type (IC NST), luminal A subtype, T1-2NxMx. The level of expression of the cytokine (TNF), growth factor genes (TGFB1, CSF1, CSF2, VEGFA) and the HIF1A gene was assessed in the samples obtained using real-time PCR, TaqMan-probes and specific oligonucleotides. Results. The study demonstrated the absence of the expression of the growth factor gene CSF2 in tumor cells of IC NST, and the expression of the gene CSF1, independent from the metastasis status and tumor structure type. The prevalence of the expression of the genes VEGFA and TGFB1 was revealed in the alveolar and solid structures along with the rare expression of the gene TNF. Conclusions. The expression of pre-metastatic niche genes in the tumors of patients with IC NST is heterogeneous. The hypoxia-mediated change in the cytokine gene expression may be expected in the alveolar and solid structures, which ultimately results in the formation of microenvironment, facilitating tumor growth and the formation of tumor metastatic potential.
Keywords: breast cancer, pre-metastatic niche, morphological heterogeneity

References

[1] Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, MacDonald DD, Jin DK, Shido K, Kerns SA, Zhu Z, Hicklin D, Wu Y, Port JL, Altorki N, Port ER, Ruggero D, Shmelkov SV, Jensen KK, Rafii S, Lyden D. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438(7069):820-7.
[2] Barcellos-Hoff MH, Lyden D, Wang TC. The evolution of the cancer niche during multistage carcinogenesis. Nat Rev Cancer. 2013;13(7):511-8.
[3] Catalano V, Turdo A, Di Franco S, Dieli F, Todaro M, Stassi G. Tumor and its microenvironment: a synergistic interplay. Semin Cancer Biol. 2013;23(6 Pt B):522-32.
[4] Marçola M, Rodrigues CE. Endothelial progenitor cells in tumor angiogenesis: another brick in the wall. Stem Cells Int. 2015;2015:832649.
[5] Danova M, Aglietta M. Cytokine receptors, growth factors and cell cycle in human bone marrow and peripheral blood hematopoietic progenitors. Haematologica. 1997;82(5):622-9.
[6] Boulais PE, Frenette PS. Making sense of hematopoietic stem cell niches. Blood. 2015;125(17):2621-9.
[7] Crane JL, Cao X. Bone marrow mesenchymal stem cells and TGF-β signaling in bone remodeling. J Clin Invest. 2014;124(2):466-72.
[8] Kidd S, Spaeth E, Watson K, Burks J, Lu H, Klopp A, Andreeff M, Marini FC. Origins of the tumor microenvironment: quantitative assessment of adipose-derived and bone marrow-derived stroma. PLoS One. 2012;7(2):e30563.
[9] Hass R, Otte A. Mesenchymal stem cells as all-round supporters in a normal and neoplastic microenvironment. Cell Commun Signal. 2012;10(1):26.
[10] Gerashchenko TS, Denisov EV, Litviakov NV, Zavyalova MV, Vtorushin SV, Tsyganov MM, Perelmuter VM, Cherdyntseva NV. Intratumor heterogeneity: nature and biological significance. Biochemistry (Mosc). 2013;78(11):1201-15.
[11] Zavjalova MV, Perelmuter VM, Slonimskaya EM, Vtorushin SV, Garbukov EYu, Gluschenko SA. Conjugation of lymphogenous metastatic spread and histologic pattern of infiltrative component of ductal breast cancer. Siberian Journal of Oncology. 2006; 1: 32–5.
[12] Zavyalova MV, Perelmuter VM, Vtorushin SV, Denisov EV, Litvyakov NV, Slonimskaya EM, Cherdyntseva NV. The presence of alveolar structures in invasive ductal NOS breast carcinoma is associated with lymph node metastasis. Diagn Cytopathol. 2013;41(3):279-82.
[13] Zavyalova MV, Denisov EV, Tashireva LA, Gerashchenko TS, Litviakov NV, Skryabin NA, Vtorushin SV, Telegina NS, Slonimskaya EM, Cherdyntseva NV, Perelmuter VM. Phenotypic drift as a cause for intratumoral morphological heterogeneity of invasive ductal breast carcinoma not otherwise specified. Biores Open Access. 2013;2(2):148-54.
[14] Denisov EV, Litviakov NV, Zavyalova MV, Perelmuter VM, Vtorushin SV, Tsyganov MM, Gerashchenko TS, Garbukov EY, Slonimskaya EM, Cherdyntseva NV. Intratumoral morphological heterogeneity of breast cancer: neoadjuvant chemotherapy efficiency and multidrug resistance gene expression. Sci Rep. 2014;4:4709.
[15] Denisov EV, Gerashchenko TS, Zavyalova MV, Litviakov NV, Tsyganov MM, Kaigorodova EV, Slonimskaya EM, Kzhyshkowska J, Cherdyntseva NV, Perelmuter VM. Invasive and drug resistant expression profile of different morphological structures of breast tumors. Neoplasma. 2015;62(3):405-11.
[16] Lakhani SR, Ellis IO, Schnitt SJ, Tan PH, van de Vijver MJ. World Health Organization (WHO) classification of tumours of the breast. 4th Ed. Lyon: IARC Press, 2012. 240 p.
[17] Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 2001;29(9):e45.
[18] Perelmuter VM, Zavyalova MV, Vtorushin SV, Slonimskaya EM, Savenkova OV. Interaction between morphologic heterogeneity of infiltrating ductal breast carcinoma and various forms of tumor progression. Siberian Journal of Oncology. 2007; 3: 58–63.
[19] Perelmuter VM, Manskikh VN. Preniche as missing link of the metastatic niche concept explaining organ-preferential metastasis of malignant tumors and the type of metastatic disease. Biochemistry (Mosc). 2012;77(1):111-8.
[20] Perelmuter VM, Manskikh VN. The Concept of a preniche for localization of future metastases. tumors of the central nervous system types of tumors, diagnosis, ultrasonography, surgery, brain metastasis, and general CNS diseases. Springer, 2014. 308 p.
[21] Turner MD, Nedjai B, Hurst T, Pennington DJ. Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease. Biochim Biophys Acta. 2014;1843(11):2563-2582.
[22] Gonzalez DM, Medici D. Signaling mechanisms of the epithelial-mesenchymal transition. Sci Signal. 2014;7(344):re8.
[23] Marie-Egyptienne DT, Lohse I, Hill RP. Cancer stem cells, the epithelial to mesenchymal transition (EMT) and radioresistance: potential role of hypoxia. Cancer Lett. 2013;341(1):63-72.
[24] Feng Z, Li R, Shi H, Bi W, Hou W, Zhang X. Combined silencing of TGF-β2 and Snail genes inhibit epithelial-mesenchymal transition of retinal pigment epithelial cells under hypoxia. Graefes Arch Clin Exp Ophthalmol. 2015;253(6):875-84.
[25] Lee SH, Jeong D, Han YS, Baek MJ. Pivotal role of vascular endothelial growth factor pathway in tumor angiogenesis. Ann Surg Treat Res. 2015;89(1):1-8.
[26] Chaturvedi P, Gilkes DM, Takano N, Semenza GL. Hypoxia-inducible factor-dependent signaling between triple-negative breast cancer cells and mesenchymal stem cells promotes macrophage recruitment. Proc Natl Acad Sci U S A. 2014;111(20):E2120-9.
[27] Fancke B, Suter M, Hochrein H, O'Keeffe M. M-CSF: a novel plasmacytoid and conventional dendritic cell poietin. Blood. 2008;111(1):150-9.
[28] Quatromoni JG, Eruslanov E. Tumor-associated macrophages: function, phenotype, and link to prognosis in human lung cancer. Am J Transl Res. 2012;4(4):376-89.