#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Osteopontin regulates proliferation, apoptosis, and migration of murine claudin-low mammary tumor cells


Background:
Osteopontin is a secreted phosphoglycoprotein that is expressed by a number of normal cells as well as a variety of tumor cells. With respect to breast cancer, osteopontin has been implicated in regulating tumor cell proliferation and migration/metastasis and may serve as a prognostic indicator. However it remains unclear whether osteopontin has the same impact in all breast cancer subtypes and in particular, osteopontin’s effects in claudin-low breast cancer are poorly understood.

Methods:
cDNA microarrays and qRT-PCR were used to evaluate osteopontin expression in mammary tumors from MTB-IGFIR transgenic mice and cell lines derived from these tumors. siRNA was then used to determine the impact of osteopontin knockdown on proliferation, apoptosis and migration in vitro in two murine claudin-low cell lines as well as identify the receptor mediating osteopontin’s physiologic effects.

Results:
Osteopontin was expressed at high levels in mammary tumors derived from MTB-IGFIR transgenic mice compared to normal mammary tissue. Evaluation of cell lines derived from different mammary tumors revealed that mammary tumor cells with claudin-low characteristic expressed high levels of osteopontin whereas mammary tumor cells with mixed luminal and basal-like features expressed lower levels of osteopontin. Reduction of osteopontin levels using siRNA significantly reduced proliferation and migration while increasing apoptosis in the claudin-low cell lines. Osteopontin’s effect appear to be mediated through a receptor containing ITGAV and not through CD44.

Conclusions:
Our data suggests that mammary tumors with a mixed luminal/basal-like phenotype express high levels of osteopontin however this osteopontin appears to be largely produced by non-tumor cells in the tumor microenvironment. In contrast tumor cells with claudin-low characteristics express high levels of osteopontin and a reduction of osteopontin in these cells impaired proliferation, survival and migration.

Keywords:
Osteopontin Breast cancer Claudin-low Proliferation Apoptosis Migration


Autoři: S. Saleh;  D. E. Thompson;  J. Mcconkey;  P. Murray;  R. A. Moorehead *
Vyšlo v časopise: BMC Cancer 2016, 16:359
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1186/s12885-016-2396-9

© The Authors. 2016
Open access
This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
The electronic version of this article is the complete one and can be found online at: http://bmccancer.biomedcentral.com/articles/10.1186/s12885-016-2396-9

Souhrn

Background:
Osteopontin is a secreted phosphoglycoprotein that is expressed by a number of normal cells as well as a variety of tumor cells. With respect to breast cancer, osteopontin has been implicated in regulating tumor cell proliferation and migration/metastasis and may serve as a prognostic indicator. However it remains unclear whether osteopontin has the same impact in all breast cancer subtypes and in particular, osteopontin’s effects in claudin-low breast cancer are poorly understood.

Methods:
cDNA microarrays and qRT-PCR were used to evaluate osteopontin expression in mammary tumors from MTB-IGFIR transgenic mice and cell lines derived from these tumors. siRNA was then used to determine the impact of osteopontin knockdown on proliferation, apoptosis and migration in vitro in two murine claudin-low cell lines as well as identify the receptor mediating osteopontin’s physiologic effects.

Results:
Osteopontin was expressed at high levels in mammary tumors derived from MTB-IGFIR transgenic mice compared to normal mammary tissue. Evaluation of cell lines derived from different mammary tumors revealed that mammary tumor cells with claudin-low characteristic expressed high levels of osteopontin whereas mammary tumor cells with mixed luminal and basal-like features expressed lower levels of osteopontin. Reduction of osteopontin levels using siRNA significantly reduced proliferation and migration while increasing apoptosis in the claudin-low cell lines. Osteopontin’s effect appear to be mediated through a receptor containing ITGAV and not through CD44.

Conclusions:
Our data suggests that mammary tumors with a mixed luminal/basal-like phenotype express high levels of osteopontin however this osteopontin appears to be largely produced by non-tumor cells in the tumor microenvironment. In contrast tumor cells with claudin-low characteristics express high levels of osteopontin and a reduction of osteopontin in these cells impaired proliferation, survival and migration.

Keywords:
Osteopontin Breast cancer Claudin-low Proliferation Apoptosis Migration


Zdroje

1. Kunii Y, Niwa S, Hagiwara Y, Maeda M, Seitoh T, Suzuki T. The immunohistochemical expression profile of osteopontin in normal human tissues using two site-specific antibodies reveals a wide distribution of positive cells and extensive expression in the central and peripheral nervous systems. Med Mol Morphol. 2009;42(3):155–61.

2. Rangaswami H, Bulbule A, Kundu GC. Osteopontin: role in cell signaling and cancer progression. Trends Cell Biol. 2006;16(2):79–87.

3. Wai PY, Kuo PC. Osteopontin: regulation in tumor metastasis. Cancer Metastasis Rev. 2008;27(1):103–18.

4. Cho HJ, Cho HJ, Kim HS. Osteopontin: a multifunctional protein at the crossroads of inflammation, atherosclerosis, and vascular calcification. Curr Atheroscler Rep. 2009;11(3):206–13.

5. Sodek J, Ganss B, McKee MD. Osteopontin. Crit Rev Oral Biol Med. 2000; 11(3):279–303.

6. Tuck AB, Chambers AF, Allan AL. Osteopontin overexpression in breast cancer: knowledge gained and possible implications for clinical management. J Cell Biochem. 2007;102(4):859–68.

7. Weber GF, Lett GS, Haubein NC. Osteopontin is a marker for cancer aggressiveness and patient survival. Br J Cancer. 2010;103(6):861–9.

8. Denhardt DT, Giachelli CM, Rittling SR. Role of osteopontin in cellular signaling and toxicant injury. Annu Rev Pharmacol Toxicol. 2001;41:723–49.

9. Denhardt DT, Noda M, O'Regan AW, Pavlin D, Berman JS. Osteopontin as a means to cope with environmental insults: regulation of inflammation, tissue remodeling, and cell survival. J Clin Invest. 2001;107(9):1055–61.

10. Yokosaki Y, Matsuura N, Sasaki T, Murakami I, Schneider H, Higashiyama S, Saitoh Y, Yamakido M, Taooka Y, Sheppard D. The integrin alpha(9)beta(1) binds to a novel recognition sequence (SVVYGLR) in the thrombin-cleaved amino-terminal fragment of osteopontin. J Biol Chem. 1999;274(51):36328–34.

11. Bellahcene A, Castronovo V. Increased expression of osteonectin and osteopontin, two bone matrix proteins, in human breast cancer. Am J Pathol. 1995;146(1):95–100.

12. Brown LF, Papadopoulos-Sergiou A, Berse B, Manseau EJ, Tognazzi K, Perruzzi CA, Dvorak HF, Senger DR. Osteopontin expression and distribution in human carcinomas. Am J Pathol. 1994;145(3):610–23.

13. Tuck AB, O'Malley FP, Singhal H, Harris JF, Tonkin KS, Kerkvliet N, Saad Z, Doig GS, Chambers AF. Osteopontin expression in a group of lymph node negative breast cancer patients. Int J Cancer. 1998;79(5):502–8.

14. Forootan SS, Foster CS, Aachi VR, Adamson J, Smith PH, Lin K, Ke Y. Prognostic significance of osteopontin expression in human prostate cancer. Int J Cancer. 2006;118(9):2255–61.

15. Hotte SJ, Winquist EW, Stitt L, Wilson SM, Chambers AF. Plasma osteopontin: associations with survival and metastasis to bone in men with hormonerefractory prostate carcinoma. Cancer. 2002;95(3):506–12.

16. Agrawal D, Chen T, Irby R, Quackenbush J, Chambers AF, Szabo M, Cantor A, Coppola D, Yeatman TJ. Osteopontin identified as lead marker of colon cancer progression, using pooled sample expression profiling. J Natl Cancer Inst. 2002;94(7):513–21.

17. Bao LH, Sakaguchi H, Fujimoto J, Tamaya T. Osteopontin in metastatic lesions as a prognostic marker in ovarian cancers. J Biomed Sci. 2007;14(3):373–81.

18. Imano M, Satou T, Itoh T, Sakai K, Ishimaru E, Yasuda A, Peng YF, Shinkai M, Akai F, Yasuda T, et al. Immunohistochemical expression of osteopontin in gastric cancer. J Gastrointest Surg. 2009;13(9):1577–82.

19. Lin F, Li Y, Cao J, Fan S, Wen J, Zhu G, Du H, Liang Y. Overexpression of osteopontin in hepatocellular carcinoma and its relationships with metastasis, invasion of tumor cells. Mol Biol Rep. 2011;38(8):5205–10.

20. Ye QH, Qin LX, Forgues M, He P, Kim JW, Peng AC, Simon R, Li Y, Robles AI, Chen Y, et al. Predicting hepatitis B virus-positive metastatic hepatocellular carcinomas using gene expression profiling and supervised machine learning. Nat Med. 2003;9(4):416–23.

21. Chambers AF, Wilson SM, Kerkvliet N, O'Malley FP, Harris JF, Casson AG. Osteopontin expression in lung cancer. Lung Cancer. 1996;15(3):311–23.

22. Zhao B, Sun T, Meng F, Qu A, Li C, Shen H, Jin Y, Li W. Osteopontin as a potential biomarker of proliferation and invasiveness for lung cancer. J Cancer Res Clin Oncol. 2011;137(7):1061–70.

23. Anborgh PH, Mutrie JC, Tuck AB, Chambers AF. Role of the metastasispromoting protein osteopontin in the tumour microenvironment. J Cell Mol Med. 2010;14(8):2037–44.

24. Pietrowska M, Marczak L, Polanska J, Behrendt K, Nowicka E, Walaszczyk A, Chmura A, Deja R, Stobiecki M, Polanski A, et al. Mass spectrometry-based serum proteome pattern analysis in molecular diagnostics of early stage breast cancer. J Transl Med. 2009;7:60.

25. Shevde LA, Samant RS, Paik JC, Metge BJ, Chambers AF, Casey G, Frost AR, Welch DR. Osteopontin knockdown suppresses tumorigenicity of human metastatic breast carcinoma, MDA-MB-435. Clin Exp Metastasis. 2006;23(2):123–33.

26. Suzuki M, Mose E, Galloy C, Tarin D. Osteopontin gene expression determines spontaneous metastatic performance of orthotopic human breast cancer xenografts. Am J Pathol. 2007;171(2):682–92.

27. Chakraborty G, Jain S, Patil TV, Kundu GC. Down-regulation of osteopontin attenuates breast tumour progression in vivo. J Cell Mol Med. 2008;12(6A): 2305–18.

28. Wang X, Chao L, Ma G, Chen L, Tian B, Zang Y, Sun J. Increased expression of osteopontin in patients with triple-negative breast cancer. Eur J Clin Invest. 2008;38(6):438–46.

29. Rodenburg W, Pennings JL, van Oostrom CT, Roodbergen M, Kuiper RV, Luijten M, de VA. Identification of breast cancer biomarkers in transgenic mouse models: A proteomics approach. Proteomics Clin Appl. 2010;4(6-7):603–12.

30. Jones RA, Campbell CI, Gunther EJ, Chodosh LA, Petrik JJ, Khokha R, Moorehead RA. Transgenic overexpression of IGF-IR disrupts mammary ductal morphogenesis and induces tumor formation. Oncogene. 2007; 26(11):1636–44.

31. Franks SE, Campbell CI, Barnett EF, Siwicky MD, Livingstone J, Cory S, Moorehead RA. Transgenic IGF-IR overexpression induces mammary tumors with basal-like characteristics while IGF-IR independent mammary tumors express a claudin-low gene signature. Oncogene. 2012;31(27):3298–309.

32. Jones RA, Moorehead RA. The impact of transgenic IGF-IR overexpression on mammary development and tumorigenesis. J Mammary Gland Biol Neoplasia. 2008;13(4):407–13.

33. Jones RA, Campbell CI, Wood GA, Petrik JJ, Moorehead RA. Reversibility and recurrence of IGF-IR-induced mammary tumors. Oncogene. 2009;13:407–13.

34. Campbell CI, Thompson DE, Siwicky MD, Moorehead RA. Murine mammary tumor cells with a claudin-low genotype. Cancer Cell Int. 2011;11:28.

35. Jones RA, Campbell CI, Petrik JJ, Moorehead RA. Characterization of a novel primary mammary tumor cell line reveals that cyclin D1 is regulated by the type I insulin-like growth factor receptor. Mol Cancer Res. 2008;6(5):819–28.

36. Watson KL, Stalker L, Jones RA, Moorehead RA. High levels of dietary soy decrease mammary tumor latency and increase incidence in MTB-IGFIR transgenic mice. BMC Cancer. 2015;15:37.

37. Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9(7):671–5.

38. Thompson DE, Siwicky MD, Moorehead RA. Caveolin-1 expression is elevated in claudin-low mammary tumor cells. Cancer Cell Int. 2012;12:6.

39. Campbell CI, Moorehead RA. Mammary tumors that become independent of the type I insulin-like growth factor receptor express elevated levels of platelet-derived growth factor receptors. BMC Cancer. 2011;11:480.

40. Campbell CI, Petrik JJ, Moorehead RA. ErbB2 enhances mammary tumorigenesis, oncogene-independent recurrence and metastasis in a model of IGF-IR-mediated mammary tumorigenesis. Mol Cancer. 2010;9:235.

41. Herschkowitz JI, Zhao W, Zhang M, Usary J, Murrow G, Edwards D, Knezevic J, Greene SB, Darr D, Troester MA, et al. Comparative oncogenomics identifies breast tumors enriched in functional tumor-initiating cells. Proc Natl Acad Sci USA. 2012;109(8):2778–83.

42. Herschkowitz JI, Simin K, Weigman VJ, Mikaelian I, Usary J, Hu Z, Rasmussen KE, Jones LP, Assefnia S, Chandrasekharan S, et al. Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors. Genome Biol. 2007;8(5):R76.

43. Prat A, Parker JS, Karginova O, Fan C, Livasy C, Herschkowitz JI, He X, Perou CM. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res. 2010;12(5):R68.

44. Ribeiro-Silva A, da Costa JP O. Osteopontin expression according to molecular profile of invasive breast cancer: a clinicopathological and immunohistochemical study. Int J Biol Markers. 2008;23(3):154–60.

45. Li NY, Weber CE, Mi Z, Wai PY, Cuevas BD, Kuo PC. Osteopontin upregulates critical epithelial-mesenchymal transition transcription factors to induce an aggressive breast cancer phenotype. J Am Coll Surg. 2013;217(1):17–26. discussion 26.

46. Chu JE, Xia Y, Chin-Yee B, Goodale D, Croker AK, Allan AL. Lung-derived factors mediate breast cancer cell migration through CD44 receptor-ligand interactions in a novel ex vivo system for analysis of organ-specific soluble proteins. Neoplasia. 2014;16(2):180–91.

47. Yang L, Wei L, Zhao W, Wang X, Zheng G, Zheng M, Song X, Zuo W. Downregulation of osteopontin expression by RNA interference affects cell proliferation and chemotherapy sensitivity of breast cancer MDA-MB-231 cells. Mol Med Rep. 2012;5(2):373–6.

48. Zhang H, Guo M, Chen JH, Wang Z, Du XF, Liu PX, Li WH. Osteopontin knockdown inhibits alphav, beta3 integrin-induced cell migration and invasion and promotes apoptosis of breast cancer cells by inducing autophagy and inactivating the PI3K/Akt/mTOR pathway. Cell Physiol Biochem. 2014;33(4):991–1002.

49. Hahnel A, Wichmann H, Kappler M, Kotzsch M, Vordermark D, Taubert H, Bache M. Effects of osteopontin inhibition on radiosensitivity of MDA-MB-231 breast cancer cells. Radiat Oncol. 2010;5:82.

50. Lu JG, Li Y, Li L, Kan X. Overexpression of osteopontin and integrin alphav in laryngeal and hypopharyngeal carcinomas associated with differentiation and metastasis. J Cancer Res Clin Oncol. 2011;137(11):1613–8.

Štítky
Detská onkológia Onkológia
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#