Skip to main content
Log in

Transcriptional regulation of osteopontin and the metastatic phenotype: Evidence for a Ras-activated enhancer in the human OPN promoter

  • Published:
Clinical & Experimental Metastasis Aims and scope Submit manuscript

Abstract

Elevated osteopontin (OPN) transcription often correlates with increased metastatic potential of transformed cells, and in several model systems OPN – whether produced by the tumor cells or by stromal cells – has been shown to enhance metastatic ability. Sequence elements in the OPN promoter have been identified on the basis of their ability to interact with protein factors associated with the tumorigenic process in one or more cell lineages. One of these is a Ras-activated enhancer (RAE) that binds a protein, the Ras-response factor (RRF), whose ability to form a complex with the RAE is stimulated by Ras signaling in fibroblasts and epithelial cells. Another is the T cell factor-4 binding site, which in the OPN promoter can retard OPN transcription when bound by the Tcf-4 protein. In Rama 37 rat mammary epithelial cells Tcf-4 suppresses OPN transcription and the metastatic phenotype. A third promoter segment consists of two sequences in the −94 to −24 region of the human OPN promoter able to bind several known transcription factors, including Sp1, Myc and Oct-1, which may act synergistically to stimulate OPN transcription in malignant astrocytic cells. Although expression of other genes may also be regulated by these transcription factors, evidence suggests that often OPN alone can stimulate metastasis. In this communication we address two issues: (1) How does OPN facilitate the metastatic phenotype? (2) What mechanisms are responsible for the increase in OPN transcription in metastatic cells?

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Senger DR, Perruzzi CA, Gracey CF et al. Secreted phosphoproteins associated with neoplastic transformation: Close homology with plasma proteins cleaved during blood coagulation. Cancer Res 1988; 48: 5770–4.

    Google Scholar 

  2. Chambers AF, Tuck AB. Ras-responsive genes and tumor metastasis. Crit Rev Oncogen 1993; 4: 95–114.

    Google Scholar 

  3. Denhardt DT, Guo X. Osteopontin a protein with diverse functions. FASEB J 1993; 7: 1475–82.

    Google Scholar 

  4. Oates AJ, Barraclough R, Rudland PS. The role of osteopontin in tumorigenesis and metastasis. Invasion Metastasis 1997; 17: 1–15.

    Google Scholar 

  5. Uede T, Katagiri Y, Iizuka J et al. Osteopontin, a coordinator of host defense system: A cytokine or an extracellular adhesive protein? Microbiol Immunol 1997; 41: 641–8.

    Google Scholar 

  6. Rittling SR, Denhardt DT. Osteopontin function in pathology: Lessons from OPN-deficient mice. Expt Nephrology 1999; 7: 103–13.

    Google Scholar 

  7. Noda M, Denhardt DT. Osteopontin. In Bilezikian JP, Raisz LG, Rodan GA (eds): Principles of Bone Biology, 2nd ed. San Diego: Academic Press 2002.

    Google Scholar 

  8. Chabas D, Baranzini SE, Mitchell D et al. The influence of the proin-flammatory cytokine, osteopontin, on autoimmune demyelinating disease. Science 2001; 294: 1731–5.

    Google Scholar 

  9. Yumoto K, Ishijima M, Rittling SR et al. Osteopontin deficiency protects joints against destruction in anti-type II collagen antibodyinduced arthritis in mice. Proc Natl Acad Sci USA 2002; 99: 4556–61.

    Google Scholar 

  10. Giachelli CM, Steitz S. Osteopontin: A versatile regulator of inflammation and biomineralization. Matrix Biol 2000; 19: 615–22.

    Google Scholar 

  11. Weber GF. The metastasis gene osteopontin: A candidate target for cancer therapy. Biochim Biophys Acta 2001; 1552: 61–85.

    Google Scholar 

  12. Denhardt DT, Noda M, O’Regan AW et al. Osteopontin-a means to cope with environmental insults: Regulation of inflammation tissue remodeling and cell survival. J Clin Invest 2001; 107: 1055–61.

    Google Scholar 

  13. Tuck AB, Chambers AF. The role of osteopontin in breast cancer: Clinical and experimental studies. J Mammary Gland Biol Neoplasia 2001; 6: 419–29.

    Google Scholar 

  14. Furger KA, Menon RK, Tuck AB et al. The functional and clinical roles of osteopontin in cancer and metastasis. Curr Mol Med 2001; 1: 621–32.

    Google Scholar 

  15. Rittling SR, Chen Y, Feng F, Wu Y. Tumor-derived osteopontin is soluble, not matrix associated. J Biol Chem 2002; 277: 9175–82.

    Google Scholar 

  16. Katagiri YU, Sleeman J, Fujii H et al. CD44 variants but not CD44s cooperate with beta1-containing integrins to permit cells to bind to osteopontin independently of arginine-glycine-aspartic acid, thereby stimulating cell motility and chemotaxis. Cancer Res 1999; 59: 219–26.

    Google Scholar 

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

    Google Scholar 

  18. Suzuki K, Zhu B, Rittling SR et al. Colocalization of intracellular osteopontin with CD44 is associated with migration, cell fusion, and resorption in osteoclasts. J Bone Miner Res 2002; 17:1486–97.

    Google Scholar 

  19. Denhardt DT, Lopez CA, Rollo EE et al. Osteopontin-induced modi-fications of cellular functions. Ann NY Acad Sci 1995; 760: 127–42.

    Google Scholar 

  20. Noiri E, Dickman K, Miller F et al. Reduced tolerance to acute renal ischemia in mice with a targeted disruption of the osteopontin gene. Kidney Int 1999; 56: 74–82.

    Google Scholar 

  21. Scatena M, Almeida M, Chaisson ML et al. NF-κB mediates ανβЗ integrin-induced endothelial cell survival. J Cell Biol 1998; 141: 1083–93.

    Google Scholar 

  22. Lin Y-H, Yang-Yen H-F. The osteopontin-CD44 survival signal involves activation of the phosphatidylinositol-3-kinase/Akt signaling pathway. J Biol Chem 2001; 276: 46024–30.

    Google Scholar 

  23. Khan SA, Lopez-Chua CA, Zhang J, Fisher LW, Sorensen ES, Denhardt DT. Soluble osteopontin inhibits apoptosis of adherent endothelial cells deprived of growth factors. J Cellul Biochem 2002; 85: 728–36.

    Google Scholar 

  24. Senger DR, Asch BB, Smith BD et al. A secreted phosphoprotein marker for neoplastic transformation of both epithelial and fibroblastic cells. Nature 1983; 302: 714–5.

    Google Scholar 

  25. Brown LF, Papadopoulos-Sergiou A, Berse B et al. Osteopontin expression and distribution in human carcinomas. Am J Pathol 1994; 145: 610–23.

    Google Scholar 

  26. Bellahcene A, Castronovo V. Increased expression of osteonectin and osteopontin two bone matrix proteins in human breast cancer. Am J Pathol 1996; 146: 95–100.

    Google Scholar 

  27. Casson AG, Wilson SM, McCart JA et al. Ras mutation and expression of the ras-regulated genes osteopontin and cathepsin L in human esophageal cancer. Int J Cancer 1997; 72: 739–45.

    Google Scholar 

  28. Gillespie MT, Thomas RJ, Zhou PU et al. Calcitonin receptors bone sialoprotein and osteopontin are expressed in primary breast cancers. Int J Cancer 1997; 10: 812–815.

    Google Scholar 

  29. Tuck AB, O’Malley FP, Singhal H et al. Osteopontin expression in a group of lymph node negative breast cancer patients. Int J Cancer 1998; 79: 502–8.

    Google Scholar 

  30. Sung V, Gilles C, Murray A et al. The LCC15-MB human breast cancer cell line expresses osteopontin and exhibits invasive and metastatic phenotype. Exp Cell Res 1998; 241: 273–84.

    Google Scholar 

  31. Sharp JA, Sung V, Slavin J et al. Tumor cells are the source of osteopontin and bone sialoprotein expression in human breast cancer. Lab Invest 1999; 79: 869–77.

    Google Scholar 

  32. Price JE, Polyzos A, Zhang RD et al. Tumorigenicity and metastasis of human breast carcinoma cell lines in nude mice. Cancer Res 1990; 50: 717–21.

    Google Scholar 

  33. Zhang RD, Fidler IJ, Price JE. Relative malignant potential of human breast carcinoma cell lines established from pleural effusions and a brain metastasis. Invasion Metastasis 1991; 11: 204–15.

    Google Scholar 

  34. Price JE. Metastasis from human breast cancer cell lines. Breast Cancer Res Treat 1996; 39: 93–102.

    Google Scholar 

  35. Bautista DS, Xuan JW, Hota C et al. Inhibition of Arg-Gly-Asp (RGD)-mediated cell adhesion to osteopontin by a monoclonal antibody against osteopontin. J Biol Chem 1994; 269: 23280–5.

    Google Scholar 

  36. Tuck AB, Arsenault DM, O’Malley FP et al. Osteopontin induces increased invasiveness and plasminogen activator expression of human mammary epithelial cells. Oncogene 1999; 18: 4237–46.

    Google Scholar 

  37. Yoneda T, Williams PJ, Niewolna M. Promotion of angiogenesis and enhancement of breast cancer metastasis to bone. Bone 1998; 23/5S: S201.

    Google Scholar 

  38. Mukhopadhyay R, Price JE. Stable expression of antisense osteopontin inhibits the growth of human breast cancer cells. Proc Am Assoc Cancer Res 1999; 40: 448.

    Google Scholar 

  39. Senger DR, Perruzzi CA, Papadopoulos A. Elevated expression of secreted phosphoprotein I (osteopontin, 2ar) as a consequence of neoplastic transformation. Anticancer Res 1989; 9: 1291–300.

    Google Scholar 

  40. Chambers AF, Tuck AB. Osteopontin: A Ras-regulated gene that contributes to tumor metastasis. InWelch DR (ed): Cancer Metastasis-Related Genes. Dordrecht, The Netherlands: Kluwer Academic Publishers 2002; 231–46.

    Google Scholar 

  41. Hwang S-M, Lopez CA, Heck DE et al. Osteopontin inhibits induction of nitric oxide synthase gene expression by inflammatory mediators in mouse kidney epithelial cells. J Biol Chem 1994; 269: 711–5.

    Google Scholar 

  42. Rollo EE, Laskin DL, Denhardt DT. Osteopontin inhibits nitric oxide production and cytotoxicity by activated RAW2647 macrophages. J Leuk Biol 1996; 60: 397–404.

    Google Scholar 

  43. Scott JA, Weir ML, Wilson SM et al. Osteopontin inhibits inducible nitric oxide synthase activity in rat vascular tissue. AmJ Physiol 1998; 275: H2258–65.

    Google Scholar 

  44. Denhardt DT, Chambers AF. Overcoming obstacles to metastasis — defenses against host defenses: Osteopontin (OPN) as a shield against attack by cytotoxic host cells. J Cell Biochem 1994; 56: 48–51.

    Google Scholar 

  45. Yue TL, McKenna PJ, Ohlstein EH et al. Osteopontin-stimulated vascular smooth muscle cell migration is mediated by β3 integrin. Exp Cell Res 1994; 214: 459–64.

    Google Scholar 

  46. Liaw L, Skinner MP, Raines EW et al. The adhesive and migratory effects of osteopontin are mediated via distinct cell surface integrins: Role of ανβ3 in smooth muscle cell migration to osteopontin in vitro. J Clin Invest 1995; 95: 713–24.

    Google Scholar 

  47. Senger DR, Perruzzi CA. Cell migration promoted by a potent GRGDS-containing thrombin-cleavage fragment of osteopontin. Biochim Biophys Acta 1996; 1314: 13–24.

    Google Scholar 

  48. Weintraub AS, Giachelli CM, Krauss RS et al. Autocrine secretion of osteopontin by vascular smooth muscle cells regulates their adhesion to collagen gels. Am J Path 1996; 149: 259–72.

    Google Scholar 

  49. Tuck AB, Elliott BE, Hota C et al. Osteopontin-induced integrindependent migration of human mammary epithelial cells involves activation of the hepatocyte growth factor receptor. J Cell Biochem 2000; 78: 465–75.

    Google Scholar 

  50. Tuck AB, Hota C, Chambers AF. Osteopontin-induced increase in human mammary epithelial cell invasiveness is urokinase (uPa)-dependent. Breast Cancer Res Treat 2001; 70: 197–204.

    Google Scholar 

  51. Takahashi F, Akutagawa S, Fukumoto H et al. Osteopontin induces angiogenesis of murine neuroblastoma cells in mice. Int J Cancer 2002; 98: 707–12.

    Google Scholar 

  52. Chambers AF, Wilson SM, Kerkvleit N et al. Osteopontin expression in lung cancer. Lung Cancer 1996; 15: 311–23.

    Google Scholar 

  53. Rudland PS, Platt-Higgins A, El-Tanani M et al. Prognostic significance of the metastasis-associated protein osteopontin in human breast cancer. Cancer Res 2002; 62: 3417–27.

    Google Scholar 

  54. Kim JH, Skates SJ, Uede T et al. Osteopontin as a potential diagnostic biomarker for ovarian cancer. JAMA 2002; 287: 1671–9.

    Google Scholar 

  55. Gotoh M, Sakamoto M, Kanetaka K et al. Overexpression of osteopontin in hepatocellular carcinoma. Pathol Int 2002; 52: 19–24.

    Google Scholar 

  56. Agrawal D, Chen T, Irby R et al. Osteopontin identified as lead marker of colon cancer progression, using pooled sample expression profiling. J Natl Cancer Inst 2002; 94: 513–21.

    Google Scholar 

  57. Bautista DS, Saad Z, Chambers AF et al. Quantification of osteopontin in human plasma with an ELISA: Basal levels in pre-and postmenopausal women. Clin Biochem 1996; 29: 231–9.

    Google Scholar 

  58. Singhal H, Bautista DS, Tonkin KS et al. Elevated plasma osteopontin in metastatic breast cancer associated with increased tumor burden and decreased survival. Clin Cancer Res 1997; 3: 605–11.

    Google Scholar 

  59. Hotte SJ, Winquist, EW, Stitt L et al. Plasma osteopontin: Associations with survival and metastasis to bone in men with hormone refractory prostate cancer. Cancer, 2002; 95: 506–512.

    Google Scholar 

  60. Fedarko NS, Fohr B, Robey PG et al. Factor H binding to bone sialoprotein and osteopontin enables tumor cell evasion of complementmediated attack. J Biol Chem 2000; 275: 16666–72.

    Google Scholar 

  61. Fedarko NS, Jain A, Karadag A et al. Elevated serum bone sialoprotein and osteopontin in colon, breast, prostate, and lung cancer. Clin Cancer Res 2001; 7: 4060–6.

    Google Scholar 

  62. O’Regan A, Fleming C. Comment on: JAMA 2002, 287: 1671-9. Osteopontin as a biomarker for ovarian cancer. JAMA 2002; 287: 3208–10.

    Google Scholar 

  63. Crawford HC, Matrisian LM, Liaw L. Distinct roles of osteopontin in host defense activity and tumor survival during squamous cell carcinoma progression in vivo. Cancer Res 1998; 58: 5206–15.

    Google Scholar 

  64. Feng F, Rittling SR. Mammary tumor development in MMTV-cmyc/ MMTV-v-Ha-ras transgenic mice is unaffected by osteopontin deficiency. Breast Cancer Res Treat 2000; 63: 71–9.

    Google Scholar 

  65. Wu Y, Denhardt DT, Rittling SR. Osteopontin expression is required for full expression of the transformed phenotype by the Ras oncogene. Br J Cancer 2000; 83: 156–63.

    Google Scholar 

  66. Nemoto H, Rittling SR, Yoshitake H et al. Osteopontin deficiency reduces experimental tumor cell metastasis to bone and soft tissues. J Bone Miner Res 2001; 16: 652–9.

    Google Scholar 

  67. Orphanides G, Reinberg DA. Unified theory of gene expression. Cell 2002; 108: 439–52.

    Google Scholar 

  68. Hunter T. Oncoprotein networks. Cell 1997; 88: 333–46.

    Google Scholar 

  69. Webb CP, Vande Woude GF. Genes that regulate metastasis and angiogenesis. J Neurooncol 2000; 50: 71–87.

    Google Scholar 

  70. Welch DR, Steeg PS, Rinker-Schaeffer CW. Molecular biology of breast cancer metastasis. Genetic regulation of human breast carcinoma metastasis. Breast Cancer Res 2000; 2: 408–16.

    Google Scholar 

  71. Denhardt DT, Noda M. Osteopontin expression and function: Role in bone remodeling. J Cell Biochem 1998; Suppl 30/31: 92–102.

    Google Scholar 

  72. Craig AM, Bowden GT, Chambers AF et al. Secreted phosphoprotein mRNA is induced during multistage carcinogenesis in mouse skin and correlates with the metastatic potiential of murine fibroblasts. Int J Cancer 1990; 46: 133–7.

    Google Scholar 

  73. Yoon JW, Kita Y, Frank DJ et al. Gene expression profiling leads to identification of GLI1-binding elements in target genes and a role for multiple downstream pathways in GLI1-induced cell transformation. J Biol Chem 2002; 277: 5548–55.

    Google Scholar 

  74. Davies BR, Barraclough R, Rudland PS. Induction of metastatic ability in a stably diploid benign rat mammary epithelial cell line by transfection with DNA from human malignant breast carcinoma cell lines. Cancer Res 1994; 54: 2785–93.

    Google Scholar 

  75. Oates AJ, Barraclough R, Rudland PS. The identification of osteopontin as a metastasis-related gene product in a rodent mammary tumour model. Oncogene 1996; 13: 97–104.

    Google Scholar 

  76. Chen H, Ke Y, Oates AJ et al. Isolation of and effector for metastasisinducing DNAs from a human metastatic carcinoma cell line. Oncogene 1997; 14: 1581–8.

    Google Scholar 

  77. El-Tanani M, Barraclough R, Wilkinson MC et al. Regulatory region of metastasis-inducing DNA is the binding site for T cell factor-4. Oncogene 2001; 20: 1793–7.

    Google Scholar 

  78. El-Tanani M, Barraclough R, Wilkinson MC et al. Metastasisinducing DNA regulates the expression of the osteopontin gene by binding the transcription factor Tcf-4. Cancer Res 2001; 61: 5619–29.

    Google Scholar 

  79. Wang D, Yamamoto S, Hijiya N et al. Transcriptional regulation of the human osteopontin promoter: Functional analysis and DNA-protein interactions. Oncogene 2000; 19: 5801–9.

    Google Scholar 

  80. Malyankar UM, Hanson R, Schwartz SM et al. Upstream stimulatory factor 1 regulates osteopontin expression in smooth muscle cells. Exp Cell Res 1999; 250:535–47.

    Google Scholar 

  81. Bos JL. Ras oncogenes in human cancer: A review. Cancer Res 1989; 49: 4682–9.

    Google Scholar 

  82. Varghese HJ, Davidson MT, MacDonald IC et al. Activated ras regulates the proliferation/apoptosis balance and early survival of developing micrometastases. Cancer Res 2002; 62: 887–91.

    Google Scholar 

  83. Rak J, Kerbel RS. Ras regulation of vascular endothelial growth factor and angiogenesis. Methods Enzymol 2001; 333: 267–83.

    Google Scholar 

  84. Guo X, Zhang YP, Mitchell DA et al. Identification of a ras-activated enhancer in the mouse osteopontin gene and its interaction with a putative ETS-related transcription factor whose activity correlates with the metastatic potential of the cell. Mol Cell Biol 1995; 15: 476–87.

    Google Scholar 

  85. Craig AM, Denhardt DT. The murine gene encoding secreted phosphoprotein (osteopontin): Promoter structure activity and induction in vivo by estrogen and progesterone. Gene 1991; 100: 163–71.

    Google Scholar 

  86. Zhang Q, Wrana JL, Sodek J. Characterization of the promoter region of the porcine opn (osteopontin secreted phosphoprotein 1) gene. Eur J Biochem 1992; 207: 649–59.

    Google Scholar 

  87. Hijiya N, Setoguchi M, Matsuura K et al. Cloning and characterization of the human osteopontin gene and its promoter. Biochem J 1994; 303: 255–62.

    Google Scholar 

  88. Rafidi K, Simkina I, Johnson E et al. Characterization of the chicken osteopontin-encoding gene. Gene 1994; 140: 163–9.

    Google Scholar 

  89. Ridall AL, Daane EL, Dickinson DP et al. Characterization of the rat osteopontin gene. Evidence for two vitamin D response elements. Ann NY Acad Sci 1995; 760: 59–66.

    Google Scholar 

  90. Webb CP, van Aelst L, Wigler MH et al. Signaling pathways in Rasmediated tumorigenicity and metastasis. Proc Natl Acad Sci USA 1998; 95: 8773–8.

    Google Scholar 

  91. Morishita R, Higaki J, Tomita N et al. Application of transcription factor ‘decoy’ strategy as means of gene therapy and study of gene expression in cardiovascular disease. Circ Res 1998; 82: 1023–8.

    Google Scholar 

  92. Tomita S, Tomita N, Yamada T et al. Transcription factor decoy to study the molecular mechanism of negative regulation of renin gene expression in the liver in vivo. Circ Res 1999; 84: 1059–66.

    Google Scholar 

  93. Van Golen KL, Wu Z-F, Qial T et al. RhoC GTPase, a novel transforming oncogene for human mammary epithelial cells that partially recapitulates the inflammatory breast cancer phenotype. Cancer Res 2000; 60: 5832–8.

    Google Scholar 

  94. Tuteja R, Tuteha N. Ku Autoantigen: A multifactorial DNA-binding protein. Crit Rev Biochem Mol Biol 2000; 35: 1–33.

    Google Scholar 

  95. Singh H, Sen R, Baltimore D et al. A nuclear factor that binds to a conserved sequence motif in transcriptional control elements of immunoglobulin genes. Nature 1986; 319: 154–8.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Denhardt, D.T., Mistretta, D., Chambers, A.F. et al. Transcriptional regulation of osteopontin and the metastatic phenotype: Evidence for a Ras-activated enhancer in the human OPN promoter. Clin Exp Metastasis 20, 77–84 (2003). https://doi.org/10.1023/A:1022550721404

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1023/A:1022550721404

Navigation