Skip to main content
Top
Published in: Tumor Biology 9/2016

01-09-2016 | Original Article

Osteopontin expression in co-cultures of human squamous cell carcinoma-derived cells and osteoblastic cells and its effects on the neoplastic cell phenotype and osteoclastic activation

Authors: Lucas Novaes Teixeira, Larissa Moreira Spinola de Castro Raucci, Gabriela Caroline Alonso, Ricardo Della Coletta, Adalberto Luiz Rosa, Paulo Tambasco de Oliveira

Published in: Tumor Biology | Issue 9/2016

Login to get access

Abstract

This study evaluated the temporal expression of osteopontin (OPN) in co-cultures of human osteoblastic cells (SAOS-2) and oral squamous cell carcinoma (OSCC)-derived cells (SCC9) and examined the effects of osteoblast-derived OPN on the neoplastic cell phenotype. Additionally, the effects of these co-cultures on subsequent osteoclastic activity were explored. SCC9 cells were plated on Transwell® membranes that were either coated or not coated with Matrigel and were then co-cultured with SAOS-2 cells during the peak of OPN expression. SCC9 cells exposed to OPN-silenced SAOS-2 cultures and SCC9 cells cultured alone served as controls. SCC9 cells were quantitatively evaluated for cell adhesion, proliferation, migration, and invasion into Matrigel. The impact of co-culturing SAOS-2 and SCC9 cells on the resorptive capacity of U-937-derived osteoclastic cells was also investigated. Furthermore, a reciprocal induction of SAOS-2 and SCC9 cells in terms of OPN expression over the co-culture interval was identified. SAOS-2-secreted OPN altered the SCC9 cell phenotype, leading to enhanced cell adhesion and proliferation and higher Matrigel invasion. This invasion was also enhanced, albeit to a lesser degree, by co-culture with OPN-silenced SAOS-2 cells. Cell migration was not affected. Co-culture with SAOS-2 cells—mainly during the period of peak OPN expression—promoted over-expression of IL-6 and IL-8 by SCC9 cells and enhanced the resorptive capacity of osteoclastic cells. Taken together, these results suggest that osteoblast-derived OPN affects the interactions among OSCC-derived epithelial cells, osteoblasts, and osteoclasts, which could contribute to the process of bone destruction during bone invasion by OSCC.
Literature
2.
go back to reference Simard EP, Torre LA, Jemal A. International trends in head and neck cancer incidence rates: differences by country, sex and anatomic site. Oral Oncol. 2014;50:387–403.CrossRefPubMed Simard EP, Torre LA, Jemal A. International trends in head and neck cancer incidence rates: differences by country, sex and anatomic site. Oral Oncol. 2014;50:387–403.CrossRefPubMed
4.
6.
go back to reference Kellermann MG, Sobral LM, da Silva SD, Zecchin KG, Graner E, Lopes MA, et al. Mutual paracrine effects of oral squamous cell carcinoma cells and normal oral fibroblasts: induction of fibroblast to myofibroblast transdifferentiation and modulation of tumor cell proliferation. Oral Oncol. 2008;44:509–17.CrossRefPubMed Kellermann MG, Sobral LM, da Silva SD, Zecchin KG, Graner E, Lopes MA, et al. Mutual paracrine effects of oral squamous cell carcinoma cells and normal oral fibroblasts: induction of fibroblast to myofibroblast transdifferentiation and modulation of tumor cell proliferation. Oral Oncol. 2008;44:509–17.CrossRefPubMed
7.
go back to reference Sobral LM, Zecchin KG, Nascimento de Aquino S, Lopes MA, Graner E, et al. Isolation and characterization of myofibroblast cell lines from oral squamous cell carcinoma. Oncol Rep. 2011;25:1013–20.PubMed Sobral LM, Zecchin KG, Nascimento de Aquino S, Lopes MA, Graner E, et al. Isolation and characterization of myofibroblast cell lines from oral squamous cell carcinoma. Oncol Rep. 2011;25:1013–20.PubMed
8.
go back to reference Sobral LM, Bufalino A, Lopes MA, Graner E, Salo T, Coletta RD. Myofibroblasts in the stroma of oral cancer promote tumorigenesis via secretion of activin A. Oral Oncol. 2011;47:840–6.CrossRefPubMed Sobral LM, Bufalino A, Lopes MA, Graner E, Salo T, Coletta RD. Myofibroblasts in the stroma of oral cancer promote tumorigenesis via secretion of activin A. Oral Oncol. 2011;47:840–6.CrossRefPubMed
9.
go back to reference Shevde LA, Samant RS, Paik JC, Metge BJ, Chambers AF, Casey G, et al. Osteopontin knockdown suppresses tumorigenicity of human metastatic breast carcinoma, MDA-MB-435. Clin Exp Metastasis. 2006;23:123–33.CrossRefPubMedPubMedCentral Shevde LA, Samant RS, Paik JC, Metge BJ, Chambers AF, Casey G, et al. Osteopontin knockdown suppresses tumorigenicity of human metastatic breast carcinoma, MDA-MB-435. Clin Exp Metastasis. 2006;23:123–33.CrossRefPubMedPubMedCentral
10.
go back to reference Senger DR, Wirth DF, Hynes RO. Transformed mammalian cells secrete specific proteins and phosphoproteins. Cell. 1979;16:885–93.CrossRefPubMed Senger DR, Wirth DF, Hynes RO. Transformed mammalian cells secrete specific proteins and phosphoproteins. Cell. 1979;16:885–93.CrossRefPubMed
11.
12.
go back to reference Weber GF, Cantor H. The immunology of Eta-1/osteopontin. Cytokine Growth Factor Rev. 1996;7:241–8.CrossRefPubMed Weber GF, Cantor H. The immunology of Eta-1/osteopontin. Cytokine Growth Factor Rev. 1996;7:241–8.CrossRefPubMed
13.
go back to reference Brown LF, Berse B, Van de Water L, Papadopoulos-Sergiou A, Perruzzi CA, Manseau EJ, et al. Expression and distribution of osteopontin in human tissues: widespread association with luminal epithelial surfaces. Mol Biol Cell. 1992;3:1169–80.CrossRefPubMedPubMedCentral Brown LF, Berse B, Van de Water L, Papadopoulos-Sergiou A, Perruzzi CA, Manseau EJ, et al. Expression and distribution of osteopontin in human tissues: widespread association with luminal epithelial surfaces. Mol Biol Cell. 1992;3:1169–80.CrossRefPubMedPubMedCentral
14.
go back to reference Denhardt DT, Guo X. Osteopontin: a protein with diverse functions. FASEB J. 1993;7:1475–82.PubMed Denhardt DT, Guo X. Osteopontin: a protein with diverse functions. FASEB J. 1993;7:1475–82.PubMed
15.
go back to reference Bautista DS, Denstedt J, Chambers AF, Harris JF. Low-molecular-weight variants of osteopontin generated by serine proteinases in urine of patients with kidney stones. J Cell Biochem. 1996;61:402–9.CrossRefPubMed Bautista DS, Denstedt J, Chambers AF, Harris JF. Low-molecular-weight variants of osteopontin generated by serine proteinases in urine of patients with kidney stones. J Cell Biochem. 1996;61:402–9.CrossRefPubMed
16.
go back to reference Chen RX, Xia YH, Xue TC, Ye SL. Osteopontin promotes hepatocellular carcinoma invasion by up-regulating MMP-2 and uPA expression. Mol Biol Rep. 2011;38:3671–7.CrossRefPubMed Chen RX, Xia YH, Xue TC, Ye SL. Osteopontin promotes hepatocellular carcinoma invasion by up-regulating MMP-2 and uPA expression. Mol Biol Rep. 2011;38:3671–7.CrossRefPubMed
17.
go back to reference Lu JG, Li Y, Li L, Kan X. Overexpression of osteopontin and integrin αv in laryngeal and hypopharyngeal carcinomas associated with differentiation and metastasis. J Cancer Res Clin Oncol. 2011;137:1613–8.CrossRefPubMed Lu JG, Li Y, Li L, Kan X. Overexpression of osteopontin and integrin αv in laryngeal and hypopharyngeal carcinomas associated with differentiation and metastasis. J Cancer Res Clin Oncol. 2011;137:1613–8.CrossRefPubMed
18.
go back to reference Wang ZM, Cui YH, Li W, Chen SY, Liu TS. Lentiviral-mediated siRNA targeted against osteopontin suppresses the growth and metastasis of gastric cancer cells. Oncol Rep. 2011;25:997–1003.PubMed Wang ZM, Cui YH, Li W, Chen SY, Liu TS. Lentiviral-mediated siRNA targeted against osteopontin suppresses the growth and metastasis of gastric cancer cells. Oncol Rep. 2011;25:997–1003.PubMed
19.
go back to reference Zhang R, Pan X, Huang Z, Weber GF, Zhang G. Osteopontin enhances the expression and activity of MMP-2 via the SDF-1/CXCR4 axis in hepatocellular carcinoma cell lines. PLoS One. 2011;6:e23831.CrossRefPubMedPubMedCentral Zhang R, Pan X, Huang Z, Weber GF, Zhang G. Osteopontin enhances the expression and activity of MMP-2 via the SDF-1/CXCR4 axis in hepatocellular carcinoma cell lines. PLoS One. 2011;6:e23831.CrossRefPubMedPubMedCentral
20.
go back to reference Matsuzaki H, Shima K, Muramatsu T, Ro Y, Hashimoto S, Shibahara T, et al. Osteopontin as biomarker in early invasion by squamous cell carcinoma in tongue. J Oral Pathol Med. 2007;36:30–4.CrossRefPubMed Matsuzaki H, Shima K, Muramatsu T, Ro Y, Hashimoto S, Shibahara T, et al. Osteopontin as biomarker in early invasion by squamous cell carcinoma in tongue. J Oral Pathol Med. 2007;36:30–4.CrossRefPubMed
21.
go back to reference Routray S, Kheur SM, Kheur M. Osteopontin: a marker for invasive oral squamous cell carcinoma but not for potentially malignant epithelial dysplasias. Ann Diagn Pathol. 2013;17:421–4.CrossRefPubMed Routray S, Kheur SM, Kheur M. Osteopontin: a marker for invasive oral squamous cell carcinoma but not for potentially malignant epithelial dysplasias. Ann Diagn Pathol. 2013;17:421–4.CrossRefPubMed
23.
go back to reference Avirović M, Matušan-Ilijaš K, Damante G, Fabrro D, Cerović R, Juretić M, et al. Osteopontin expression is an independent factor for poor survival in oral squamous cell carcinoma: a computer-assisted analysis on TMA sections. J Oral Pathol Med. 2013;42:620–6.CrossRefPubMed Avirović M, Matušan-Ilijaš K, Damante G, Fabrro D, Cerović R, Juretić M, et al. Osteopontin expression is an independent factor for poor survival in oral squamous cell carcinoma: a computer-assisted analysis on TMA sections. J Oral Pathol Med. 2013;42:620–6.CrossRefPubMed
24.
go back to reference Ingale Y, Routray S, Kheur SM, Kheur M, Mohanty N. Evaluating the efficacy of osteopontin expression as a prognostic marker in oral squamous cell carcinoma in the Indian subpopulation. J Oral Maxillofac Pathol. 2014;18(Suppl 1):S11–5.PubMedPubMedCentral Ingale Y, Routray S, Kheur SM, Kheur M, Mohanty N. Evaluating the efficacy of osteopontin expression as a prognostic marker in oral squamous cell carcinoma in the Indian subpopulation. J Oral Maxillofac Pathol. 2014;18(Suppl 1):S11–5.PubMedPubMedCentral
26.
go back to reference Standal T, Borset M, Sundan A. Role of osteopontin in adhesion, migration, cell survival and bone remodeling. Exp Oncol. 2004;26:179–84.PubMed Standal T, Borset M, Sundan A. Role of osteopontin in adhesion, migration, cell survival and bone remodeling. Exp Oncol. 2004;26:179–84.PubMed
27.
go back to reference Hunter GK. Role of osteopontin in modulation of hydroxyapatite formation. Calcif Tissue Int. 2013;93:348–54.CrossRefPubMed Hunter GK. Role of osteopontin in modulation of hydroxyapatite formation. Calcif Tissue Int. 2013;93:348–54.CrossRefPubMed
28.
go back to reference Semba I, Matsuuchi H, Miura Y. Histomorphometric analysis of osteoclastic resorption in bone directly invaded by gingival squamous cell carcinoma. J Oral Pathol Med. 1996;25:429–35.CrossRefPubMed Semba I, Matsuuchi H, Miura Y. Histomorphometric analysis of osteoclastic resorption in bone directly invaded by gingival squamous cell carcinoma. J Oral Pathol Med. 1996;25:429–35.CrossRefPubMed
29.
30.
go back to reference Chellaiah MA, Kizer N, Biswas R, Alvarez U, Strauss-Schoenberger J, Rifas L, et al. Osteopontin deficiency produces osteoclast dysfunction due to reduced CD44 surface expression. Mol Biol Cell. 2003;14:173–89.CrossRefPubMedPubMedCentral Chellaiah MA, Kizer N, Biswas R, Alvarez U, Strauss-Schoenberger J, Rifas L, et al. Osteopontin deficiency produces osteoclast dysfunction due to reduced CD44 surface expression. Mol Biol Cell. 2003;14:173–89.CrossRefPubMedPubMedCentral
31.
go back to reference Li M, Amizuka N, Takeuchi K, Freitas PH, Kawano Y, Hoshino M, et al. Histochemical evidence of osteoclastic degradation of extracellular matrix in osteolytic metastasis originating from human lung small carcinoma (SBC-5) cells. Microsc Res Tech. 2006;69:73–83.CrossRefPubMed Li M, Amizuka N, Takeuchi K, Freitas PH, Kawano Y, Hoshino M, et al. Histochemical evidence of osteoclastic degradation of extracellular matrix in osteolytic metastasis originating from human lung small carcinoma (SBC-5) cells. Microsc Res Tech. 2006;69:73–83.CrossRefPubMed
32.
go back to reference Kruger TE, Miller AH, Godwin AK, Wang J. Bone sialoprotein and osteopontin in bone metastasis of osteotropic cancers. Crit Rev Oncol Hematol. 2014;89:330–41.CrossRefPubMed Kruger TE, Miller AH, Godwin AK, Wang J. Bone sialoprotein and osteopontin in bone metastasis of osteotropic cancers. Crit Rev Oncol Hematol. 2014;89:330–41.CrossRefPubMed
33.
go back to reference Jimi E, Furuta H, Matsuo K, Tominaga K, Takahashi T, Nakanishi O. The cellular and molecular mechanisms of bone invasion by oral squamous cell carcinoma. Oral Dis. 2011;17:462–8.CrossRefPubMed Jimi E, Furuta H, Matsuo K, Tominaga K, Takahashi T, Nakanishi O. The cellular and molecular mechanisms of bone invasion by oral squamous cell carcinoma. Oral Dis. 2011;17:462–8.CrossRefPubMed
34.
go back to reference Amoui M, Suhr SM, Baylink DJ, Lau KH. An osteoclastic protein-tyrosine phosphatase may play a role in differentiation and activity of human monocytic U-937 cell-derived, osteoclast-like cells. Am J Phys Cell Phys. 2004;287:C874–84.CrossRef Amoui M, Suhr SM, Baylink DJ, Lau KH. An osteoclastic protein-tyrosine phosphatase may play a role in differentiation and activity of human monocytic U-937 cell-derived, osteoclast-like cells. Am J Phys Cell Phys. 2004;287:C874–84.CrossRef
35.
go back to reference Schwartz Fo HO, Novaes Jr AB, de Castro LM, Rosa AL, de Oliveira PT. In vitro osteogenesis on a microstructured titanium surface with additional submicron-scale topography. Clin Oral Implants Res. 2007;18:333–44.CrossRefPubMed Schwartz Fo HO, Novaes Jr AB, de Castro LM, Rosa AL, de Oliveira PT. In vitro osteogenesis on a microstructured titanium surface with additional submicron-scale topography. Clin Oral Implants Res. 2007;18:333–44.CrossRefPubMed
36.
go back to reference Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 2001;25:402–8.CrossRefPubMed Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 2001;25:402–8.CrossRefPubMed
37.
go back to reference de Oliva MA, Maximiano WM, de Castro LM, da Silva Jr PE, Fernandes RR, Ciancaglini P, et al. Treatment with a growth factor-protein mixture inhibits formation of mineralized nodules in osteogenic cell cultures grown on titanium. J Histochem Cytochem. 2009;57:265–76.CrossRefPubMedPubMedCentral de Oliva MA, Maximiano WM, de Castro LM, da Silva Jr PE, Fernandes RR, Ciancaglini P, et al. Treatment with a growth factor-protein mixture inhibits formation of mineralized nodules in osteogenic cell cultures grown on titanium. J Histochem Cytochem. 2009;57:265–76.CrossRefPubMedPubMedCentral
38.
go back to reference Wai PY, Kuo PC. Osteopontin: regulation in tumor metastasis. Cancer Metastasis Rev. 2008;27:103–18.CrossRefPubMed Wai PY, Kuo PC. Osteopontin: regulation in tumor metastasis. Cancer Metastasis Rev. 2008;27:103–18.CrossRefPubMed
39.
go back to reference Terpos E, Kiagia M, Karapanagiotou EM, Charpidou A, Dilana KD, Nasothimiou E, et al. The clinical significance of serum markers of bone turnover in NSCLC patients: surveillance, management and prognostic implications. Anticancer Res. 2009;29:1651–7.PubMed Terpos E, Kiagia M, Karapanagiotou EM, Charpidou A, Dilana KD, Nasothimiou E, et al. The clinical significance of serum markers of bone turnover in NSCLC patients: surveillance, management and prognostic implications. Anticancer Res. 2009;29:1651–7.PubMed
40.
go back to reference Weber GF, Lett GS, Haubein NC. Categorical meta-analysis of osteopontin as a clinical cancer marker. Oncol Rep. 2011;25:433–41.CrossRefPubMed Weber GF, Lett GS, Haubein NC. Categorical meta-analysis of osteopontin as a clinical cancer marker. Oncol Rep. 2011;25:433–41.CrossRefPubMed
41.
go back to reference Mardani M, Andisheh-Tadbir A, Khademi B, Fattahi MJ, Shafiee S, Asad-Zadeh M. Serum levels of osteopontin as a prognostic factor in patients with oral squamous cell carcinoma. Tumour Biol. 2014;35:3827–9.CrossRefPubMed Mardani M, Andisheh-Tadbir A, Khademi B, Fattahi MJ, Shafiee S, Asad-Zadeh M. Serum levels of osteopontin as a prognostic factor in patients with oral squamous cell carcinoma. Tumour Biol. 2014;35:3827–9.CrossRefPubMed
42.
go back to reference Buijs JT, van der Pluijm G. Osteotropic cancers: from primary tumor to bone. Cancer Lett. 2009;273:177–93.CrossRefPubMed Buijs JT, van der Pluijm G. Osteotropic cancers: from primary tumor to bone. Cancer Lett. 2009;273:177–93.CrossRefPubMed
43.
go back to reference Samuvel DJ, Sundararaj KP, Li Y, Lopes-Virella MF, Huang Y. Adipocyte-mononuclear cell interaction, toll-like receptor 4 activation, and high glucose synergistically up-regulate osteopontin expression via an interleukin 6-mediated mechanism. J Biol Chem. 2010;285:3916–27.CrossRefPubMed Samuvel DJ, Sundararaj KP, Li Y, Lopes-Virella MF, Huang Y. Adipocyte-mononuclear cell interaction, toll-like receptor 4 activation, and high glucose synergistically up-regulate osteopontin expression via an interleukin 6-mediated mechanism. J Biol Chem. 2010;285:3916–27.CrossRefPubMed
44.
go back to reference Fried D, Mullins B, Weissler M, Shores C, Zanation A, Hackman T, et al. Prognostic significance of bone invasion for oral cavity squamous cell carcinoma considered T1/T2 by American joint committee on cancer size criteria. Head Neck. 2014;36:776–81.CrossRefPubMed Fried D, Mullins B, Weissler M, Shores C, Zanation A, Hackman T, et al. Prognostic significance of bone invasion for oral cavity squamous cell carcinoma considered T1/T2 by American joint committee on cancer size criteria. Head Neck. 2014;36:776–81.CrossRefPubMed
45.
go back to reference Miki Y, Ono K, Hata S, Suzuki T, Kumamoto H, Sasano H. The advantages of co-culture over mono cell culture in simulating in vivo environment. J Steroid Biochem Mol Biol. 2012;131:68–75.CrossRefPubMed Miki Y, Ono K, Hata S, Suzuki T, Kumamoto H, Sasano H. The advantages of co-culture over mono cell culture in simulating in vivo environment. J Steroid Biochem Mol Biol. 2012;131:68–75.CrossRefPubMed
46.
go back to reference Gao C, Guo H, Downey L, Marroquin C, Wei J, Kuo PC. Osteopontin-dependent CD44v6 expression and cell adhesion in HepG2 cells. Carcinogenesis. 2003;24:1871–8.CrossRefPubMed Gao C, Guo H, Downey L, Marroquin C, Wei J, Kuo PC. Osteopontin-dependent CD44v6 expression and cell adhesion in HepG2 cells. Carcinogenesis. 2003;24:1871–8.CrossRefPubMed
47.
go back to reference Lyons AJ, Jones J. Cell adhesion molecules, the extracellular matrix and oral squamous carcinoma. Int J Oral Maxillofac Surg. 2007;36:671–9.CrossRefPubMed Lyons AJ, Jones J. Cell adhesion molecules, the extracellular matrix and oral squamous carcinoma. Int J Oral Maxillofac Surg. 2007;36:671–9.CrossRefPubMed
48.
go back to reference Harada T, Shinohara M, Nakamura S, Oka M. An immunohistochemical study of the extracellular matrix in oral squamous cell carcinoma and its association with invasive and metastatic potential. Virchows Arch. 1994;424:257–66.CrossRefPubMed Harada T, Shinohara M, Nakamura S, Oka M. An immunohistochemical study of the extracellular matrix in oral squamous cell carcinoma and its association with invasive and metastatic potential. Virchows Arch. 1994;424:257–66.CrossRefPubMed
49.
go back to reference Clëzardin P. Integrins in bone metastasis formation and potential therapeutic implications. Curr Cancer Drug Targets. 2009;9:801–6.CrossRefPubMed Clëzardin P. Integrins in bone metastasis formation and potential therapeutic implications. Curr Cancer Drug Targets. 2009;9:801–6.CrossRefPubMed
50.
go back to reference Schneider JG, Amend SR, Weilbaecher KN. Integrins and bone metastasis: integrating tumor cell and stromal cell interactions. Bone. 2011;48:54–65.CrossRefPubMed Schneider JG, Amend SR, Weilbaecher KN. Integrins and bone metastasis: integrating tumor cell and stromal cell interactions. Bone. 2011;48:54–65.CrossRefPubMed
51.
go back to reference Angelucci A, Festuccia C, Gravina GL, Muzi P, Bonghi L, Vicentini C, et al. Osteopontin enhances the cell proliferation induced by the epidermal growth factor in human prostate cancer cells. Prostate. 2004;59:157–66.CrossRefPubMed Angelucci A, Festuccia C, Gravina GL, Muzi P, Bonghi L, Vicentini C, et al. Osteopontin enhances the cell proliferation induced by the epidermal growth factor in human prostate cancer cells. Prostate. 2004;59:157–66.CrossRefPubMed
52.
go back to reference Nam TJ, Busby Jr WH, Rees C, Clemmons DR. Thrombospondin and osteopontin bind to insulin-like growth factor (IGF)-binding protein-5 leading to an alteration in IGF-I-stimulated cell growth. Endocrinology. 2000;141:1100–6.PubMed Nam TJ, Busby Jr WH, Rees C, Clemmons DR. Thrombospondin and osteopontin bind to insulin-like growth factor (IGF)-binding protein-5 leading to an alteration in IGF-I-stimulated cell growth. Endocrinology. 2000;141:1100–6.PubMed
53.
go back to reference Duan C, Xu Q. Roles of insulin-like growth factor (IGF) binding proteins in regulating IGF actions. Gen Comp Endocrinol. 2005;142:44–52.CrossRefPubMed Duan C, Xu Q. Roles of insulin-like growth factor (IGF) binding proteins in regulating IGF actions. Gen Comp Endocrinol. 2005;142:44–52.CrossRefPubMed
54.
go back to reference Zhi X, Lamperska K, Golusinski P, Schork NJ, Luczewski L, Golusinski W, et al. Expression levels of insulin-like growth factors 1 and 2 in head and neck squamous cell carcinoma. Growth Hormon IGF Res. 2014;24:137–41.CrossRef Zhi X, Lamperska K, Golusinski P, Schork NJ, Luczewski L, Golusinski W, et al. Expression levels of insulin-like growth factors 1 and 2 in head and neck squamous cell carcinoma. Growth Hormon IGF Res. 2014;24:137–41.CrossRef
55.
go back to reference Martinez C, Churchman M, Freeman T, Ilyas M. Osteopontin provides early proliferative drive and may be dependent upon aberrant c-myc signalling in murine intestinal tumours. Exp Mol Pathol. 2010;88:272–7.CrossRefPubMed Martinez C, Churchman M, Freeman T, Ilyas M. Osteopontin provides early proliferative drive and may be dependent upon aberrant c-myc signalling in murine intestinal tumours. Exp Mol Pathol. 2010;88:272–7.CrossRefPubMed
56.
go back to reference Fukuda M, Hamao A, Tanaka A, Kitada M, Suzuki S, Kusama K, et al. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/APO2L) and its receptors expression in human squamous cell carcinoma of the oral cavity. Oncol Rep. 2003;10:1113–9.PubMed Fukuda M, Hamao A, Tanaka A, Kitada M, Suzuki S, Kusama K, et al. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/APO2L) and its receptors expression in human squamous cell carcinoma of the oral cavity. Oncol Rep. 2003;10:1113–9.PubMed
57.
go back to reference Kassouf N, Thornhill MH. Oral cancer cell lines can use multiple ligands, including Fas-L, TRAIL and TNF-alpha, to induce apoptosis in Jurkat T cells: possible mechanisms for immune escape by head and neck cancers. Oral Oncol. 2008;44:672–82.CrossRefPubMed Kassouf N, Thornhill MH. Oral cancer cell lines can use multiple ligands, including Fas-L, TRAIL and TNF-alpha, to induce apoptosis in Jurkat T cells: possible mechanisms for immune escape by head and neck cancers. Oral Oncol. 2008;44:672–82.CrossRefPubMed
58.
go back to reference Polat B, Wohlleben G, Katzer A, Djuzenova CS, Technau A, Flentje M. Influence of osteopontin silencing on survival and migration of lung cancer cells. Strahlenther Onkol. 2013;189:62–7.CrossRefPubMed Polat B, Wohlleben G, Katzer A, Djuzenova CS, Technau A, Flentje M. Influence of osteopontin silencing on survival and migration of lung cancer cells. Strahlenther Onkol. 2013;189:62–7.CrossRefPubMed
59.
go back to reference Zhang H, Guo M, Chen JH, Wang Z, Du XF, Liu PX, et al. Osteopontin knockdown inhibits αv,β3 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;33:991–1002.CrossRefPubMed Zhang H, Guo M, Chen JH, Wang Z, Du XF, Liu PX, et al. Osteopontin knockdown inhibits αv,β3 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;33:991–1002.CrossRefPubMed
60.
go back to reference Agnihotri R, Crawford HC, Haro H, Matrisian LM, Havrda MC, Liaw L. Osteopontin, a novel substrate for matrix metalloproteinase-3 (stromelysin-1) and matrix metalloproteinase-7 (matrilysin). J Biol Chem. 2001;276:28261–7.CrossRefPubMed Agnihotri R, Crawford HC, Haro H, Matrisian LM, Havrda MC, Liaw L. Osteopontin, a novel substrate for matrix metalloproteinase-3 (stromelysin-1) and matrix metalloproteinase-7 (matrilysin). J Biol Chem. 2001;276:28261–7.CrossRefPubMed
61.
go back to reference Gao YA, Agnihotri R, Vary CP, Liaw L. Expression and characterization of recombinant osteopontin peptides representing matrix metalloproteinase proteolytic fragments. Matrix Biol. 2004;23:457–66.CrossRefPubMed Gao YA, Agnihotri R, Vary CP, Liaw L. Expression and characterization of recombinant osteopontin peptides representing matrix metalloproteinase proteolytic fragments. Matrix Biol. 2004;23:457–66.CrossRefPubMed
62.
go back to reference Chuang HC, Su CY, Huang HY, Huang CC, Chien CY, Du YY, et al. Active matrix metalloproteinase-7 is associated with invasion in buccal squamous cell carcinoma. Mod Pathol. 2008;21:1444–50.CrossRefPubMed Chuang HC, Su CY, Huang HY, Huang CC, Chien CY, Du YY, et al. Active matrix metalloproteinase-7 is associated with invasion in buccal squamous cell carcinoma. Mod Pathol. 2008;21:1444–50.CrossRefPubMed
63.
go back to reference Li TJ, Cui J. COX-2, MMP-7 expression in oral lichen planus and oral squamous cell carcinoma. Asian Pac J Trop Med. 2013;6:640–3.CrossRefPubMed Li TJ, Cui J. COX-2, MMP-7 expression in oral lichen planus and oral squamous cell carcinoma. Asian Pac J Trop Med. 2013;6:640–3.CrossRefPubMed
64.
go back to reference Mäkinen LK, Häyry V, Hagström J, Sorsa T, Passador-Santos F, Keski-Säntti H, et al. Matrix metalloproteinase-7 and matrix metalloproteinase-25 in oral tongue squamous cell carcinoma. Head Neck. 2014;36:1783–8.CrossRefPubMed Mäkinen LK, Häyry V, Hagström J, Sorsa T, Passador-Santos F, Keski-Säntti H, et al. Matrix metalloproteinase-7 and matrix metalloproteinase-25 in oral tongue squamous cell carcinoma. Head Neck. 2014;36:1783–8.CrossRefPubMed
65.
go back to reference Mashhadiabbas F, Mahjour F, Mahjour SB, Fereidooni F, Hosseini FS. The immunohistochemical characterization of MMP-2, MMP-10, TIMP-1, TIMP-2, and podoplanin in oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol. 2012;114:240–50.CrossRefPubMed Mashhadiabbas F, Mahjour F, Mahjour SB, Fereidooni F, Hosseini FS. The immunohistochemical characterization of MMP-2, MMP-10, TIMP-1, TIMP-2, and podoplanin in oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol. 2012;114:240–50.CrossRefPubMed
66.
go back to reference Gao ZB, Duan YQ, Zhang L, Chen DW, Ding PT. Expression of matrix metalloproteinase 2 and its tissue inhibitor in oral squamous cell carcinoma. Int J Mol Med. 2005;16:599–603.PubMed Gao ZB, Duan YQ, Zhang L, Chen DW, Ding PT. Expression of matrix metalloproteinase 2 and its tissue inhibitor in oral squamous cell carcinoma. Int J Mol Med. 2005;16:599–603.PubMed
67.
go back to reference Singh RD, Haridas N, Patel JB, Shah FD, Shukla SN, Shah PM, et al. Matrix metalloproteinases and their inhibitors: correlation with invasion and metastasis in oral cancer. Indian J Clin Biochem. 2010;25:250–9.CrossRefPubMedPubMedCentral Singh RD, Haridas N, Patel JB, Shah FD, Shukla SN, Shah PM, et al. Matrix metalloproteinases and their inhibitors: correlation with invasion and metastasis in oral cancer. Indian J Clin Biochem. 2010;25:250–9.CrossRefPubMedPubMedCentral
68.
go back to reference Singh RD, Nilayangode H, Patel JB, Shah FD, Shukla SN, Shah PM, et al. Combined evaluation of matrix metalloproteinases and their inhibitors has better clinical utility in oral cancer. Int J Biol Markers. 2011;26:27–36.CrossRefPubMed Singh RD, Nilayangode H, Patel JB, Shah FD, Shukla SN, Shah PM, et al. Combined evaluation of matrix metalloproteinases and their inhibitors has better clinical utility in oral cancer. Int J Biol Markers. 2011;26:27–36.CrossRefPubMed
69.
go back to reference Pérez-Sayáns García M, Suárez-Peñaranda JM, Gayoso-Diz P, Barros-Angueira F, Gándara-Rey JM, García-García A. Tissue inhibitor of metalloproteinases in oral squamous cell carcinomas—a therapeutic target? Cancer Lett. 2012;323:11–9.CrossRefPubMed Pérez-Sayáns García M, Suárez-Peñaranda JM, Gayoso-Diz P, Barros-Angueira F, Gándara-Rey JM, García-García A. Tissue inhibitor of metalloproteinases in oral squamous cell carcinomas—a therapeutic target? Cancer Lett. 2012;323:11–9.CrossRefPubMed
70.
go back to reference Bodnar M, Szylberg Ł, Kazmierczak W, Marszalek A. Tumor progression driven by pathways activating matrix metalloproteinases and their inhibitors. J Oral Pathol Med. 2015;44:437–43.CrossRefPubMed Bodnar M, Szylberg Ł, Kazmierczak W, Marszalek A. Tumor progression driven by pathways activating matrix metalloproteinases and their inhibitors. J Oral Pathol Med. 2015;44:437–43.CrossRefPubMed
71.
go back to reference Li M, Wang Z, Xing Y, Yu J, Tian L, Zhang D, et al. A multicenter study on expressions of vascular endothelial growth factor, matrix metallopeptidase-9 and tissue inhibitor of metalloproteinase-2 in oral and maxillofacial squamous cell carcinoma. Iran Red Crescent Med J. 2014;16:e13185.CrossRefPubMedPubMedCentral Li M, Wang Z, Xing Y, Yu J, Tian L, Zhang D, et al. A multicenter study on expressions of vascular endothelial growth factor, matrix metallopeptidase-9 and tissue inhibitor of metalloproteinase-2 in oral and maxillofacial squamous cell carcinoma. Iran Red Crescent Med J. 2014;16:e13185.CrossRefPubMedPubMedCentral
72.
go back to reference Brew K, Nagase H. The tissue inhibitors of metalloproteinases (TIMPs): an ancient family with structural and functional diversity. Biochim Biophys Acta. 1803;2010:55–71. Brew K, Nagase H. The tissue inhibitors of metalloproteinases (TIMPs): an ancient family with structural and functional diversity. Biochim Biophys Acta. 1803;2010:55–71.
73.
go back to reference Erdem NF, Carlson ER, Gerard DA, Ichiki AT. Characterization of 3 oral squamous cell carcinoma cell lines with different invasion and/or metastatic potentials. J Oral Maxillofac Surg. 2007;65:1725–33.CrossRefPubMed Erdem NF, Carlson ER, Gerard DA, Ichiki AT. Characterization of 3 oral squamous cell carcinoma cell lines with different invasion and/or metastatic potentials. J Oral Maxillofac Surg. 2007;65:1725–33.CrossRefPubMed
74.
go back to reference Nakashima T, Yasumatsu R, Masuda M, Clayman GL, Komune S. Prognostic value of cathepsin L and its inhibitor headpin in oral squamous cell carcinoma. J Laryngol Otol. 2012;126:1134–7.CrossRefPubMed Nakashima T, Yasumatsu R, Masuda M, Clayman GL, Komune S. Prognostic value of cathepsin L and its inhibitor headpin in oral squamous cell carcinoma. J Laryngol Otol. 2012;126:1134–7.CrossRefPubMed
75.
go back to reference Bitu CC, Kauppila JH, Bufalino A, Nurmenniemi S, Teppo S, Keinänen M, et al. Cathepsin K is present in invasive oral tongue squamous cell carcinoma in vivo and in vitro. PLoS One. 2013;8:e70925.CrossRefPubMedPubMedCentral Bitu CC, Kauppila JH, Bufalino A, Nurmenniemi S, Teppo S, Keinänen M, et al. Cathepsin K is present in invasive oral tongue squamous cell carcinoma in vivo and in vitro. PLoS One. 2013;8:e70925.CrossRefPubMedPubMedCentral
76.
go back to reference Duong LT, Wesolowski GA, Leung P, Oballa R, Pickarski M. Efficacy of a cathepsin k inhibitor in a preclinical model for prevention and treatment of breast cancer bone metastasis. Mol Cancer Ther. 2014;13:2898–909.CrossRefPubMed Duong LT, Wesolowski GA, Leung P, Oballa R, Pickarski M. Efficacy of a cathepsin k inhibitor in a preclinical model for prevention and treatment of breast cancer bone metastasis. Mol Cancer Ther. 2014;13:2898–909.CrossRefPubMed
77.
go back to reference Deyama Y, Tei K, Yoshimura Y, Izumiyama Y, Takeyama S, Hatta M, et al. Oral squamous cell carcinomas stimulate osteoclast differentiation. Oncol Rep. 2008;20:663–8.PubMed Deyama Y, Tei K, Yoshimura Y, Izumiyama Y, Takeyama S, Hatta M, et al. Oral squamous cell carcinomas stimulate osteoclast differentiation. Oncol Rep. 2008;20:663–8.PubMed
78.
go back to reference Kayamori K, Sakamoto K, Nakashima T, Takayanagi H, Morita K, Omura K, et al. Roles of interleukin-6 and parathyroid hormone-related peptide in osteoclast formation associated with oral cancers: significance of interleukin-6 synthesized by stromal cells in response to cancer cells. Am J Pathol. 2010;176:968–80.CrossRefPubMedPubMedCentral Kayamori K, Sakamoto K, Nakashima T, Takayanagi H, Morita K, Omura K, et al. Roles of interleukin-6 and parathyroid hormone-related peptide in osteoclast formation associated with oral cancers: significance of interleukin-6 synthesized by stromal cells in response to cancer cells. Am J Pathol. 2010;176:968–80.CrossRefPubMedPubMedCentral
79.
go back to reference Hwang YS, Lee SK, Park KK, Chung WY. Secretion of IL-6 and IL-8 from lysophosphatidic acid-stimulated oral squamous cell carcinoma promotes osteoclastogenesis and bone resorption. Oral Oncol. 2012;48:40–8. Hwang YS, Lee SK, Park KK, Chung WY. Secretion of IL-6 and IL-8 from lysophosphatidic acid-stimulated oral squamous cell carcinoma promotes osteoclastogenesis and bone resorption. Oral Oncol. 2012;48:40–8.
80.
go back to reference Bendre MS, Montague DC, Peery T, Akel NS, Gaddy D, Suva LJ. Interleukin-8 stimulation of osteoclastogenesis and bone resorption is a mechanism for the increased osteolysis of metastatic bone disease. Bone. 2003;33:28–37.CrossRefPubMed Bendre MS, Montague DC, Peery T, Akel NS, Gaddy D, Suva LJ. Interleukin-8 stimulation of osteoclastogenesis and bone resorption is a mechanism for the increased osteolysis of metastatic bone disease. Bone. 2003;33:28–37.CrossRefPubMed
81.
go back to reference Kudo O, Sabokbar A, Pocock A, Itonaga I, Fujikawa Y, Athanasou NA. Interleukin-6 and interleukin-11 support human osteoclast formation by a RANKL-independent mechanism. Bone. 2003;32:1–7.CrossRefPubMed Kudo O, Sabokbar A, Pocock A, Itonaga I, Fujikawa Y, Athanasou NA. Interleukin-6 and interleukin-11 support human osteoclast formation by a RANKL-independent mechanism. Bone. 2003;32:1–7.CrossRefPubMed
82.
go back to reference Kwan Tat S, Padrines M, Théoleyre S, Heymann D, Fortun Y. IL-6, RANKL, TNF-alpha/IL-1: interrelations in bone resorption pathophysiology. Cytokine Growth Factor Rev. 2004;15:49–60.CrossRefPubMed Kwan Tat S, Padrines M, Théoleyre S, Heymann D, Fortun Y. IL-6, RANKL, TNF-alpha/IL-1: interrelations in bone resorption pathophysiology. Cytokine Growth Factor Rev. 2004;15:49–60.CrossRefPubMed
83.
go back to reference Gilbert M, Giachelli CM, Stayton PS. Biomimetic peptides that engage specific integrin-dependent signaling pathways and bind to calcium phosphate surfaces. J Biomed Mater Res A. 2003;67:69–77.CrossRefPubMed Gilbert M, Giachelli CM, Stayton PS. Biomimetic peptides that engage specific integrin-dependent signaling pathways and bind to calcium phosphate surfaces. J Biomed Mater Res A. 2003;67:69–77.CrossRefPubMed
84.
go back to reference Gilbert M, Shaw WJ, Long JR, Nelson K, Drobny GP, Giachelli CM, Stayton PS. Chimeric peptides of statherin and osteopontin that bind hydroxyapatite and mediate cell adhesion. J Biol Chem. 2000;275:16213–8.CrossRefPubMed Gilbert M, Shaw WJ, Long JR, Nelson K, Drobny GP, Giachelli CM, Stayton PS. Chimeric peptides of statherin and osteopontin that bind hydroxyapatite and mediate cell adhesion. J Biol Chem. 2000;275:16213–8.CrossRefPubMed
85.
go back to reference Pietak AM, Sayer M. Functional atomic force microscopy investigation of osteopontin affinity for silicon stabilized tricalcium phosphate bioceramic surfaces. Biomaterials. 2006;27:3–14.CrossRefPubMed Pietak AM, Sayer M. Functional atomic force microscopy investigation of osteopontin affinity for silicon stabilized tricalcium phosphate bioceramic surfaces. Biomaterials. 2006;27:3–14.CrossRefPubMed
Metadata
Title
Osteopontin expression in co-cultures of human squamous cell carcinoma-derived cells and osteoblastic cells and its effects on the neoplastic cell phenotype and osteoclastic activation
Authors
Lucas Novaes Teixeira
Larissa Moreira Spinola de Castro Raucci
Gabriela Caroline Alonso
Ricardo Della Coletta
Adalberto Luiz Rosa
Paulo Tambasco de Oliveira
Publication date
01-09-2016
Publisher
Springer Netherlands
Published in
Tumor Biology / Issue 9/2016
Print ISSN: 1010-4283
Electronic ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-016-5104-0

Other articles of this Issue 9/2016

Tumor Biology 9/2016 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine