Skip to main content
Top
Published in: Clinical & Experimental Metastasis 1/2013

01-01-2013 | Research Paper

Expression of TGFβ3 and its effects on migratory and invasive behavior of prostate cancer cells: involvement of PI3-kinase/AKT signaling pathway

Authors: Lindsey Walker, Ana C. Millena, Nicole Strong, Shafiq A. Khan

Published in: Clinical & Experimental Metastasis | Issue 1/2013

Login to get access

Abstract

Transforming growth factor-β (TGFβ) is a secreted cytokine implicated as a factor in cancer cell migration and invasion. Previous studies have indicated that TGFβ isoforms may exert differential effects on cancer cells during different stages of the disease, however very little is known about the expression patterns and activity of the three isoforms in prostate cancer. Non-traditional signaling pathways including the PI3-Kinase have been associated with TGFβ-mediated effects on cancer cell invasion. In the present study, we have carried out expression analysis of TGFβ isoforms and signaling components in cell line models representing different stages of prostate cancer and studied the differential effects of specific isoforms on migratory and invasive behavior and induction of the PI3-kinase pathway. TGFβ1 and TGFβ3 were expressed in all cell lines, with TGFβ3 expression increasing in metastatic cell lines. Both TGFβ1 and TGFβ3 induced motility and invasive behavior in PC3 cells, however, TGFβ3 was significantly more potent than TGFβ1. TGFβRI and Smad3 inhibitors blocked TGFβ1 and TGFβ3 induced motility and invasion. TGFβ3 caused a significant increase in pAKTser473 in PC3 cells and PI3-kinase inhibitor LY294002 blocked TGFβ3 induced migration, invasion and phosphorylation of AKT. Both TGFβRI and Smad3 inhibitors blocked TGFβ3 induced pAKTser473. Based on these results, we conclude that TGFβ3 is expressed in metastatic prostate cancer cell lines and is involved in induction of invasive behavior in these cells. Furthermore, these effects of TGFβ3 are TGFβRI and Smad3 dependent and mediated via the PI3-kinase pathway.
Literature
1.
go back to reference Wakefield LM, Roberts AB (2002) TGF-beta signaling: positive and negative effects on tumorigenesis. Curr Opin Genet Dev 12(1):22–29PubMedCrossRef Wakefield LM, Roberts AB (2002) TGF-beta signaling: positive and negative effects on tumorigenesis. Curr Opin Genet Dev 12(1):22–29PubMedCrossRef
3.
go back to reference Massague J, Blain SW, Lo RS (2000) TGFbeta signaling in growth control, cancer, and heritable disorders. Cell 103(2):295–309PubMedCrossRef Massague J, Blain SW, Lo RS (2000) TGFbeta signaling in growth control, cancer, and heritable disorders. Cell 103(2):295–309PubMedCrossRef
4.
go back to reference Verrecchia F, Mauviel A (2002) Transforming growth factor-beta signaling through the Smad pathway: role in extracellular matrix gene expression and regulation. J Invest Dermatol 118(2):211–215PubMedCrossRef Verrecchia F, Mauviel A (2002) Transforming growth factor-beta signaling through the Smad pathway: role in extracellular matrix gene expression and regulation. J Invest Dermatol 118(2):211–215PubMedCrossRef
5.
go back to reference Seoane J (2006) Escaping from the TGFbeta anti-proliferative control. Carcinogenesis 27(11):2148–2156PubMedCrossRef Seoane J (2006) Escaping from the TGFbeta anti-proliferative control. Carcinogenesis 27(11):2148–2156PubMedCrossRef
6.
go back to reference Derynck R, Akhurst RJ, Balmain A (2001) TGF-beta signaling in tumor suppression and cancer progression. Nat Genet 29(2):117–129PubMedCrossRef Derynck R, Akhurst RJ, Balmain A (2001) TGF-beta signaling in tumor suppression and cancer progression. Nat Genet 29(2):117–129PubMedCrossRef
7.
go back to reference Bierie B, Moses HL (2009) Transforming growth factor beta (TGF-beta) and inflammation in cancer. Cytokine Growth Factor Rev 21(1):49–59PubMedCrossRef Bierie B, Moses HL (2009) Transforming growth factor beta (TGF-beta) and inflammation in cancer. Cytokine Growth Factor Rev 21(1):49–59PubMedCrossRef
8.
go back to reference Joshi A, Cao D TGF-beta signaling, tumor microenvironment and tumor progression: the butterfly effect. Front Biosci 15: 180-94 Joshi A, Cao D TGF-beta signaling, tumor microenvironment and tumor progression: the butterfly effect. Front Biosci 15: 180-94
9.
go back to reference Baardsnes J et al (2009) TbetaR-II discriminates the high- and low-affinity TGF-beta isoforms via two hydrogen-bonded ion pairs. Biochemistry 48(10):2146–2155PubMedCrossRef Baardsnes J et al (2009) TbetaR-II discriminates the high- and low-affinity TGF-beta isoforms via two hydrogen-bonded ion pairs. Biochemistry 48(10):2146–2155PubMedCrossRef
10.
go back to reference Piek E, Heldin CH, Ten Dijke P (1999) Specificity, diversity, and regulation in TGF-beta superfamily signaling. Faseb J 13(15):2105–2124PubMed Piek E, Heldin CH, Ten Dijke P (1999) Specificity, diversity, and regulation in TGF-beta superfamily signaling. Faseb J 13(15):2105–2124PubMed
11.
go back to reference Rahimi RA, Leof EB (2007) TGF-beta signaling: a tale of two responses. J Cell Biochem 102(3):593–608PubMedCrossRef Rahimi RA, Leof EB (2007) TGF-beta signaling: a tale of two responses. J Cell Biochem 102(3):593–608PubMedCrossRef
12.
13.
go back to reference Dickson MC et al (1995) Defective haematopoiesis and vasculogenesis in transforming growth factor-beta 1 knock out mice. Development 121(6):1845–1854PubMed Dickson MC et al (1995) Defective haematopoiesis and vasculogenesis in transforming growth factor-beta 1 knock out mice. Development 121(6):1845–1854PubMed
14.
go back to reference Sanford LP et al (1997) TGFbeta2 knockout mice have multiple developmental defects that are non-overlapping with other TGFbeta knockout phenotypes. Development 124(13):2659–2670PubMed Sanford LP et al (1997) TGFbeta2 knockout mice have multiple developmental defects that are non-overlapping with other TGFbeta knockout phenotypes. Development 124(13):2659–2670PubMed
15.
go back to reference Proetzel G et al (1995) Transforming growth factor-beta 3 is required for secondary palate fusion. Nat Genet 11(4):409–414PubMedCrossRef Proetzel G et al (1995) Transforming growth factor-beta 3 is required for secondary palate fusion. Nat Genet 11(4):409–414PubMedCrossRef
16.
go back to reference Kaartinen V et al (1995) Abnormal lung development and cleft palate in mice lacking TGF-beta 3 indicates defects of epithelial-mesenchymal interaction. Nat Genet 11(4):415–421PubMedCrossRef Kaartinen V et al (1995) Abnormal lung development and cleft palate in mice lacking TGF-beta 3 indicates defects of epithelial-mesenchymal interaction. Nat Genet 11(4):415–421PubMedCrossRef
17.
go back to reference Wrana JL et al (1992) TGF beta signals through a heteromeric protein kinase receptor complex. Cell 71(6):1003–1014PubMedCrossRef Wrana JL et al (1992) TGF beta signals through a heteromeric protein kinase receptor complex. Cell 71(6):1003–1014PubMedCrossRef
18.
go back to reference Cheifetz S et al (1990) Distinct transforming growth factor-beta (TGF-beta) receptor subsets as determinants of cellular responsiveness to three TGF-beta isoforms. J Biol Chem 265(33):20533–20538PubMed Cheifetz S et al (1990) Distinct transforming growth factor-beta (TGF-beta) receptor subsets as determinants of cellular responsiveness to three TGF-beta isoforms. J Biol Chem 265(33):20533–20538PubMed
19.
go back to reference De Crescenzo G et al (2003) Real-time monitoring of the interactions of two-stranded de novo designed coiled-coils: effect of chain length on the kinetic and thermodynamic constants of binding. Biochemistry 42(6):1754–1763PubMedCrossRef De Crescenzo G et al (2003) Real-time monitoring of the interactions of two-stranded de novo designed coiled-coils: effect of chain length on the kinetic and thermodynamic constants of binding. Biochemistry 42(6):1754–1763PubMedCrossRef
20.
go back to reference De Crescenzo G et al (2004) Enhancement of the antagonistic potency of transforming growth factor-beta receptor extracellular domains by coiled coil-induced homo- and heterodimerization. J Biol Chem 279(25):26013–26018PubMedCrossRef De Crescenzo G et al (2004) Enhancement of the antagonistic potency of transforming growth factor-beta receptor extracellular domains by coiled coil-induced homo- and heterodimerization. J Biol Chem 279(25):26013–26018PubMedCrossRef
21.
go back to reference Lopez-Casillas F, Wrana JL, Massague J (1993) Betaglycan presents ligand to the TGF beta signaling receptor. Cell 73(7):1435–1444PubMedCrossRef Lopez-Casillas F, Wrana JL, Massague J (1993) Betaglycan presents ligand to the TGF beta signaling receptor. Cell 73(7):1435–1444PubMedCrossRef
22.
go back to reference Vilchis-Landeros MM et al (2001) Recombinant soluble betaglycan is a potent and isoform-selective transforming growth factor-beta neutralizing agent. Biochem J 355(Pt 1):215–222PubMedCrossRef Vilchis-Landeros MM et al (2001) Recombinant soluble betaglycan is a potent and isoform-selective transforming growth factor-beta neutralizing agent. Biochem J 355(Pt 1):215–222PubMedCrossRef
23.
go back to reference Hart PJ et al (2002) Crystal structure of the human TbetaR2 ectodomain–TGF-beta3 complex. Nat Struct Biol 9(3):203–208PubMed Hart PJ et al (2002) Crystal structure of the human TbetaR2 ectodomain–TGF-beta3 complex. Nat Struct Biol 9(3):203–208PubMed
24.
go back to reference Van Themsche C et al (2007) Transforming growth factor-beta3 increases the invasiveness of endometrial carcinoma cells through phosphatidylinositol 3-kinase-dependent up-regulation of X-linked inhibitor of apoptosis and protein kinase c-dependent induction of matrix metalloproteinase-9. J Biol Chem 282(7):4794–4802PubMedCrossRef Van Themsche C et al (2007) Transforming growth factor-beta3 increases the invasiveness of endometrial carcinoma cells through phosphatidylinositol 3-kinase-dependent up-regulation of X-linked inhibitor of apoptosis and protein kinase c-dependent induction of matrix metalloproteinase-9. J Biol Chem 282(7):4794–4802PubMedCrossRef
25.
go back to reference Karan D et al (2002) Expression profile of differentially-regulated genes during progression of androgen-independent growth in human prostate cancer cells. Carcinogenesis 23(6):967–975PubMedCrossRef Karan D et al (2002) Expression profile of differentially-regulated genes during progression of androgen-independent growth in human prostate cancer cells. Carcinogenesis 23(6):967–975PubMedCrossRef
26.
go back to reference Assinder SJ et al (2009) The TGF-beta, PI3 K/Akt and PTEN pathways: established and proposed biochemical integration in prostate cancer. Biochem J 417(2):411–421PubMedCrossRef Assinder SJ et al (2009) The TGF-beta, PI3 K/Akt and PTEN pathways: established and proposed biochemical integration in prostate cancer. Biochem J 417(2):411–421PubMedCrossRef
27.
go back to reference Jiang BH, Liu LZ (2009) PI3 K/PTEN signaling in angiogenesis and tumorigenesis. Adv Cancer Res 102:19–65PubMedCrossRef Jiang BH, Liu LZ (2009) PI3 K/PTEN signaling in angiogenesis and tumorigenesis. Adv Cancer Res 102:19–65PubMedCrossRef
28.
29.
go back to reference Bakin AV et al (2000) Phosphatidylinositol 3-kinase function is required for transforming growth factor beta-mediated epithelial to mesenchymal transition and cell migration. J Biol Chem 275(47):36803–36810PubMedCrossRef Bakin AV et al (2000) Phosphatidylinositol 3-kinase function is required for transforming growth factor beta-mediated epithelial to mesenchymal transition and cell migration. J Biol Chem 275(47):36803–36810PubMedCrossRef
30.
go back to reference Lamouille S, Derynck R (2007) Cell size and invasion in TGF-beta-induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway. J Cell Biol 178(3):437–451PubMedCrossRef Lamouille S, Derynck R (2007) Cell size and invasion in TGF-beta-induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway. J Cell Biol 178(3):437–451PubMedCrossRef
31.
go back to reference Shin I et al (2001) Transforming growth factor beta enhances epithelial cell survival via Akt-dependent regulation of FKHRL1. Mol Biol Cell 12(11):3328–3329PubMed Shin I et al (2001) Transforming growth factor beta enhances epithelial cell survival via Akt-dependent regulation of FKHRL1. Mol Biol Cell 12(11):3328–3329PubMed
32.
go back to reference Vinals F, Pouyssegur J (2001) Transforming growth factor beta1 (TGF-beta1) promotes endothelial cell survival during in vitro angiogenesis via an autocrine mechanism implicating TGF-alpha signaling. Mol Cell Biol 21(21):7218–7230PubMedCrossRef Vinals F, Pouyssegur J (2001) Transforming growth factor beta1 (TGF-beta1) promotes endothelial cell survival during in vitro angiogenesis via an autocrine mechanism implicating TGF-alpha signaling. Mol Cell Biol 21(21):7218–7230PubMedCrossRef
33.
go back to reference Wilkes MC et al (2005) Transforming growth factor-beta activation of phosphatidylinositol 3-kinase is independent of Smad2 and Smad3 and regulates fibroblast responses via p21-activated kinase-2. Cancer Res 65(22):10431–10440PubMedCrossRef Wilkes MC et al (2005) Transforming growth factor-beta activation of phosphatidylinositol 3-kinase is independent of Smad2 and Smad3 and regulates fibroblast responses via p21-activated kinase-2. Cancer Res 65(22):10431–10440PubMedCrossRef
34.
go back to reference Zhong M et al (2010) Oxytocin induces the migration of prostate cancer cells: involvement of the Gi-coupled signaling pathway. Mol Cancer Res 8(8):1164–1172PubMedCrossRef Zhong M et al (2010) Oxytocin induces the migration of prostate cancer cells: involvement of the Gi-coupled signaling pathway. Mol Cancer Res 8(8):1164–1172PubMedCrossRef
35.
go back to reference Vo BT, Khan SA (2011) Expression of nodal and nodal receptors in prostate stem cells and prostate cancer cells: autocrine effects on cell proliferation and migration. Prostate 71(10):1084–1096PubMedCrossRef Vo BT, Khan SA (2011) Expression of nodal and nodal receptors in prostate stem cells and prostate cancer cells: autocrine effects on cell proliferation and migration. Prostate 71(10):1084–1096PubMedCrossRef
36.
go back to reference McDonald CA et al (2006) Follicle-stimulating hormone-induced aromatase in immature rat Sertoli cells requires an active phosphatidylinositol 3-kinase pathway and is inhibited via the mitogen-activated protein kinase signaling pathway. Mol Endocrinol 20(3):608–618PubMedCrossRef McDonald CA et al (2006) Follicle-stimulating hormone-induced aromatase in immature rat Sertoli cells requires an active phosphatidylinositol 3-kinase pathway and is inhibited via the mitogen-activated protein kinase signaling pathway. Mol Endocrinol 20(3):608–618PubMedCrossRef
37.
go back to reference Massague J (1984) Type beta transforming growth factor from feline sarcoma virus-transformed rat cells. Isolation and biological properties. J Biol Chem 259(15):9756–9761PubMed Massague J (1984) Type beta transforming growth factor from feline sarcoma virus-transformed rat cells. Isolation and biological properties. J Biol Chem 259(15):9756–9761PubMed
38.
go back to reference Reinhold D et al (1997) Inhibitors of dipeptidyl peptidase IV induce secretion of transforming growth factor-beta 1 in PWM-stimulated PBMC and T cells. Immunology 91(3):354–360PubMedCrossRef Reinhold D et al (1997) Inhibitors of dipeptidyl peptidase IV induce secretion of transforming growth factor-beta 1 in PWM-stimulated PBMC and T cells. Immunology 91(3):354–360PubMedCrossRef
39.
go back to reference Jones E, Pu H, Kyprianou N (2009) Targeting TGF-beta in prostate cancer: therapeutic possibilities during tumor progression. Expert Opin Ther Targets 13(2):227–234PubMedCrossRef Jones E, Pu H, Kyprianou N (2009) Targeting TGF-beta in prostate cancer: therapeutic possibilities during tumor progression. Expert Opin Ther Targets 13(2):227–234PubMedCrossRef
40.
go back to reference Amatschek S et al (2004) Tissue-wide expression profiling using cDNA subtraction and microarrays to identify tumor-specific genes. Cancer Res 64(3):844–856PubMedCrossRef Amatschek S et al (2004) Tissue-wide expression profiling using cDNA subtraction and microarrays to identify tumor-specific genes. Cancer Res 64(3):844–856PubMedCrossRef
41.
go back to reference Li C et al (1998) Role of transforming growth factor beta3 in lymphatic metastasis in breast cancer. Int J Cancer 79(5):455–459PubMedCrossRef Li C et al (1998) Role of transforming growth factor beta3 in lymphatic metastasis in breast cancer. Int J Cancer 79(5):455–459PubMedCrossRef
42.
44.
go back to reference Dubrovska A et al (2009) The role of PTEN/Akt/PI3 K signaling in the maintenance and viability of prostate cancer stem-like cell populations. Proc Natl Acad Sci USA 106(1):268–273PubMedCrossRef Dubrovska A et al (2009) The role of PTEN/Akt/PI3 K signaling in the maintenance and viability of prostate cancer stem-like cell populations. Proc Natl Acad Sci USA 106(1):268–273PubMedCrossRef
45.
go back to reference Cao C et al (2006) Inhibition of mammalian target of rapamycin or apoptotic pathway induces autophagy and radiosensitizes PTEN null prostate cancer cells. Cancer Res 66(20):10040–10047PubMedCrossRef Cao C et al (2006) Inhibition of mammalian target of rapamycin or apoptotic pathway induces autophagy and radiosensitizes PTEN null prostate cancer cells. Cancer Res 66(20):10040–10047PubMedCrossRef
46.
go back to reference Festuccia C et al (2005) Molecular aspects of gefitinib antiproliferative and pro-apoptotic effects in PTEN-positive and PTEN-negative prostate cancer cell lines. Endocr Relat Cancer 12(4):983–998PubMedCrossRef Festuccia C et al (2005) Molecular aspects of gefitinib antiproliferative and pro-apoptotic effects in PTEN-positive and PTEN-negative prostate cancer cell lines. Endocr Relat Cancer 12(4):983–998PubMedCrossRef
47.
go back to reference Huang H et al (2001) PTEN induces chemosensitivity in PTEN-mutated prostate cancer cells by suppression of Bcl-2 expression. J Biol Chem 276(42):38830–38836PubMedCrossRef Huang H et al (2001) PTEN induces chemosensitivity in PTEN-mutated prostate cancer cells by suppression of Bcl-2 expression. J Biol Chem 276(42):38830–38836PubMedCrossRef
Metadata
Title
Expression of TGFβ3 and its effects on migratory and invasive behavior of prostate cancer cells: involvement of PI3-kinase/AKT signaling pathway
Authors
Lindsey Walker
Ana C. Millena
Nicole Strong
Shafiq A. Khan
Publication date
01-01-2013
Publisher
Springer Netherlands
Published in
Clinical & Experimental Metastasis / Issue 1/2013
Print ISSN: 0262-0898
Electronic ISSN: 1573-7276
DOI
https://doi.org/10.1007/s10585-012-9494-0

Other articles of this Issue 1/2013

Clinical & Experimental Metastasis 1/2013 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