Skip to main content
Top
Published in: Cellular Oncology 6/2013

01-12-2013 | Original Paper

Interleukin-32β stimulates migration of MDA-MB-231 and MCF-7cells via the VEGF-STAT3 signaling pathway

Authors: Jeong Su Park, Su Yun Choi, Jeong-Hyung Lee, Maria Lee, Eun Sook Nam, Ae Lee Jeong, Sunyi Lee, Sora Han, Myeong-Sok Lee, Jong-Seok Lim, Do Young Yoon, Yongil Kwon, Young Yang

Published in: Cellular Oncology | Issue 6/2013

Login to get access

Abstract

Background

IL-32 is known to play an important role in inflammatory and autoimmune disease responses. In addition to its role in these responses, IL-32 and its different isoforms have in recent years been implicated in the development of various cancers. As of yet, the role of IL-32 in breast cancer has remained largely unknown.

Results

By performing immunohistochemical assays on primary breast cancer samples, we found that the level of IL-32β expression was positively correlated with tumor size, number of lymph node metastases and tumor stage. In addition, we found that breast cancer-derived MDA-MB-231 cells exogenously expressing IL-32β exhibited increased migration and invasion capacities. These increased capacities were found to be associated with an increased expression of the epithelial mesenchymal transition (EMT) markers vimentin and Slug, the latter of which is responsible for the increase in vimentin transcription. To next investigate whether IL-32β enhances migration and invasion through a soluble factor, we determined the levels of several migration-stimulating ligands, and found that the production of VEGF was increased by IL-32β. In addition, we found that IL-32β-induced VEGF increased migration and invasion through STAT3 activation.

Conclusion

The IL-32β-VEGF-STAT3 pathway represents an additional pathway that mediates the migration and invasion of breast cancer cells under the conditions of normoxia and hypoxia.
Literature
1.
go back to reference S.H. Kim, S.Y. Han, T. Azam, D.Y. Yoon, C.A. Dinarello, Interleukin-32: a cytokine and inducer of TNFalpha. Immunity 22(1), 131–142 (2005)PubMed S.H. Kim, S.Y. Han, T. Azam, D.Y. Yoon, C.A. Dinarello, Interleukin-32: a cytokine and inducer of TNFalpha. Immunity 22(1), 131–142 (2005)PubMed
2.
go back to reference P. Felaco, M.L. Castellani, M.A. De Lutiis, M. Felaco, F. Pandolfi, V. Salini, D. De Amicis, J. Vecchiet, S. Tete, C. Ciampoli, F. Conti, G. Cerulli, A. Caraffa, P. Antinolfi, C. Cuccurullo, A. Perrella, T.C. Theoharides, P. Conti, E. Toniato, D. Kempuraj, Y.B. Shaik, IL-32: a newly-discovered proinflammatory cytokine. J. Biol. Regul. Homeost. Agents 23(3), 141–147 (2009)PubMed P. Felaco, M.L. Castellani, M.A. De Lutiis, M. Felaco, F. Pandolfi, V. Salini, D. De Amicis, J. Vecchiet, S. Tete, C. Ciampoli, F. Conti, G. Cerulli, A. Caraffa, P. Antinolfi, C. Cuccurullo, A. Perrella, T.C. Theoharides, P. Conti, E. Toniato, D. Kempuraj, Y.B. Shaik, IL-32: a newly-discovered proinflammatory cytokine. J. Biol. Regul. Homeost. Agents 23(3), 141–147 (2009)PubMed
3.
go back to reference S.J. Na, S.H. So, K.O. Lee, Y.C. Choi, Elevated serum level of interleukin-32alpha in the patients with myasthenia gravis. J. Neurol. 258(10), 1865–1870 (2011)PubMedCrossRef S.J. Na, S.H. So, K.O. Lee, Y.C. Choi, Elevated serum level of interleukin-32alpha in the patients with myasthenia gravis. J. Neurol. 258(10), 1865–1870 (2011)PubMedCrossRef
4.
go back to reference P. Conti, P. Youinou, T.C. Theoharides, Modulation of autoimmunity by the latest interleukins (with special emphasis on IL-32). Autoimmun. Rev. 6(3), 131–137 (2007)PubMedCrossRef P. Conti, P. Youinou, T.C. Theoharides, Modulation of autoimmunity by the latest interleukins (with special emphasis on IL-32). Autoimmun. Rev. 6(3), 131–137 (2007)PubMedCrossRef
5.
go back to reference L.A. Joosten, M.G. Netea, S.H. Kim, D.Y. Yoon, B. Oppers-Walgreen, T.R. Radstake, P. Barrera, F.A. van de Loo, C.A. Dinarello, W.B. van den Berg, IL-32, a proinflammatory cytokine in rheumatoid arthritis. Proc. Natl. Acad. Sci. U. S. A. 103(9), 3298–3303 (2006)PubMedCrossRef L.A. Joosten, M.G. Netea, S.H. Kim, D.Y. Yoon, B. Oppers-Walgreen, T.R. Radstake, P. Barrera, F.A. van de Loo, C.A. Dinarello, W.B. van den Berg, IL-32, a proinflammatory cytokine in rheumatoid arthritis. Proc. Natl. Acad. Sci. U. S. A. 103(9), 3298–3303 (2006)PubMedCrossRef
6.
go back to reference N. Meyer, M. Zimmermann, S. Burgler, C. Bassin, S. Woehrl, K. Moritz, C. Rhyner, P. Indermitte, P. Schmid-Grendelmeier, M. Akdis, G. Menz, C.A. Akdis, IL-32 is expressed by human primary keratinocytes and modulates keratinocyte apoptosis in atopic dermatitis. J. Allergy Clin. Immunol. 125(4), 858–865 (2010). e810PubMedCrossRef N. Meyer, M. Zimmermann, S. Burgler, C. Bassin, S. Woehrl, K. Moritz, C. Rhyner, P. Indermitte, P. Schmid-Grendelmeier, M. Akdis, G. Menz, C.A. Akdis, IL-32 is expressed by human primary keratinocytes and modulates keratinocyte apoptosis in atopic dermatitis. J. Allergy Clin. Immunol. 125(4), 858–865 (2010). e810PubMedCrossRef
7.
go back to reference M.G. Netea, T. Azam, E.C. Lewis, L.A. Joosten, M. Wang, D. Langenberg, X. Meng, E.D. Chan, D.Y. Yoon, T. Ottenhoff, S.H. Kim, C.A. Dinarello, Mycobacterium tuberculosis induces interleukin-32 production through a caspase- 1/IL-18/interferon-gamma-dependent mechanism. PLoS Med. 3(8), e277 (2006)PubMedCrossRef M.G. Netea, T. Azam, E.C. Lewis, L.A. Joosten, M. Wang, D. Langenberg, X. Meng, E.D. Chan, D.Y. Yoon, T. Ottenhoff, S.H. Kim, C.A. Dinarello, Mycobacterium tuberculosis induces interleukin-32 production through a caspase- 1/IL-18/interferon-gamma-dependent mechanism. PLoS Med. 3(8), e277 (2006)PubMedCrossRef
8.
go back to reference H. J. Jeong, S. Y. Shin, H. A. Oh, M. H. Kim, J. S. Cho, H. M. Kim, IL-32 up-regulation is associated with inflammatory cytokine production in allergic rhinitis. J. Pathol 224(4), 553–563 (2011) H. J. Jeong, S. Y. Shin, H. A. Oh, M. H. Kim, J. S. Cho, H. M. Kim, IL-32 up-regulation is associated with inflammatory cytokine production in allergic rhinitis. J. Pathol 224(4), 553–563 (2011)
9.
go back to reference B. Heinhuis, M.I. Koenders, F.A. van de Loo, M.G. Netea, W.B. van den Berg, L.A. Joosten, Inflammation-dependent secretion and splicing of IL-32{gamma} in rheumatoid arthritis. Proc. Natl. Acad. Sci. U. S. A. 108(12), 4962–4967 (2011)PubMedCrossRef B. Heinhuis, M.I. Koenders, F.A. van de Loo, M.G. Netea, W.B. van den Berg, L.A. Joosten, Inflammation-dependent secretion and splicing of IL-32{gamma} in rheumatoid arthritis. Proc. Natl. Acad. Sci. U. S. A. 108(12), 4962–4967 (2011)PubMedCrossRef
10.
go back to reference A. R. Moschen, T. Fritz, A. D. Clouston, I. Rebhan, O. Bauhofer, H. D. Barrie, E. E. Powell, S. H. Kim, C. A. Dinarello, R. Bartenschlager, J. R. Jonsson, H. Tilg, Interleukin-32: a new proinflammatory cytokine involved in hepatitis C virus-related liver inflammation and fibrosis. Hepatology 53(6), 1819–1829 (2011) A. R. Moschen, T. Fritz, A. D. Clouston, I. Rebhan, O. Bauhofer, H. D. Barrie, E. E. Powell, S. H. Kim, C. A. Dinarello, R. Bartenschlager, J. R. Jonsson, H. Tilg, Interleukin-32: a new proinflammatory cytokine involved in hepatitis C virus-related liver inflammation and fibrosis. Hepatology 53(6), 1819–1829 (2011)
11.
go back to reference C. Goda, T. Kanaji, S. Kanaji, G. Tanaka, K. Arima, S. Ohno, K. Izuhara, Involvement of IL-32 in activation-induced cell death in T cells. Int. Immunol. 18(2), 233–240 (2006)PubMedCrossRef C. Goda, T. Kanaji, S. Kanaji, G. Tanaka, K. Arima, S. Ohno, K. Izuhara, Involvement of IL-32 in activation-induced cell death in T cells. Int. Immunol. 18(2), 233–240 (2006)PubMedCrossRef
12.
go back to reference N. Meyer, J. Christoph, H. Makrinioti, P. Indermitte, C. Rhyner, M. Soyka, T. Eiwegger, M. Chalubinski, K. Wanke, H. Fujita, P. Wawrzyniak, S. Burgler, S. Zhang, M. Akdis, G. Menz, C. Akdis, Inhibition of angiogenesis by IL-32: possible role in asthma. J. Allergy. Clin. Immunol. 129(4), 964–973 (2012) N. Meyer, J. Christoph, H. Makrinioti, P. Indermitte, C. Rhyner, M. Soyka, T. Eiwegger, M. Chalubinski, K. Wanke, H. Fujita, P. Wawrzyniak, S. Burgler, S. Zhang, M. Akdis, G. Menz, C. Akdis, Inhibition of angiogenesis by IL-32: possible role in asthma. J. Allergy. Clin. Immunol. 129(4), 964–973 (2012)
13.
go back to reference B. Heinhuis, M.I. Koenders, W.B. van den Berg, M.G. Netea, C.A. Dinarello, L.A. Joosten, Interleukin 32 (IL-32) contains a typical alpha-helix bundle structure that resembles focal adhesion targeting region of focal adhesion kinase-1. J. Biol. Chem. 287(8), 5733–5743 (2012)PubMedCrossRef B. Heinhuis, M.I. Koenders, W.B. van den Berg, M.G. Netea, C.A. Dinarello, L.A. Joosten, Interleukin 32 (IL-32) contains a typical alpha-helix bundle structure that resembles focal adhesion targeting region of focal adhesion kinase-1. J. Biol. Chem. 287(8), 5733–5743 (2012)PubMedCrossRef
14.
go back to reference A. Nishida, A. Andoh, O. Inatomi, Y. Fujiyama, Interleukin-32 expression in the pancreas. J. Biol. Chem. 284(26), 17868–17876 (2009)PubMedCrossRef A. Nishida, A. Andoh, O. Inatomi, Y. Fujiyama, Interleukin-32 expression in the pancreas. J. Biol. Chem. 284(26), 17868–17876 (2009)PubMedCrossRef
15.
go back to reference E.H. Seo, J. Kang, K.H. Kim, M.C. Cho, S. Lee, H.J. Kim, J.H. Kim, E.J. Kim, D.K. Park, S.H. Kim, Y.K. Choi, J.M. Kim, J.T. Hong, D.Y. Yoon, Detection of expressed IL-32 in human stomach cancer using ELISA and immunostaining. J. Microbiol. Biotechnol. 18(9), 1606–1612 (2008)PubMed E.H. Seo, J. Kang, K.H. Kim, M.C. Cho, S. Lee, H.J. Kim, J.H. Kim, E.J. Kim, D.K. Park, S.H. Kim, Y.K. Choi, J.M. Kim, J.T. Hong, D.Y. Yoon, Detection of expressed IL-32 in human stomach cancer using ELISA and immunostaining. J. Microbiol. Biotechnol. 18(9), 1606–1612 (2008)PubMed
16.
go back to reference C. Sorrentino, E. Di Carlo, Expression of IL-32 in human lung cancer is related to the histotype and metastatic phenotype. Am. J. Respir. Crit. Care Med. 180(8), 769–779 (2009)PubMedCrossRef C. Sorrentino, E. Di Carlo, Expression of IL-32 in human lung cancer is related to the histotype and metastatic phenotype. Am. J. Respir. Crit. Care Med. 180(8), 769–779 (2009)PubMedCrossRef
17.
go back to reference H. Kobayashi, P.C. Lin, Molecular characterization of IL-32 in human endothelial cells. Cytokine 46(3), 351–358 (2009)PubMedCrossRef H. Kobayashi, P.C. Lin, Molecular characterization of IL-32 in human endothelial cells. Cytokine 46(3), 351–358 (2009)PubMedCrossRef
18.
go back to reference Y.H. Kang, M.Y. Park, D.Y. Yoon, S.R. Han, C.I. Lee, N.Y. Ji, P.K. Myung, H.G. Lee, J.W. Kim, Y.I. Yeom, Y.J. Jang, D.K. Ahn, E.Y. Song, Dysregulation of overexpressed IL-32alpha in hepatocellular carcinoma suppresses cell growth and induces apoptosis through inactivation of NF-kappaB and Bcl-2. Cancer Lett. 318(2), 226–233 (2012)PubMedCrossRef Y.H. Kang, M.Y. Park, D.Y. Yoon, S.R. Han, C.I. Lee, N.Y. Ji, P.K. Myung, H.G. Lee, J.W. Kim, Y.I. Yeom, Y.J. Jang, D.K. Ahn, E.Y. Song, Dysregulation of overexpressed IL-32alpha in hepatocellular carcinoma suppresses cell growth and induces apoptosis through inactivation of NF-kappaB and Bcl-2. Cancer Lett. 318(2), 226–233 (2012)PubMedCrossRef
19.
go back to reference J.H. Oh, M.C. Cho, J.H. Kim, S.Y. Lee, H.J. Kim, E.S. Park, J.O. Ban, J.W. Kang, D.H. Lee, J.H. Shim, S.B. Han, D.C. Moon, Y.H. Park, D.Y. Yu, J.M. Kim, S.H. Kim, D.Y. Yoon, J.T. Hong, IL-32gamma inhibits cancer cell growth through inactivation of NF-kappaB and STAT3 signals. Oncogene 30(30), 3345–3359 (2011)PubMedCrossRef J.H. Oh, M.C. Cho, J.H. Kim, S.Y. Lee, H.J. Kim, E.S. Park, J.O. Ban, J.W. Kang, D.H. Lee, J.H. Shim, S.B. Han, D.C. Moon, Y.H. Park, D.Y. Yu, J.M. Kim, S.H. Kim, D.Y. Yoon, J.T. Hong, IL-32gamma inhibits cancer cell growth through inactivation of NF-kappaB and STAT3 signals. Oncogene 30(30), 3345–3359 (2011)PubMedCrossRef
20.
go back to reference W. Remmele, H.E. Stegner, [Recommendation for uniform definition of an immunoreactive score (IRS) for immunohistochemical estrogen receptor detection (ER-ICA) in breast cancer tissue]. Pathologe 8(3), 138–140 (1987)PubMed W. Remmele, H.E. Stegner, [Recommendation for uniform definition of an immunoreactive score (IRS) for immunohistochemical estrogen receptor detection (ER-ICA) in breast cancer tissue]. Pathologe 8(3), 138–140 (1987)PubMed
21.
go back to reference K. Vuoriluoto, H. Haugen, S. Kiviluoto, J.P. Mpindi, J. Nevo, C. Gjerdrum, C. Tiron, J.B. Lorens, J. Ivaska, Vimentin regulates EMT induction by Slug and oncogenic H-Ras and migration by governing Axl expression in breast cancer. Oncogene 30(12), 1436–1448 (2011)PubMedCrossRef K. Vuoriluoto, H. Haugen, S. Kiviluoto, J.P. Mpindi, J. Nevo, C. Gjerdrum, C. Tiron, J.B. Lorens, J. Ivaska, Vimentin regulates EMT induction by Slug and oncogenic H-Ras and migration by governing Axl expression in breast cancer. Oncogene 30(12), 1436–1448 (2011)PubMedCrossRef
22.
go back to reference A. Shibata, T. Nagaya, T. Imai, H. Funahashi, A. Nakao, H. Seo, Inhibition of NF-kappaB activity decreases the VEGF mRNA expression in MDA-MB-231 breast cancer cells. Breast Cancer Res. Treat. 73(3), 237–243 (2002)PubMedCrossRef A. Shibata, T. Nagaya, T. Imai, H. Funahashi, A. Nakao, H. Seo, Inhibition of NF-kappaB activity decreases the VEGF mRNA expression in MDA-MB-231 breast cancer cells. Breast Cancer Res. Treat. 73(3), 237–243 (2002)PubMedCrossRef
23.
go back to reference S. Weis, J. Cui, L. Barnes, D. Cheresh, Endothelial barrier disruption by VEGF-mediated Src activity potentiates tumor cell extravasation and metastasis. J. Cell Biol. 167(2), 223–229 (2004)PubMedCrossRef S. Weis, J. Cui, L. Barnes, D. Cheresh, Endothelial barrier disruption by VEGF-mediated Src activity potentiates tumor cell extravasation and metastasis. J. Cell Biol. 167(2), 223–229 (2004)PubMedCrossRef
24.
go back to reference N.S. Brown, R. Bicknell, Hypoxia and oxidative stress in breast cancer. Oxidative stress: its effects on the growth, metastatic potential and response to therapy of breast cancer. Breast Cancer Res. 3(5), 323–327 (2001)PubMedCrossRef N.S. Brown, R. Bicknell, Hypoxia and oxidative stress in breast cancer. Oxidative stress: its effects on the growth, metastatic potential and response to therapy of breast cancer. Breast Cancer Res. 3(5), 323–327 (2001)PubMedCrossRef
25.
go back to reference L. Li, Y. Lu, Inhibition of Hypoxia-Induced Cell Motility by p16 in MDA-MB-231 Breast Cancer Cells. J. Cancer 1, 126–135 (2010)PubMedCrossRef L. Li, Y. Lu, Inhibition of Hypoxia-Induced Cell Motility by p16 in MDA-MB-231 Breast Cancer Cells. J. Cancer 1, 126–135 (2010)PubMedCrossRef
26.
go back to reference M. Krol, K.M. Pawlowski, I. Dolka, O. Musielak, K. Majchrzak, J. Mucha, T. Motyl, Density of Gr1-positive myeloid precursor cells, p-STAT3 expression and gene expression pattern in canine mammary cancer metastasis. Vet. Res. Commun. 35(7), 409–423 (2011)PubMedCrossRef M. Krol, K.M. Pawlowski, I. Dolka, O. Musielak, K. Majchrzak, J. Mucha, T. Motyl, Density of Gr1-positive myeloid precursor cells, p-STAT3 expression and gene expression pattern in canine mammary cancer metastasis. Vet. Res. Commun. 35(7), 409–423 (2011)PubMedCrossRef
27.
go back to reference S. Zhang, H. E. Zhau, A. O. Osunkoya, S. Iqbal, X. Yang, S. Fan, Z. Chen, R. Wang, F. F. Marshall, L. W. Chung, D. Wu, Vascular endothelial growth factor regulates myeloid cell leukemia-1 expression through neuropilin-1-dependent activation of c-MET signaling in human prostate cancer cells. Mol. Cancer 9, 9 (2010) S. Zhang, H. E. Zhau, A. O. Osunkoya, S. Iqbal, X. Yang, S. Fan, Z. Chen, R. Wang, F. F. Marshall, L. W. Chung, D. Wu, Vascular endothelial growth factor regulates myeloid cell leukemia-1 expression through neuropilin-1-dependent activation of c-MET signaling in human prostate cancer cells. Mol. Cancer 9, 9 (2010)
28.
go back to reference S. Ishigami, T. Arigami, Y. Uchikado, T. Setoyama, Y. Kita, K. Sasaki, H. Okumura, H. Kurahara, Y. Kijima, A. Harada, S. Ueno, S. Natsugoe, IL-32 expression is an independent prognostic marker for gastric cancer. Med. Oncol. 30(2), 472 (2013)PubMedCrossRef S. Ishigami, T. Arigami, Y. Uchikado, T. Setoyama, Y. Kita, K. Sasaki, H. Okumura, H. Kurahara, Y. Kijima, A. Harada, S. Ueno, S. Natsugoe, IL-32 expression is an independent prognostic marker for gastric cancer. Med. Oncol. 30(2), 472 (2013)PubMedCrossRef
29.
go back to reference M.G. Netea, T. Azam, G. Ferwerda, S.E. Girardin, M. Walsh, J.S. Park, E. Abraham, J.M. Kim, D.Y. Yoon, C.A. Dinarello, S.H. Kim, IL-32 synergizes with nucleotide oligomerization domain (NOD) 1 and NOD2 ligands for IL-1beta and IL-6 production through a caspase 1-dependent mechanism. Proc. Natl. Acad. Sci. U. S. A. 102(45), 16309–16314 (2005)PubMedCrossRef M.G. Netea, T. Azam, G. Ferwerda, S.E. Girardin, M. Walsh, J.S. Park, E. Abraham, J.M. Kim, D.Y. Yoon, C.A. Dinarello, S.H. Kim, IL-32 synergizes with nucleotide oligomerization domain (NOD) 1 and NOD2 ligands for IL-1beta and IL-6 production through a caspase 1-dependent mechanism. Proc. Natl. Acad. Sci. U. S. A. 102(45), 16309–16314 (2005)PubMedCrossRef
30.
go back to reference C.A. Nold-Petry, M.F. Nold, J.A. Zepp, S.H. Kim, N.F. Voelkel, C.A. Dinarello, IL-32-dependent effects of IL-1beta on endothelial cell functions. Proc. Natl. Acad. Sci. U. S. A. 106(10), 3883–3888 (2009)PubMedCrossRef C.A. Nold-Petry, M.F. Nold, J.A. Zepp, S.H. Kim, N.F. Voelkel, C.A. Dinarello, IL-32-dependent effects of IL-1beta on endothelial cell functions. Proc. Natl. Acad. Sci. U. S. A. 106(10), 3883–3888 (2009)PubMedCrossRef
31.
go back to reference A.M. Marcondes, A.J. Mhyre, D.L. Stirewalt, S.H. Kim, C.A. Dinarello, H.J. Deeg, Dysregulation of IL-32 in myelodysplastic syndrome and chronic myelomonocytic leukemia modulates apoptosis and impairs NK function. Proc. Natl. Acad. Sci. U. S. A. 105(8), 2865–2870 (2008)PubMedCrossRef A.M. Marcondes, A.J. Mhyre, D.L. Stirewalt, S.H. Kim, C.A. Dinarello, H.J. Deeg, Dysregulation of IL-32 in myelodysplastic syndrome and chronic myelomonocytic leukemia modulates apoptosis and impairs NK function. Proc. Natl. Acad. Sci. U. S. A. 105(8), 2865–2870 (2008)PubMedCrossRef
32.
go back to reference R. Aesoy, B.C. Sanchez, J.H. Norum, R. Lewensohn, K. Viktorsson, B. Linderholm, An autocrine VEGF/VEGFR2 and p38 signaling loop confers resistance to 4-hydroxytamoxifen in MCF-7 breast cancer cells. Mol. Cancer Res. 6(10), 1630–1638 (2008)PubMedCrossRef R. Aesoy, B.C. Sanchez, J.H. Norum, R. Lewensohn, K. Viktorsson, B. Linderholm, An autocrine VEGF/VEGFR2 and p38 signaling loop confers resistance to 4-hydroxytamoxifen in MCF-7 breast cancer cells. Mol. Cancer Res. 6(10), 1630–1638 (2008)PubMedCrossRef
33.
go back to reference M. Dal Monte, D. Martini, C. Ristori, D. Azara, C. Armani, A. Balbarini, P. Bagnoli, Hypoxia effects on proangiogenic factors in human umbilical vein endothelial cells: functional role of the peptide somatostatin. Naunyn Schmiedebergs Arch. Pharmacol. 383(6), 593–612 (2011) M. Dal Monte, D. Martini, C. Ristori, D. Azara, C. Armani, A. Balbarini, P. Bagnoli, Hypoxia effects on proangiogenic factors in human umbilical vein endothelial cells: functional role of the peptide somatostatin. Naunyn Schmiedebergs Arch. Pharmacol. 383(6), 593–612 (2011)
34.
go back to reference G. Niu, K.L. Wright, M. Huang, L. Song, E. Haura, J. Turkson, S. Zhang, T. Wang, D. Sinibaldi, D. Coppola, R. Heller, L.M. Ellis, J. Karras, J. Bromberg, D. Pardoll, R. Jove, H. Yu, Constitutive Stat3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene 21(13), 2000–2008 (2002)PubMedCrossRef G. Niu, K.L. Wright, M. Huang, L. Song, E. Haura, J. Turkson, S. Zhang, T. Wang, D. Sinibaldi, D. Coppola, R. Heller, L.M. Ellis, J. Karras, J. Bromberg, D. Pardoll, R. Jove, H. Yu, Constitutive Stat3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene 21(13), 2000–2008 (2002)PubMedCrossRef
35.
go back to reference R.M. Neve, K. Chin, J. Fridlyand, J. Yeh, F.L. Baehner, T. Fevr, L. Clark, N. Bayani, J.P. Coppe, F. Tong, T. Speed, P.T. Spellman, S. DeVries, A. Lapuk, N.J. Wang, W.L. Kuo, J.L. Stilwell, D. Pinkel, D.G. Albertson, F.M. Waldman, F. McCormick, R.B. Dickson, M.D. Johnson, M. Lippman, S. Ethier, A. Gazdar, J.W. Gray, A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 10(6), 515–527 (2006)PubMedCrossRef R.M. Neve, K. Chin, J. Fridlyand, J. Yeh, F.L. Baehner, T. Fevr, L. Clark, N. Bayani, J.P. Coppe, F. Tong, T. Speed, P.T. Spellman, S. DeVries, A. Lapuk, N.J. Wang, W.L. Kuo, J.L. Stilwell, D. Pinkel, D.G. Albertson, F.M. Waldman, F. McCormick, R.B. Dickson, M.D. Johnson, M. Lippman, S. Ethier, A. Gazdar, J.W. Gray, A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 10(6), 515–527 (2006)PubMedCrossRef
36.
go back to reference L. McInroy, A. Maatta, Down-regulation of vimentin expression inhibits carcinoma cell migration and adhesion. Biochem. Biophys. Res. Commun. 360(1), 109–114 (2007)PubMedCrossRef L. McInroy, A. Maatta, Down-regulation of vimentin expression inhibits carcinoma cell migration and adhesion. Biochem. Biophys. Res. Commun. 360(1), 109–114 (2007)PubMedCrossRef
37.
go back to reference A.D. Yang, E.R. Camp, F. Fan, L. Shen, M.J. Gray, W. Liu, R. Somcio, T.W. Bauer, Y. Wu, D.J. Hicklin, L.M. Ellis, Vascular endothelial growth factor receptor-1 activation mediates epithelial to mesenchymal transition in human pancreatic carcinoma cells. Cancer Res. 66(1), 46–51 (2006)PubMedCrossRef A.D. Yang, E.R. Camp, F. Fan, L. Shen, M.J. Gray, W. Liu, R. Somcio, T.W. Bauer, Y. Wu, D.J. Hicklin, L.M. Ellis, Vascular endothelial growth factor receptor-1 activation mediates epithelial to mesenchymal transition in human pancreatic carcinoma cells. Cancer Res. 66(1), 46–51 (2006)PubMedCrossRef
38.
go back to reference A.K. Sasser, N.J. Sullivan, A.W. Studebaker, L.F. Hendey, A.E. Axel, B.M. Hall, Interleukin-6 is a potent growth factor for ER-alpha-positive human breast cancer. FASEB J. 21(13), 3763–3770 (2007)PubMedCrossRef A.K. Sasser, N.J. Sullivan, A.W. Studebaker, L.F. Hendey, A.E. Axel, B.M. Hall, Interleukin-6 is a potent growth factor for ER-alpha-positive human breast cancer. FASEB J. 21(13), 3763–3770 (2007)PubMedCrossRef
39.
go back to reference B. Wegiel, A. Bjartell, Z. Culig, J.L. Persson, Interleukin-6 activates PI3K/Akt pathway and regulates cyclin A1 to promote prostate cancer cell survival. Int. J. Cancer 122(7), 1521–1529 (2008)PubMedCrossRef B. Wegiel, A. Bjartell, Z. Culig, J.L. Persson, Interleukin-6 activates PI3K/Akt pathway and regulates cyclin A1 to promote prostate cancer cell survival. Int. J. Cancer 122(7), 1521–1529 (2008)PubMedCrossRef
40.
go back to reference T. Luedde, N. Beraza, V. Kotsikoris, G. van Loo, A. Nenci, R. De Vos, T. Roskams, C. Trautwein, M. Pasparakis, Deletion of NEMO/IKKgamma in liver parenchymal cells causes steatohepatitis and hepatocellular carcinoma. Cancer Cell 11(2), 119–132 (2007)PubMedCrossRef T. Luedde, N. Beraza, V. Kotsikoris, G. van Loo, A. Nenci, R. De Vos, T. Roskams, C. Trautwein, M. Pasparakis, Deletion of NEMO/IKKgamma in liver parenchymal cells causes steatohepatitis and hepatocellular carcinoma. Cancer Cell 11(2), 119–132 (2007)PubMedCrossRef
41.
go back to reference T. Sakurai, G. He, A. Matsuzawa, G.Y. Yu, S. Maeda, G. Hardiman, M. Karin, Hepatocyte necrosis induced by oxidative stress and IL-1 alpha release mediate carcinogen-induced compensatory proliferation and liver tumorigenesis. Cancer Cell 14(2), 156–165 (2008)PubMedCrossRef T. Sakurai, G. He, A. Matsuzawa, G.Y. Yu, S. Maeda, G. Hardiman, M. Karin, Hepatocyte necrosis induced by oxidative stress and IL-1 alpha release mediate carcinogen-induced compensatory proliferation and liver tumorigenesis. Cancer Cell 14(2), 156–165 (2008)PubMedCrossRef
Metadata
Title
Interleukin-32β stimulates migration of MDA-MB-231 and MCF-7cells via the VEGF-STAT3 signaling pathway
Authors
Jeong Su Park
Su Yun Choi
Jeong-Hyung Lee
Maria Lee
Eun Sook Nam
Ae Lee Jeong
Sunyi Lee
Sora Han
Myeong-Sok Lee
Jong-Seok Lim
Do Young Yoon
Yongil Kwon
Young Yang
Publication date
01-12-2013
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 6/2013
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-013-0154-4

Other articles of this Issue 6/2013

Cellular Oncology 6/2013 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