Skip to main content
Top
Published in: Cellular Oncology 2/2017

01-04-2017 | Original Paper

HPIP promotes epithelial-mesenchymal transition and cisplatin resistance in ovarian cancer cells through PI3K/AKT pathway activation

Authors: Suresh Bugide, Vijay Kumar Gonugunta, Vasudevarao Penugurti, Vijaya Lakshmi Malisetty, Ratna K. Vadlamudi, Bramanandam Manavathi

Published in: Cellular Oncology | Issue 2/2017

Login to get access

Abstract

Purpose

Hematopoietic PBX interacting protein (HPIP), a scaffold protein, is known to regulate the proliferation, migration and invasion in different cancer cell types. The aim of this study was to assess the role of HPIP in ovarian cancer cell migration, invasion and epithelial-mesenchymal transition (EMT), and to unravel the mechanism by which it regulates these processes.

Methods

HPIP expression was assessed by immunohistochemistry of tissue microarrays containing primary ovarian tumor samples of different grades. OAW42, an ovarian carcinoma-derived cell line exhibiting a high HPIP expression, was used to study the role of HPIP in cell migration, invasion and EMT. HPIP knockdown in these cells was achieved using a small hairpin RNA (shRNA) approach. Cell migration and invasion were assessed using scratch wound and transwell invasion assays, respectively. The extent of EMT was assessed by determining the expression levels of Snail, Vimentin and E-cadherin using Western blotting. The effect of HPIP expression on AKT and MAPK activation was also investigated by Western blotting. Cell viabilities in response to cisplatin treatment were assessed using a MTT assay, whereas apoptosis was assessed by determining caspase-3 and PARP cleavage in ovarian carcinoma-derived SKOV3 cells.

Results

We found that HPIP is highly expressed in high-grade primary ovarian tumors. In addition, we found that HPIP promotes the migration, invasion and EMT in OAW42 cells and induces EMT in these cells via activation of the PI3K/AKT pathway. The latter was found to lead to stabilization of the Snail protein and to repression of E-cadherin expression through inactivation of GSK-3β. We also found that HPIP expression confers cisplatin resistance to SKOV3 cells after prolonged exposure and that its subsequent knockdown decreases the viability of these cells and increases caspase-3 activation and PARP proteolysis in these cells following cisplatin treatment.

Conclusions

From these results we conclude that HPIP expression is associated with high-grade ovarian tumors and may promote their migration, invasion and EMT, a process that is associated with metastasis. In addition, we conclude that HPIP may serve as a potential therapeutic target for cisplatin resistant ovarian tumors.
Appendix
Available only for authorised users
Literature
1.
go back to reference R.P. Perez, A.K. Godwin, T.C. Hamilton, R.F. Ozols, Ovarian cancer biology. Semin Oncol 18, 186–204 (1991)PubMed R.P. Perez, A.K. Godwin, T.C. Hamilton, R.F. Ozols, Ovarian cancer biology. Semin Oncol 18, 186–204 (1991)PubMed
6.
9.
go back to reference J. Mikula-Pietrasik, P. Uruski, K. Matuszkiewicz, S. Szubert, R. Moszynski, D. Szpurek, S. Sajdak, A. Tykarski, K. Ksiazek, Ovarian cancer-derived ascitic fluids induce a senescence-dependent pro-cancerogenic phenotype in normal peritoneal mesothelial cells. Cell Oncol 39, 473–481 (2016). doi:10.1007/s13402-016-0289-1 CrossRef J. Mikula-Pietrasik, P. Uruski, K. Matuszkiewicz, S. Szubert, R. Moszynski, D. Szpurek, S. Sajdak, A. Tykarski, K. Ksiazek, Ovarian cancer-derived ascitic fluids induce a senescence-dependent pro-cancerogenic phenotype in normal peritoneal mesothelial cells. Cell Oncol 39, 473–481 (2016). doi:10.​1007/​s13402-016-0289-1 CrossRef
10.
go back to reference Y. Kuwabara, T. Yamada, K. Yamazaki, W.L. Du, K. Banno, D. Aoki, M. Sakamoto, Establishment of an ovarian metastasis model and possible involvement of E-cadherin down-regulation in the metastasis. Cancer Sci 99, 1933–1939 (2008). doi:10.1111/j.1349-7006.2008.00946.x PubMed Y. Kuwabara, T. Yamada, K. Yamazaki, W.L. Du, K. Banno, D. Aoki, M. Sakamoto, Establishment of an ovarian metastasis model and possible involvement of E-cadherin down-regulation in the metastasis. Cancer Sci 99, 1933–1939 (2008). doi:10.​1111/​j.​1349-7006.​2008.​00946.​x PubMed
12.
15.
go back to reference X. Xu, Z. Fan, L. Kang, J. Han, C. Jiang, X. Zheng, Z. Zhu, H. Jiao, J. Lin, K. Jiang, L. Ding, H. Zhang, L. Cheng, H. Fu, Y. Song, Y. Jiang, J. Liu, R. Wang, N. Du, Q. Ye, Hepatitis B virus X protein represses miRNA-148a to enhance tumorigenesis. J Clin Invest 123, 630–645 (2013). doi:10.1172/JCI64265 PubMedPubMedCentral X. Xu, Z. Fan, L. Kang, J. Han, C. Jiang, X. Zheng, Z. Zhu, H. Jiao, J. Lin, K. Jiang, L. Ding, H. Zhang, L. Cheng, H. Fu, Y. Song, Y. Jiang, J. Liu, R. Wang, N. Du, Q. Ye, Hepatitis B virus X protein represses miRNA-148a to enhance tumorigenesis. J Clin Invest 123, 630–645 (2013). doi:10.​1172/​JCI64265 PubMedPubMedCentral
16.
go back to reference S. Bugide, D. David, A. Nair, N. Kannan, V.S. Samanthapudi, J. Prabhakar, B. Manavathi, Hematopoietic PBX-interacting protein (HPIP) is over expressed in breast infiltrative ductal carcinoma and regulates cell adhesion and migration through modulation of focal adhesion dynamics. Oncogene 34, 4601–4612 (2015). doi:10.1038/onc.2014.389 CrossRefPubMed S. Bugide, D. David, A. Nair, N. Kannan, V.S. Samanthapudi, J. Prabhakar, B. Manavathi, Hematopoietic PBX-interacting protein (HPIP) is over expressed in breast infiltrative ductal carcinoma and regulates cell adhesion and migration through modulation of focal adhesion dynamics. Oncogene 34, 4601–4612 (2015). doi:10.​1038/​onc.​2014.​389 CrossRefPubMed
17.
go back to reference Y. Feng, X. Xu, Y. Zhang, J. Ding, Y. Wang, X. Zhang, Z. Wu, L. Kang, Y. Liang, L. Zhou, S. Song, K. Zhao, Q. Ye, HPIP is upregulated in colorectal cancer and regulates colorectal cancer cell proliferation, apoptosis and invasion. Sci Rep 5, 9429 (2015). doi:10.1038/srep09429 CrossRefPubMedPubMedCentral Y. Feng, X. Xu, Y. Zhang, J. Ding, Y. Wang, X. Zhang, Z. Wu, L. Kang, Y. Liang, L. Zhou, S. Song, K. Zhao, Q. Ye, HPIP is upregulated in colorectal cancer and regulates colorectal cancer cell proliferation, apoptosis and invasion. Sci Rep 5, 9429 (2015). doi:10.​1038/​srep09429 CrossRefPubMedPubMedCentral
18.
go back to reference Y. Feng, L. Li, X. Zhang, Y. Zhang, Y. Liang, J. Lv, Z. Fan, J. Guo, T. Hong, B. Ji, Q. Ji, G. Mei, L. Ding, S. Zhang, X. Xu, Q. Ye, HPIP is overexpressed in gastric cancer and promotes gastric cancer cell proliferation, migration and invasion. Cancer Sci 106, 1313–1322 (2015). doi:10.1111/cas.12754 CrossRefPubMedPubMedCentral Y. Feng, L. Li, X. Zhang, Y. Zhang, Y. Liang, J. Lv, Z. Fan, J. Guo, T. Hong, B. Ji, Q. Ji, G. Mei, L. Ding, S. Zhang, X. Xu, Q. Ye, HPIP is overexpressed in gastric cancer and promotes gastric cancer cell proliferation, migration and invasion. Cancer Sci 106, 1313–1322 (2015). doi:10.​1111/​cas.​12754 CrossRefPubMedPubMedCentral
20.
go back to reference S. Okada, T. Irie, J. Tanaka, R. Yasuhara, G. Yamamoto, T. Isobe, C. Hokazono, T. Tachikawa, Y. Kohno, K. Mishima, Potential role of hematopoietic pre-B-cell leukemia transcription factor-interacting protein in oral carcinogenesis. J Oral Pathol Med 44, 115–125 (2015). doi:10.1111/jop.12210 CrossRefPubMed S. Okada, T. Irie, J. Tanaka, R. Yasuhara, G. Yamamoto, T. Isobe, C. Hokazono, T. Tachikawa, Y. Kohno, K. Mishima, Potential role of hematopoietic pre-B-cell leukemia transcription factor-interacting protein in oral carcinogenesis. J Oral Pathol Med 44, 115–125 (2015). doi:10.​1111/​jop.​12210 CrossRefPubMed
22.
go back to reference D.G. van Vuurden, E. Aronica, E. Hulleman, L.E. Wedekind, D. Biesmans, A. Malekzadeh, M. Bugiani, D. Geerts, D.P. Noske, W.P. Vandertop, G.J. Kaspers, J. Cloos, T. Wurdinger, P.P. van der Stoop, Pre-B-cell leukemia homeobox interacting protein 1 is overexpressed in astrocytoma and promotes tumor cell growth and migration. Neuro-Oncology 16, 946–959 (2014). doi:10.1093/neuonc/not308 CrossRefPubMedPubMedCentral D.G. van Vuurden, E. Aronica, E. Hulleman, L.E. Wedekind, D. Biesmans, A. Malekzadeh, M. Bugiani, D. Geerts, D.P. Noske, W.P. Vandertop, G.J. Kaspers, J. Cloos, T. Wurdinger, P.P. van der Stoop, Pre-B-cell leukemia homeobox interacting protein 1 is overexpressed in astrocytoma and promotes tumor cell growth and migration. Neuro-Oncology 16, 946–959 (2014). doi:10.​1093/​neuonc/​not308 CrossRefPubMedPubMedCentral
23.
go back to reference K. Shostak, F. Patrascu, S.I. Goktuna, P. Close, L. Borgs, L. Nguyen, F. Olivier, A. Rammal, H. Brinkhaus, M. Bentires-Alj, J.C. Marine, A. Chariot, MDM2 restrains estrogen-mediated AKT activation by promoting TBK1-dependent HPIP degradation. Cell Death Differ 21, 811–824 (2014). doi:10.1038/cdd.2014.2 CrossRefPubMedPubMedCentral K. Shostak, F. Patrascu, S.I. Goktuna, P. Close, L. Borgs, L. Nguyen, F. Olivier, A. Rammal, H. Brinkhaus, M. Bentires-Alj, J.C. Marine, A. Chariot, MDM2 restrains estrogen-mediated AKT activation by promoting TBK1-dependent HPIP degradation. Cell Death Differ 21, 811–824 (2014). doi:10.​1038/​cdd.​2014.​2 CrossRefPubMedPubMedCentral
24.
go back to reference V.N. Gajulapalli, V.S. Samanthapudi, M. Pulaganti, S.S. Khumukcham, V.L. Malisetty, L. Guruprasad, S.K. Chitta, B. Manavathi, A transcriptional repressive role for epithelial-specific ETS factor ELF3 on oestrogen receptor alpha in breast cancer cells. Biochem J 473, 1047–1061 (2016). doi:10.1042/BCJ20160019 CrossRefPubMed V.N. Gajulapalli, V.S. Samanthapudi, M. Pulaganti, S.S. Khumukcham, V.L. Malisetty, L. Guruprasad, S.K. Chitta, B. Manavathi, A transcriptional repressive role for epithelial-specific ETS factor ELF3 on oestrogen receptor alpha in breast cancer cells. Biochem J 473, 1047–1061 (2016). doi:10.​1042/​BCJ20160019 CrossRefPubMed
25.
go back to reference G.R. Sareddy, B.C. Nair, S.K. Krishnan, V.K. Gonugunta, Q.G. Zhang, T. Suzuki, N. Miyata, A.J. Brenner, D.W. Brann, R.K. Vadlamudi, KDM1 is a novel therapeutic target for the treatment of gliomas. Oncotarget 4, 18–28 (2013). doi:10.18632/oncotarget.725 PubMed G.R. Sareddy, B.C. Nair, S.K. Krishnan, V.K. Gonugunta, Q.G. Zhang, T. Suzuki, N. Miyata, A.J. Brenner, D.W. Brann, R.K. Vadlamudi, KDM1 is a novel therapeutic target for the treatment of gliomas. Oncotarget 4, 18–28 (2013). doi:10.​18632/​oncotarget.​725 PubMed
26.
go back to reference T. Bonome, D.A. Levine, J. Shih, M. Randonovich, C.A. Pise-Masison, F. Bogomolniy, L. Ozbun, J. Brady, J.C. Barrett, J. Boyd, M.J. Birrer, A gene signature predicting for survival in suboptimally debulked patients with ovarian cancer. Cancer Res 68, 5478–5486 (2008). doi:10.1158/0008-5472.CAN-07-6595 CrossRefPubMed T. Bonome, D.A. Levine, J. Shih, M. Randonovich, C.A. Pise-Masison, F. Bogomolniy, L. Ozbun, J. Brady, J.C. Barrett, J. Boyd, M.J. Birrer, A gene signature predicting for survival in suboptimally debulked patients with ovarian cancer. Cancer Res 68, 5478–5486 (2008). doi:10.​1158/​0008-5472.​CAN-07-6595 CrossRefPubMed
29.
go back to reference E. Batlle, E. Sancho, C. Franci, D. Dominguez, M. Monfar, J. Baulida, A. Garcia De Herreros, The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol 2, 84–89 (2000). doi:10.1038/35000034 CrossRefPubMed E. Batlle, E. Sancho, C. Franci, D. Dominguez, M. Monfar, J. Baulida, A. Garcia De Herreros, The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol 2, 84–89 (2000). doi:10.​1038/​35000034 CrossRefPubMed
31.
go back to reference B.P. Zhou, J. Deng, W. Xia, J. Xu, Y.M. Li, M. Gunduz, M.C. Hung, Dual regulation of snail by GSK-3beta-mediated phosphorylation in control of epithelial-mesenchymal transition. Nat Cell Biol 6, 931–940 (2004). doi:10.1038/ncb1173 CrossRefPubMed B.P. Zhou, J. Deng, W. Xia, J. Xu, Y.M. Li, M. Gunduz, M.C. Hung, Dual regulation of snail by GSK-3beta-mediated phosphorylation in control of epithelial-mesenchymal transition. Nat Cell Biol 6, 931–940 (2004). doi:10.​1038/​ncb1173 CrossRefPubMed
32.
go back to reference D.A. Cross, D.R. Alessi, P. Cohen, M. Andjelkovich, B.A. Hemmings, Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature 378, 785–789 (1995). doi:10.1038/378785a0 CrossRefPubMed D.A. Cross, D.R. Alessi, P. Cohen, M. Andjelkovich, B.A. Hemmings, Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature 378, 785–789 (1995). doi:10.​1038/​378785a0 CrossRefPubMed
33.
go back to reference A. Cano, M.A. Perez-Moreno, I. Rodrigo, A. Locascio, M.J. Blanco, M.G. del Barrio, F. Portillo, M.A. Nieto, The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol 2, 76–83 (2000). doi:10.1038/35000025 CrossRefPubMed A. Cano, M.A. Perez-Moreno, I. Rodrigo, A. Locascio, M.J. Blanco, M.G. del Barrio, F. Portillo, M.A. Nieto, The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol 2, 76–83 (2000). doi:10.​1038/​35000025 CrossRefPubMed
35.
go back to reference H. Asechi, E. Hatano, T. Nitta, M. Tada, K. Iwaisako, N. Tamaki, H. Nagata, M. Narita, A. Yanagida, I. Ikai, S. Uemoto, Resistance to cisplatin-induced apoptosis via PI3K-dependent survivin expression in a rat hepatoma cell line. Int J Oncol 37, 89–96 (2010)PubMed H. Asechi, E. Hatano, T. Nitta, M. Tada, K. Iwaisako, N. Tamaki, H. Nagata, M. Narita, A. Yanagida, I. Ikai, S. Uemoto, Resistance to cisplatin-induced apoptosis via PI3K-dependent survivin expression in a rat hepatoma cell line. Int J Oncol 37, 89–96 (2010)PubMed
39.
go back to reference R.G. Verhaak, P. Tamayo, J.Y. Yang, D. Hubbard, H. Zhang, C.J. Creighton, S. Fereday, M. Lawrence, S.L. Carter, C.H. Mermel, A.D. Kostic, D. Etemadmoghadam, G. Saksena, K. Cibulskis, S. Duraisamy, K. Levanon, C. Sougnez, A. Tsherniak, S. Gomez, R. Onofrio, S. Gabriel, L. Chin, N. Zhang, P.T. Spellman, Y. Zhang, R. Akbani, K.A. Hoadley, A. Kahn, M. Kobel, D. Huntsman, R.A. Soslow, A. Defazio, M.J. Birrer, J.W. Gray, J.N. Weinstein, D.D. Bowtell, R. Drapkin, J.P. Mesirov, G. Getz, D.A. Levine, M. Meyerson, Cancer genome atlas research, Prognostically relevant gene signatures of high-grade serous ovarian carcinoma. J Clin Invest 123, 517–525 (2013). doi:10.1172/JCI65833 PubMed R.G. Verhaak, P. Tamayo, J.Y. Yang, D. Hubbard, H. Zhang, C.J. Creighton, S. Fereday, M. Lawrence, S.L. Carter, C.H. Mermel, A.D. Kostic, D. Etemadmoghadam, G. Saksena, K. Cibulskis, S. Duraisamy, K. Levanon, C. Sougnez, A. Tsherniak, S. Gomez, R. Onofrio, S. Gabriel, L. Chin, N. Zhang, P.T. Spellman, Y. Zhang, R. Akbani, K.A. Hoadley, A. Kahn, M. Kobel, D. Huntsman, R.A. Soslow, A. Defazio, M.J. Birrer, J.W. Gray, J.N. Weinstein, D.D. Bowtell, R. Drapkin, J.P. Mesirov, G. Getz, D.A. Levine, M. Meyerson, Cancer genome atlas research, Prognostically relevant gene signatures of high-grade serous ovarian carcinoma. J Clin Invest 123, 517–525 (2013). doi:10.​1172/​JCI65833 PubMed
41.
go back to reference D.A. Altomare, H.Q. Wang, K.L. Skele, A. De Rienzo, A.J. Klein-Szanto, A.K. Godwin, J.R. Testa, AKT and mTOR phosphorylation is frequently detected in ovarian cancer and can be targeted to disrupt ovarian tumor cell growth. Oncogene 23, 5853–5857 (2004). doi:10.1038/sj.onc.1207721 CrossRefPubMed D.A. Altomare, H.Q. Wang, K.L. Skele, A. De Rienzo, A.J. Klein-Szanto, A.K. Godwin, J.R. Testa, AKT and mTOR phosphorylation is frequently detected in ovarian cancer and can be targeted to disrupt ovarian tumor cell growth. Oncogene 23, 5853–5857 (2004). doi:10.​1038/​sj.​onc.​1207721 CrossRefPubMed
42.
go back to reference A.J. Hanrahan, N. Schultz, M.L. Westfal, R.A. Sakr, D.D. Giri, S. Scarperi, M. Janakiraman, N. Olvera, E.V. Stevens, Q.B. She, C. Aghajanian, T.A. King, E. Stanchina, D.R. Spriggs, A. Heguy, B.S. Taylor, C. Sander, N. Rosen, D.A. Levine, D.B. Solit, Genomic complexity and AKT dependence in serous ovarian cancer. Cancer Discov 2, 56–67 (2012). doi:10.1158/2159-8290.CD-11-0170 CrossRefPubMedPubMedCentral A.J. Hanrahan, N. Schultz, M.L. Westfal, R.A. Sakr, D.D. Giri, S. Scarperi, M. Janakiraman, N. Olvera, E.V. Stevens, Q.B. She, C. Aghajanian, T.A. King, E. Stanchina, D.R. Spriggs, A. Heguy, B.S. Taylor, C. Sander, N. Rosen, D.A. Levine, D.B. Solit, Genomic complexity and AKT dependence in serous ovarian cancer. Cancer Discov 2, 56–67 (2012). doi:10.​1158/​2159-8290.​CD-11-0170 CrossRefPubMedPubMedCentral
43.
go back to reference K. Kurose, X.P. Zhou, T. Araki, S.A. Cannistra, E.R. Maher, C. Eng, Frequent loss of PTEN expression is linked to elevated phosphorylated Akt levels, but not associated with p27 and cyclin D1 expression, in primary epithelial ovarian carcinomas. Am J Pathol 158, 2097–2106 (2001). doi:10.1016/S0002-9440(10)64681-0 CrossRefPubMedPubMedCentral K. Kurose, X.P. Zhou, T. Araki, S.A. Cannistra, E.R. Maher, C. Eng, Frequent loss of PTEN expression is linked to elevated phosphorylated Akt levels, but not associated with p27 and cyclin D1 expression, in primary epithelial ovarian carcinomas. Am J Pathol 158, 2097–2106 (2001). doi:10.​1016/​S0002-9440(10)64681-0 CrossRefPubMedPubMedCentral
44.
go back to reference M. Sun, G. Wang, J.E. Paciga, R.I. Feldman, Z.Q. Yuan, X.L. Ma, S.A. Shelley, R. Jove, P.N. Tsichlis, S.V. Nicosia, J.Q. Cheng, AKT1/PKBalpha kinase is frequently elevated in human cancers and its constitutive activation is required for oncogenic transformation in NIH3T3 cells. Am J Pathol 159, 431–437 (2001)CrossRefPubMedPubMedCentral M. Sun, G. Wang, J.E. Paciga, R.I. Feldman, Z.Q. Yuan, X.L. Ma, S.A. Shelley, R. Jove, P.N. Tsichlis, S.V. Nicosia, J.Q. Cheng, AKT1/PKBalpha kinase is frequently elevated in human cancers and its constitutive activation is required for oncogenic transformation in NIH3T3 cells. Am J Pathol 159, 431–437 (2001)CrossRefPubMedPubMedCentral
45.
go back to reference Z.Q. Yuan, M. Sun, R.I. Feldman, G. Wang, X. Ma, C. Jiang, D. Coppola, S.V. Nicosia, J.Q. Cheng, Frequent activation of AKT2 and induction of apoptosis by inhibition of phosphoinositide-3-OH kinase/Akt pathway in human ovarian cancer. Oncogene 19, 2324–2330 (2000). doi:10.1038/sj.onc.1203598 CrossRefPubMed Z.Q. Yuan, M. Sun, R.I. Feldman, G. Wang, X. Ma, C. Jiang, D. Coppola, S.V. Nicosia, J.Q. Cheng, Frequent activation of AKT2 and induction of apoptosis by inhibition of phosphoinositide-3-OH kinase/Akt pathway in human ovarian cancer. Oncogene 19, 2324–2330 (2000). doi:10.​1038/​sj.​onc.​1203598 CrossRefPubMed
46.
go back to reference L. Shayesteh, Y. Lu, W.L. Kuo, R. Baldocchi, T. Godfrey, C. Collins, D. Pinkel, B. Powell, G.B. Mills, J.W. Gray, PIK3CA is implicated as an oncogene in ovarian cancer. Nat Genet 21, 99–102 (1999). doi:10.1038/5042 CrossRefPubMed L. Shayesteh, Y. Lu, W.L. Kuo, R. Baldocchi, T. Godfrey, C. Collins, D. Pinkel, B. Powell, G.B. Mills, J.W. Gray, PIK3CA is implicated as an oncogene in ovarian cancer. Nat Genet 21, 99–102 (1999). doi:10.​1038/​5042 CrossRefPubMed
47.
go back to reference K.E. Sheppard, C. Cullinane, K.M. Hannan, M. Wall, J. Chan, F. Barber, J. Foo, D. Cameron, A. Neilsen, P. Ng, J. Ellul, M. Kleinschmidt, K.M. Kinross, D.D. Bowtell, J.G. Christensen, R.J. Hicks, R.W. Johnstone, G.A. McArthur, R.D. Hannan, W.A. Phillips, R.B. Pearson, Synergistic inhibition of ovarian cancer cell growth by combining selective PI3K/mTOR and RAS/ERK pathway inhibitors. Eur J Cancer 49, 3936–3944 (2013). doi:10.1016/j.ejca.2013.08.007 CrossRefPubMed K.E. Sheppard, C. Cullinane, K.M. Hannan, M. Wall, J. Chan, F. Barber, J. Foo, D. Cameron, A. Neilsen, P. Ng, J. Ellul, M. Kleinschmidt, K.M. Kinross, D.D. Bowtell, J.G. Christensen, R.J. Hicks, R.W. Johnstone, G.A. McArthur, R.D. Hannan, W.A. Phillips, R.B. Pearson, Synergistic inhibition of ovarian cancer cell growth by combining selective PI3K/mTOR and RAS/ERK pathway inhibitors. Eur J Cancer 49, 3936–3944 (2013). doi:10.​1016/​j.​ejca.​2013.​08.​007 CrossRefPubMed
50.
go back to reference G. Singer, R. Oldt 3rd, Y. Cohen, B.G. Wang, D. Sidransky, R.J. Kurman, M. Shih Ie, Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J Natl Cancer Inst 95, 484–486 (2003)CrossRefPubMed G. Singer, R. Oldt 3rd, Y. Cohen, B.G. Wang, D. Sidransky, R.J. Kurman, M. Shih Ie, Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J Natl Cancer Inst 95, 484–486 (2003)CrossRefPubMed
51.
52.
go back to reference Y. Yang, J. Zhang, Y. Zhu, Z. Zhang, H. Sun, Y. Feng, Follicle-stimulating hormone induced epithelial-mesenchymal transition of epithelial ovarian cancer cells through follicle-stimulating hormone receptor PI3K/Akt-snail signaling pathway. Int J Gynecol Cancer 24, 1564–1574 (2014). doi:10.1097/IGC.0000000000000279 CrossRefPubMed Y. Yang, J. Zhang, Y. Zhu, Z. Zhang, H. Sun, Y. Feng, Follicle-stimulating hormone induced epithelial-mesenchymal transition of epithelial ovarian cancer cells through follicle-stimulating hormone receptor PI3K/Akt-snail signaling pathway. Int J Gynecol Cancer 24, 1564–1574 (2014). doi:10.​1097/​IGC.​0000000000000279​ CrossRefPubMed
56.
go back to reference Y. Zhang, C. Bao, Q. Mu, J. Chen, J. Wang, Y. Mi, A.J. Sayari, Y. Chen, M. Guo, Reversal of cisplatin resistance by inhibiting PI3K/Akt signal pathway in human lung cancer cells. Neoplasma 63, 362–370 (2016). doi:10.4149/304_150806N433 CrossRefPubMed Y. Zhang, C. Bao, Q. Mu, J. Chen, J. Wang, Y. Mi, A.J. Sayari, Y. Chen, M. Guo, Reversal of cisplatin resistance by inhibiting PI3K/Akt signal pathway in human lung cancer cells. Neoplasma 63, 362–370 (2016). doi:10.​4149/​304_​150806N433 CrossRefPubMed
57.
go back to reference C.F. Hu, Y.Y. Huang, Y.J. Wang, F.G. Gao, Upregulation of ABCG2 via the PI3K-Akt pathway contributes to acidic microenvironment-induced cisplatin resistance in A549 and LTEP-a-2 lung cancer cells. Oncol Rep 36, 455–461 (2016). doi:10.3892/or.2016.4827 CrossRefPubMed C.F. Hu, Y.Y. Huang, Y.J. Wang, F.G. Gao, Upregulation of ABCG2 via the PI3K-Akt pathway contributes to acidic microenvironment-induced cisplatin resistance in A549 and LTEP-a-2 lung cancer cells. Oncol Rep 36, 455–461 (2016). doi:10.​3892/​or.​2016.​4827 CrossRefPubMed
Metadata
Title
HPIP promotes epithelial-mesenchymal transition and cisplatin resistance in ovarian cancer cells through PI3K/AKT pathway activation
Authors
Suresh Bugide
Vijay Kumar Gonugunta
Vasudevarao Penugurti
Vijaya Lakshmi Malisetty
Ratna K. Vadlamudi
Bramanandam Manavathi
Publication date
01-04-2017
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 2/2017
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-016-0308-2

Other articles of this Issue 2/2017

Cellular Oncology 2/2017 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