Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2016

Open Access 01-12-2016 | Research

CCDC88A, a prognostic factor for human pancreatic cancers, promotes the motility and invasiveness of pancreatic cancer cells

Authors: Aki Tanouchi, Keisuke Taniuchi, Mutsuo Furihata, Seiji Naganuma, Ken Dabanaka, Masashi Kimura, Ryohei Watanabe, Takuhiro Kohsaki, Takahiro Shimizu, Motoaki Saito, Kazuhiro Hanazaki, Toshiji Saibara

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2016

Login to get access

Abstract

Background

Coiled-Coil Domain Containing 88A (CCDC88A) was identified as a substrate of the serine/threonine kinase Akt that is capable of binding to the actin cytoskeleton. The aim of this study was to investigate the potential role of CCDC88A in the migration and invasiveness of pancreatic ductal adenocarcinoma (PDAC) cells.

Methods

Immunohistochemistry was performed to determine whether high CCDC88A expression in human PDAC tissues is correlated with poor prognosis. Immunoprecipitation, immunoblotting and immunocytochemistry were performed to determine the intracellular distribution of CCDC88A, and its association with the serine/threonine kinase Akt and actin-filaments in PDAC cells. Phosphoprotein array analysis was performed to determine CCDC88A-associated intracellular signaling pathways. Finally, immunofluorescence analyses and Matrigel invasion assays were performed to examine the effects of CCDC88A on the formation of cell protrusions and PDAC cell invasion.

Results

Expression of CCDC88A in PDAC tissue was significantly correlated with overall survival. CCDC88A was co-localized with peripheral actin structures in cell protrusions of migrating PDAC cells. Knockdown of CCDC88A inhibited the migration and invasiveness of PDAC cells through a decrease in cell protrusions. Although CCDC88A has been previously reported to be a binding partner and substrate of Akt, the level of active Akt was not associated with the translocation of CCDC88A towards cell protrusions. CCDC88A-dependent promotion of cell migration and invasiveness was not modulated by Akt signaling. Knockdown of CCDC88A decreased phosphorylated Src and ERK1/2 and increased phosphorylated AMPK1 in PDAC cells. Knockdown of AMPK1 inhibited the migration and invasiveness of PDAC cells. The combined data suggest that CCDC88A may be a useful marker for predicting the outcome of patients with PDAC and that CCDC88A can promote PDAC cell migration and invasion through a signaling pathway that involves phosphorylation of Src and ERK1/2 and/or dephosphorylation of AMPK1.

Conclusions

CCDC88A was accumulated in cell protrusions, contributed to the formation of membrane protrusions, and increased the migration and invasiveness of PDAC cells.
Appendix
Available only for authorised users
Literature
1.
go back to reference Enomoto A, Murakami H, Asai N, et al. Akt/PKB regulates actin organization and cell motility via Girdin/APE. Dev Cell. 2005;9(3):389–402.CrossRefPubMed Enomoto A, Murakami H, Asai N, et al. Akt/PKB regulates actin organization and cell motility via Girdin/APE. Dev Cell. 2005;9(3):389–402.CrossRefPubMed
2.
go back to reference Weng L, Enomoto A, Ishida-Takagishi M, et al. Girding for migratory cues: roles of the Akt substrate Girdin in cancer progression and angiogenesis. Cancer Sci. 2010;101(4):836–42.CrossRefPubMed Weng L, Enomoto A, Ishida-Takagishi M, et al. Girding for migratory cues: roles of the Akt substrate Girdin in cancer progression and angiogenesis. Cancer Sci. 2010;101(4):836–42.CrossRefPubMed
3.
go back to reference Kim D, Kim S, Koh H, et al. Akt/PKB promotes cancer cell invasion via increased motility and metalloproteinase production. FASEB J. 2001;15(11):1953–62.CrossRefPubMed Kim D, Kim S, Koh H, et al. Akt/PKB promotes cancer cell invasion via increased motility and metalloproteinase production. FASEB J. 2001;15(11):1953–62.CrossRefPubMed
4.
go back to reference Enomoto A, Ping J, Takahashi M. Girdin, a novel actin-binding protein, and its family of proteins possess versatile functions in the Akt and Wnt signaling pathways. Ann N Y Acad Sci. 2006;1086:169–84.CrossRefPubMed Enomoto A, Ping J, Takahashi M. Girdin, a novel actin-binding protein, and its family of proteins possess versatile functions in the Akt and Wnt signaling pathways. Ann N Y Acad Sci. 2006;1086:169–84.CrossRefPubMed
5.
go back to reference Ghosh P, Garcia-Marcos M, Bornheimer SJ, et al. Activation of Galphai3 triggers cell migration via regulation of GIV. J Cell Biol. 2008;182(2):381–93.CrossRefPubMedPubMedCentral Ghosh P, Garcia-Marcos M, Bornheimer SJ, et al. Activation of Galphai3 triggers cell migration via regulation of GIV. J Cell Biol. 2008;182(2):381–93.CrossRefPubMedPubMedCentral
6.
go back to reference Kitamura T, Asai N, Enomoto A, et al. Regulation of VEGF-mediated angiogenesis by the Akt/PKB substrate Girdin. Nat Cell Biol. 2008;103(3):329–37.CrossRef Kitamura T, Asai N, Enomoto A, et al. Regulation of VEGF-mediated angiogenesis by the Akt/PKB substrate Girdin. Nat Cell Biol. 2008;103(3):329–37.CrossRef
7.
go back to reference Jiang P, Enomoto A, Jijiwa M, et al. An actin-binding protein Girdin regulates the motility of breast cancer cells. Cancer Res. 2008;68(5):1310–8.CrossRefPubMed Jiang P, Enomoto A, Jijiwa M, et al. An actin-binding protein Girdin regulates the motility of breast cancer cells. Cancer Res. 2008;68(5):1310–8.CrossRefPubMed
8.
go back to reference Pierce KL, Premont RT, Lefkowitz RJ. Seven-transmembrane receptors. Nat Rev Mol Cell Biol. 2002;3(9):639–50.CrossRefPubMed Pierce KL, Premont RT, Lefkowitz RJ. Seven-transmembrane receptors. Nat Rev Mol Cell Biol. 2002;3(9):639–50.CrossRefPubMed
9.
go back to reference Garcia-Marcos M, Ghosh P, Farquhar MG. GIV is a nonreceptor GEF for G alpha i with a unique motif that regulates Akt signaling. Proc Natl Acad Sci U S A. 2009;106(9):3178–83.CrossRefPubMedPubMedCentral Garcia-Marcos M, Ghosh P, Farquhar MG. GIV is a nonreceptor GEF for G alpha i with a unique motif that regulates Akt signaling. Proc Natl Acad Sci U S A. 2009;106(9):3178–83.CrossRefPubMedPubMedCentral
10.
go back to reference Baumgart M, Heinmöller E, Horstmann O, et al. The genetic basis of sporadic pancreatic cancer. Cell Oncol. 2005;27(1):3–13.PubMedPubMedCentral Baumgart M, Heinmöller E, Horstmann O, et al. The genetic basis of sporadic pancreatic cancer. Cell Oncol. 2005;27(1):3–13.PubMedPubMedCentral
11.
go back to reference Taniuchi K, Furihata M, Hanazaki K, et al. IGF2BP3-mediated translation in cell protrusions promotes cell invasiveness and metastasis of pancreatic cancer. Oncotarget. 2014;5(16):6832–45.CrossRefPubMedPubMedCentral Taniuchi K, Furihata M, Hanazaki K, et al. IGF2BP3-mediated translation in cell protrusions promotes cell invasiveness and metastasis of pancreatic cancer. Oncotarget. 2014;5(16):6832–45.CrossRefPubMedPubMedCentral
12.
go back to reference Taniuchi K, Furihata M, Saibara T. KIF20A-mediated RNA granule transport system promotes the invasiveness of pancreatic cancer cells. Neoplasia. 2014;16(12):1082–93.CrossRefPubMedPubMedCentral Taniuchi K, Furihata M, Saibara T. KIF20A-mediated RNA granule transport system promotes the invasiveness of pancreatic cancer cells. Neoplasia. 2014;16(12):1082–93.CrossRefPubMedPubMedCentral
13.
go back to reference Miyazawa Y, Uekita T, Hiraoka N, et al. CUB domain-containing protein 1, a prognostic factor for human pancreatic cancers, promotes cell migration and extracellular matrix degradation. Cancer Res. 2010;70(12):5136–46.CrossRefPubMed Miyazawa Y, Uekita T, Hiraoka N, et al. CUB domain-containing protein 1, a prognostic factor for human pancreatic cancers, promotes cell migration and extracellular matrix degradation. Cancer Res. 2010;70(12):5136–46.CrossRefPubMed
14.
go back to reference Koch HJ, Hau P. ROC analysis as an additional method to characterize time to event data. Pathol Oncol Res. 2005;11(1):50–2.CrossRefPubMed Koch HJ, Hau P. ROC analysis as an additional method to characterize time to event data. Pathol Oncol Res. 2005;11(1):50–2.CrossRefPubMed
15.
go back to reference Taniuchi K, Nishimori I, Hollingsworth MA. Intracellular CD24 inhibits cell invasion by posttranscriptional regulation of BART through interaction with G3BP. Cancer Res. 2011;71(3):895–905.CrossRefPubMed Taniuchi K, Nishimori I, Hollingsworth MA. Intracellular CD24 inhibits cell invasion by posttranscriptional regulation of BART through interaction with G3BP. Cancer Res. 2011;71(3):895–905.CrossRefPubMed
16.
go back to reference Taniuchi K, Yokotani K, Saibara T. BART inhibits pancreatic cancer cell invasion by Rac1 inactivation through direct binding to active Rac1. Neoplasia. 2012;14(5):440–50.CrossRefPubMedPubMedCentral Taniuchi K, Yokotani K, Saibara T. BART inhibits pancreatic cancer cell invasion by Rac1 inactivation through direct binding to active Rac1. Neoplasia. 2012;14(5):440–50.CrossRefPubMedPubMedCentral
17.
go back to reference Tsuboi M, Taniuchi K, Furihata M, et al. Vav3 is linked to poor prognosis of pancreatic cancers and promotes the motility and invasiveness of pancreatic cancer cells. Pancreatology. 2016;16(5):905–16.CrossRefPubMed Tsuboi M, Taniuchi K, Furihata M, et al. Vav3 is linked to poor prognosis of pancreatic cancers and promotes the motility and invasiveness of pancreatic cancer cells. Pancreatology. 2016;16(5):905–16.CrossRefPubMed
18.
go back to reference Gallo G, Letourneau PC. Regulation of growth cone actin filaments by guidance cues. J Neurobiol. 2004;58(1):92–102.CrossRefPubMed Gallo G, Letourneau PC. Regulation of growth cone actin filaments by guidance cues. J Neurobiol. 2004;58(1):92–102.CrossRefPubMed
19.
go back to reference Roberts Jr CT. Control of insulin-like growth factor (IGF) action by regulation of IGF-I receptor expression. Endocr J. 1996;43(Suppl):S49–55.CrossRefPubMed Roberts Jr CT. Control of insulin-like growth factor (IGF) action by regulation of IGF-I receptor expression. Endocr J. 1996;43(Suppl):S49–55.CrossRefPubMed
20.
go back to reference Goto M, Iwase A, Harata T, et al. IGF1-induced AKT phosphorylation and cell proliferation are suppressed with the increase in PTEN during luteinization in human granulosa cells. Reproduction. 2009;137(5):835–42.CrossRefPubMed Goto M, Iwase A, Harata T, et al. IGF1-induced AKT phosphorylation and cell proliferation are suppressed with the increase in PTEN during luteinization in human granulosa cells. Reproduction. 2009;137(5):835–42.CrossRefPubMed
21.
go back to reference Ghosh P, Beas AO, Bornheimer SJ, et al. A G{alpha}i-GIV molecular complex binds epidermal growth factor receptor and determines whether cells migrate or proliferate. Mol Biol Cell. 2010;21(13):2338–54.CrossRefPubMedPubMedCentral Ghosh P, Beas AO, Bornheimer SJ, et al. A G{alpha}i-GIV molecular complex binds epidermal growth factor receptor and determines whether cells migrate or proliferate. Mol Biol Cell. 2010;21(13):2338–54.CrossRefPubMedPubMedCentral
22.
go back to reference Bamburg JR, McGough A, Ono S. Putting a new twist on actin: ADF/cofilins modulate actin dynamics. Trends Cell Biol. 1999;9(9):364–70.CrossRefPubMed Bamburg JR, McGough A, Ono S. Putting a new twist on actin: ADF/cofilins modulate actin dynamics. Trends Cell Biol. 1999;9(9):364–70.CrossRefPubMed
23.
go back to reference Eiseler T, Döppler H, Yan IK, et al. Protein kinase D1 regulates cofilin-mediated F-actin reorganization and cell motility through slingshot. Nat Cell Biol. 2009;11(5):545–56.CrossRefPubMedPubMedCentral Eiseler T, Döppler H, Yan IK, et al. Protein kinase D1 regulates cofilin-mediated F-actin reorganization and cell motility through slingshot. Nat Cell Biol. 2009;11(5):545–56.CrossRefPubMedPubMedCentral
24.
go back to reference Farkas A, Szatmari E, Orbok A, et al. Hyperosmotic mannitol induces Src kinase-dependent phosphorylation of beta-catenin in cerebral endothelial cells. J Neurosci Res. 2005;80(6):855–61.CrossRefPubMed Farkas A, Szatmari E, Orbok A, et al. Hyperosmotic mannitol induces Src kinase-dependent phosphorylation of beta-catenin in cerebral endothelial cells. J Neurosci Res. 2005;80(6):855–61.CrossRefPubMed
25.
go back to reference Bowman T, Broome MA, Sinibaldi D, et al. Stat3-mediated Myc expression is required for Src transformation and PDGF-induced mitogenesis. Proc Natl Acad Sci U S A. 2001;98(13):7319–24.CrossRefPubMedPubMedCentral Bowman T, Broome MA, Sinibaldi D, et al. Stat3-mediated Myc expression is required for Src transformation and PDGF-induced mitogenesis. Proc Natl Acad Sci U S A. 2001;98(13):7319–24.CrossRefPubMedPubMedCentral
26.
29.
go back to reference Park HU, Suy S, Danner M, et al. AMP-activated protein kinase promotes human prostate cancer cell growth and survival. Mol Cancer Ther. 2009;8(4):733–41.CrossRefPubMedPubMedCentral Park HU, Suy S, Danner M, et al. AMP-activated protein kinase promotes human prostate cancer cell growth and survival. Mol Cancer Ther. 2009;8(4):733–41.CrossRefPubMedPubMedCentral
30.
go back to reference Japan Pancreatic Society. Classification of pancreatic carcinoma. 2nd English ed., Kanehara & Co Tokyo; 2003. Japan Pancreatic Society. Classification of pancreatic carcinoma. 2nd English ed., Kanehara & Co Tokyo; 2003.
31.
go back to reference Sobin LH, Gospodarowicz MK, Witteknd C, editors. TNM classification of malignant tumors. 7th ed. New York: Wiley-Blackwell; 2009. p. 132–5. Sobin LH, Gospodarowicz MK, Witteknd C, editors. TNM classification of malignant tumors. 7th ed. New York: Wiley-Blackwell; 2009. p. 132–5.
32.
go back to reference Iwamura T, Katsuki T, Ide K. Establishment and characterization of a human pancreatic cancer cell line (SUIT-2) producing carcinoembryonic antigen and carbohydrate antigen 19–9. Jpn J Cancer Res. 1987;78(1):54–62.PubMed Iwamura T, Katsuki T, Ide K. Establishment and characterization of a human pancreatic cancer cell line (SUIT-2) producing carcinoembryonic antigen and carbohydrate antigen 19–9. Jpn J Cancer Res. 1987;78(1):54–62.PubMed
Metadata
Title
CCDC88A, a prognostic factor for human pancreatic cancers, promotes the motility and invasiveness of pancreatic cancer cells
Authors
Aki Tanouchi
Keisuke Taniuchi
Mutsuo Furihata
Seiji Naganuma
Ken Dabanaka
Masashi Kimura
Ryohei Watanabe
Takuhiro Kohsaki
Takahiro Shimizu
Motoaki Saito
Kazuhiro Hanazaki
Toshiji Saibara
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2016
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-016-0466-0

Other articles of this Issue 1/2016

Journal of Experimental & Clinical Cancer Research 1/2016 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