Skip to main content
Top
Published in: BMC Cancer 1/2016

Open Access 01-12-2016 | Research article

HDAC 1 and 6 modulate cell invasion and migration in clear cell renal cell carcinoma

Authors: Swathi Ramakrishnan, ShengYu Ku, Eric Ciamporcero, Kiersten Marie Miles, Kris Attwood, Sreenivasulu Chintala, Li Shen, Leigh Ellis, Paula Sotomayor, Wendy Swetzig, Ray Huang, Dylan Conroy, Ashley Orillion, Gokul Das, Roberto Pili

Published in: BMC Cancer | Issue 1/2016

Login to get access

Abstract

Background

Class I histone deacetylases (HDACs) have been reported to be overexpressed in clear cell renal cell carcinoma (ccRCC), whereas the expression of class II HDACs is unknown.

Methods

Four isogenic cell lines C2/C2VHL and 786-O/786-OVHL with differential VHL expression are used in our studies. Cobalt chloride is used to mimic hypoxia in vitro. HIF-2α knockdowns in C2 and 786-O cells is used to evaluate the effect on HDAC 1 expression and activity. Invasion and migration assays are used to investigate the role of HDAC 1 and HDAC 6 expression in ccRCC cells. Comparisons are made between experimental groups using the paired T-test, the two-sample Student’s T-test or one-way ANOVA, as appropriate. ccRCC and the TCGA dataset are used to observe the clinical correlation between HDAC 1 and HDAC 6 overexpression and overall and progression free survival.

Results

Our analysis of tumor and matched non-tumor tissues from radical nephrectomies showed overexpression of class I and II HDACs (HDAC6 only in a subset of patients). In vitro, both HDAC1 and HDAC6 over-expression increased cell invasion and motility, respectively, in ccRCC cells. HDAC1 regulated invasiveness by increasing matrix metalloproteinase (MMP) expression. Furthermore, hypoxia stimulation in VHL-reconstituted cell lines increased HIF isoforms and HDAC1 expression. Presence of hypoxia response elements in the HDAC1 promoter along with chromatin immunoprecipitation data suggests that HIF-2α is a transcriptional regulator of HDAC1 gene. Conversely, HDAC6 and estrogen receptor alpha (ERα) were co-localized in cytoplasm of ccRCC cells and HDAC6 enhanced cell motility by decreasing acetylated α-tubulin expression, and this biological effect was attenuated by either biochemical or pharmacological inhibition. Finally, analysis of human ccRCC specimens revealed positive correlation between HIF isoforms and HDAC. HDAC1 mRNA upregulation was associated with worse overall survival in the TCGA dataset.

Conclusions

Taking together, these results suggest that HDAC1 and HDAC6 may play a role in ccRCC biology and could represent rational therapeutic targets.
Appendix
Available only for authorised users
Literature
1.
go back to reference Gossage L, Eisen T, Maher ER. VHL, the story of a tumour suppressor gene. Nat Rev Cancer. 2015;15(1):55–64.CrossRefPubMed Gossage L, Eisen T, Maher ER. VHL, the story of a tumour suppressor gene. Nat Rev Cancer. 2015;15(1):55–64.CrossRefPubMed
2.
go back to reference Ohh M, Park CW, Ivan M, Hoffman MA, Kim TY, Huang LE, Pavletich N, Chau V, Kaelin WG. Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nat Cell Biol. 2000;2(7):423–7.CrossRefPubMed Ohh M, Park CW, Ivan M, Hoffman MA, Kim TY, Huang LE, Pavletich N, Chau V, Kaelin WG. Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nat Cell Biol. 2000;2(7):423–7.CrossRefPubMed
3.
go back to reference Cockman ME, Masson N, Mole DR, Jaakkola P, Chang G-W, Clifford SC, Maher ER, Pugh CW, Ratcliffe PJ, Maxwell PH. Hypoxia Inducible Factor-α Binding and Ubiquitylation by the von Hippel-Lindau Tumor Suppressor Protein. J Biol Chem. 2000;275(33):25733–41.CrossRefPubMed Cockman ME, Masson N, Mole DR, Jaakkola P, Chang G-W, Clifford SC, Maher ER, Pugh CW, Ratcliffe PJ, Maxwell PH. Hypoxia Inducible Factor-α Binding and Ubiquitylation by the von Hippel-Lindau Tumor Suppressor Protein. J Biol Chem. 2000;275(33):25733–41.CrossRefPubMed
4.
go back to reference Koul H, Huh JS, Rove KO, Crompton L, Koul S, Meacham RB, Kim FJ. Molecular aspects of renal cell carcinoma: a review. Am J Cancer Res. 2011;1(2):240–54.PubMed Koul H, Huh JS, Rove KO, Crompton L, Koul S, Meacham RB, Kim FJ. Molecular aspects of renal cell carcinoma: a review. Am J Cancer Res. 2011;1(2):240–54.PubMed
5.
go back to reference Sharma NL, Groselj B, Hamdy FC, Kiltie AE. The emerging role of histone deacetylase (HDAC) inhibitors in urological cancers. BJU Int. 2013;111(4):537–42.CrossRefPubMed Sharma NL, Groselj B, Hamdy FC, Kiltie AE. The emerging role of histone deacetylase (HDAC) inhibitors in urological cancers. BJU Int. 2013;111(4):537–42.CrossRefPubMed
6.
go back to reference Fritzsche F, Weichert W, Roske A, Gekeler V, Beckers T, Stephan C, Jung K, Scholman K, Denkert C, Dietel M, et al. Class I histone deacetylases 1, 2 and 3 are highly expressed in renal cell cancer. BMC Cancer. 2008;8(1):381.CrossRefPubMed Fritzsche F, Weichert W, Roske A, Gekeler V, Beckers T, Stephan C, Jung K, Scholman K, Denkert C, Dietel M, et al. Class I histone deacetylases 1, 2 and 3 are highly expressed in renal cell cancer. BMC Cancer. 2008;8(1):381.CrossRefPubMed
7.
go back to reference Qian DZ, Kachhap SK, Collis SJ, Verheul HMW, Carducci MA, Atadja P, Pili R. Class II Histone Deacetylases Are Associated with VHL-Independent Regulation of Hypoxia-Inducible Factor 1α. Cancer Res. 2006;66(17):8814–21.CrossRefPubMed Qian DZ, Kachhap SK, Collis SJ, Verheul HMW, Carducci MA, Atadja P, Pili R. Class II Histone Deacetylases Are Associated with VHL-Independent Regulation of Hypoxia-Inducible Factor 1α. Cancer Res. 2006;66(17):8814–21.CrossRefPubMed
8.
go back to reference Qian DZ, Wang X, Kachhap SK, Kato Y, Wei Y, Zhang L, Atadja P, Pili R. The histone deacetylase inhibitor NVP-LAQ824 inhibits angiogenesis and has a greater antitumor effect in combination with the vascular endothelial growth factor receptor tyrosine kinase inhibitor PTK787/ZK222584. Cancer Res. 2004;64(18):6626–34.CrossRefPubMed Qian DZ, Wang X, Kachhap SK, Kato Y, Wei Y, Zhang L, Atadja P, Pili R. The histone deacetylase inhibitor NVP-LAQ824 inhibits angiogenesis and has a greater antitumor effect in combination with the vascular endothelial growth factor receptor tyrosine kinase inhibitor PTK787/ZK222584. Cancer Res. 2004;64(18):6626–34.CrossRefPubMed
9.
go back to reference Kim MS, Kwon HJ, Lee YM, Baek JH, Jang J-E, Lee S-W, Moon E-J, Kim H-S, Lee S-K, Chung HY, et al. Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat Med. 2001;7(4):437–43.CrossRefPubMed Kim MS, Kwon HJ, Lee YM, Baek JH, Jang J-E, Lee S-W, Moon E-J, Kim H-S, Lee S-K, Chung HY, et al. Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat Med. 2001;7(4):437–43.CrossRefPubMed
10.
go back to reference Kim SH, Jeong JW, Park JA, Lee JW, Seo JH, Jung BK, Bae MK, Kim KW. Regulation of the HIF-1alpha stability by histone deacetylases. Oncol Rep. 2007;17(3):647–51.PubMed Kim SH, Jeong JW, Park JA, Lee JW, Seo JH, Jung BK, Bae MK, Kim KW. Regulation of the HIF-1alpha stability by histone deacetylases. Oncol Rep. 2007;17(3):647–51.PubMed
11.
go back to reference Verheul HMW, Salumbides B, Van Erp K, Hammers H, Qian DZ, Sanni T, Atadja P, Pili R. Combination Strategy Targeting the Hypoxia Inducible Factor-1α with Mammalian Target of Rapamycin and Histone Deacetylase Inhibitors. Clin Cancer Res. 2008;14(11):3589–97.CrossRefPubMed Verheul HMW, Salumbides B, Van Erp K, Hammers H, Qian DZ, Sanni T, Atadja P, Pili R. Combination Strategy Targeting the Hypoxia Inducible Factor-1α with Mammalian Target of Rapamycin and Histone Deacetylase Inhibitors. Clin Cancer Res. 2008;14(11):3589–97.CrossRefPubMed
12.
go back to reference Tran AD-A, Marmo TP, Salam AA, Che S, Finkelstein E, Kabarriti R, Xenias HS, Mazitschek R, Hubbert C, Kawaguchi Y, et al. HDAC6 deacetylation of tubulin modulates dynamics of cellular adhesions. J Cell Sci. 2007;120(8):1469–79.CrossRefPubMed Tran AD-A, Marmo TP, Salam AA, Che S, Finkelstein E, Kabarriti R, Xenias HS, Mazitschek R, Hubbert C, Kawaguchi Y, et al. HDAC6 deacetylation of tubulin modulates dynamics of cellular adhesions. J Cell Sci. 2007;120(8):1469–79.CrossRefPubMed
13.
go back to reference Azuma K, Urano T, Horie-Inoue K, Hayashi S, Sakai R, Ouchi Y, Inoue S. Association of estrogen receptor alpha and histone deacetylase 6 causes rapid deacetylation of tubulin in breast cancer cells. Cancer Res. 2009;69(7):2935–40.CrossRefPubMed Azuma K, Urano T, Horie-Inoue K, Hayashi S, Sakai R, Ouchi Y, Inoue S. Association of estrogen receptor alpha and histone deacetylase 6 causes rapid deacetylation of tubulin in breast cancer cells. Cancer Res. 2009;69(7):2935–40.CrossRefPubMed
14.
go back to reference Saji S, Kawakami M, Hayashi S-I, Yoshida N, Hirose M, Horiguchi S-I, Itoh A, Funata N, Schreiber SL, Yoshida M, et al. Significance of HDAC6 regulation via estrogen signaling for cell motility and prognosis in estrogen receptor-positive breast cancer. Oncogene. 2005;24(28):4531–9.CrossRefPubMed Saji S, Kawakami M, Hayashi S-I, Yoshida N, Hirose M, Horiguchi S-I, Itoh A, Funata N, Schreiber SL, Yoshida M, et al. Significance of HDAC6 regulation via estrogen signaling for cell motility and prognosis in estrogen receptor-positive breast cancer. Oncogene. 2005;24(28):4531–9.CrossRefPubMed
15.
go back to reference Bhat HK, Hacker HJ, Bannasch P, Thompson EA, Liehr JG. Localization of estrogen receptors in interstitial cells of hamster kidney and in estradiol-induced renal tumors as evidence of the mesenchymal origin of this neoplasm. Cancer Res. 1993;53(22):5447–51.PubMed Bhat HK, Hacker HJ, Bannasch P, Thompson EA, Liehr JG. Localization of estrogen receptors in interstitial cells of hamster kidney and in estradiol-induced renal tumors as evidence of the mesenchymal origin of this neoplasm. Cancer Res. 1993;53(22):5447–51.PubMed
16.
go back to reference Fiskus W, Ren Y, Mohapatra A, Bali P, Mandawat A, Rao R, Herger B, Yang Y, Atadja P, Wu J, et al. Hydroxamic Acid Analogue Histone Deacetylase Inhibitors Attenuate Estrogen Receptor-α Levels and Transcriptional Activity: A Result of Hyperacetylation and Inhibition of Chaperone Function of Heat Shock Protein 90. Clin Cancer Res. 2007;13(16):4882–90.CrossRefPubMed Fiskus W, Ren Y, Mohapatra A, Bali P, Mandawat A, Rao R, Herger B, Yang Y, Atadja P, Wu J, et al. Hydroxamic Acid Analogue Histone Deacetylase Inhibitors Attenuate Estrogen Receptor-α Levels and Transcriptional Activity: A Result of Hyperacetylation and Inhibition of Chaperone Function of Heat Shock Protein 90. Clin Cancer Res. 2007;13(16):4882–90.CrossRefPubMed
17.
go back to reference Hodges-Gallagher L, Valentine CD, Bader SE, Kushner PJ. Inhibition of histone deacetylase enhances the anti-proliferative action of antiestrogens on breast cancer cells and blocks tamoxifen-induced proliferation of uterine cells. Breast Cancer Res Treat. 2007;105(3):297–309.CrossRefPubMed Hodges-Gallagher L, Valentine CD, Bader SE, Kushner PJ. Inhibition of histone deacetylase enhances the anti-proliferative action of antiestrogens on breast cancer cells and blocks tamoxifen-induced proliferation of uterine cells. Breast Cancer Res Treat. 2007;105(3):297–309.CrossRefPubMed
18.
go back to reference Jang ER, Lim S-J, Lee ES, Jeong G, Kim T-Y, Bang Y-J, Lee J-S. The histone deacetylase inhibitor trichostatin A sensitizes estrogen receptor [alpha]-negative breast cancer cells to tamoxifen. Oncogene. 2003;23(9):1724–36.CrossRef Jang ER, Lim S-J, Lee ES, Jeong G, Kim T-Y, Bang Y-J, Lee J-S. The histone deacetylase inhibitor trichostatin A sensitizes estrogen receptor [alpha]-negative breast cancer cells to tamoxifen. Oncogene. 2003;23(9):1724–36.CrossRef
19.
go back to reference Yu C-P, Ho J-Y, Huang Y-T, Cha T-L, Sun G-H, Yu D-S, Chang F-W, Chen S-P, Hsu R-J. Estrogen Inhibits Renal Cell Carcinoma Cell Progression through Estrogen Receptor-β Activation. PLoS One. 2013;8(2):e56667.CrossRefPubMed Yu C-P, Ho J-Y, Huang Y-T, Cha T-L, Sun G-H, Yu D-S, Chang F-W, Chen S-P, Hsu R-J. Estrogen Inhibits Renal Cell Carcinoma Cell Progression through Estrogen Receptor-β Activation. PLoS One. 2013;8(2):e56667.CrossRefPubMed
20.
go back to reference Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al. The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data. Cancer Discov. 2012;2(5):401–4.CrossRefPubMed Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al. The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data. Cancer Discov. 2012;2(5):401–4.CrossRefPubMed
21.
go back to reference Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6(269):l1.CrossRef Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6(269):l1.CrossRef
22.
go back to reference Jeon HW, Lee YM. Inhibition of Histone Deacetylase Attenuates Hypoxia-Induced Migration and Invasion of Cancer Cells via the Restoration of RECK Expression. Mol Cancer Ther. 2010;9(5):1361–70.CrossRefPubMed Jeon HW, Lee YM. Inhibition of Histone Deacetylase Attenuates Hypoxia-Induced Migration and Invasion of Cancer Cells via the Restoration of RECK Expression. Mol Cancer Ther. 2010;9(5):1361–70.CrossRefPubMed
23.
go back to reference Liu L-T, Chang H-C, Chiang L-C, Hung W-C. Histone Deacetylase Inhibitor Up-Regulates RECK to Inhibit MMP-2 Activation and Cancer Cell Invasion. Cancer Res. 2003;63(12):3069–72.PubMed Liu L-T, Chang H-C, Chiang L-C, Hung W-C. Histone Deacetylase Inhibitor Up-Regulates RECK to Inhibit MMP-2 Activation and Cancer Cell Invasion. Cancer Res. 2003;63(12):3069–72.PubMed
24.
go back to reference Lee KJ, Lee KY, Lee YM. Downregulation of a tumor suppressor RECK by hypoxia through recruitment of HDAC1 and HIF-1α to reverse HRE site in the promoter. Biochim Biophys Acta. 2010;1803(5):608–16.CrossRefPubMed Lee KJ, Lee KY, Lee YM. Downregulation of a tumor suppressor RECK by hypoxia through recruitment of HDAC1 and HIF-1α to reverse HRE site in the promoter. Biochim Biophys Acta. 2010;1803(5):608–16.CrossRefPubMed
25.
go back to reference Chintala S, Najrana T, Toth K, Cao S, Durrani FA, Pili R, Rustum YM. Prolyl hydroxylase 2 dependent and Von-Hippel-Lindau independent degradation of Hypoxia-inducible factor 1 and 2 alpha by selenium in clear cell renal cell carcinoma leads to tumor growth inhibition. BMC Cancer. 2012;12:293.CrossRefPubMed Chintala S, Najrana T, Toth K, Cao S, Durrani FA, Pili R, Rustum YM. Prolyl hydroxylase 2 dependent and Von-Hippel-Lindau independent degradation of Hypoxia-inducible factor 1 and 2 alpha by selenium in clear cell renal cell carcinoma leads to tumor growth inhibition. BMC Cancer. 2012;12:293.CrossRefPubMed
26.
go back to reference Baldewijns MM, van Vlodrop IJH, Vermeulen PB, Soetekouw PMMB, van Engeland M, de Bruïne AP. VHL and HIF signalling in renal cell carcinogenesis. J Pathol. 2010;221(2):125–38.CrossRefPubMed Baldewijns MM, van Vlodrop IJH, Vermeulen PB, Soetekouw PMMB, van Engeland M, de Bruïne AP. VHL and HIF signalling in renal cell carcinogenesis. J Pathol. 2010;221(2):125–38.CrossRefPubMed
27.
go back to reference Haase VH. The VHL//HIF oxygen-sensing pathway and its relevance to kidney disease. Kidney Int. 2006;69(8):1302–7.CrossRefPubMed Haase VH. The VHL//HIF oxygen-sensing pathway and its relevance to kidney disease. Kidney Int. 2006;69(8):1302–7.CrossRefPubMed
28.
go back to reference Mahon PC, Hirota K, Semenza GL. FIH-1: a novel protein that interacts with HIF-1alpha and VHL to mediate repression of HIF-1 transcriptional activity. Genes Dev. 2001;15(20):2675–86.CrossRefPubMed Mahon PC, Hirota K, Semenza GL. FIH-1: a novel protein that interacts with HIF-1alpha and VHL to mediate repression of HIF-1 transcriptional activity. Genes Dev. 2001;15(20):2675–86.CrossRefPubMed
29.
go back to reference Gordan JD, Lal P, Dondeti VR, Letrero R, Parekh KN, Oquendo CE, Greenberg RA, Flaherty KT, Rathmell WK, Keith B, et al. HIF-α Effects on c-Myc Distinguish Two Subtypes of Sporadic VHL-Deficient Clear Cell Renal Carcinoma. Cancer Cell. 2008;14(6):435–46.CrossRefPubMed Gordan JD, Lal P, Dondeti VR, Letrero R, Parekh KN, Oquendo CE, Greenberg RA, Flaherty KT, Rathmell WK, Keith B, et al. HIF-α Effects on c-Myc Distinguish Two Subtypes of Sporadic VHL-Deficient Clear Cell Renal Carcinoma. Cancer Cell. 2008;14(6):435–46.CrossRefPubMed
30.
go back to reference Hwang IY, Roe JS, Seol JH, Kim HR, Cho EJ, Youn HD. pVHL-mediated transcriptional repression of c-Myc by recruitment of histone deacetylases. Mol Cells. 2012;33(2):195–201.CrossRefPubMed Hwang IY, Roe JS, Seol JH, Kim HR, Cho EJ, Youn HD. pVHL-mediated transcriptional repression of c-Myc by recruitment of histone deacetylases. Mol Cells. 2012;33(2):195–201.CrossRefPubMed
31.
go back to reference Selvarajah J, Nathawat K, Moumen A, Ashcroft M, Carroll VA. Chemotherapy-mediated p53-dependent DNA damage response in clear cell renal cell carcinoma: role of the mTORC1/2 and hypoxia-inducible factor pathways. Cell Death Dis. 2013;4:e865.CrossRefPubMed Selvarajah J, Nathawat K, Moumen A, Ashcroft M, Carroll VA. Chemotherapy-mediated p53-dependent DNA damage response in clear cell renal cell carcinoma: role of the mTORC1/2 and hypoxia-inducible factor pathways. Cell Death Dis. 2013;4:e865.CrossRefPubMed
32.
go back to reference Kong X, Lin Z, Liang D, Fath D, Sang N, Caro J. Histone deacetylase inhibitors induce VHL and ubiquitin-independent proteasomal degradation of hypoxia-inducible factor 1alpha. Mol Cell Biol. 2006;26(6):2019–28.CrossRefPubMed Kong X, Lin Z, Liang D, Fath D, Sang N, Caro J. Histone deacetylase inhibitors induce VHL and ubiquitin-independent proteasomal degradation of hypoxia-inducible factor 1alpha. Mol Cell Biol. 2006;26(6):2019–28.CrossRefPubMed
33.
go back to reference Wang F, Qi Y, Li X, He W, Fan Q-X, Zong H. HDAC inhibitor trichostatin A suppresses esophageal squamous cell carcinoma metastasis through HADC2 reduced MMP-2/9. Clin Invest Med. 2013;36(2):E87–94.PubMed Wang F, Qi Y, Li X, He W, Fan Q-X, Zong H. HDAC inhibitor trichostatin A suppresses esophageal squamous cell carcinoma metastasis through HADC2 reduced MMP-2/9. Clin Invest Med. 2013;36(2):E87–94.PubMed
34.
go back to reference Lee JH, Mahendran A, Yao Y, Ngo L, Venta-Perez G, Choy ML, Kim N, Ham WS, Breslow R, Marks PA. Development of a histone deacetylase 6 inhibitor and its biological effects. Proc Natl Acad Sci U S A. 2013;110(39):15704–9.CrossRefPubMed Lee JH, Mahendran A, Yao Y, Ngo L, Venta-Perez G, Choy ML, Kim N, Ham WS, Breslow R, Marks PA. Development of a histone deacetylase 6 inhibitor and its biological effects. Proc Natl Acad Sci U S A. 2013;110(39):15704–9.CrossRefPubMed
35.
go back to reference Lee Y-S, Lim K-H, Guo X, Kawaguchi Y, Gao Y, Barrientos T, Ordentlich P, Wang X-F, Counter CM, Yao T-P. The Cytoplasmic Deacetylase HDAC6 Is Required for Efficient Oncogenic Tumorigenesis. Cancer Res. 2008;68(18):7561–9.CrossRefPubMed Lee Y-S, Lim K-H, Guo X, Kawaguchi Y, Gao Y, Barrientos T, Ordentlich P, Wang X-F, Counter CM, Yao T-P. The Cytoplasmic Deacetylase HDAC6 Is Required for Efficient Oncogenic Tumorigenesis. Cancer Res. 2008;68(18):7561–9.CrossRefPubMed
36.
go back to reference Liu W, Fan LX, Zhou X, Sweeney Jr WE, Avner ED, Li X. HDAC6 Regulates Epidermal Growth Factor Receptor (EGFR) Endocytic Trafficking and Degradation in Renal Epithelial Cells. PLoS One. 2012;7(11):e49418.CrossRefPubMed Liu W, Fan LX, Zhou X, Sweeney Jr WE, Avner ED, Li X. HDAC6 Regulates Epidermal Growth Factor Receptor (EGFR) Endocytic Trafficking and Degradation in Renal Epithelial Cells. PLoS One. 2012;7(11):e49418.CrossRefPubMed
37.
go back to reference Sakuma T, Uzawa K, Onda T, Shiiba M, Yokoe H, Shibahara T, Tanzawa H. Aberrant expression of histone deacetylase 6 in oral squamous cell carcinoma. Int J Oncol. 2006;29(1):117–24.PubMed Sakuma T, Uzawa K, Onda T, Shiiba M, Yokoe H, Shibahara T, Tanzawa H. Aberrant expression of histone deacetylase 6 in oral squamous cell carcinoma. Int J Oncol. 2006;29(1):117–24.PubMed
38.
go back to reference Shen L, Ciesielski M, Ramakrishnan S, Miles KM, Ellis L, Sotomayor P, Shrikant P, Fenstermaker R, Pili R. Class I Histone Deacetylase Inhibitor Entinostat Suppresses Regulatory T Cells and Enhances Immunotherapies in Renal and Prostate Cancer Models. PLoS One. 2012;7(1):e30815.CrossRefPubMed Shen L, Ciesielski M, Ramakrishnan S, Miles KM, Ellis L, Sotomayor P, Shrikant P, Fenstermaker R, Pili R. Class I Histone Deacetylase Inhibitor Entinostat Suppresses Regulatory T Cells and Enhances Immunotherapies in Renal and Prostate Cancer Models. PLoS One. 2012;7(1):e30815.CrossRefPubMed
39.
go back to reference Kim MJ, Kim DE, Jeong IG, Choi J, Jang S, Lee JH, Ro S, Hwang JJ, Kim CS. HDAC inhibitors synergize antiproliferative effect of sorafenib in renal cell carcinoma cells. Anticancer Res. 2012;32(8):3161–8.PubMed Kim MJ, Kim DE, Jeong IG, Choi J, Jang S, Lee JH, Ro S, Hwang JJ, Kim CS. HDAC inhibitors synergize antiproliferative effect of sorafenib in renal cell carcinoma cells. Anticancer Res. 2012;32(8):3161–8.PubMed
Metadata
Title
HDAC 1 and 6 modulate cell invasion and migration in clear cell renal cell carcinoma
Authors
Swathi Ramakrishnan
ShengYu Ku
Eric Ciamporcero
Kiersten Marie Miles
Kris Attwood
Sreenivasulu Chintala
Li Shen
Leigh Ellis
Paula Sotomayor
Wendy Swetzig
Ray Huang
Dylan Conroy
Ashley Orillion
Gokul Das
Roberto Pili
Publication date
01-12-2016
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2016
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-016-2604-7

Other articles of this Issue 1/2016

BMC Cancer 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