Skip to main content
Top
Published in: Breast Cancer Research 3/2014

Open Access 01-06-2014 | Research article

JMJD2A contributes to breast cancer progression through transcriptional repression of the tumor suppressor ARHI

Authors: Li-Liang Li, Ai-Min Xue, Bei-Xu Li, Yi-Wen Shen, Yu-Hua Li, Cheng-Liang Luo, Ming-Chang Zhang, Jie-Qing Jiang, Zu-De Xu, Jian-Hui Xie, Zi-Qin Zhao

Published in: Breast Cancer Research | Issue 3/2014

Login to get access

Abstract

Introduction

Breast cancer is a worldwide health problem and the leading cause of cancer death among females. We previously identified Jumonji domain containing 2A (JMJD2A) as a critical mediator of breast cancer proliferation, migration and invasion. We now report that JMJD2A could promote breast cancer progression through transcriptional repression of the tumor suppressor aplasia Ras homolog member I (ARHI).

Methods

Immunohistochemistry was performed to examine protein expressions in 155 cases of breast cancer and 30 non-neoplastic tissues. Spearman correlation analysis was used to analyze the correlation between JMJD2A expression and clinical parameters as well as several tumor regulators in 155 cases of breast cancer. Gene and protein expressions were monitored by quantitative polymerase chain reaction (qPCR) and Western blot. Results from knockdown of JMJD2A, overexpression of JMJD2A, Co-immunoprecipitation (Co-IP) assay, dual luciferase reporter gene assay and chromatin immunoprecipitation (ChIP) elucidated molecular mechanisms of JMJD2A action in breast cancer progression. Furthermore, the effects of ARHI overexpression on JMJD2A-mediated tumor progression were investigated in vitro and in vivo. For in vitro experiments, cell proliferation, wound-healing, migration and invasion were monitored by cell counting, scratch and Boyden Chamber assays. For in vivo experiments, control cells and cells stably expressing JMJD2A alone or together with ARHI were inoculated into mammary fat pads of mice. Tumor volume, tumor weight and metastatic nodules were measured by caliper, electronic balance and nodule counting, respectively.

Results

JMJD2A was highly expressed in human breast cancers and positively correlated with tumor progression. Knockdown of JMJD2A increased ARHI expression whereas overexpression of JMJD2A decreased ARHI expression at both protein and mRNA levels. Furthermore, E2Fs and histone deacetylases were involved in the transcriptional repression of ARHI expression by JMJD2A. And the aggressive behavior of JMJD2A in breast cancers could be reversed by re-expression of ARHI in vitro and in vivo.

Conclusion

We demonstrated a cancer-promoting effect of JMJD2A and defined a novel molecular pathway contributing to JMJD2A-mediated breast cancer progression.
Appendix
Available only for authorised users
Literature
1.
go back to reference Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D: Global cancer statistics. CA Cancer J Clin. 2011, 61: 69-90. 10.3322/caac.20107.CrossRefPubMed Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D: Global cancer statistics. CA Cancer J Clin. 2011, 61: 69-90. 10.3322/caac.20107.CrossRefPubMed
2.
go back to reference Meads C, Ahmed I, Riley RD: A systematic review of breast cancer incidence risk prediction models with meta-analysis of their performance. Breast Cancer Res Treat. 2012, 132: 365-377. 10.1007/s10549-011-1818-2.CrossRefPubMed Meads C, Ahmed I, Riley RD: A systematic review of breast cancer incidence risk prediction models with meta-analysis of their performance. Breast Cancer Res Treat. 2012, 132: 365-377. 10.1007/s10549-011-1818-2.CrossRefPubMed
3.
go back to reference Palacios J, Robles-Frias MJ, Castilla MA, Lopez-Garcia MA, Benitez J: The molecular pathology of hereditary breast cancer. Pathobiology. 2008, 75: 85-94. 10.1159/000123846.CrossRefPubMed Palacios J, Robles-Frias MJ, Castilla MA, Lopez-Garcia MA, Benitez J: The molecular pathology of hereditary breast cancer. Pathobiology. 2008, 75: 85-94. 10.1159/000123846.CrossRefPubMed
4.
go back to reference Lee EY, Muller WJ: Oncogenes and tumor suppressor genes. Cold Spring Harb Perspect Biol. 2010, 2: a3236-10.1101/cshperspect.a003236.CrossRef Lee EY, Muller WJ: Oncogenes and tumor suppressor genes. Cold Spring Harb Perspect Biol. 2010, 2: a3236-10.1101/cshperspect.a003236.CrossRef
5.
go back to reference Banerji S, Cibulskis K, Rangel-Escareno C, Brown KK, Carter SL, Frederick AM, Lawrence MS, Sivachenko AY, Sougnez C, Zou L, Cortes ML, Fernandez-Lopez JC, Peng S, Ardlie KG, Auclair D, Bautista-Piña V, Duke F, Francis J, Jung J, Maffuz-Aziz A, Onofrio RC, Parkin M, Pho NH, Quintanar-Jurado V, Ramos AH, Rebollar-Vega R, Rodriguez-Cuevas S, Romero-Cordoba SL, Schumacher SE, Stransky N, et al: Sequence analysis of mutations and translocations across breast cancer subtypes. Nature. 2012, 486: 405-409. 10.1038/nature11154.CrossRefPubMedPubMedCentral Banerji S, Cibulskis K, Rangel-Escareno C, Brown KK, Carter SL, Frederick AM, Lawrence MS, Sivachenko AY, Sougnez C, Zou L, Cortes ML, Fernandez-Lopez JC, Peng S, Ardlie KG, Auclair D, Bautista-Piña V, Duke F, Francis J, Jung J, Maffuz-Aziz A, Onofrio RC, Parkin M, Pho NH, Quintanar-Jurado V, Ramos AH, Rebollar-Vega R, Rodriguez-Cuevas S, Romero-Cordoba SL, Schumacher SE, Stransky N, et al: Sequence analysis of mutations and translocations across breast cancer subtypes. Nature. 2012, 486: 405-409. 10.1038/nature11154.CrossRefPubMedPubMedCentral
6.
go back to reference Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, Nik-Zainal S, Martin S, Varela I, Bignell GR, Yates LR, Papaemmanuil E, Beare D, Butler A, Cheverton A, Gamble J, Hinton J, Jia M, Jayakumar A, Jones D, Latimer C, Lau KW, McLaren S, McBride DJ, Menzies A, Mudie L, Raine K, Rad R, Chapman MS, Teague J, et al: The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012, 486: 400-404.PubMedPubMedCentral Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, Nik-Zainal S, Martin S, Varela I, Bignell GR, Yates LR, Papaemmanuil E, Beare D, Butler A, Cheverton A, Gamble J, Hinton J, Jia M, Jayakumar A, Jones D, Latimer C, Lau KW, McLaren S, McBride DJ, Menzies A, Mudie L, Raine K, Rad R, Chapman MS, Teague J, et al: The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012, 486: 400-404.PubMedPubMedCentral
7.
go back to reference Klose RJ, Zhang Y: Regulation of histone methylation by demethylimination and demethylation. Nat Rev Mol Cell Biol. 2007, 8: 307-318. 10.1038/nrm2143.CrossRefPubMed Klose RJ, Zhang Y: Regulation of histone methylation by demethylimination and demethylation. Nat Rev Mol Cell Biol. 2007, 8: 307-318. 10.1038/nrm2143.CrossRefPubMed
8.
go back to reference Mosammaparast N, Shi Y: Reversal of histone methylation: biochemical and molecular mechanisms of histone demethylases. Annu Rev Biochem. 2010, 79: 155-179. 10.1146/annurev.biochem.78.070907.103946.CrossRefPubMed Mosammaparast N, Shi Y: Reversal of histone methylation: biochemical and molecular mechanisms of histone demethylases. Annu Rev Biochem. 2010, 79: 155-179. 10.1146/annurev.biochem.78.070907.103946.CrossRefPubMed
9.
go back to reference Yamane K, Toumazou C, Tsukada Y, Erdjument-Bromage H, Tempst P, Wong J, Zhang Y: JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor. Cell. 2006, 125: 483-495. 10.1016/j.cell.2006.03.027.CrossRefPubMed Yamane K, Toumazou C, Tsukada Y, Erdjument-Bromage H, Tempst P, Wong J, Zhang Y: JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor. Cell. 2006, 125: 483-495. 10.1016/j.cell.2006.03.027.CrossRefPubMed
10.
go back to reference Peterson CL, Laniel MA: Histones and histone modifications. Curr Biol. 2004, 14: R546-R551. 10.1016/j.cub.2004.07.007.CrossRefPubMed Peterson CL, Laniel MA: Histones and histone modifications. Curr Biol. 2004, 14: R546-R551. 10.1016/j.cub.2004.07.007.CrossRefPubMed
11.
go back to reference Klose RJ, Yamane K, Bae Y, Zhang D, Erdjument-Bromage H, Tempst P, Wong J, Zhang Y: The transcriptional repressor JHDM3A demethylates trimethyl histone H3 lysine 9 and lysine 36. Nature. 2006, 442: 312-316. 10.1038/nature04853.CrossRefPubMed Klose RJ, Yamane K, Bae Y, Zhang D, Erdjument-Bromage H, Tempst P, Wong J, Zhang Y: The transcriptional repressor JHDM3A demethylates trimethyl histone H3 lysine 9 and lysine 36. Nature. 2006, 442: 312-316. 10.1038/nature04853.CrossRefPubMed
12.
go back to reference Huang Y, Fang J, Bedford MT, Zhang Y, Xu RM: Recognition of histone H3 lysine-4 methylation by the double tudor domain of JMJD2A. Science. 2006, 312: 748-751. 10.1126/science.1125162.CrossRefPubMed Huang Y, Fang J, Bedford MT, Zhang Y, Xu RM: Recognition of histone H3 lysine-4 methylation by the double tudor domain of JMJD2A. Science. 2006, 312: 748-751. 10.1126/science.1125162.CrossRefPubMed
13.
go back to reference Lee J, Thompson JR, Botuyan MV, Mer G: Distinct binding modes specify the recognition of methylated histones H3K4 and H4K20 by JMJD2A-tudor. Nat Struct Mol Biol. 2008, 15: 109-111. 10.1038/nsmb1326.CrossRefPubMed Lee J, Thompson JR, Botuyan MV, Mer G: Distinct binding modes specify the recognition of methylated histones H3K4 and H4K20 by JMJD2A-tudor. Nat Struct Mol Biol. 2008, 15: 109-111. 10.1038/nsmb1326.CrossRefPubMed
14.
go back to reference Shin S, Janknecht R: Diversity within the JMJD2 histone demethylase family. Biochem Biophys Res Commun. 2007, 353: 973-977. 10.1016/j.bbrc.2006.12.147.CrossRefPubMed Shin S, Janknecht R: Diversity within the JMJD2 histone demethylase family. Biochem Biophys Res Commun. 2007, 353: 973-977. 10.1016/j.bbrc.2006.12.147.CrossRefPubMed
15.
go back to reference Kogure M, Takawa M, Cho HS, Toyokawa G, Hayashi K, Tsunoda T, Kobayashi T, Daigo Y, Sugiyama M, Atomi Y, Nakamura Y, Hamamoto R: Deregulation of the histone demethylase JMJD2A is involved in human carcinogenesis through regulation of the G1/S transition. Cancer Lett. 2013, 336: 76-84. 10.1016/j.canlet.2013.04.009.CrossRefPubMed Kogure M, Takawa M, Cho HS, Toyokawa G, Hayashi K, Tsunoda T, Kobayashi T, Daigo Y, Sugiyama M, Atomi Y, Nakamura Y, Hamamoto R: Deregulation of the histone demethylase JMJD2A is involved in human carcinogenesis through regulation of the G1/S transition. Cancer Lett. 2013, 336: 76-84. 10.1016/j.canlet.2013.04.009.CrossRefPubMed
16.
go back to reference Zhang D, Yoon HG, Wong J: JMJD2A is a novel N-CoR-interacting protein and is involved in repression of the human transcription factor achaete scute-like homologue 2 (ASCL2/Hash2). Mol Cell Biol. 2005, 25: 6404-6414. 10.1128/MCB.25.15.6404-6414.2005.CrossRefPubMedPubMedCentral Zhang D, Yoon HG, Wong J: JMJD2A is a novel N-CoR-interacting protein and is involved in repression of the human transcription factor achaete scute-like homologue 2 (ASCL2/Hash2). Mol Cell Biol. 2005, 25: 6404-6414. 10.1128/MCB.25.15.6404-6414.2005.CrossRefPubMedPubMedCentral
17.
go back to reference Mallette FA, Richard S: JMJD2A promotes cellular transformation by blocking cellular senescence through transcriptional repression of the tumor suppressor CHD5. Cell Rep. 2012, 2: 1233-1243. 10.1016/j.celrep.2012.09.033.CrossRefPubMed Mallette FA, Richard S: JMJD2A promotes cellular transformation by blocking cellular senescence through transcriptional repression of the tumor suppressor CHD5. Cell Rep. 2012, 2: 1233-1243. 10.1016/j.celrep.2012.09.033.CrossRefPubMed
18.
go back to reference Berry WL, Shin S, Lightfoot SA, Janknecht R: Oncogenic features of the JMJD2A histone demethylase in breast cancer. Int J Oncol. 2012, 41: 1701-1706.PubMed Berry WL, Shin S, Lightfoot SA, Janknecht R: Oncogenic features of the JMJD2A histone demethylase in breast cancer. Int J Oncol. 2012, 41: 1701-1706.PubMed
19.
go back to reference Kim TD, Shin S, Berry WL, Oh S, Janknecht R: The JMJD2A demethylase regulates apoptosis and proliferation in colon cancer cells. J Cell Biochem. 2012, 113: 1368-1376. 10.1002/jcb.24009.CrossRefPubMed Kim TD, Shin S, Berry WL, Oh S, Janknecht R: The JMJD2A demethylase regulates apoptosis and proliferation in colon cancer cells. J Cell Biochem. 2012, 113: 1368-1376. 10.1002/jcb.24009.CrossRefPubMed
20.
go back to reference Li BX, Li J, Luo CL, Zhang MC, Li H, Li LL, Xu HF, Shen YW, Xue AM, Zhao ZQ: Expression of JMJD2A in infiltrating duct carcinoma was markedly higher than fibroadenoma, and associated with expression of ARHI, p53 and ER in infiltrating duct carcinoma. Indian J Exp Biol. 2013, 51: 208-217.PubMed Li BX, Li J, Luo CL, Zhang MC, Li H, Li LL, Xu HF, Shen YW, Xue AM, Zhao ZQ: Expression of JMJD2A in infiltrating duct carcinoma was markedly higher than fibroadenoma, and associated with expression of ARHI, p53 and ER in infiltrating duct carcinoma. Indian J Exp Biol. 2013, 51: 208-217.PubMed
21.
go back to reference Gray SG, Iglesias AH, Lizcano F, Villanueva R, Camelo S, Jingu H, Teh BT, Koibuchi N, Chin WW, Kokkotou E, Dangond F: Functional characterization of JMJD2A, a histone deacetylase- and retinoblastoma-binding protein. J Biol Chem. 2005, 280: 28507-28518. 10.1074/jbc.M413687200.CrossRefPubMed Gray SG, Iglesias AH, Lizcano F, Villanueva R, Camelo S, Jingu H, Teh BT, Koibuchi N, Chin WW, Kokkotou E, Dangond F: Functional characterization of JMJD2A, a histone deacetylase- and retinoblastoma-binding protein. J Biol Chem. 2005, 280: 28507-28518. 10.1074/jbc.M413687200.CrossRefPubMed
22.
go back to reference Luo RZ, Fang X, Marquez R, Liu SY, Mills GB, Liao WS, Yu Y, Bast RC: ARHI is a Ras-related small G-protein with a novel N-terminal extension that inhibits growth of ovarian and breast cancers. Oncogene. 2003, 22: 2897-2909. 10.1038/sj.onc.1206380.CrossRefPubMed Luo RZ, Fang X, Marquez R, Liu SY, Mills GB, Liao WS, Yu Y, Bast RC: ARHI is a Ras-related small G-protein with a novel N-terminal extension that inhibits growth of ovarian and breast cancers. Oncogene. 2003, 22: 2897-2909. 10.1038/sj.onc.1206380.CrossRefPubMed
23.
go back to reference Yu Y, Xu F, Peng H, Fang X, Zhao S, Li Y, Cuevas B, Kuo WL, Gray JW, Siciliano M, Mills GB, Bast RJ: NOEY2 (ARHI), an imprinted putative tumor suppressor gene in ovarian and breast carcinomas. Proc Natl Acad Sci U S A. 1999, 96: 214-219. 10.1073/pnas.96.1.214.CrossRefPubMedPubMedCentral Yu Y, Xu F, Peng H, Fang X, Zhao S, Li Y, Cuevas B, Kuo WL, Gray JW, Siciliano M, Mills GB, Bast RJ: NOEY2 (ARHI), an imprinted putative tumor suppressor gene in ovarian and breast carcinomas. Proc Natl Acad Sci U S A. 1999, 96: 214-219. 10.1073/pnas.96.1.214.CrossRefPubMedPubMedCentral
24.
go back to reference Hisatomi H, Nagao K, Wakita K, Kohno N: ARHI/NOEY2 inactivation may be important in breast tumor pathogenesis. Oncology-Basel. 2002, 62: 136-140. 10.1159/000048259.CrossRef Hisatomi H, Nagao K, Wakita K, Kohno N: ARHI/NOEY2 inactivation may be important in breast tumor pathogenesis. Oncology-Basel. 2002, 62: 136-140. 10.1159/000048259.CrossRef
25.
go back to reference Wang L, Hoque A, Luo RZ, Yuan J, Lu Z, Nishimoto A, Liu J, Sahin AA, Lippman SM, Bast RJ, Yu Y: Loss of the expression of the tumor suppressor gene ARHI is associated with progression of breast cancer. Clin Cancer Res. 2003, 9: 3660-3666.PubMed Wang L, Hoque A, Luo RZ, Yuan J, Lu Z, Nishimoto A, Liu J, Sahin AA, Lippman SM, Bast RJ, Yu Y: Loss of the expression of the tumor suppressor gene ARHI is associated with progression of breast cancer. Clin Cancer Res. 2003, 9: 3660-3666.PubMed
26.
go back to reference Janssen EA, Ovestad IT, Skaland I, Soiland H, Gudlaugsson E, Kjellevold KH, Nysted A, Soreide JA, Baak JP: LOH at 1p31 (ARHI) and proliferation in lymph node-negative breast cancer. Cell Oncol. 2009, 31: 335-343.PubMedPubMedCentral Janssen EA, Ovestad IT, Skaland I, Soiland H, Gudlaugsson E, Kjellevold KH, Nysted A, Soreide JA, Baak JP: LOH at 1p31 (ARHI) and proliferation in lymph node-negative breast cancer. Cell Oncol. 2009, 31: 335-343.PubMedPubMedCentral
27.
go back to reference Li Y, Liu M, Zhang Y, Han C, You J, Yang J, Cao C, Jiao S: Effects of ARHI on breast cancer cell biological behavior regulated by microRNA-221. Tumour Biol. 2013, 34: 3545-3554. 10.1007/s13277-013-0933-6.CrossRefPubMed Li Y, Liu M, Zhang Y, Han C, You J, Yang J, Cao C, Jiao S: Effects of ARHI on breast cancer cell biological behavior regulated by microRNA-221. Tumour Biol. 2013, 34: 3545-3554. 10.1007/s13277-013-0933-6.CrossRefPubMed
28.
go back to reference Yu Y, Luo R, Lu Z, Wei FW, Badgwell D, Issa JP, Rosen DG, Liu J, Bast RJ: Biochemistry and biology of ARHI (DIRAS3), an imprinted tumor suppressor gene whose expression is lost in ovarian and breast cancers. Methods Enzymol. 2006, 407: 455-468.CrossRefPubMed Yu Y, Luo R, Lu Z, Wei FW, Badgwell D, Issa JP, Rosen DG, Liu J, Bast RJ: Biochemistry and biology of ARHI (DIRAS3), an imprinted tumor suppressor gene whose expression is lost in ovarian and breast cancers. Methods Enzymol. 2006, 407: 455-468.CrossRefPubMed
29.
go back to reference Klingauf M, Beck M, Berge U, Turgay Y, Heinzer S, Horvath P, Kroschewski R: The tumour suppressor DiRas3 interacts with C-RAF and downregulates MEK activity to restrict cell migration. Biol Cell. 2013, 105: 91-107. 10.1111/boc.201200030.CrossRefPubMed Klingauf M, Beck M, Berge U, Turgay Y, Heinzer S, Horvath P, Kroschewski R: The tumour suppressor DiRas3 interacts with C-RAF and downregulates MEK activity to restrict cell migration. Biol Cell. 2013, 105: 91-107. 10.1111/boc.201200030.CrossRefPubMed
30.
go back to reference Badgwell DB, Lu Z, Le K, Gao F, Yang M, Suh GK, Bao JJ, Das P, Andreeff M, Chen W, Yu Y, Ahmed AA, S-L Liao W, Bast RJ: The tumor-suppressor gene ARHI (DIRAS3) suppresses ovarian cancer cell migration through inhibition of the Stat3 and FAK/Rho signaling pathways. Oncogene. 2012, 31: 68-79. 10.1038/onc.2011.213.CrossRefPubMed Badgwell DB, Lu Z, Le K, Gao F, Yang M, Suh GK, Bao JJ, Das P, Andreeff M, Chen W, Yu Y, Ahmed AA, S-L Liao W, Bast RJ: The tumor-suppressor gene ARHI (DIRAS3) suppresses ovarian cancer cell migration through inhibition of the Stat3 and FAK/Rho signaling pathways. Oncogene. 2012, 31: 68-79. 10.1038/onc.2011.213.CrossRefPubMed
31.
go back to reference Feng W, Marquez RT, Lu Z, Liu J, Lu KH, Issa JP, Fishman DM, Yu Y, Bast RJ: Imprinted tumor suppressor genes ARHI and PEG3 are the most frequently down-regulated in human ovarian cancers by loss of heterozygosity and promoter methylation. Cancer. 2008, 112: 1489-1502. 10.1002/cncr.23323.CrossRefPubMed Feng W, Marquez RT, Lu Z, Liu J, Lu KH, Issa JP, Fishman DM, Yu Y, Bast RJ: Imprinted tumor suppressor genes ARHI and PEG3 are the most frequently down-regulated in human ovarian cancers by loss of heterozygosity and promoter methylation. Cancer. 2008, 112: 1489-1502. 10.1002/cncr.23323.CrossRefPubMed
32.
go back to reference Feng W, Lu Z, Luo RZ, Zhang X, Seto E, Liao WS, Yu Y: Multiple histone deacetylases repress tumor suppressor gene ARHI in breast cancer. Int J Cancer. 2007, 120: 1664-1668. 10.1002/ijc.22474.CrossRefPubMed Feng W, Lu Z, Luo RZ, Zhang X, Seto E, Liao WS, Yu Y: Multiple histone deacetylases repress tumor suppressor gene ARHI in breast cancer. Int J Cancer. 2007, 120: 1664-1668. 10.1002/ijc.22474.CrossRefPubMed
33.
go back to reference Lu Z, Luo RZ, Peng H, Huang M, Nishmoto A, Hunt KK, Helin K, Liao WS, Yu Y: E2F-HDAC complexes negatively regulate the tumor suppressor gene ARHI in breast cancer. Oncogene. 2006, 25: 230-239.CrossRefPubMed Lu Z, Luo RZ, Peng H, Huang M, Nishmoto A, Hunt KK, Helin K, Liao WS, Yu Y: E2F-HDAC complexes negatively regulate the tumor suppressor gene ARHI in breast cancer. Oncogene. 2006, 25: 230-239.CrossRefPubMed
34.
go back to reference Li BX, Luo CL, Li H, Yang P, Zhang MC, Xu HM, Xu HF, Shen YW, Xue AM, Zhao ZQ: Effects of siRNA-mediated knockdown of jumonji domain containing 2A on proliferation, migration and invasion of the human breast cancer cell line MCF-7. Exp Ther Med. 2012, 4: 755-761.PubMedPubMedCentral Li BX, Luo CL, Li H, Yang P, Zhang MC, Xu HM, Xu HF, Shen YW, Xue AM, Zhao ZQ: Effects of siRNA-mediated knockdown of jumonji domain containing 2A on proliferation, migration and invasion of the human breast cancer cell line MCF-7. Exp Ther Med. 2012, 4: 755-761.PubMedPubMedCentral
35.
go back to reference Li BX, Zhang MC, Luo CL, Yang P, Li H, Xu HM, Xu HF, Shen YW, Xue AM, Zhao ZQ: Effects of RNA interference-mediated gene silencing of JMJD2A on human breast cancer cell line MDA-MB-231 in vitro. J Exp Clin Cancer Res. 2011, 30: 90-10.1186/1756-9966-30-90.CrossRefPubMedPubMedCentral Li BX, Zhang MC, Luo CL, Yang P, Li H, Xu HM, Xu HF, Shen YW, Xue AM, Zhao ZQ: Effects of RNA interference-mediated gene silencing of JMJD2A on human breast cancer cell line MDA-MB-231 in vitro. J Exp Clin Cancer Res. 2011, 30: 90-10.1186/1756-9966-30-90.CrossRefPubMedPubMedCentral
36.
go back to reference Guo R, Chen J, Mitchell DL, Johnson DG: GCN5 and E2F1 stimulate nucleotide excision repair by promoting H3K9 acetylation at sites of damage. Nucleic Acids Res. 2011, 39: 1390-1397. 10.1093/nar/gkq983.CrossRefPubMed Guo R, Chen J, Mitchell DL, Johnson DG: GCN5 and E2F1 stimulate nucleotide excision repair by promoting H3K9 acetylation at sites of damage. Nucleic Acids Res. 2011, 39: 1390-1397. 10.1093/nar/gkq983.CrossRefPubMed
37.
go back to reference DuPree EL, Mazumder S, Almasan A: Genotoxic stress induces expression of E2F4, leading to its association with p130 in prostate carcinoma cells. Cancer Res. 2004, 64: 4390-4393. 10.1158/0008-5472.CAN-03-3695.CrossRefPubMed DuPree EL, Mazumder S, Almasan A: Genotoxic stress induces expression of E2F4, leading to its association with p130 in prostate carcinoma cells. Cancer Res. 2004, 64: 4390-4393. 10.1158/0008-5472.CAN-03-3695.CrossRefPubMed
38.
go back to reference Luo CL, Chen XP, Li LL, Li QQ, Li BX, Xue AM, Xu HF, Dai DK, Shen YW, Tao LY, Zhao ZQ: Poloxamer 188 Attenuates in vitro Traumatic Brain Injury-Induced Mitochondrial and Lysosomal Membrane Permeabilization Damage in Cultured Primary Neurons. J Neurotrauma. 2013, 30: 597-607. 10.1089/neu.2012.2425.CrossRefPubMed Luo CL, Chen XP, Li LL, Li QQ, Li BX, Xue AM, Xu HF, Dai DK, Shen YW, Tao LY, Zhao ZQ: Poloxamer 188 Attenuates in vitro Traumatic Brain Injury-Induced Mitochondrial and Lysosomal Membrane Permeabilization Damage in Cultured Primary Neurons. J Neurotrauma. 2013, 30: 597-607. 10.1089/neu.2012.2425.CrossRefPubMed
39.
go back to reference Wang Y, Han R, Liang ZQ, Wu JC, Zhang XD, Gu ZL, Qin ZH: An autophagic mechanism is involved in apoptotic death of rat striatal neurons induced by the non-N-methyl-D-aspartate receptor agonist kainic acid. Autophagy. 2008, 4: 214-226.CrossRefPubMed Wang Y, Han R, Liang ZQ, Wu JC, Zhang XD, Gu ZL, Qin ZH: An autophagic mechanism is involved in apoptotic death of rat striatal neurons induced by the non-N-methyl-D-aspartate receptor agonist kainic acid. Autophagy. 2008, 4: 214-226.CrossRefPubMed
40.
go back to reference Holen I, Whitworth J, Nutter F, Evans A, Brown HK, Lefley DV, Barbaric I, Jones M, Ottewell PD: Loss of plakoglobin promotes decreased cell-cell contact, increased invasion, and breast cancer cell dissemination in vivo. Breast Cancer Res. 2012, 14: R86-10.1186/bcr3201.CrossRefPubMedPubMedCentral Holen I, Whitworth J, Nutter F, Evans A, Brown HK, Lefley DV, Barbaric I, Jones M, Ottewell PD: Loss of plakoglobin promotes decreased cell-cell contact, increased invasion, and breast cancer cell dissemination in vivo. Breast Cancer Res. 2012, 14: R86-10.1186/bcr3201.CrossRefPubMedPubMedCentral
41.
go back to reference Whetstine JR, Nottke A, Lan F, Huarte M, Smolikov S, Chen Z, Spooner E, Li E, Zhang G, Colaiacovo M, Shi Y: Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases. Cell. 2006, 125: 467-481. 10.1016/j.cell.2006.03.028.CrossRefPubMed Whetstine JR, Nottke A, Lan F, Huarte M, Smolikov S, Chen Z, Spooner E, Li E, Zhang G, Colaiacovo M, Shi Y: Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases. Cell. 2006, 125: 467-481. 10.1016/j.cell.2006.03.028.CrossRefPubMed
42.
go back to reference Cheng C, Kong X, Wang H, Gan H, Hao Y, Zou W, Wu J, Chi Y, Yang J, Hong Y, Chen K, Gu J: Trihydrophobin 1 Interacts with PAK1 and Regulates ERK/MAPK Activation and Cell Migration. J Biol Chem. 2009, 284: 8786-8796. 10.1074/jbc.M806144200.CrossRefPubMedPubMedCentral Cheng C, Kong X, Wang H, Gan H, Hao Y, Zou W, Wu J, Chi Y, Yang J, Hong Y, Chen K, Gu J: Trihydrophobin 1 Interacts with PAK1 and Regulates ERK/MAPK Activation and Cell Migration. J Biol Chem. 2009, 284: 8786-8796. 10.1074/jbc.M806144200.CrossRefPubMedPubMedCentral
43.
go back to reference Lee YF, Miller LD, Chan XB, Black MA, Pang B, Ong CW, Salto-Tellez M, Liu ET, Desai KV: JMJD6 is a driver of cellular proliferation and motility and a marker of poor prognosis in breast cancer. Breast Cancer Res. 2012, 14: R85-10.1186/bcr3200.CrossRefPubMedPubMedCentral Lee YF, Miller LD, Chan XB, Black MA, Pang B, Ong CW, Salto-Tellez M, Liu ET, Desai KV: JMJD6 is a driver of cellular proliferation and motility and a marker of poor prognosis in breast cancer. Breast Cancer Res. 2012, 14: R85-10.1186/bcr3200.CrossRefPubMedPubMedCentral
44.
go back to reference Zou CF, Jia L, Jin H, Yao M, Zhao N, Huan J, Lu Z, Bast RJ, Feng Y, Yu Y: Re-expression of ARHI (DIRAS3) induces autophagy in breast cancer cells and enhances the inhibitory effect of paclitaxel. BMC Cancer. 2011, 11: 22-10.1186/1471-2407-11-22.CrossRefPubMedPubMedCentral Zou CF, Jia L, Jin H, Yao M, Zhao N, Huan J, Lu Z, Bast RJ, Feng Y, Yu Y: Re-expression of ARHI (DIRAS3) induces autophagy in breast cancer cells and enhances the inhibitory effect of paclitaxel. BMC Cancer. 2011, 11: 22-10.1186/1471-2407-11-22.CrossRefPubMedPubMedCentral
45.
go back to reference Blander G, Guarente L: The Sir2 family of protein deacetylases. Annu Rev Biochem. 2004, 73: 417-435. 10.1146/annurev.biochem.73.011303.073651.CrossRefPubMed Blander G, Guarente L: The Sir2 family of protein deacetylases. Annu Rev Biochem. 2004, 73: 417-435. 10.1146/annurev.biochem.73.011303.073651.CrossRefPubMed
46.
go back to reference Bowen NJ, Fujita N, Kajita M, Wade PA: Mi-2/NuRD: multiple complexes for many purposes. Biochim Biophys Acta. 2004, 1677: 52-57. 10.1016/j.bbaexp.2003.10.010.CrossRefPubMed Bowen NJ, Fujita N, Kajita M, Wade PA: Mi-2/NuRD: multiple complexes for many purposes. Biochim Biophys Acta. 2004, 1677: 52-57. 10.1016/j.bbaexp.2003.10.010.CrossRefPubMed
47.
go back to reference Feng Q, Zhang Y: The NuRD complex: linking histone modification to nucleosome remodeling. Curr Top Microbiol Immunol. 2003, 274: 269-290.PubMed Feng Q, Zhang Y: The NuRD complex: linking histone modification to nucleosome remodeling. Curr Top Microbiol Immunol. 2003, 274: 269-290.PubMed
48.
go back to reference Glass CK, Rosenfeld MG: The coregulator exchange in transcriptional functions of nuclear receptors. Genes Dev. 2000, 14: 121-141.PubMed Glass CK, Rosenfeld MG: The coregulator exchange in transcriptional functions of nuclear receptors. Genes Dev. 2000, 14: 121-141.PubMed
49.
go back to reference Wolffe AP, Urnov FD, Guschin D: Co-repressor complexes and remodelling chromatin for repression. Biochem Soc Trans. 2000, 28: 379-386. 10.1042/0300-5127:0280379.CrossRefPubMed Wolffe AP, Urnov FD, Guschin D: Co-repressor complexes and remodelling chromatin for repression. Biochem Soc Trans. 2000, 28: 379-386. 10.1042/0300-5127:0280379.CrossRefPubMed
50.
go back to reference Luo RZ, Peng H, Xu F, Bao J, Pang Y, Pershad R, Issa JP, Liao WS, Bast RJ, Yu Y: Genomic structure and promoter characterization of an imprinted tumor suppressor gene ARHI. Biochim Biophys Acta. 2001, 1519: 216-222. 10.1016/S0167-4781(01)00226-3.CrossRefPubMed Luo RZ, Peng H, Xu F, Bao J, Pang Y, Pershad R, Issa JP, Liao WS, Bast RJ, Yu Y: Genomic structure and promoter characterization of an imprinted tumor suppressor gene ARHI. Biochim Biophys Acta. 2001, 1519: 216-222. 10.1016/S0167-4781(01)00226-3.CrossRefPubMed
51.
go back to reference Kawazu M, Saso K, Tong KI, McQuire T, Goto K, Son DO, Wakeham A, Miyagishi M, Mak TW, Okada H: Histone demethylase JMJD2B functions as a co-factor of estrogen receptor in breast cancer proliferation and mammary gland development. PLoS One. 2011, 6: e17830-10.1371/journal.pone.0017830.CrossRefPubMedPubMedCentral Kawazu M, Saso K, Tong KI, McQuire T, Goto K, Son DO, Wakeham A, Miyagishi M, Mak TW, Okada H: Histone demethylase JMJD2B functions as a co-factor of estrogen receptor in breast cancer proliferation and mammary gland development. PLoS One. 2011, 6: e17830-10.1371/journal.pone.0017830.CrossRefPubMedPubMedCentral
52.
go back to reference Kouzarides T: Histone acetylases and deacetylases in cell proliferation. Curr Opin Genet Dev. 1999, 9: 40-48. 10.1016/S0959-437X(99)80006-9.CrossRefPubMed Kouzarides T: Histone acetylases and deacetylases in cell proliferation. Curr Opin Genet Dev. 1999, 9: 40-48. 10.1016/S0959-437X(99)80006-9.CrossRefPubMed
53.
go back to reference Hess KR, Varadhachary GR, Taylor SH, Wei W, Raber MN, Lenzi R, Abbruzzese JL: Metastatic patterns in adenocarcinoma. Cancer. 2006, 106: 1624-1633. 10.1002/cncr.21778.CrossRefPubMed Hess KR, Varadhachary GR, Taylor SH, Wei W, Raber MN, Lenzi R, Abbruzzese JL: Metastatic patterns in adenocarcinoma. Cancer. 2006, 106: 1624-1633. 10.1002/cncr.21778.CrossRefPubMed
54.
go back to reference Joyce JA, Pollard JW: Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009, 9: 239-252. 10.1038/nrc2618.CrossRefPubMed Joyce JA, Pollard JW: Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009, 9: 239-252. 10.1038/nrc2618.CrossRefPubMed
55.
go back to reference Langley RR, Fidler IJ: The seed and soil hypothesis revisited–the role of tumor-stroma interactions in metastasis to different organs. Int J Cancer. 2011, 128: 2527-2535. 10.1002/ijc.26031.CrossRefPubMedPubMedCentral Langley RR, Fidler IJ: The seed and soil hypothesis revisited–the role of tumor-stroma interactions in metastasis to different organs. Int J Cancer. 2011, 128: 2527-2535. 10.1002/ijc.26031.CrossRefPubMedPubMedCentral
56.
go back to reference Mook OR, van Marle J, Jonges R, Vreeling-Sindelarova H, Frederiks WM, Van Noorden CJ: Interactions between colon cancer cells and hepatocytes in rats in relation to metastasis. J Cell Mol Med. 2008, 12: 2052-2061. 10.1111/j.1582-4934.2008.00242.x.CrossRefPubMedPubMedCentral Mook OR, van Marle J, Jonges R, Vreeling-Sindelarova H, Frederiks WM, Van Noorden CJ: Interactions between colon cancer cells and hepatocytes in rats in relation to metastasis. J Cell Mol Med. 2008, 12: 2052-2061. 10.1111/j.1582-4934.2008.00242.x.CrossRefPubMedPubMedCentral
57.
go back to reference Mook OR, Van Marle J, Vreeling-Sindelarova H, Jonges R, Frederiks WM, Van Noorden CJ: Visualization of early events in tumor formation of eGFP-transfected rat colon cancer cells in liver. Hepatology. 2003, 38: 295-304.CrossRefPubMed Mook OR, Van Marle J, Vreeling-Sindelarova H, Jonges R, Frederiks WM, Van Noorden CJ: Visualization of early events in tumor formation of eGFP-transfected rat colon cancer cells in liver. Hepatology. 2003, 38: 295-304.CrossRefPubMed
58.
go back to reference Tabaries S, Dong Z, Annis MG, Omeroglu A, Pepin F, Ouellet V, Russo C, Hassanain M, Metrakos P, Diaz Z, Basik M, Bertos N, Park M, Guettier C, Adam R, Hallett M, Siegel PM: Claudin-2 is selectively enriched in and promotes the formation of breast cancer liver metastases through engagement of integrin complexes. Oncogene. 2011, 30: 1318-1328. 10.1038/onc.2010.518.CrossRefPubMed Tabaries S, Dong Z, Annis MG, Omeroglu A, Pepin F, Ouellet V, Russo C, Hassanain M, Metrakos P, Diaz Z, Basik M, Bertos N, Park M, Guettier C, Adam R, Hallett M, Siegel PM: Claudin-2 is selectively enriched in and promotes the formation of breast cancer liver metastases through engagement of integrin complexes. Oncogene. 2011, 30: 1318-1328. 10.1038/onc.2010.518.CrossRefPubMed
59.
go back to reference Tabaries S, Dupuy F, Dong Z, Monast A, Annis MG, Spicer J, Ferri LE, Omeroglu A, Basik M, Amir E, Clemons M, Siegel PM: Claudin-2 promotes breast cancer liver metastasis by facilitating tumor cell interactions with hepatocytes. Mol Cell Biol. 2012, 32: 2979-2991. 10.1128/MCB.00299-12.CrossRefPubMedPubMedCentral Tabaries S, Dupuy F, Dong Z, Monast A, Annis MG, Spicer J, Ferri LE, Omeroglu A, Basik M, Amir E, Clemons M, Siegel PM: Claudin-2 promotes breast cancer liver metastasis by facilitating tumor cell interactions with hepatocytes. Mol Cell Biol. 2012, 32: 2979-2991. 10.1128/MCB.00299-12.CrossRefPubMedPubMedCentral
Metadata
Title
JMJD2A contributes to breast cancer progression through transcriptional repression of the tumor suppressor ARHI
Authors
Li-Liang Li
Ai-Min Xue
Bei-Xu Li
Yi-Wen Shen
Yu-Hua Li
Cheng-Liang Luo
Ming-Chang Zhang
Jie-Qing Jiang
Zu-De Xu
Jian-Hui Xie
Zi-Qin Zhao
Publication date
01-06-2014
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 3/2014
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/bcr3667

Other articles of this Issue 3/2014

Breast Cancer Research 3/2014 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