Stimulation of β-catenin and colon cancer cell growth by the KDM4B histone demethylase

  • Authors:
    • William L. Berry
    • Tae-Dong Kim
    • Ralf Janknecht
  • View Affiliations

  • Published online on: January 27, 2014     https://doi.org/10.3892/ijo.2014.2279
  • Pages: 1341-1348
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The linchpin of colorectal cancer is the oncoprotein and transcriptional cofactor β-catenin, whose overexpression is causative for the neoplastic transformation of colon cells. However, the molecular details of β-catenin dependent gene transcription in cancer cells are still not comprehensively explored. Here, we show that the histone demethylase KDM4B was upregulated in colon and rectal adenocarcinomas and required for efficient growth and clonogenic activity of human HT-29 colon cancer cells. Moreover, KDM4B formed complexes with β-catenin in vitro and in vivo, which involved its central amino acids 353-740. In addition, KDM4B also interacted with the DNA-binding protein TCF4, which is the main factor recruiting β-catenin to chromatin in the intestine. KDM4B downregulation resulted in reduced expression of the β-catenin/TCF4 target genes JUN, MYC and Cyclin D1, all of which encode for oncoproteins. Collectively, our data indicate that KDM4B overexpression supports β-catenin mediated gene transcription and thereby contributes to the genesis of colorectal tumors. Accordingly, inhibition of the KDM4B histone demethylase may represent a novel avenue of fighting colorectal cancer, one of the major causes of cancer death throughout the world.

Introduction

Colorectal cancer is a major health issue and over 50,000 US residents alone are expected to die from this disease this year (1). A crucial defect in the vast majority of colorectal tumors is the overexpression of the oncoprotein β-catenin. This is primarily due to the loss of the tumor suppressor adenomatous polyposis coli (APC), which normally directs the intracellular destruction of β-catenin, or activating mutations in β-catenin itself (2). Unfortunately, knowledge on the devastating impact of these genetic mutations has not yet translated into improved therapy.

Aside from genetic mutations, epigenetic changes are an underlying cause of tumorigenesis. Most prominently, such epigenetic changes involve the methylation of DNA on cytosine residues and the modification of histones by acetylation and methylation (3,4). In contrast to DNA methylation and histone acetylation, the methylation of histones was only recently validated as a major epigenetic mechanism. Methylation occurs on lysine and arginine residues at multiple sites on histones and the tight regulation of the histone methylation status is absolutely required for normal cell physiology and safeguards against aberrant cell growth. Thus, enzymes affecting histone methylation play seminal roles in cellular homeostasis and, accordingly, dysregulation of both histone methyltransferases as well as the opposing demethylases is thought to be capable of inducing cancer (5,6). However, the roles of the enzymes determining histone methylation in colorectal tumors are largely unexplored.

The family of human Jumonji C domain containing proteins comprises 30 members, many of which have been shown to function as histone demethylases (7). These include the four related lysine-specific demethylase 4 (KDM4) proteins, KDM4A-D (8). They can demethylate histone H3 on lysines 9 and 36 as well as histone H1.4 on lysine 26 (915). Notably, KDM4A, B and C are overexpressed in human breast tumors and promote proliferation of breast tumor cells, in part due to their ability to function as cofactors of estrogen receptor α (1621). Likewise, all four KDM4 proteins were shown to coactivate the androgen receptor and may thereby contribute to prostate tumor formation (2224). Despite these examples of overlapping function, KDM4 proteins can also behave differently from each other (8). For instance, KDM4B seems to be less catalytically active (9,12) and is the only KDM4 protein that is robustly overexpressed under hypoxia (25,26). Here, we explored the role of KDM4B in colorectal cancer.

Materials and methods

Lentivirus mediated KDM4B downregulation with shRNA

To generate an inducible miRshRNA entry vector, annealed oligonucleotides KDM4B-shRNA sense (5′-AGCGAGCGCTGACACTGTATTCTTATTAGTGAAGCCACAGATGTAATAAGAATACAGTGTCAGCGC-3′) and KDM4B-shRNA antisense (5′-GGCAGGCGCTGACACTGTATTCTTATTACATCTGTGGCTTCACTAATAAGAATACAGTGTCAGCGCT-3′) were cloned into pEN_TTRmiRc2 (Addgene). To transfer sequences into the lentiviral destination vector, the miRshKDM4B entry vector was incubated together with pSLIK Hygro (Addgene) and Clonase 2 (Invitrogen) per company instructions. The resulting miRshKDM4B lentiviral expression vector was cotransfected along with packaging plasmids pMD2.G (Addgene), pMDL/RRE g/p (Addgene) and pRSV-Rev (Addgene) into 293T cells. Cells were seeded at ∼60% confluence and DNA was delivered using the polyethylenimine transfection method overnight. Polyethylenimine to DNA ratio was 3:1. Forty-eight and 72 h post-transfection, supernatant was harvested and filtered through a 0.45-μm membrane. Cleared viral supernatant was concentrated using poly(ethylene glycol)-8000 and used to infect HT-29 cells (27).

Cell proliferation assay

HT-29 cells that inducibly expressed shRNA targeting KDM4B were seeded into 96-wells and grown in DMEM medium plus 10% fetal calf serum. One day thereafter, the cell number was determined for the first time and defined as number of cells at day 0. Then, cells were mock treated or with 500 ng/ml doxycycline and cell numbers measured at the indicated days thereafter with the TACS (Trevigen) MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) kit (28). Averages with standard errors of triplicate experiments were determined.

Clonogenic assay

Cells were seeded at 5,000 cells per well. Thereafter, cells were mock treated or with 500 ng/ml doxycycline and grown in DMEM medium plus 10% fetal calf serum. Media without and with doxycycline were replenished every four days for twelve days. The cells were then fixed in 3.7% formaldehyde for 10 min and stained with 1% crystal violet blue for 30 min. After rinsing with distilled water, cells were photographed.

Coimmunoprecipitation assays

Human embryonic kidney 293T cells were grown in a humidified atmosphere in 10% CO2 at 37°C (29). Cells were seeded into 6-cm dishes that were coated with poly-L-lysine 12–24 h before transfection (30). Cells were then transiently transfected by the calcium phosphate coprecipitation method (31) and lysed 36 h after transfection with 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 50 mM NaF, 0.25 mM Na3VO4, 0.5% Igepal CA-630, 10 μg/ml leupeptin, 2 μg/ml aprotinin, 1 μg/ml pepstatin A, 0.5 mM PMSF, 0.2 mM DTT. Immunoprecipitation was performed with anti-Flag M2 or anti-Myc 9E10 monoclonal antibodies as described (32). Coprecipitated proteins were detected by western blotting employing enhanced chemiluminescence (33).

Glutathione S-transferase (GST) pull-downs

GST fusion proteins were produced in Escherichia coli and purified as previously described (34). Cell extract containing Myc-tagged β-catenin protein was prepared from transiently transfected 293T cells (35). This extract was then incubated for 3 h at 4°C with GST fusion proteins bound to glutathione agarose beads utilizing 20 mM HEPES (pH 7.4), 50 mM NaCl, 1 mM DTT, 0.01% Tween-20, 0.5 mM PMSF as a binding buffer. After three washes in the same buffer, any bound Myc-tagged β-catenin was revealed by western blotting employing anti-Myc 9E10 monoclonal antibodies.

Reporter gene assay

HT-29 cells inducibly expressing KDM4B shRNA were infected with pBARLS lentivirus (kind gift from Dr Randall Moon) that encodes 12 TCF4 binding elements upstream of luciferase cDNA. Equal numbers of cells were then split into two groups of triplicates and treated without and with 500 ng/ml doxycycline for 72 h. Then, cells were lysed as described before (36) and luciferase activities derived from pBARLS were measured in a Berthold LB9507 luminometer (37).

Reverse transcription-polymerase chain reaction (RT-PCR)

RNA was isolated employing TRIzol (Invitrogen) and utilized in the Access RT-PCR kit (Promega) as described before (38). Oligonucleotides used were 5′-GTGACCGCGACTTTTCAAAGC-3′ and 5′-CGTTGCTGGACTG GATTATCAG-3′ for JUN, 5′-TGAGGAGACACCGCCCAC-3′ and 5′-CAACATCGATTTCTTCCTCATCTTC-3′ for MYC, 5′-AAGGCGGAGGAGACCTGCGCG-3′ and 5′-ATCGTGCGGGGTCATTGCGGC-3′ for Cyclin D1, and 5′-GAGCCACATCGCTCAGACACC-3′ and 5′-TGACAAGCTTCCGCTTCTCAGC-3′ for GAPDH. Resulting PCR products were separated on agarose gels and stained with ethidium bromide (39).

Chromatin immunoprecipitations

These were performed essentially as described before (40) with rabbit KDM4B antibodies either from Bethyl (A301-478A) in case of the JUN promoter or from Novus Biologicals (NBP1-67802) in case of the Cyclin D1 promoter. For promoter fragment amplification, nested PCR was employed using the temperature program 98°C for 2 min; 8 cycles of 98°C for 30 sec, 65°C (−1°C per cycle) for 30 sec, 72°C for 25 sec; 18 cycles (in the first PCR) or 15 cycles (in the second PCR) of 98°C for 30 sec, 57°C for 30 sec, 72°C for 25 sec (+ 1 sec per cycle), followed by a final 4-min extension at 72°C (41). The iProof high fidelity DNA polymerase (Bio-Rad) was used in the first PCR and GoTaq polymerase with 5X Green buffer (Promega) in the second PCR. The primers for the first PCR were: Jun-ChIP-2805-for (5′-GGCAGCCACCGTCACTAGACAGTC-3′) and Jun-ChIP-3184-rev (5′-GCCACACTCAGTGCAACTCTGAGC-3′), or D1-pro-for6 (5′-GTAACGTCACACGGACTACAGG-3′) and D1-pro-rev5 (5′-GCACACATTTGAAGTAGGACACC-3′). The primers for the second PCR were: Jun-ChIP-2834-for (5′-CCAAGACGTCAGCCCACAATGCACC-3′) and Jun-ChIP-3145-rev (5′-GCTCAACACTTATCTGCTACCAGTC-3′), or D1-ChIP-2726-for (5′-GTTGCAAAGTCCTGGAGCCTCCAG-3′) and D1-ChIP-2982-rev (5′-CGGTCGTTGAGGAGGTTGGCATCG-3′). The resultant 312 bp JUN promoter and 257 bp Cyclin D1 promoter fragments were revealed by agarose gel electrophoresis (42).

Results

Overexpression of KDM4B in colorectal tumors

To comprehensively assess the expression pattern of KDM4 genes in human colorectal tumors, we performed in silico analyses comparing normal and cancer tissues with the help of Oncomine (www.oncomine.org). In two independent data sets (43,44), KDM4B mRNA was significantly upregulated in both colon and rectal adenocarcinomas compared to normal colon tissue, whereas no relevant upregulation of KDM4A, KDM4C or KDM4D was observable (Fig. 1). These data implicate that elevated KDM4B levels, but not overexpression of any of the other three KDM4 family members, play a role in the development of colorectal tumors.

Growth deficit of colon cancer cells following loss of KDM4B

To test the impact of KDM4B on cell physiology, we infected human HT-29 colon cancer cells with lentivirus that doxycycline-inducibly expressed KDM4B shRNA. After selection for viral integration, these cells were treated with and without doxycycline, which resulted in efficient downregulation of KDM4B (Fig. 2A). Importantly, doxycycline treatment significantly reduced HT-29 cell growth (Fig. 2A), suggesting that KDM4B has a pro-growth effect. We also determined the importance of KDM4B for growing colonies from single cells, another litmus test for oncogenic activity. When treated with doxycycline to ablate KDM4B expression, HT-29 cells displayed a markedly reduced ability to establish colonies (Fig. 2B). Altogether, these results support the notion that KDM4B promotes neoplastic growth of colon cells.

Complex formation of KDM4B with β-catenin

Overexpression of the β-catenin oncoprotein, the linchpin of colon tumorigenesis, is observed in >80% of sporadic colorectal tumors (2). Like KDM4B, β-catenin is a transcriptional cofactor (45). Therefore, we tested whether KDM4B might interact with β-catenin. To this end, we coexpressed Flag-tagged β-catenin with Myc-tagged KDM4B in 293T cells and performed coimmunoprecipitation experiments. We found that β-catenin coprecipitated with KDM4B (Fig. 3A). Similarly, in a reverse order coimmunoprecipitation experiment, KDM4B coprecipitated with β-catenin (Fig. 3B). These data demonstrate that KDM4B and β-catenin form complexes in vivo.

Next, we explored whether β-catenin would bind to KDM4B in vitro and which domains of KDM4B would be involved. To this end, we divided KDM4B into three parts and purified respective GST fusion proteins. These were then bound to glutathione agarose beads, which were subsequently incubated with a cell extract containing Myc-tagged β-catenin. No binding of β-catenin to the GST moiety itself was detectable, but it interacted with KDM4B amino acids 353–740 (Fig. 4A). These amino acids are devoid of any known structural motifs (Fig. 4B). In contrast, β-catenin did not appreciably bind to the N-terminal KDM4B amino acids 2–352 encompassing the catalytic JmjC and JmjN domains or the C-terminal amino acids 741–1130, which contain the double PHD and TUDOR domains that are involved in recognizing methylated histone residues (8,46). Collectively, our data reveal that KDM4B and β-catenin interact both in vitro and in vivo.

Coactivation of gene transcription by KDM4B

The β-catenin protein itself does not directly bind to DNA. Rather, it is primarily recruited to chromatin in the intestine by the DNA-binding transcription factor TCF4 (47). Therefore, we reasoned that not only β-catenin, but also TCF4 might form a complex with KDM4B. To test this hypothesis, we coexpressed Flag-tagged KDM4B and Myc-tagged TCF4 in 293T cells, immunoprecipitated with Flag antibodies and then tested for coprecipitated TCF4 by anti-Myc western blotting. Indeed, TCF4 coprecipitated with KDM4B (Fig. 5A), implicating that a tripartite complex of TCF4, β-catenin and KDM4B can be formed.

To determine whether KDM4B stimulates or represses β-catenin/TCF4-dependent transcription, we employed a TOPflash luciferase reporter system that specifically measures β-catenin/TCF4-dependent transcription in colon cells (48). A lentiviral vector was employed to stably introduce the luciferase reporter gene into our HT-29 colon cancer cells that doxycycline-inducibly expressed KDM4B shRNA. In the absence of doxycycline, high luciferase activity was expectedly observable (Fig. 5B). However, upon depletion of KDM4B after doxycyline treatment, luciferase activity was ∼3-fold reduced, indicating that KDM4B can stimulate β-catenin/TCF4-dependent gene transcription.

We next sought to assess how endogenous target genes of β-catenin/TCF4 would be affected by KDM4B downregulation. Specifically, we studied three oncogenes encoding the JUN or MYC transcription factor or the cell cycle regulator Cyclin D1, all of which are bona fide targets of β-catenin/TCF4 (4851). When utilizing our doxycycline-inducible KDM4B shRNA expressing HT-29 cells, we observed that JUN, MYC and Cyclin D1 transcription became reduced upon KDM4B downregulation, whereas the control GAPDH was unaffected (Fig. 6A). In addition, western blotting showed that this also held true at the protein level for JUN and Cyclin D1 (Fig. 6B), whereas MYC protein expression was below our detection level (not shown); as controls, neither β-catenin nor actin protein levels were affected by KDM4B downregulation (Fig. 6B).

Finally, we assessed whether KDM4B would bind to the JUN or Cyclin D1 gene promoter. To this end, we performed chromatin immunoprecipitation experiments. While control IgG antibodies barely or not at all led to precipitation of promoter fragments, KDM4B antibodies were able to robustly precipitate both JUN and Cyclin D1 promoter fragments (Fig. 6C). This indicates that JUN and Cyclin D1 are directly regulated by KDM4B. Altogether, our data provide evidence that KDM4B coactivates β-catenin/TCF4-dependent gene transcription.

Discussion

The data described in this report provide novel information. First, we discovered that the KDM4B histone demethylase is appreciably upregulated at the mRNA level in colorectal tumors, whereas none of the other three KDM4 family members is. Second, we showed that growth and colony formation ability of HT-29 colon cancer cells is stimulated by KDM4B, and third, we revealed that KDM4B is capable of forming complexes with β-catenin and TCF4 and can thereby enhance transcription of oncogenes such as JUN, MYC and Cyclin D1. Altogether, these data strongly suggest that KDM4B has oncogenic properties in colorectal cells.

KDM4B is a histone demethylase that especially demethylates trimethylated H3K9, H3K36 and H1.4K26 (9,12,14). In general, both trimethylated H3K9 and H1.4K26 at a gene promoter are associated with a transcriptionally repressed status of chromatin (52,53), providing an explanation how KDM4B may stimulate gene transcription by removing these repressive marks. On the other hand, trimethylated H3K36 has a variety of functions: it may promote transcription elongation but suppress transcription initiation at the promoter (54). Thus, the net effect on gene expression upon removal of H3K36 trimethylation by KDM4B is debatable. However, since KDM4 proteins appear to more efficiently target H3K9 than H3K36 (8), it is likely that the effect of KDM4B on gene transcription is primarily governed by its ability to demethylate trimethylated H3K9 (and H1.4K26) and accordingly KDM4B overexpression should result in enhanced transcription of target genes like JUN, MYC and Cyclin D1.

MYC and JUN encode for DNA-binding transcription factors and are prominent oncogenes (55,56). They are also β-catenin target genes (48,49) and upregulated in human colorectal tumors (57,58). Interestingly, JUN can form complexes with β-catenin and TCF4, which stabilizes the β-catenin/TCF4 complex and enhances β-catenin dependent transcription (59,60). Furthermore, a dominant-negative version of JUN was able to suppress the tumorigenic potential of HT-29 colon cancer cells and JUN inactivation also reduced gastrointestinal tumor formation in the APCMin mouse model, in which inactivation of APC leads to β-catenin overexpression (59,61). Likewise, the cell cycle regulator Cyclin D1 is upregulated in colorectal tumors (62) and its ablation suppressed tumor formation in APCMin mice (63). Thus, stimulation of JUN, MYC and Cyclin D1 expression by KDM4B is predicted to promote colorectal tumor formation.

Cooperation with β-catenin may not be the only mechanism by which KDM4B contributes to the causation of colorectal cancer. Recently, it was reported that KDM4B is involved in the stimulation of hypoxia-inducible genes in colon cancer (64). Possibly, this may involve complex formation of KDM4B with the hypoxia-inducible factor 1α, the master mediator of the hypoxic response, since a relative of KDM4B, the KDM4C protein, can actually bind to hypoxia-inducible factor 1α (65). Moreover, KDM4A is capable of binding to the p53 tumor suppressor in colon cancer cells and inhibit p53-dependent transcription (66), raising the possibility that also KDM4B might do so.

Apart from transcription regulation, KDM4B is also involved in DNA repair. Its TUDOR domains can bind to dimethylated H4K20, which normally recruits 53BP1 to sites of DNA damage and thereby facilitates repair. Notably, this activity of KDM4B is independent of its enzymatic activity (67). Accordingly, KDM4B overexpression could impair DNA repair by preventing the recruitment of 53BP1 and thereby induce genomic instability, another mechanism by which KDM4B could contribute to tumor formation. However, a recent report posits that KDM4B enhances DNA repair in a manner dependent on its enzymatic activity and thus promotes cell survival (68). This may be relevant during therapy, when KDM4B overexpression could help cancer cells to repair the DNA damage caused by radiotherapy and many chemotherapeutic agents.

In conclusion, our study highlighted the pro-growth function of KDM4B in colorectal tumors and provided a mechanism by which KDM4B may attain this through interaction with β-catenin/TCF4. Given that β-catenin is involved in many other types of cancer (2,45), including breast and bladder cancer where KDM4B is also overexpressed (18,69,70), the oncogenic functions of KDM4B may be of widespread importance. Thus, inhibition of KDM4B enzymatic activity or preventing its interaction with β-catenin by small molecule drugs, similar to the inhibition of chromatin recruitment of the BRD4 epigenetic regulator in acute myeloid leukemia (71,72), may hold promise in cancer therapy.

References

1. 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar

2. 

Clevers H: Wnt/beta-catenin signaling in development and disease. Cell. 127:469–480. 2006. View Article : Google Scholar : PubMed/NCBI

3. 

Chi P, Allis CD and Wang GG: Covalent histone modifications--miswritten, misinterpreted and mis-erased in human cancers. Nat Rev Cancer. 10:457–469. 2010. View Article : Google Scholar : PubMed/NCBI

4. 

Kulis M and Esteller M: DNA methylation and cancer. Adv Genet. 70:27–56. 2010. View Article : Google Scholar

5. 

Dawson MA and Kouzarides T: Cancer epigenetics: from mechanism to therapy. Cell. 150:12–27. 2012. View Article : Google Scholar : PubMed/NCBI

6. 

Black JC, Van Rechem C and Whetstine JR: Histone lysine methylation dynamics: establishment, regulation, and biological impact. Mol Cell. 48:491–507. 2012. View Article : Google Scholar

7. 

Kooistra SM and Helin K: Molecular mechanisms and potential functions of histone demethylases. Nat Rev Mol Cell Biol. 13:297–311. 2012.PubMed/NCBI

8. 

Berry WL and Janknecht R: KDM4/JMJD2 histone demethylases: epigenetic regulators in cancer cells. Cancer Res. 73:2936–2942. 2013. View Article : Google Scholar : PubMed/NCBI

9. 

Whetstine JR, Nottke A, Lan F, Huarte M, Smolikov S, Chen Z, Spooner E, Li E, Zhang G, Colaiacovo M and Shi Y: Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases. Cell. 125:467–481. 2006. View Article : Google Scholar : PubMed/NCBI

10. 

Cloos PA, Christensen J, Agger K, Maiolica A, Rappsilber J, Antal T, Hansen KH and Helin K: The putative oncogene GASC1 demethylates tri- and dimethylated lysine 9 on histone H3. Nature. 442:307–311. 2006. View Article : Google Scholar : PubMed/NCBI

11. 

Klose RJ, Yamane K, Bae Y, Zhang D, Erdjument-Bromage H, Tempst P, Wong J and Zhang Y: The transcriptional repressor JHDM3A demethylates trimethyl histone H3 lysine 9 and lysine 36. Nature. 442:312–316. 2006. View Article : Google Scholar

12. 

Fodor BD, Kubicek S, Yonezawa M, O’Sullivan RJ, Sengupta R, Perez-Burgos L, Opravil S, Mechtler K, Schotta G and Jenuwein T: Jmjd2b antagonizes H3K9 trimethylation at pericentric heterochromatin in mammalian cells. Genes Dev. 20:1557–1562. 2006. View Article : Google Scholar : PubMed/NCBI

13. 

Shin S and Janknecht R: Diversity within the JMJD2 histone demethylase family. Biochem Biophys Res Commun. 353:973–977. 2007. View Article : Google Scholar : PubMed/NCBI

14. 

Trojer P, Zhang J, Yonezawa M, Schmidt A, Zheng H, Jenuwein T and Reinberg D: Dynamic histone H1 isotype 4 methylation and demethylation by histone lysine methyltransferase G9a/KMT1C and the Jumonji domain-containing JMJD2/KDM4 proteins. J Biol Chem. 284:8395–8405. 2009. View Article : Google Scholar : PubMed/NCBI

15. 

Weiss T, Hergeth S, Zeissler U, Izzo A, Tropberger P, Zee BM, Dundr M, Garcia BA, Daujat S and Schneider R: Histone H1 variant-specific lysine methylation by G9a/KMT1C and Glp1/KMT1D. Epigenetics Chromatin. 3:72010. View Article : Google Scholar : PubMed/NCBI

16. 

Liu G, Bollig-Fischer A, Kreike B, van de Vijver MJ, Abrams J, Ethier SP and Yang ZQ: Genomic amplification and oncogenic properties of the GASC1 histone demethylase gene in breast cancer. Oncogene. 28:4491–4500. 2009. View Article : Google Scholar : PubMed/NCBI

17. 

Yang J, Jubb AM, Pike L, Buffa FM, Turley H, Baban D, Leek R, Gatter KC, Ragoussis J and Harris AL: The histone demethylase JMJD2B is regulated by estrogen receptor alpha and hypoxia, and is a key mediator of estrogen induced growth. Cancer Res. 70:6456–6466. 2010. View Article : Google Scholar : PubMed/NCBI

18. 

Kawazu M, Saso K, Tong KI, McQuire T, Goto K, Son DO, Wakeham A, Miyagishi M, Mak TW and Okada H: Histone demethylase JMJD2B functions as a co-factor of estrogen receptor in breast cancer proliferation and mammary gland development. PLoS One. 6:e178302011. View Article : Google Scholar : PubMed/NCBI

19. 

Shi L, Sun L, Li Q, Liang J, Yu W, Yi X, Yang X, Li Y, Han X, Zhang Y, Xuan C, Yao Z and Shang Y: Histone demethylase JMJD2B coordinates H3K4/H3K9 methylation and promotes hormonally responsive breast carcinogenesis. Proc Natl Acad Sci USA. 108:7541–7546. 2011. View Article : Google Scholar : PubMed/NCBI

20. 

Berry WL, Shin S, Lightfoot SA and Janknecht R: Oncogenic features of the JMJD2A histone demethylase in breast cancer. Int J Oncol. 41:1701–1706. 2012.

21. 

Gaughan L, Stockley J, Coffey K, O’Neill D, Jones DL, Wade M, Wright J, Moore M, Tse S, Rogerson L and Robson CN: KDM4B is a master regulator of the estrogen receptor signalling cascade. Nucleic Acids Res. 41:6892–6904. 2013. View Article : Google Scholar

22. 

Wissmann M, Yin N, Muller JM, Greschik H, Fodor BD, Jenuwein T, Vogler C, Schneider R, Gunther T, Buettner R, Metzger E and Schule R: Cooperative demethylation by JMJD2C and LSD1 promotes androgen receptor-dependent gene expression. Nat Cell Biol. 9:347–353. 2007. View Article : Google Scholar

23. 

Shin S and Janknecht R: Activation of androgen receptor by histone demethylases JMJD2A and JMJD2D. Biochem Biophys Res Commun. 359:742–746. 2007. View Article : Google Scholar : PubMed/NCBI

24. 

Coffey K, Rogerson L, Ryan-Munden C, Alkharaif D, Stockley J, Heer R, Sahadevan K, O’Neill D, Jones D, Darby S, Staller P, Mantilla A, Gaughan L and Robson CN: The lysine demethylase, KDM4B, is a key molecule in androgen receptor signalling and turnover. Nucleic Acids Res. 41:4433–4446. 2013. View Article : Google Scholar : PubMed/NCBI

25. 

Pollard PJ, Loenarz C, Mole DR, McDonough MA, Gleadle JM, Schofield CJ and Ratcliffe PJ: Regulation of Jumonji-domain-containing histone demethylases by hypoxia-inducible factor (HIF)-1alpha. Biochem J. 416:387–394. 2008. View Article : Google Scholar : PubMed/NCBI

26. 

Beyer S, Kristensen MM, Jensen KS, Johansen JV and Staller P: The histone demethylases JMJD1A and JMJD2B are transcriptional targets of hypoxia-inducible factor HIF. J Biol Chem. 283:36542–36552. 2008. View Article : Google Scholar

27. 

Kim TD, Oh S, Shin S and Janknecht R: Regulation of tumor suppressor p53 and HCT116 cell physiology by histone demethylase JMJD2D/KDM4D. PLoS One. 7:e346182012. View Article : Google Scholar

28. 

Oh S, Shin S, Lightfoot SA and Janknecht R: 14-3-3 proteins modulate the ETS transcription factor ETV1 in prostate cancer. Cancer Res. 73:5110–5119. 2013. View Article : Google Scholar

29. 

Mooney SM, Grande JP, Salisbury JL and Janknecht R: Sumoylation of p68 and p72 RNA helicases affects protein stability and transactivation potential. Biochemistry. 49:1–10. 2010. View Article : Google Scholar : PubMed/NCBI

30. 

Janknecht R: Regulation of the ER81 transcription factor and its coactivators by mitogen- and stress-activated protein kinase 1 (MSK1). Oncogene. 22:746–755. 2003. View Article : Google Scholar : PubMed/NCBI

31. 

Dowdy SC, Mariani A and Janknecht R: HER2/Neu- and TAK1-mediated up-regulation of the transforming growth factor beta inhibitor Smad7 via the ETS protein ER81. J Biol Chem. 278:44377–44384. 2003. View Article : Google Scholar

32. 

Goel A and Janknecht R: Concerted activation of ETS protein ER81 by p160 coactivators, the acetyltransferase p300 and the receptor tyrosine kinase HER2/Neu. J Biol Chem. 279:14909–14916. 2004. View Article : Google Scholar : PubMed/NCBI

33. 

Papoutsopoulou S and Janknecht R: Phosphorylation of ETS transcription factor ER81 in a complex with its coactivators CREB-binding protein and p300. Mol Cell Biol. 20:7300–7310. 2000. View Article : Google Scholar : PubMed/NCBI

34. 

Knebel J, De Haro L and Janknecht R: Repression of transcription by TSGA/Jmjd1a, a novel interaction partner of the ETS protein ER71. J Cell Biochem. 99:319–329. 2006. View Article : Google Scholar : PubMed/NCBI

35. 

Wu J and Janknecht R: Regulation of the ETS transcription factor ER81 by the 90-kDa ribosomal S6 kinase 1 and protein kinase A. J Biol Chem. 277:42669–42679. 2002. View Article : Google Scholar : PubMed/NCBI

36. 

Rossow KL and Janknecht R: The Ewing’s sarcoma gene product functions as a transcriptional activator. Cancer Res. 61:2690–2695. 2001.

37. 

Mooney SM, Goel A, D’Assoro AB, Salisbury JL and Janknecht R: Pleiotropic effects of p300-mediated acetylation on p68 and p72 RNA helicase. J Biol Chem. 285:30443–30452. 2010. View Article : Google Scholar : PubMed/NCBI

38. 

Goel A and Janknecht R: Acetylation-mediated transcriptional activation of the ETS protein ER81 by p300, P/CAF, and HER2/Neu. Mol Cell Biol. 23:6243–6254. 2003. View Article : Google Scholar : PubMed/NCBI

39. 

Oh S and Janknecht R: Histone demethylase JMJD5 is essential for embryonic development. Biochem Biophys Res Commun. 420:61–65. 2012. View Article : Google Scholar : PubMed/NCBI

40. 

Shin S, Oh S, An S and Janknecht R: ETS variant 1 regulates matrix metalloproteinase-7 transcription in LNCaP prostate cancer cells. Oncol Rep. 29:306–314. 2013.PubMed/NCBI

41. 

DiTacchio L, Bowles J, Shin S, Lim DS, Koopman P and Janknecht R: Transcription factors ER71/ETV2 and SOX9 participate in a positive feedback loop in fetal and adult mouse testis. J Biol Chem. 287:23657–23666. 2012. View Article : Google Scholar : PubMed/NCBI

42. 

Shin S, Kim TD, Jin F, van Deursen JM, Dehm SM, Tindall DJ, Grande JP, Munz JM, Vasmatzis G and Janknecht R: Induction of prostatic intraepithelial neoplasia and modulation of androgen receptor by ETS variant 1/ETS-related protein 81. Cancer Res. 69:8102–8110. 2009. View Article : Google Scholar

43. 

Kaiser S, Park YK, Franklin JL, Halberg RB, Yu M, Jessen WJ, Freudenberg J, Chen X, Haigis K, Jegga AG, Kong S, Sakthivel B, Xu H, Reichling T, Azhar M, Boivin GP, Roberts RB, Bissahoyo AC, Gonzales F, Bloom GC, Eschrich S, Carter SL, Aronow JE, Kleimeyer J, Kleimeyer M, Ramaswamy V, Settle SH, Boone B, Levy S, Graff JM, Doetschman T, Groden J, Dove WF, Threadgill DW, Yeatman TJ, Coffey RJ Jr and Aronow BJ: Transcriptional recapitulation and subversion of embryonic colon development by mouse colon tumor models and human colon cancer. Genome Biol. 8:R1312007. View Article : Google Scholar

44. 

Kurashina K, Yamashita Y, Ueno T, Koinuma K, Ohashi J, Horie H, Miyakura Y, Hamada T, Haruta H, Hatanaka H, Soda M, Choi YL, Takada S, Yasuda Y, Nagai H and Mano H: Chromosome copy number analysis in screening for prognosis-related genomic regions in colorectal carcinoma. Cancer Sci. 99:1835–1840. 2008. View Article : Google Scholar : PubMed/NCBI

45. 

Clevers H and Nusse R: Wnt/beta-catenin signaling and disease. Cell. 149:1192–1205. 2012. View Article : Google Scholar : PubMed/NCBI

46. 

Yap KL and Zhou MM: Keeping it in the family: diverse histone recognition by conserved structural folds. Crit Rev Biochem Mol Biol. 45:488–505. 2010. View Article : Google Scholar : PubMed/NCBI

47. 

Mosimann C, Hausmann G and Basler K: Beta-catenin hits chromatin: regulation of Wnt target gene activation. Nat Rev Mol Cell Biol. 10:276–286. 2009. View Article : Google Scholar

48. 

He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B and Kinzler KW: Identification of c-MYC as a target of the APC pathway. Science. 281:1509–1512. 1998. View Article : Google Scholar : PubMed/NCBI

49. 

Mann B, Gelos M, Siedow A, Hanski ML, Gratchev A, Ilyas M, Bodmer WF, Moyer MP, Riecken EO, Buhr HJ and Hanski C: Target genes of beta-catenin-T cell-factor/lymphoid-enhancer-factor signaling in human colorectal carcinomas. Proc Natl Acad Sci USA. 96:1603–1608. 1999. View Article : Google Scholar

50. 

Shtutman M, Zhurinsky J, Simcha I, Albanese C, D’Amico M, Pestell R and Ben-Ze’ev A: The Cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway. Proc Natl Acad Sci USA. 96:5522–5527. 1999. View Article : Google Scholar : PubMed/NCBI

51. 

Tetsu O and McCormick F: Beta-catenin regulates expression of Cyclin D1 in colon carcinoma cells. Nature. 398:422–426. 1999. View Article : Google Scholar : PubMed/NCBI

52. 

Kouzarides T: Chromatin modifications and their function. Cell. 128:693–705. 2007. View Article : Google Scholar : PubMed/NCBI

53. 

Daujat S, Zeissler U, Waldmann T, Happel N and Schneider R: HP1 binds specifically to Lys26-methylated histone H1.4, whereas simultaneous Ser27 phosphorylation blocks HP1 binding. J Biol Chem. 280:38090–38095. 2005. View Article : Google Scholar : PubMed/NCBI

54. 

Wagner EJ and Carpenter PB: Understanding the language of Lys36 methylation at histone H3. Nat Rev Mol Cell Biol. 13:115–126. 2012. View Article : Google Scholar : PubMed/NCBI

55. 

Eilers M and Eisenman RN: Myc’s broad reach. Genes Dev. 22:2755–2766. 2008.

56. 

Eferl R and Wagner EF: AP-1: a double-edged sword in tumorigenesis. Nat Rev Cancer. 3:859–868. 2003. View Article : Google Scholar : PubMed/NCBI

57. 

Smith DR, Myint T and Goh HS: Over-expression of the c-myc proto-oncogene in colorectal carcinoma. Br J Cancer. 68:407–413. 1993. View Article : Google Scholar : PubMed/NCBI

58. 

Magrisso IJ, Richmond RE, Carter JH, Pross CB, Gilfillen RA and Carter HW: Immunohistochemical detection of RAS, JUN, FOS, and p53 oncoprotein expression in human colorectal adenomas and carcinomas. Lab Invest. 69:674–681. 1993.PubMed/NCBI

59. 

Nateri AS, Spencer-Dene B and Behrens A: Interaction of phosphorylated c-Jun with TCF4 regulates intestinal cancer development. Nature. 437:281–285. 2005. View Article : Google Scholar : PubMed/NCBI

60. 

Gan XQ, Wang JY, Xi Y, Wu ZL, Li YP and Li L: Nuclear Dvl, c-Jun, beta-catenin, and TCF form a complex leading to stabilization of beta-catenin-TCF interaction. J Cell Biol. 180:1087–1100. 2008. View Article : Google Scholar : PubMed/NCBI

61. 

Suto R, Tominaga K, Mizuguchi H, Sasaki E, Higuchi K, Kim S, Iwao H and Arakawa T: Dominant-negative mutant of c-Jun gene transfer: a novel therapeutic strategy for colorectal cancer. Gene Ther. 11:187–193. 2004. View Article : Google Scholar : PubMed/NCBI

62. 

Bartkova J, Lukas J, Strauss M and Bartek J: The PRAD-1/Cyclin D1 oncogene product accumulates aberrantly in a subset of colorectal carcinomas. Int J Cancer. 58:568–573. 1994. View Article : Google Scholar

63. 

Hulit J, Wang C, Li Z, Albanese C, Rao M, Di Vizio D, Shah S, Byers SW, Mahmood R, Augenlicht LH, Russell R and Pestell RG: Cyclin D1 genetic heterozygosity regulates colonic epithelial cell differentiation and tumor number in ApcMin mice. Mol Cell Biol. 24:7598–7611. 2004. View Article : Google Scholar : PubMed/NCBI

64. 

Fu L, Chen L, Yang J, Ye T, Chen Y and Fang J: HIF-1alpha-induced histone demethylase JMJD2B contributes to the malignant phenotype of colorectal cancer cells via an epigenetic mechanism. Carcinogenesis. 33:1664–1673. 2012. View Article : Google Scholar : PubMed/NCBI

65. 

Luo W, Chang R, Zhong J, Pandey A and Semenza GL: Histone demethylase JMJD2C is a coactivator for hypoxia-inducible factor 1 that is required for breast cancer progression. Proc Natl Acad Sci USA. 109:E3367–E3376. 2012. View Article : Google Scholar : PubMed/NCBI

66. 

Kim TD, Shin S, Berry WL, Oh S and Janknecht R: The JMJD2A demethylase regulates apoptosis and proliferation in colon cancer cells. J Cell Biochem. 113:1368–1376. 2012. View Article : Google Scholar : PubMed/NCBI

67. 

Mallette FA, Mattiroli F, Cui G, Young LC, Hendzel MJ, Mer G, Sixma TK and Richard S: RNF8- and RNF168-dependent degradation of KDM4A/JMJD2A triggers 53BP1 recruitment to DNA damage sites. EMBO J. 31:1865–1878. 2012. View Article : Google Scholar : PubMed/NCBI

68. 

Young LC, McDonald DW and Hendzel MJ: Kdm4b histone demethylase is a DNA damage response protein and confers a survival advantage following gamma-irradiation. J Biol Chem. 288:21376–21388. 2013. View Article : Google Scholar

69. 

Slee RB, Steiner CM, Herbert BS, Vance GH, Hickey RJ, Schwarz T, Christan S, Radovich M, Schneider BP, Schindelhauer D and Grimes BR: Cancer-associated alteration of pericentromeric heterochromatin may contribute to chromosome instability. Oncogene. 31:3244–3253. 2012. View Article : Google Scholar

70. 

Toyokawa G, Cho HS, Iwai Y, Yoshimatsu M, Takawa M, Hayami S, Maejima K, Shimizu N, Tanaka H, Tsunoda T, Field HI, Kelly JD, Neal DE, Ponder BA, Maehara Y, Nakamura Y and Hamamoto R: The histone demethylase JMJD2B plays an essential role in human carcinogenesis through positive regulation of cyclin-dependent kinase 6. Cancer Prev Res. 4:2051–2061. 2011. View Article : Google Scholar : PubMed/NCBI

71. 

Dawson MA, Prinjha RK, Dittmann A, Giotopoulos G, Bantscheff M, Chan WI, Robson SC, Chung CW, Hopf C, Savitski MM, Huthmacher C, Gudgin E, Lugo D, Beinke S, Chapman TD, Roberts EJ, Soden PE, Auger KR, Mirguet O, Doehner K, Delwel R, Burnett AK, Jeffrey P, Drewes G, Lee K, Huntly BJ and Kouzarides T: Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature. 478:529–533. 2011. View Article : Google Scholar

72. 

Zuber J, Shi J, Wang E, Rappaport AR, Herrmann H, Sison EA, Magoon D, Qi J, Blatt K, Wunderlich M, Taylor MJ, Johns C, Chicas A, Mulloy JC, Kogan SC, Brown P, Valent P, Bradner JE, Lowe SW and Vakoc CR: RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature. 478:524–528. 2011. View Article : Google Scholar

Related Articles

Journal Cover

2014-April
Volume 44 Issue 4

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Berry WL, Kim T and Janknecht R: Stimulation of β-catenin and colon cancer cell growth by the KDM4B histone demethylase. Int J Oncol 44: 1341-1348, 2014
APA
Berry, W.L., Kim, T., & Janknecht, R. (2014). Stimulation of β-catenin and colon cancer cell growth by the KDM4B histone demethylase. International Journal of Oncology, 44, 1341-1348. https://doi.org/10.3892/ijo.2014.2279
MLA
Berry, W. L., Kim, T., Janknecht, R."Stimulation of β-catenin and colon cancer cell growth by the KDM4B histone demethylase". International Journal of Oncology 44.4 (2014): 1341-1348.
Chicago
Berry, W. L., Kim, T., Janknecht, R."Stimulation of β-catenin and colon cancer cell growth by the KDM4B histone demethylase". International Journal of Oncology 44, no. 4 (2014): 1341-1348. https://doi.org/10.3892/ijo.2014.2279