You are on page 1of 11

Published OnlineFirst August 18, 2011; DOI: 10.1158/0008-5472.

CAN-11-0782

Cancer
Therapeutics, Targets, and Chemical Biology Research

Oncogene AEG-1 Promotes Glioma-Induced


Neurodegeneration by Increasing Glutamate Excitotoxicity
Seok-Geun Lee1, Keetae Kim3, Timothy P. Kegelman3, Rupesh Dash3, Swadesh K. Das3,
Jung Kyoung Choi1, Luni Emdad3,4, Eric L. Howlett3,5, Hyun Yong Jeon3, Zhao Zhong Su3,
Byoung Kwon Yoo3, Devanand Sarkar3,4,5, Sung-Hoon Kim1, Dong-Chul Kang2, and Paul B. Fisher3,4,5

Abstract
Aggressive tumor growth, diffuse tissue invasion, and neurodegeneration are hallmarks of malignant glioma.
Although glutamate excitotoxicity is considered to play a key role in glioma-induced neurodegeneration, the
mechanism(s) controlling this process is poorly understood. Astrocyte elevated gene-1 (AEG-1) is an oncogene
that is overexpressed in several types of human cancers, including more than 90% of brain tumors. In addition,
AEG-1 promotes gliomagenesis, particularly in the context of tumor growth and invasion, 2 primary char-
acteristics of glioma. In the present study, we investigated the contribution of AEG-1 to glioma-induced
neurodegeneration. Pearson correlation coefficient analysis in normal brain tissues and samples from glioma
patients indicated a strong negative correlation between expression of AEG-1 and a primary glutamate
transporter of astrocytes EAAT2. Gain- and loss-of-function studies in normal primary human fetal astrocytes
and T98G glioblastoma multiforme cells revealed that AEG-1 repressed EAAT2 expression at a transcriptional
level by inducing YY1 activity to inhibit CBP function as a coactivator on the EAAT2 promoter. In addition, AEG-
1–mediated EAAT2 repression caused a reduction of glutamate uptake by glial cells, resulting in induction of
neuronal cell death. These findings were also confirmed in samples from glioma patients showing that AEG-1
expression negatively correlated with NeuN expression. Taken together, our findings suggest that AEG-1
contributes to glioma-induced neurodegeneration, a hallmark of this fatal tumor, through regulation of EAAT2
expression. Cancer Res; 71(20); 6514–23. 2011 AACR.

Introduction oligodendroglial, ependymal, and choroid plexus tumors


(2–4). Astrocytomas, composed predominantly of neoplastic
Tumors of the central nervous system (CNS) are the most astrocytes, account for 80% to 85% of all gliomas and are
prevalent solid neoplasms of childhood and the second staged as low grade (grade I) to high grade (grade IV) on the
leading cancer-related cause of death in adults between basis of nuclear atypia, mitotic activity, endothelial hyper-
the ages of 20 and 39 years (1, 2). Gliomas, the most common plasia, and necrosis (4). Glioblastoma multiforme (grade IV
brain tumors of the adult CNS, originate from neuroepithe- astrocytoma; GBM) is an extremely aggressive, invasive, and
lial tissue and are classified morphologically as astrocytic, destructive malignancy with a proliferation rate that is 2 to
5 times faster than grade III tumors (2, 5). Extensive surgical
resection is not curative due to the highly invasive capacity
Authors' Affiliations: 1Cancer Preventive Material Development Research of GBM cells into normal brain parenchyma (3). Moreover,
Center, Institute of Oriental Medicine, College of Oriental Medicine, Kyung glioblastoma multiforme is largely resistant to currently
Hee University, Seoul; 2Ilsong Institute of Life Science, Hallym University, available treatments that are based on cytotoxic approaches
Anyang, Kyonggi-do, Republic of Korea; 3Department of Human and
Molecular Genetics, 4VCU Institute of Molecular Medicine, 5VCU Massey targeting replicating DNA, such as chemotherapy or radio-
Cancer Center, Virginia Commonwealth University, School of Medicine, therapy (6–8). In addition to uncontrolled proliferation and
Richmond, Virginia
diffuse tissue invasion, neurodegeneration is another attri-
Note: Supplementary data for this article are available at Cancer Research bute of malignant gliomas (2, 9–11).
Online (http://cancerres.aacrjournals.org/).
The mechanisms of glioma-induced neurodegeneration are
Corresponding Authors: Paul B. Fisher, Department of Human and
Molecular Genetics, and VCU Institute of Molecular Medicine, Virginia
poorly understood although excitotoxic levels of glutamate
Commonwealth University School of Medicine, Sanger Hall 11-015, 1101 play a key role in this phenomenon (10, 11). Although gluta-
East Marshall Street, Richmond, VA 23298. Phone: 804-828-9632; Fax: mate is a major neurotransmitter implicated in most aspects
804-827-1124; E-mail: pbfisher@vcu.edu or Seok-Geun Lee, Cancer Pre-
ventive Material Development Research Center, Institute of Oriental Med- of normal brain functions, it is a potent neurotoxin at high
icine, College of Oriental Medicine, Kyung Hee University, Seoul, Republic concentration, indicating that glutamate must be constantly
of Korea; E-mail: seokgeun@khu.ac.kr removed for maintenance at a low level (12, 13). The excitatory
doi: 10.1158/0008-5472.CAN-11-0782 amino acid transporters 1 and 2 (EAAT1 and EAAT2), pre-
2011 American Association for Cancer Research. dominantly expressed on astrocytes, are responsible for the

6514 Cancer Res; 71(20) October 15, 2011

Downloaded from cancerres.aacrjournals.org on March 12, 2015. © 2011 American Association for Cancer
Research.
Published OnlineFirst August 18, 2011; DOI: 10.1158/0008-5472.CAN-11-0782

Role of AEG-1 in Glioma-Induced Neurodegeneration

clearance of excitotoxic levels of glutamate from synapses, and glioblastoma) cells were previously described (26, 28). PC-12
an impaired glutamate uptake by glial cells causes widespread (rat pheochromocytoma) cells were purchased from the
neurodegeneration and lethal epilepsy (14–16). Furthermore, American Type Tissue Culture and Collection and cultured
a number of studies have reported that glioma cells release in Dulbecco's modified Eagle medium (DMEM) with 5% FBS
high levels of glutamate, which causes neuronal cell death and and 10% heat-inactivated horse serum at 37 C. The normal
promotes malignant glioma progression (17–20). control sh (NCsh), AEG-1sh-2, and AEG-1sh-4 cell lines were
Astrocyte elevated gene-1 (AEG-1) is a multifunctional established by transfection with control short-hairpin RNA
oncogene that is overexpressed in a variety of human cancers, (shRNA), AEG-1 shRNA #2, and AEG-1 shRNA #4 expression
although it was originally isolated as a novel HIV-1- and TNF- plasmids (SA Biosciences; catalogue no. KH18459H) in T98G
a–induced transcript from primary human fetal astrocytes cells, respectively, and selected with hygromycin.
(PHFA; refs. 21, 22). As a target of Ras, AEG-1 activates multiple
oncogenic signaling pathways including phosphoinositide-3 Recombinant adenovirus, siRNA, and plasmids
kinase (PI3K)–Akt, mitogen-activated protein kinase (MAPK), Both Ad.vec and Ad.AEG-1 have been previously des-
Wnt, and NF-kB involved in regulation of proliferation, inva- cribed (26). Control and YY1 siRNAs were purchased from
sion, chemoresistance, angiogenesis, and metastasis (21, 23– Santa Cruz Biotechnology. The expression plasmids of AEG-1
30). In particular, AEG-1 showed higher expression, compared and AEG-1 deletion mutants tagged with hemagglutinin (HA)
with that in normal brain tissues, in tumors of the CNS, such were described previously (25). The N0 -deletion mutants N1–
as neuroblastoma, glioblastoma multiforme, and oligodendro- N5 include amino acids 71–582, 101–582, 205–582, 232–582,
glioma (28, 30–32). Gain- and loss-of-function studies in and 262–582, respectively. The C0 -deletion mutants C1–C4
glioma cells revealed crucial roles in proliferation and invasive include amino acids 1–513, 1–404, 1–356, and 10-289, respec-
ability of glioma cells (28, 31). In addition, in vivo experiments tively. The 50 -deletion mutants of the human EAAT2 promot-
using orthotopic glioma models confirmed the role of AEG-1 er–reporter (EAAT2Pro) constructs and NF-kB–Luc have
in glioma progression (28, 31). Furthermore, we observed an been described previously (25, 35, 38). The EAAT2Pro-
interesting inverse correlation between the expression levels 954mYY1 and EAAT2Prom-954mNFkB1 constructs were
of AEG-1 and EAAT2. AEG-1 expression is elevated following made using the QuickChange Site-Directed Mutagenesis
HIV-1 and TNF-a treatment of astrocytes, whereas EAAT2 Kit (Stratagene) in the context of the EAAT2Pro-954 con-
expression is downregulated (22, 33–36). In the setting of struct. The sequences used for PCR primers include: EAAT2
glioma progression, AEG-1 gradually increases as astrocytes Pro-954mYY1, 50 -TCGGAGCCCCCGGAGCTCCCCGCCAAG-
evolve into malignant glioma while, in parallel, EAAT2 ex- CATTATCCCCGCG-30 , and EAAT2Prom-954mNFkB1, 50 -
pression decreases (11, 17, 18, 28, 31, 32, 37). Both HIV-1 TCGGAGCCCCCGGAGCTCAAAGCCAAGCGCCATCCCCG-
infection and glioma progression are associated with neuro- CG-30 . The mutated sequences are underlined.
degenerative changes, and glutamate excitotoxicity is one of
the predominant mechanisms mediating neurodegeneration. Western blotting and immunoprecipitation assays
On the basis of these considerations, we presently investigated Whole-cell lysates were prepared, and coimmunoprecipita-
the role of AEG-1 in glioma-induced neurodegeneration with a tion and Western blot analyses were carried out as described
focus on regulation of the glutamate transporter and its previously (25, 38). The antibodies for YY1 (Santa Cruz Bio-
concomitant control of glutamate levels. technology), CBP (Abcam), and EF1a (Upstate) were pur-
chased. Whole-cell lysates from human tissue samples were
Materials and Methods previously described (28).

Tissue array and immunostaining Northern blotting, real-time PCR, and nuclear run-on
Immunofluorescence analyses in human glioma tissue assays
arrays (GL806) obtained from Tissue Array Networks were Total RNA was extracted using the RNeasy Mini Kit (Qia-
conducted as previously described (28). Anti-AEG-1, anti- gen). Real-time PCR (RT-PCR) was conducted using the ABI
EAAT1, and anti-EAAT2 antibodies were described (34, 38) 7900 Fast Real-Time PCR System and TaqMan Gene Expres-
and the anti-NeuN antibody was purchased from Millipore. sion Assays for individual mRNAs (Applied Biosystems). The
Images were captured with a confocal laser scanning nuclei were extracted using NP40 lysis buffer, and nuclear run-
microscope LSM multiphton 510 META (Zeiss), and ana- on assays were conducted (38).
lyzed using ImageJ (NIH). For analyzing localization of AEG-
1, PHFA cells seeded onto 4-well chamber slides were Transient transfection and luciferase assays
transfected with pcDNA, AEG-1, or each AEG-1 deletion Cells were plated in 24-well plates, infected with Ad.vec
construct. Two days later, the cells were fixed and immu- or Ad.AEG-1, and transfected with the indicated plasmids
nostaining was carried out with anti-HA antibody (Covance) and Renilla luciferase plasmid (Promega) together with
as described (27). 20 nmol/L of control or YY1 siRNA using LipofecAMINE
2000 (Invitrogen); luciferase activities were measured
Cell lines using a Dual-Luciferase Reporter Assay Kit (Promega).
Normal PHFAs, human glioma cell lines H4 (neuroglioma), Firefly luciferase activity was normalized by Renilla luci-
T98G (glioblastoma multiforme), and U251-MG (neuronal ferase activity.

www.aacrjournals.org Cancer Res; 71(20) October 15, 2011 6515

Downloaded from cancerres.aacrjournals.org on March 12, 2015. © 2011 American Association for Cancer
Research.
Published OnlineFirst August 18, 2011; DOI: 10.1158/0008-5472.CAN-11-0782

Lee et al.

Electrophoretic mobility shift assays media for 1 day. Cell viability was measured by an MTT
Electrophoretic mobility shift assays (EMSA) were con- assay (24).
ducted as described (38). The sequences of oligonucleotide
used as probe include 50 -CGCCAAGCGCCATCCCCGCG-30 Statistical analysis
(the YY1 binding site is underlined) and the mutant oligonu- Data were presented as mean  SEM and analyzed for
cleotide 50 -CGCCAAGCATTATCCCCGCG-30 (the mutated statistical significance using the unpaired Student's t test.
sequences are in bold type). Pearson correlation coefficient (r) analysis was used to com-
pare gene expressions between 2 genes.
Chromatin immunoprecipitation assays
Chromatin immunoprecipitation (ChIP) assays were con- Results
ducted using the ChIP-IT Kit (Active Motif). The primers for
the human EAAT2 promoter used are as following: sense, 50 - AEG-1 expression negatively correlates with EAAT2
ATCGCTCTCTCGGGGAAGCCA-30 ; antisense, 50 -TAAGCCCT- expression and the number of neuronal cells in glioma
TTAGCGCCTCAA-30 . patients
To examine a possible correlation between the expression
Glutamate uptake assays of AEG-1 and EAAT2 in glioma, we first carried out immu-
Assays to determine glutamate uptake were conducted as nofluorescence staining of AEG-1 and EAAT2 in samples
described (38). from glioma patients using a tissue array containing 35 cases
of glioma and 5 normal brain tissues in duplicate (Fig. 1A–
Cell viability assays C). The specificity of anti-AEG-1 and anti-EAAT2 antibodies
Cells were treated with 1% heat-inactivated horse serum for immunostaining was confirmed by competition with
containing 100 mmol/L of glutamate for 10 or less min- each immunogen (Supplementary Fig. S1). Although the
utes, and the conditioned media were collected. PC-12 cells expression of AEG-1 greatly increased in samples from
(1  104 cells/well) were seeded in 24-well plates, and glioma patients compared with that in normal human
treated with neuronal differentiation media [DMEM with brains, EAAT2 expression significantly decreased (Fig. 1A
1% heat-inactivated horse serum supplemented with and C). A scatter-plot and Pearson correlation coefficient
1 mmol/L di-butyryl cAMP, Sigma; and 50 ng/mL of nerve analysis revealed a strong negative correlation (r ¼ 0.725)
growth factor (NGF), Promega] for 3 to 6 days. Then, the between expression of AEG-1 and EAAT2 (Fig. 1B). However,
PC-12–derived neuron cells were treated with conditioned analysis of expression patterns of EAAT1, another glial

Figure 1. Correlation among expression of AEG-1, EAAT2, and NeuN in samples from glioma patients. A, comparison of AEG-1 and EAAT2 expression
in samples from glioma patients compared with NC. B, correlation between AEG-1 and EAAT2 expression. C, expression of AEG-1, EAAT2, and
NeuN. Scale bar, 100 mm. D, expression of AEG-1, EAAT1, and EAAT2 in 3 NB samples and 6 samples from glioblastoma multiforme patients.
E, quantification and comparison of NeuN expression in samples from glioma patients compared with NC. F and G, correlation between NeuN and EAAT2
(F) or AEG1 (G) expression analyzed using a scatter-plot. Data in graphs represent mean  SD. *, P < 0.01 versus NC. NC, normal cerebrum tissues;
NB, normal brain tissue.

6516 Cancer Res; 71(20) October 15, 2011 Cancer Research

Downloaded from cancerres.aacrjournals.org on March 12, 2015. © 2011 American Association for Cancer
Research.
Published OnlineFirst August 18, 2011; DOI: 10.1158/0008-5472.CAN-11-0782

Role of AEG-1 in Glioma-Induced Neurodegeneration

glutamate transporter, indicated almost no difference negatively regulate EAAT2 expression and glutamate uptake,
between glioma and normal brain tissues and little corre- thereby causing neuronal cell death in patients with glioma.
lation between expressions of AEG-1 and EAAT1 (Supple-
mentary Fig. S2A–C). These results were also confirmed in AEG-1 represses expression of EAAT2
samples from glioblastoma multiforme patients compared To determine whether AEG-1 could repress EAAT2 expres-
with normal brain tissues by Western blot analysis (Fig. 1D). sion, we first confirmed the negative relationship between
These results indicate an inverse correlation between expression of AEG-1 and EAAT2 in PHFA and human glioma
expressions of AEG-1 and EAAT2, but not EAAT1 in glioma. cell lines (Fig. 2A). Then, we analyzed PHFA cells infected with
Dysregulation of EAAT2 causes glutamate excitotoxicity, Ad.vec or Ad.AEG-1. As shown in Figure 2B (left), overexpres-
which is implicated in various types of neurodegenerative sion of AEG-1 significantly reduced EAAT2 expression in
diseases and glioma-induced neurodegeneration (10–14, 17– PHFA. In addition, EAAT2 mRNA was strongly reduced by
20, 37). Accordingly, we further quantified and compared the AEG-1 (Fig. 2B, middle). This reduction in EAAT2 mRNA
expression of AEG-1 and EAAT2 with NeuN, a neuron- expression resulted from decreased transcription, as con-
specific marker in glioma patient samples. NeuN expression firmed using nuclear run-on assays (Fig. 2B, right). These
was decreased in glioma samples compared with normal observations document that AEG-1 negatively regulates
cerebrum tissues, and a scatter-plot of the data and Pearson EAAT2 expression at a transcriptional level. To further confirm
correlation coefficient analysis showed a strong positive the negative regulation of EAAT2 by AEG-1 in glioma, we
correlation (r ¼ 0.798) between expression of EAAT2 and cloned AEG-1 knockdown glioma cell lines (NCsh, AEG-1sh-2,
NeuN (Fig. 1E and F). Furthermore, the comparison analysis and AEG-1sh-4) by stable transfection with a control, AEG-1
revealed a strong negative correlation (r ¼ 0.649) between shRNA-2 or AEG-1 shRNA-4 plasmid in T98G cells, a highly
expression of AEG-1 and NeuN (Fig. 1G), indicating that aggressive glioma cell line with high levels of AEG-1 and low
samples from glioma patients with their higher levels of levels of EAAT2 (as shown in Fig. 2A). RT-PCR and Western
AEG-1 have fewer neurons. These observations and the blot analyses verified AEG-1 knockdown in the clones. AEG-1
negative correlation between AEG-1 and EAAT2 expression knockdown increased EAAT2 expression (Fig. 2C) and recov-
(Fig. 1B) together with previous studies showing that AEG-1 ery of AEG-1 expression in the knockdown cells reduced
has the ability to regulate promoter activity of its target gene EAAT2 expression (Supplementary Fig. S3), confirming that
(21, 25, 31, 34) suggested that AEG-1 in glioma might AEG-1 is a negative regulator of EAAT2. In addition, to

Figure 2. AEG-1 represses EAAT2 expression. A, AEG-1 and EAAT2 expression in various cell lines. B, PHFA cells were infected with multiplicity of
infection (MOI) of 20 for Ad.vec or Ad.AEG-1 and Western blot analysis was carried out with the indicated antibodies, and EF1a served as an internal control
(left). Using total RNAs from the infected cells, Northern blotting assays were conducted, and the levels of GAPDH mRNA served as a control (middle). Nuclear
run-on assays with nuclei from infected cells were conducted, and the transcription rate of GAPDH was used as a control (right). C, real-time PCR (RT-PCR)
analysis of AEG-1 and EAAT2 mRNA expression in NCsh (normal control sh), AEG-1sh-2, or AEG-1sh-4 cells (left). *, P < 0.01 versus NCsh. Cell lysates
from NCsh, AEG-1sh-2, and AEG-1sh-4 were immunoblotted with the indicated antibodies (right). D, Ad.vec- or Ad.AEG-1–infected PHFA cells were
transfected with EAAT2Pro–Luc. Transient transfection and luciferase assays were conducted at least 3 times in triplicate. Data: fold-normalized activity
relative to that of Ad-vec–infected PHFA was taken as 1. *, P < 0.01 versus Ad.vec.

www.aacrjournals.org Cancer Res; 71(20) October 15, 2011 6517

Downloaded from cancerres.aacrjournals.org on March 12, 2015. © 2011 American Association for Cancer
Research.
Published OnlineFirst August 18, 2011; DOI: 10.1158/0008-5472.CAN-11-0782

Lee et al.

investigate the role of AEG-1 in regulating the EAAT2 pro- activators or repressors, depending on cellular binding con-
moter, an EAAT2 promoter–reporter plasmid (EAAT2Pro– text of a target promoter and the presence of other transcrip-
Luc) was transiently transfected into Ad.vec- or Ad.AEG-1– tion factor binding sites in the promoter (39, 40). To determine
infected PHFA cells. AEG-1 potently induced NF-kB activity as which transcription factor was responsible for EAAT2 pro-
previously shown (25; Supplementary Fig. S4), whereas it moter repression, we engineered 2 mutant EAAT2 promoter–
repressed EAAT2 promoter activity (Fig. 2D). Taken together, reporter plasmids containing a site-directed mutation of the
these results show that AEG-1 negatively regulates EAAT2 putative YY1 or NF-kB binding site in the 954 EAAT2
expression at a transcriptional level. promoter construct (mYY1 and mNF-kB). These constructs
were transfected into PHFA cells, and each promoter acti-
YY-1 is responsible for AEG-1–mediated EAAT2 vity was compared with that of the wild-type plasmid
repression 954EAAT2Pro–Luc. As shown in Figure 3B, mutation in
To determine the mechanism(s) by which AEG-1 regulates the YY1 binding site abolished the EAAT2 promoter response
transcription of EAAT2, a series of 50 -deletion mutants of the to AEG-1, whereas mutation in the NF-kB site had no effect.
EAAT2 promoter–reporter construct were transfected into Ad. These experiments indicate that YY1 is responsible for AEG-1–
vec- or Ad.AEG-1–infected PHFA cells. Serial deletions from mediated EAAT2 promoter repression. To further clarify the
964 to 37 showed a similar pattern of AEG-1–mediated role of YY1, we conducted EMSA using a 20-bp double-
repression in promoter activity (Fig. 3A). These results suggest stranded probe containing the YY1 binding site of the EAAT2
that transcription factors binding to the 37/+43 region are promoter. As shown in Figure 3C (lanes 2 and 3), the intensity
capable of regulating a reduction of EAAT2 promoter activity of a DNA–protein complex was significantly higher in Ad.AEG-
in response to AEG-1. Accordingly, we analyzed the 37/+44 1–infected PHFA nuclear extracts in comparison with Ad.vec–
region of the EAAT2 promoter using Transcription Element infected PHFA nuclear extracts. To further characterize this
Search System (www.cbil.upenn.edu/cgi-bin/tess) to identify nucleoprotein complex, competition assays were conducted
putative transcription factor binding sites potentially respon- using an unlabeled probe or a mutant probe containing
sible for AEG-1–mediated repression. This analysis located 2 mutations in the YY1 binding site. As shown in Figure 3C,
putative transcription factor binding sites, YY1 and NF-kB1, the cold probe (lane 4) completely competed with the DNA–
which are well-known transcription factors that function as protein complex, whereas the mutant probe (lane 5) had

Figure 3. YY1 is responsible for AEG-1–mediated EAAT2 repression. A, Ad.vec- or Ad.AEG-1–infected PHFA cells were transfected with each deletion
EAAT2 promoter–reporter construct. Data: fold-normalized activity relative to that of the 954 EAAT2Pro–Luc in Ad-vec–infected PHFA taken as 1. B, the
infected PHFA cells were transfected with EAAT2Pro-954, mNFkB1, or mYY1. Data: fold-normalized activity relative to that of the 954 EAAT2Pro–Luc in
Ad-vec–infected PHFA was taken as 1. C, nuclear extracts from the infected PHFA cells were mixed with the probe containing the YY1 site of EAAT2
promoter in the following order: 1, no extracts; 2, Ad.vec; 3, Ad.AEG-1; 4, Ad.AEG-1 þ cold wild probe (100); 5, Ad.AEG-1 þ unlabeled mutant probe
containing mutated YY1 site (100); 6, Ad.AEG-1 þ anti-YY-1 antibody; and 7, Ad.AEG-1 þ control immunoglobulin G (IgG). D, the infected cells were
cotransfected with EAAT2–Luc and control or YY1 siRNA. Data: fold-normalized activity relative to that of Ad-vec–infected and control siRNA-transected
PHFA was taken as 1. E, the infected and transfected PHFA cell lysates were immunoblotted with the indicated antibodies. All transient transfection
and luciferase assays were conducted at least 3 times in triplicate. Data in graphs present mean  SEM. *, P < 0.01 versus Ad.vec.

6518 Cancer Res; 71(20) October 15, 2011 Cancer Research

Downloaded from cancerres.aacrjournals.org on March 12, 2015. © 2011 American Association for Cancer
Research.
Published OnlineFirst August 18, 2011; DOI: 10.1158/0008-5472.CAN-11-0782

Role of AEG-1 in Glioma-Induced Neurodegeneration

little effect on complex formation. In addition, supershift cells, all 3 of the proteins, AEG-1, YY1, and CBP as a complex,
experiments with an anti-YY1 antibody resulted in a retarded bound to the EAAT2 promoter in Ad.AEG-1–infected PHFA
mobility of the complex, whereas a control immunoglobulin G cells (Fig. 4B). These results were further confirmed in T98G
(IgG) did not (Fig. 3C, lanes 6 and 7). In addition, YY1 siRNA glioma cells that were highly expressing AEG-1. AEG-1, YY1,
inhibited the AEG-1–mediated EAAT2 promoter repression and CBP in NCsh cells were bound to the EAAT2 promoter as a
(Fig. 3D). These results were also confirmed by Western blot complex, but AEG-1 knockdown in these cells abolished AEG-
analysis using PHFA cell lysates treated with Ad.AEG-1 and 1 as well as YY1 association with the EAAT2 promoter
YY1 siRNA (Fig. 3E). In total, these results indicate that AEG-1 (Fig. 4B). These data indicate that AEG-1 increases YY1
increases YY1 binding to the EAAT2 promoter, which binding to the EAAT2 promoter and suggest that AEG-1 might
is responsible for AEG-1–mediated repression of EAAT2 function as a bridge molecule between YY1 and CBP and the
expression. basal transcription machinery, thus facilitating YY1 inhibition
of CBP function as a co-activator on the EAAT2 promoter.
AEG-1 directly interacts with YY-1 and CBP To further clarify these interactions, we analyzed which
AEG-1 physically interacts with a coactivator, cyclic AMP- domain of AEG-1 was responsible for the interactions by using
responsive element binding protein (CREB)-binding protein AEG-1 deletion constructs. As shown in Figure 4C, all C0 -
(CBP), and YY1 interaction with CBP is 1 mechanism by which deletion mutants were associated with both YY1 and CBP as
YY1 negatively or positively regulates gene expression (25, 39). were wild-type AEG-1. However, only the N1 construct among
On the basis of this consideration, we hypothesized that AEG- the N0 -deletion mutants interacted with YY1, and none of the
1 might play a role as a bridge between YY1 and CBP on the N0 -deletion constructs physically interacted with CBP
EAAT2 promoter, causing YY1 to function as a negative (Fig. 4C), suggesting that, in each region, amino acids 1–70
regulator of EAAT2 expression by inhibiting CBP. To examine and 71–100 of AEG-1 are responsible for interaction with CBP
this possibility, we first established whether these proteins and YY1, respectively. We next examined whether these
directly physically interact with each other by immunopre- interactions would be crucial for AEG-1–mediated EAAT2
cipitation assays using Ad.vec- or Ad.AEG-1–infected PHFA repression. As shown in Figure 4D, as expected only wild-
cell lysates. As shown in Figure 4A, anti-HA, anti-YY1, and anti- type AEG-1 repressed the EAAT2 promoter activity whereas
CBP antibodies effectively immunoprecipitated YY1 and CBP, none of N'-deletion constructs induced repression. Intriguing-
AEG-1 and CBP, and AEG-1 and YY1, respectively. To clarify ly, all of the C'-deletion mutants failed to modify EAAT2
whether these associations occur on the EAAT2 promoter, promoter activity (Fig. 4D). A recent study suggested that
PHFA cells were infected with Ad.vec or Ad.AEG-1, and ChIP the predominant nuclear localization signal of AEG-1 is
assays were carried out using either control IgG or anti-HA, located at the end region of the C terminus (amino acids
anti-YY1, or anti-CBP antibodies. Although only CBP was 546–582; ref. 41), which is missing in all of the C0 -deletion
associated with the EAAT2 promoter in Ad.vec–infected PHFA mutants we analyzed. In addition, we confirmed that AEG-1

Figure 4. AEG-1 interacts with CBP and YY-1 on the EAAT2 promoter. A, the Ad.vec (V)- or Ad.AEG-1 (A)-infected PHFA cell lysates were used for
immunoprecipitation (IP) analysis using anti-HA, anti-YY1, or anti-CBP antibody and the same antibodies for immunoblotting. B, the nuclear pellet containing
chromatin isolated from the infected PHFA cells, NCsh, and AEG-1sh-2 cells were immunoprecipitated with control IgG, anti-AEG-1, anti-YY1, or anti-CBP
antibody, and then the eluted DNAs were subjected to PCR. C, PHFA cells were transfected with control vector (pcDNA), full-length AEG-1, or each AEG-1
deletion-expression construct as indicated. The cell lysates were immunoprecipitated and immunoblotted as indicated. D, PHFA cells were cotransfected with
EAAT2–Luc and each AEG-1 deletion mutant. Data (mean  SEM): fold-normalized activity relative to that of PHFA cells transfected with pcDNA (denoted
as "") was taken as 1. *, P < 0.01 versus pcDNA. E, PHFA cells were transfected as indicated. The AEG-1 (HA) and 40 ,6-diamidino-2-phenylindole
staining were evaluated by confocal microscopy. Scale bar, 20 mm. V, Ad.vec; A, Ad.AEG-1.

www.aacrjournals.org Cancer Res; 71(20) October 15, 2011 6519

Downloaded from cancerres.aacrjournals.org on March 12, 2015. © 2011 American Association for Cancer
Research.
Published OnlineFirst August 18, 2011; DOI: 10.1158/0008-5472.CAN-11-0782

Lee et al.

and the N1 mutant were located in the nucleus, but the C1 mate uptake of astrocytes. In addition, AEG-1 knockdown in
mutant that did not contain the nuclear localization signal T98G cells increased the glutamate uptake in these cells
was present in the cytoplasm (Fig. 4E). Specificity of anti-HA (Fig. 5C). Expression of AEG-1 and EAAT2 in each gain-of-
antibody for immunostaining was confirmed by competition function and loss-of-function study was confirmed by West-
with HA peptide (Supplementary Fig. S5). Taken together, ern blot analysis (Supplementary Fig. S6). These results
these results indicate that interactions among AEG-1, YY1, indicate that AEG-1 causes glutamate excitotoxicity. To
and CBP are crucial for AEG-1–mediated EAAT2 repression. examine whether decreased uptake of glutamate causes
neuronal cell death, we cultured PC-12–differentiated rat
AEG-1 promotes glioma-induced neurodegeneration by neuronal cells in conditioned media prepared from Ad.vec-
blocking EAAT2 function or Ad.AEG-1–infected PHFA cells treated with glutamate.
EAAT2 is the predominant glial glutamate transporter in The conditioned media from Ad.vec–infected PHFA cells
the brain and it functions to remove glutamate from the containing intact glial glutamate transporters did not
synapse to prevent excitotoxicity. Dysregulation of EAAT2 cause neuronal cell death. In contrast, the conditioned
causes glutamate excitotoxicity, which is implicated in var- media from Ad.AEG-1–infected cells induced severe neuro-
ious types of neurodegenerative diseases and glioma-in- nal cell death (Fig. 5B). Furthermore, AEG-1 knockdown in
duced neurodegeneration (10–14, 17–20, 37). In addition, glioma cells inhibited glutamate-induced neuronal cell
we found a negative correlation between AEG-1 expression death (Fig. 5D). However, these conditioned media from
and both EAAT2 and NeuN expression in glioma patients glioma cells and AEG-1–overexpressing PHFA cells had no
(Fig. 1). For these reasons, we hypothesized and investigated cytotoxic effect on PHFA cells (Supplementary Fig. S7),
whether AEG-1–mediated EAAT2 repression is essential for indicating that this glutamate excitotoxicity causes cell
glioma-induced neurodegeneration. We first measured glu- death of neurons, but not astrocytes. Considered together
tamate uptake in PHFA cells infected with Ad.vec or Ad.AEG- with results obtained from patient samples shown in Fig-
1 to determine the functional significance of AEG-1–medi- ure 1, these observations indicate that AEG-1 represses
ated EAAT2 repression in astrocytes. As shown in Figure 5A, EAAT2 expression and glutamate uptake, thereby causing
AEG-1 decreased glutamate uptake in PHFA cells, suggesting neuronal cell death in glioma patients. These provocative
that the AEG-1–mediated EAAT2 repression impairs gluta- findings highlight a novel mechanism by which gliomas
induce neurodegeneration as summarized in Figure 6.

Discussion

Brain tumors induce pathogenic changes by rapidly pro-


liferating and invading surrounding normal tissues and by
promoting neuronal cell death through glutamate excitotoxi-
city (10). Consequently, a primary therapeutic focus to limit
brain tumor-induced damage is by inhibiting cancer cell
proliferation and invasion and potentially altering defects
in glutamate homeostasis. To achieve these objectives
requires an enhanced understanding of the genetic and epi-
genetic changes that promote development, progression, and
pathogenesis of brain cancers. Previous studies have docu-
mented that AEG-1 plays crucial roles in malignant glioma
progression, and its expression level significantly correlates
with clinicopathologic stages of glioma (28, 31). In the present
study, we report that AEG-1 expression also significantly
correlates with reduction of EAAT2 expression and neuronal
cells in glioma patient samples. In addition, AEG-1 overex-
pression in glioma impairs glutamate uptake by reducing
EAAT2 expression, a primary glutamate transporter, culmi-
Figure 5. AEG-1 induces neuronal cell death via inhibiting glutamate nating in glioma-induced neurodegeneration. These newer
uptake in glial cells. A, glutamate uptake levels (pmol/mg protein/min) were data when combined with previous studies suggest that
measured in Ad.vec- or Ad.AEG-1–infected PHFA cells. *, P < 0.01 versus
AEG-1 is involved in the majority of features of glioma
Ad.vec. B, the differentiated PC-12 rat neuron cells were treated with
conditioned media prepared from Ad.vec- or Ad.AEG-1–infected PHFA progression, that is, rapid tumor growth, destructive invasion
cells following treatment with 100 mmol/L glutamate. One day later, MTT of surrounding normal brain tissue, and glioma-induced neu-
assays were conducted. C, glutamate uptake levels (pmol/mg protein/min) rodegeneration. Accordingly, we hypothesize that AEG-1
were measured in NCsh and AEG-1sh cells. *, P < 0.01 versus NCsh. D, the could be a primary regulator of glioma progression and thus
differentiated PC-12 neuron cells were treated with conditioned media
prepared from NCsh and AEG-1sh cells following treatment with 100
could be a potential therapeutic target for this fatal disease.
mmol/L glutamate. One day later, MTT assays were conducted. *, P < 0.01 Developing pharmacologic agents and/or small molecule
in MTT assays (B and D) versus Mock-treated cells. drugs that target AEG-1 for extinction would be predicted

6520 Cancer Res; 71(20) October 15, 2011 Cancer Research

Downloaded from cancerres.aacrjournals.org on March 12, 2015. © 2011 American Association for Cancer
Research.
Published OnlineFirst August 18, 2011; DOI: 10.1158/0008-5472.CAN-11-0782

Role of AEG-1 in Glioma-Induced Neurodegeneration

A B
Neuron Neuron

Neurotoxicity

Figure 6. Potential molecular


mechanism by which EAAT2
reduction mediated by AEG-1
promotes neurodegeneration in
human glioma. A, in normal
astrocytes, EAAT2 functions as a Glutamate
primary glutamate transporter. B, Glutamate
in glioma cells, increased AEG-1
negatively regulates EAAT2
expression and induces glutamate
EAAT2
excitotoxicity and neuronal cell
death.
AEG-1 EAAT2
EAAT2
promoter
EAAT2
promoter

to delimit the pathogenesis and toxicity of glioma. In addition, facilitating interaction among NF-kB, CBP, and the basal
restoring glutamate transporter function in glioma might also transcriptional machinery, and therefore, functions as a coac-
provide a means of reducing neuronal damage in patients with tivator facilitating the induction of NF-kB–dependent genes
malignant glioma, particularly when combined with inhibition (25). In the present study, we document that AEG-1 plays a
of AEG-1. critical role as a link between YY1 and CBP on the EAAT2
Gliomas release glutamate at levels that are neurotoxic promoter, causing YY1 to function as a negative regulator of
(10, 11, 17, 20). Clearance of extracellular glutamate is marked- EAAT2 expression by inhibiting CBP. These results indicate
ly impaired in glioma cells compared with that in normal that interactions among AEG-1, YY1, and CBP are crucial for
astrocytes mainly due to a loss of the predominant astroglial AEG-1–mediated EAAT2 repression, and also suggest that
glutamate transporter EAAT2 (10, 16). This glutamate release AEG-1 functions in the nucleus as a bona fide transcriptional
also promotes growth of malignant gliomas (20). We previ- cofactor.
ously reported that TNF-a induces AEG-1 expression, and Excitotoxicity caused by impaired glutamate uptake by
AEG-1 functions as a coactivator for NF-kB by its direct glial cells has been implicated in various neurodegenerative
interaction with p65 (22, 25, 33). However, TNF-a reduces conditions such as ischemia, stroke, epilepsy, amyotrophic
EAAT2 expression in an NF-kB–dependent manner (35, 42). lateral sclerosis, and HIV-associated dementia (41), in psy-
These results suggest that NF-kB might be a signaling path- chiatric disorders like depression and schizophrenia, as well
way in AEG-1–mediated EAAT2 repression. However, we have as in certain forms of pain (12, 14–16). Although most studies
now showed that AEG-1 employs YY1 to repress EAAT2 of AEG-1 have focused on its functions in tumor progression,
expression. TNF-a–mediated NF-kB repression is not a AEG-1 was originally isolated as a HIV-1–inducible gene
general phenomenon in all contexts, because it can also (22, 33), implying a possible role for it in HIV-associated
function as a positive regulator of NF-kB expression, and this dementia. A recent genome-wide association study implicat-
process is poorly understood. We previously observed that ed AEG-1 in migraine (43). In addition, in this article we
TNF-a preferentially recruits N-myc to the EAAT2 promoter document that AEG-1 reduces expression of EAAT2 in astro-
resulting in a repression of NF-kB–mediated EAAT2 promoter cytes, causing neuronal cell death, suggesting that AEG-1
activation, indicating a mechanism by which TNF-a over- might also play crucial roles in neurodegenerative diseases,
comes intrinsic NF-kB–mediated activation through a path- not only in glioma-induced neurodegeneration. We have also
way not involving the direct inhibition of NF-kB (42). Instead, observed that AEG-1 expression is negatively correlated with
we now showed that AEG-1 recruits YY1 to the EAAT2 EAAT2 expression and neuronal cell survival in a transient
promoter resulting in reduction of EAAT2 expression. A focal ischemia animal model (unpublished data), indicating
previous report showed that AEG-1 acts as a bridge molecule its role in ischemia associated with EAAT2 and glutamate

www.aacrjournals.org Cancer Res; 71(20) October 15, 2011 6521

Downloaded from cancerres.aacrjournals.org on March 12, 2015. © 2011 American Association for Cancer
Research.
Published OnlineFirst August 18, 2011; DOI: 10.1158/0008-5472.CAN-11-0782

Lee et al.

excitotoxicity. In this context, our present studies are focused Acknowledgments


on developing a conditional transgenic animal model to
express AEG-1 specifically in astrocytes using the GFAP We thank Professor David J. Volsky (Molecular Virology Division, St. Luke's-
Roosevelt Hospital Center, Columbia University, New York, NY) for providing
and nestin promoters, which would be beneficial in more primary human fetal astrocytes. D. Sarkar is the Harrison Endowed Scholar in
precisely defining AEG-1 functions in astrocytes for examin- the VCU Massey Cancer Center. P.B. Fisher holds the Thelma Newmeyer
Corman Endowed Chair in Cancer Research at the VCU Massey Cancer Center
ing in vivo brain tumor development/progression as well as and is an SWCRF investigator.
for neurodegeneration/glioma-induced neurodegeneration.
In view of the assortment of effects of AEG-1 in the context
Grant Support
of brain tumors, such as glioblastoma multiforme, this gene
provides a viable target not only for delimiting the direct This study was financially supported by the NIH grants R01 CA134721, P01
pathogenesis of brain tumors but also for reducing the CA104177, and P01 NS31492, the Thelma Newmeyer Corman Endowment, the
Samuel Waxman Cancer Research Foundation, and the National Foundation for
indirect toxicity to neurons promoted by defects in glutamate Cancer Research (PB. Fisher); the Goldhirsh Foundation for Brain Cancer
transport observed in glioblastoma multiforme. Further stud- Research, the Dana Foundation, and the McDonnell Foundation (D. Sarkar);
ies are warranted and some are currently in progress to test the National Research Foundation Basic Science Research Program (2010-
0008219; S.G. Lee); and the Medical Research Center Program (2011-0006220;
these possibilities and to develop improved therapies for S.G. Lee and S.H. Kim) of the Korean Ministry of Education, Science and
glioblastoma multiforme and methods for ameliorating its Technology.
pathogenesis. The costs of publication of this article were defrayed in part by the payment
of page charges. This article must therefore be hereby marked advertisement in
accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
Disclosure of Potential Conflicts of Interest
Received March 8, 2011; revised July 11, 2011; accepted July 18, 2011;
No potential conflicts of interest were disclosed. published OnlineFirst August 18, 2011.

References
1. Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J 17. Ye ZC, Sontheimer H. Glioma cells release excitotoxic concentrations
Clin 2010;60:277–300. of glutamate. Cancer Res 1999;59:4383–91.
2. DeAngelis LM. Brain tumors. N Engl J Med 2001;344:114–23. 18. Ye ZC, Rothstein JD, Sontheimer H. Compromised gluta-
3. Nakada M, Nakada S, Demuth T, Tran NL, Hoelzinger DB, Berens ME. mate transport in human glioma cells: reduction-mislocalization
Molecular targets of glioma invasion. Cell Mol Life Sci 2007;64:458– of sodium-dependent glutamate transporters and enhanced
78. activity of cystine-glutamate exchange. J Neurosci 1999;19:
4. Kleihues P, Louis DN, Scheithauer BW, Rorke LB, Reifenberger G, 10767–77.
Burger PC, et al. The WHO classification of tumors of the nervous 19. Marcus HJ, Carpenter KL, Price SJ, Hutchinson PJ. In vivo assess-
system. J Neuropathol Exp Neurol 2002;61:215–25; discussion 26–9. ment of high-grade glioma biochemistry using microdialysis: a study
5. Rao JS. Molecular mechanisms of glioma invasiveness: the role of of energy-related molecules, growth factors and cytokines. J Neu-
proteases. Nat Rev Cancer 2003;3:489–501. rooncol 2010;97:11–23.
6. Brandes AA, Turazzi S, Basso U, Pasetto LM, Guglielmi B, Volpin L, 20. Takano T, Lin JH, Arcuino G, Gao Q, Yang J, Nedergaard M. Gluta-
et al. A multidrug combination designed for reversing resistance to mate release promotes growth of malignant gliomas. Nat Med
BCNU in glioblastoma multiforme. Neurology 2002;58:1759–64. 2001;7:1010–5.
7. Hofer S, Herrmann R. Chemotherapy for malignant brain tumors of 21. Sarkar D, Emdad L, Lee SG, Yoo BK, Su ZZ, Fisher PB. Astrocyte
astrocytic and oligodendroglial lineage. J Cancer Res Clin Oncol elevated gene-1: far more than just a gene regulated in astrocytes.
2001;127:91–5. Cancer Res 2009;69:8529–35.
8. Kappelle AC, Postma TJ, Taphoorn MJ, Groeneveld GJ, van den Bent 22. Su ZZ, Kang DC, Chen Y, Pekarskaya O, Chao W, Volsky DJ, et al.
MJ, van Groeningen CJ, et al. PCV chemotherapy for recurrent Identification and cloning of human astrocyte genes displaying ele-
glioblastoma multiforme. Neurology 2001;56:118–20. vated expression after infection with HIV-1 or exposure to HIV-1
9. Maher EA, Furnari FB, Bachoo RM, Rowitch DH, Louis DN, Cavenee envelope glycoprotein by rapid subtraction hybridization, RaSH. On-
WK, et al. Malignant glioma: genetics and biology of a grave matter. cogene 2002;21:3592–602.
Genes Dev 2001;15:1311–33. 23. Lee SG, Su ZZ, Emdad L, Sarkar D, Fisher PB. Astrocyte elevated
10. Rothstein JD. Paving new pathways. Nat Med 2002;8:938–40. gene-1 (AEG-1) is a target gene of oncogenic Ha-ras requiring phos-
11. Sontheimer H. A role for glutamate in growth and invasion of primary phatidylinositol 3-kinase and c-Myc. Proc Natl Acad Sci U S A
brain tumors. J Neurochem 2008;105:287–95. 2006;103:17390–5.
12. Anderson CM, Swanson RA. Astrocyte glutamate transport: review of 24. Lee SG, Su ZZ, Emdad L, Sarkar D, Franke TF, Fisher PB. Astrocyte
properties, regulation, and physiological functions. Glia 2000;32:1– elevated gene-1 activates cell survival pathways through PI3K-Akt
14. signaling. Oncogene 2008;27:1114–21.
13. Headley PM, Grillner S. Excitatory amino acids and synaptic trans- 25. Sarkar D, Park ES, Emdad L, Lee SG, Su ZZ, Fisher PB. Molecular
mission: the evidence for a physiological function. Trends Pharmacol basis of nuclear factor-kappaB activation by astrocyte elevated gene-
Sci 1990;11:205–11. 1. Cancer Res 2008;68:1478–84.
14. Bunch L, Erichsen MN, Jensen AA. Excitatory amino acid trans- 26. Emdad L, Lee SG, Su ZZ, Jeon HY, Boukerche H, Sarkar D, et al.
porters as potential drug targets. Expert Opin Ther Targets Astrocyte elevated gene-1 (AEG-1) functions as an oncogene
2009;13:719–31. and regulates angiogenesis. Proc Natl Acad Sci U S A 2009;106:
15. Kim K, Lee SG, Kegelman TP, Su ZZ, Das SK, Dash R, et al. Role of 21300–5.
excitatory amino acid transporter-2 (EAAT2) and glutamate in neuro- 27. Yoo BK, Chen D, Su ZZ, Gredler R, Yoo J, Shah K, et al. Molecular
degeneration: Opportunities for developing novel therapeutics. J Cell mechanism of chemoresistance by astrocyte elevated gene-1. Can-
Physiol 2011;226:2484–93. cer Res 2010;70:3249–58.
16. Rothstein JD, Patel S, Regan MR, Haenggeli C, Huang YH, Bergles 28. Emdad L, Sarkar D, Lee SG, Su ZZ, Yoo BK, Dash R, et al. Astrocyte
DE, et al. Beta-lactam antibiotics offer neuroprotection by increasing elevated gene-1: a novel target for human glioma therapy. Mol Cancer
glutamate transporter expression. Nature 2005;433:73–7. Ther 2010;9:79–88.

6522 Cancer Res; 71(20) October 15, 2011 Cancer Research

Downloaded from cancerres.aacrjournals.org on March 12, 2015. © 2011 American Association for Cancer
Research.
Published OnlineFirst August 18, 2011; DOI: 10.1158/0008-5472.CAN-11-0782

Role of AEG-1 in Glioma-Induced Neurodegeneration

29. Yoo BK, Emdad L, Su ZZ, Villanueva A, Chiang DY, Mukhopadhyay expression and function: potential role in neuropathogenesis. J Neu-
ND, et al. Astrocyte elevated gene-1 regulates hepatocellular carci- rovirol 2004;10 Suppl 1:25–32.
noma development and progression. J Clin Invest 2009;119:465–77. 37. de Groot JF, Liu TJ, Fuller G, Yung WK. The excitatory amino acid
30. Lee SG, Jeon HY, Su ZZ, Richards JE, Vozhilla N, Sarkar D, et al. transporter-2 induces apoptosis and decreases glioma growth in vitro
Astrocyte elevated gene-1 contributes to the pathogenesis of neu- and in vivo. Cancer Res 2005;65:1934–40.
roblastoma. Oncogene 2009;28:2476–84. 38. Lee SG, Su ZZ, Emdad L, Gupta P, Sarkar D, Borjabad A, et al.
31. Liu L, Wu J, Ying Z, Chen B, Han A, Liang Y, et al. Astrocyte elevated Mechanism of ceftriaxone induction of excitatory amino acid trans-
gene-1 upregulates matrix metalloproteinase-9 and induces human porter-2 expression and glutamate uptake in primary human astro-
glioma invasion. Cancer Res 2010;70:3750–9. cytes. J Biol Chem 2008;283:13116–23.
32. Xia Z, Zhang N, Jin H, Yu Z, Xu G, Huang Z. Clinical significance of 39. Wang CC, Chen JJ, Yang PC. Multifunctional transcription factor YY1:
astrocyte elevated gene-1 expression in human oligodendrogliomas. a therapeutic target in human cancer? Expert Opin Ther Targets
Clin Neurol Neurosurg 2010;112:413–9. 2006;10:253–66.
33. Su ZZ, Chen Y, Kang DC, Chao W, Simm M, Volsky DJ, et al. 40. Vallabhapurapu S, Karin M. Regulation and function of NF-kappaB
Customized rapid subtraction hybridization (RaSH) gene microarrays transcription factors in the immune system. Annu Rev Immunol
identify overlapping expression changes in human fetal astrocytes 2009;27:693–733.
resulting from human immunodeficiency virus-1 infection or tumor 41. Thirkettle HJ, Girling J, Warren AY, Mills IG, Sahadevan K, Leung H,
necrosis factor-alpha treatment. Gene 2003;306:67–78. et al. LYRIC/AEG-1 is targeted to different subcellular compartments
34. Kang DC, Su ZZ, Sarkar D, Emdad L, Volsky DJ, Fisher PB. Cloning by ubiquitinylation and intrinsic nuclear localization signals. Clin
and characterization of HIV-1-inducible astrocyte elevated gene-1, Cancer Res 2009;15:3003–13.
AEG-1. Gene 2005;353:8–15. 42. Sitcheran R, Gupta P, Fisher PB, Baldwin AS. Positive and negative
35. Su ZZ, Leszczyniecka M, Kang DC, Sarkar D, Chao W, Volsky DJ, et al. regulation of EAAT2 by NF-kappaB: a role for N-myc in TNF-alpha-
Insights into glutamate transport regulation in human astrocytes: controlled repression. EMBO J 2005;24:510–20.
cloning of the promoter for excitatory amino acid transporter 2 43. Anttila V, Stefansson H, Kallela M, Todt U, Terwindt GM, Calafato
(EAAT2). Proc Natl Acad Sci U S A 2003;100:1955–60. MS, et al. Genome-wide association study of migraine implicates
36. Wang Z, Trillo-Pazos G, Kim SY, Canki M, Morgello S, Sharer LR, et al. a common susceptibility variant on 8q22.1. Nat Genet 2010;42:
Effects of human immunodeficiency virus type 1 on astrocyte gene 869–73.

www.aacrjournals.org Cancer Res; 71(20) October 15, 2011 6523

Downloaded from cancerres.aacrjournals.org on March 12, 2015. © 2011 American Association for Cancer
Research.
Published OnlineFirst August 18, 2011; DOI: 10.1158/0008-5472.CAN-11-0782

Oncogene AEG-1 Promotes Glioma-Induced


Neurodegeneration by Increasing Glutamate Excitotoxicity
Seok-Geun Lee, Keetae Kim, Timothy P. Kegelman, et al.

Cancer Res 2011;71:6514-6523. Published OnlineFirst August 18, 2011.

Updated version Access the most recent version of this article at:
doi:10.1158/0008-5472.CAN-11-0782

Supplementary Access the most recent supplemental material at:


Material http://cancerres.aacrjournals.org/content/suppl/2011/08/18/0008-5472.CAN-11-0782.DC1.ht
ml

Cited Articles This article cites by 43 articles, 16 of which you can access for free at:
http://cancerres.aacrjournals.org/content/71/20/6514.full.html#ref-list-1

Citing articles This article has been cited by 9 HighWire-hosted articles. Access the articles at:
http://cancerres.aacrjournals.org/content/71/20/6514.full.html#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, contact the AACR Publications
Department at permissions@aacr.org.

Downloaded from cancerres.aacrjournals.org on March 12, 2015. © 2011 American Association for Cancer
Research.

You might also like