ArticlePDF Available

The Proneural Molecular Signature Is Enriched in Oligodendrogliomas and Predicts Improved Survival among Diffuse Gliomas

PLOS
PLOS ONE
Authors:
  • Emory University / Georgia Institute of Technology

Abstract and Figures

The Cancer Genome Atlas Project (TCGA) has produced an extensive collection of '-omic' data on glioblastoma (GBM), resulting in several key insights on expression signatures. Despite the richness of TCGA GBM data, the absence of lower grade gliomas in this data set prevents analysis genes related to progression and the uncovering of predictive signatures. A complementary dataset exists in the form of the NCI Repository for Molecular Brain Neoplasia Data (Rembrandt), which contains molecular and clinical data for diffuse gliomas across the full spectrum of histologic class and grade. Here we present an investigation of the significance of the TCGA consortium's expression classification when applied to Rembrandt gliomas. We demonstrate that the proneural signature predicts improved clinical outcome among 176 Rembrandt gliomas that includes all histologies and grades, including GBMs (log rank test p = 1.16e-6), but also among 75 grade II and grade III samples (p = 2.65e-4). This gene expression signature was enriched in tumors with oligodendroglioma histology and also predicted improved survival in this tumor type (n = 43, p = 1.25e-4). Thus, expression signatures identified in the TCGA analysis of GBMs also have intrinsic prognostic value for lower grade oligodendrogliomas, and likely represent important differences in tumor biology with implications for treatment and therapy. Integrated DNA and RNA analysis of low-grade and high-grade proneural gliomas identified increased expression and gene amplification of several genes including GLIS3, TGFB2, TNC, AURKA, and VEGFA in proneural GBMs, with corresponding loss of DLL3 and HEY2. Pathway analysis highlights the importance of the Notch and Hedgehog pathways in the proneural subtype. This demonstrates that the expression signatures identified in the TCGA analysis of GBMs also have intrinsic prognostic value for low-grade oligodendrogliomas, and likely represent important differences in tumor biology with implications for treatment and therapy.
Content may be subject to copyright.
The Proneural Molecular Signature Is Enriched in
Oligodendrogliomas and Predicts Improved Survival
among Diffuse Gliomas
Lee A. D. Cooper
1
, David A. Gutman
1
,QiLong
3,4
, Brent A. Johnson
4
, Sharath R. Cholleti
1
, Tahsin Kurc
1
,
Joel H. Saltz
1,2,3
, Daniel J. Brat
1,2,3
, Carlos S. Moreno
1,2,3
*
1 Center for Comprehensive Informatics, Emory University, Atlanta, Georgia, United States of America, 2 Pathology and Laboratory Medicine, Emory University School of
Medicine, Atlanta, Georgia, United States of America, 3 Emory Winship Cancer Institute, Atlanta, Georgia, United States of America, 4 Department of Biostatistics and
Bioinformatics, Emory University, Atlanta, Georgia, United States of America
Abstract
The Cancer Genome Atlas Project (TCGA) has produced an extensive collection of ‘-omic’ data on glioblastoma (GBM),
resulting in several key insights on expression signatures. Despite the richness of TCGA GBM data, the absence of lower
grade gliomas in this data set prevents analysis genes related to progression and the uncovering of predictive signatures. A
complementary dataset exists in the form of the NCI Repository for Molecular Brain Neoplasia Data (Rembrandt), which
contains molecular and clinical data for diffuse gliomas across the full spectrum of histologic class and grade. Here we
present an investigation of the significance of the TCGA consortium’s expression classification when applied to Rembrandt
gliomas. We demonstrate that the proneural signature predicts improved clinical outcome among 176 Rembrandt gliomas
that includes all histologies and grades, including GBMs (log rank test p = 1.16e-6), but also among 75 grade II and grade III
samples (p = 2.65e-4). This gene expression signature was enriched in tumors with oligodendroglioma histology and also
predicted improved survival in this tumor type (n = 43, p = 1.25e-4). Thus, expression signatures identified in the TCGA
analysis of GBMs also have intrinsic prognostic value for lower grade oligodendrogliomas, and likely represent important
differences in tumor biology with implications for treatment and therapy. Integrated DNA and RNA analysis of low-grade
and high-grade proneural gliomas identified increased expression and gene amplification of several genes including GLIS3,
TGFB2, TNC, AURKA, and VEGFA in proneural GBMs, with corresponding loss of DLL3 and HEY2. Pathway analysis highlights
the importance of the Notch and Hedgehog pathways in the proneural subtype. This demonstrates that the expression
signatures identified in the TCGA analysis of GBMs also have intrinsic prognostic value for low-grade oligodendrogliomas,
and likely represent important differences in tumor biology with implications for treatment and therapy.
Citation: Cooper LAD, Gutman DA, Long Q, Johnson BA, Cholleti SR, et al. (2010) The Proneura l Molecular Signature Is Enriched in Oligodendrogliomas and
Predicts Improved Survival among Diffuse Gliomas. PLoS ONE 5(9): e12548. doi:10.1371/journal.pone.0012548
Editor: Maciej Lesniak, The University of Chicago, United States of America
Received May 24, 2010; Accepted August 11, 2010; Published September 3, 2010
Copyright: ß 2010 Cooper et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provide d the original author and source are credited.
Funding: This work was supported in part by contract S09-094 from the National Cancer Institute (NCI) administered through NCI Science Applications
International Corporation (SAIC) Frederick campus, and the C enter for Comprehensive Informatics, Emory University. The funders had no role in study design, data
collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: cmoreno@emory.edu
Introduction
Glioblastoma (GBM) is the most common primary brain tumor,
with 8700 new cases per year in the United States [1]. It is also the
highest grade astrocytoma (WHO grade IV), with a truly dismal
prognosis [2]. Following surgical resection, radiation therapy and
temozolamide chemotherapy–the current therapeutic gold stan-
dard–mean survival is 60 weeks [3]. Perhaps more telling of its
aggressive clinical behavior, survival of patients treated by surgical
resection alone averages 13 weeks [4]. Lower grade gliomas (i.e.
WHO grade II and III) account for an additional 2500 cases/year
and are also ultimately fatal, but have slower growth rates and
longer survival times (3–8 years). With rare exception, patients
with lower grade tumors will eventually die due to progression to
GBM.
The analysis of gene expression patterns in GBMs suggests that
this histologic category may include distinct subtypes. Several
groups have developed approaches for subtyping GBMs by gene
expression signatures [5,6]. Verhaak et al [6] used data from The
Cancer Genome Atlas (TCGA) to describe four distinct subtypes
of GBM (Proneural, Neural, Classic, and Mesenchymal) that are
defined by gene expression signatures following unsupervised
hierarchical clustering. TCGA gene expression subtypes have
moderate similarity to those described by Phillips et al [7], who
defined three transcriptional subtypes (proneural, proliferative,
and mesenchymal). Although the TCGA analysis did not reveal
differences in survival between subtypes, this may reflect the
overall short survival period for this highly malignant neoplasm
[8]. The TCGA data at this point does not contain any lower-
grade gliomas. To assess whether the gene expression patterns
uncovered by TCGA data could be used to segregate or predict
survival of lower-grade gliomas, we applied these gene signatures
to the NCI Repository for Molecular Brain Neoplasia Data
(Rembrandt) dataset, which includes infiltrative gliomas with
diverse histologies and includes grades II, III, and IV. The
Rembrandt dataset used here contains 176 samples with long-term
PLoS ONE | www.plosone.org 1 September 2010 | Volume 5 | Issue 9 | e12548
survival data, including 32 astrocytomas (WHO grades II and III),
and 43 oligodendrogliomas (WHO grades II and III). Here, we
demonstrate that the Proneural gene expression signature as
defined by TCGA is enriched in gliomas with oligodendrogliomal
differentiation. This signature also predicts improved outcome for
oligodendrogliomas. Moreover, we have compared the expression
patterns and DNA copy number of low-grade and high-grade
proneural gliomas and identified genes and pathways associated
with progression of this disease. Genes increased in proneural
GBMs (PN-GBM) at both the DNA and RNA level included
GLIS3, TGFB2, TNC, AURKA, and VEGFA. Network analysis
of these changes in gene expression identified several regulatory
hubs, including GLI1, RUNX2, MYC, BMP2, and NOTCH1.
Materials and Methods
Ethics Statement
All human subjects data was publicly available de-identified
data from The Cancer Genome Atlas project and the Rembrandt
database, and thus, not designated as human subjects research. No
Institutional Review Board approval was required.
Patient Datasets
Microarray and clinical data were acquired in un-normalized
form from the Rembrandt [9] public data repository (https://
caintegrator.nci.nih.gov/rembrandt/) using data available on
November 11, 2009. Clinical data were derived from contributing
center institutional diagnoses at Henry Ford Hospital, UCSF, Lee
Moffitt Cancer Center, Dana Farber Cancer Center, University of
Wisconsin, and NCI and are available as Table S1. Affymetrix
HGU133 Plus 2.0 CEL files were normalized using the robust
multi-array average (RMA) method [10] with default parameters
from the Matlab Bioinformatics Toolbox. Of the 296 samples
downloaded, 176 had associated survival data, 75 were oligoden-
drogliomas or astrocytomas with grades II or III, and 101 were
GBMs. Copy number data was derived using Affymetrix 100K
SNP arrays available from the Rembrandt public data repository.
Of the 176 samples with survival data, 147 have associated SNP
arrays for tumor tissue (85 GBMs, 34 Oligodendrogliomas grades
II and III, and 28 Astrocytomas grades II and III).
Data Analysis
To classify Rembrandt samples within the TCGA classification
schema, Rembrandt data for the probes from the Affymetrix U133
Plus 2.0 GeneChip were mapped to the TCGA class signature
genes using HUGO gene symbol and Entrez gene ID number.
This comparison yielded an intersection of 1486 Affymetrix probe
sets. Classification of the Rembrandt samples was then performed
using prediction analysis for microarrays using the signature gene
class centroids [11].
Comparisons of survival between different classes were
performed using the log-rank test [12] as implemented in
GenePattern 3.0 [13]. Survival was compared between each
expression subtype and all others for all 176 samples, all grade II
and III gliomas, and the subsets of the 32 astrocytomas, and 43
oligodendrogliomas. Unsupervised hierarchical clustering was
performed using uncentered correlation and average linkage with
gene and array centering and normalization using cluster [14] and
javatreeview [15].
Differences in gene expression between subtypes were determined
using the comparative selection marker module of GenePattern 3.0.
Cutoffs for statistical significance were a Benjamini- Hochberg
corrected False Discovery Rate (FDR) ,0.05 and a minimum fold
change .1.8. Statistical significance of Gene Ontology overrepresen-
tation were determined by hypergeometric distribution using the
DAVID database [16] and Ingenuity Pathway Analysis [17]. Gene
Set Enrichment Analysis used the GSEA software [18] with an FDR
cutoff of 0.25. Classes were analyzed using the curated c2.biocar-
ta.v2.5.symbols and c2.kegg.v2.5.symbols databases and the gene
ontology c5.all.v2.5.symbols and c5.bp.v2.5.symbols databases from
the Molecular Signatures Database (MSigDB) at the Broad Institute
[18]. Literature Lab software [19] version 2.9 (Acumenta, Inc.,
Boston, MA) was used to identify statistically significant associations of
gene lists with Pathways and MESH terms, and to compare gene lists
against one another.
Cox proportional hazards (PH) models were used to examine
the association between patient survival and four subtypes after
adjusting for patient age at diagnosis as well as 1p/19q status
whenever applicable. We note that 1p/19q deletion is present only
in Oligodendrogliomas; hence, 1p/19q deletion status was not
adjusted for in cases with Astrocytoma or GBM.
Results
Unsupervised hierarchical cluster analysis of gene expression
data derived from TCGA GBM samples resulted in four distinct
gene expression subtypes: Neural, Proneural, Classical, and
Mesenchymal. This classification utilized an integrated analysis
of three gene expression platforms to identify a set of genes that is
consistently and variably expressed among the TCGA samples.
Expression measurements from the Affymetrix HT-HG-U133A,
the Affymetrix Human Exon 1.0 ST, and a custom Agilent array,
were combined by mapping to a transcript database 11,861 total
genes. Of these, 1740 were reliably expressed across platforms,
with 840 identified as class signature genes by ClaNC analysis
[20]. The full details of the TCGA cluster analysis are available
elsewhere [8].
Total gene expression and clinical survival data were downloaded
from the Rembrandt website (https://caintegrator.nci.nih.gov/
rembrandt/). To map the 176 Rembrandt samples with survival
data to the relevant TCGA molecular subtypes, we extracted the
gene symbols from the 840 gene centroid developed previously [6].
Gene symbols and corresponding entrez gene ID numbers were
used to filter the Rembrandt Affymetrix U133Plus2.0 GeneChip
data, resulting in a total of 1486 probes corresponding to 819 genes.
These data were analyzed by supervised hierarchical clustering
(Figure 1) and visual inspection indicated that the four subtypes are
readily discernible in the Rembrandt dataset. A composite dataset
corresponding to these 819 genes was then assembled including
both the TCGA and Rembrandt datasets. A shrunken centroid
model was trained on the TCGA data using PAM software [11] and
tested by leave-one-out-cross-validation (Table 1, 148/160 cor-
rect = 92.5% classification accuracy). We next applied the centroid
model to the Rembrandt dataset to classify each of the samples to
one of the four molecular subtypes (Table 2 and Figure 1A).
Classification of GBM samples in the Rembrandt dataset mirrored
those previously observed in the TCGA analysis. We observed that
the proneural class dominated the oligodendrogliomas grade II and
III samples (32/43 = 74%), and that the mesenchymal class was
completely absent in the oligodendrogliomas (Figure 1A). Although
the proneural class was also enriched somewhat in astrocytomas, it
was less dramatic, and all gene expression subtypes, including
mesenchymal, were represented. The classic signature was more
common in GBM samples than in grade II and III samples.
Classifications, class probabilities, pathology, and survival data for
all 176 samples included in this analysis are provided in Table S2.
The complete set of clinical data for all of the Rembrandt samples
used in this analysis downloaded in November, 2009 is given in
Proneural Glioma Signature
PLoS ONE | www.plosone.org 2 September 2010 | Volume 5 | Issue 9 | e12548
Table S1. Unsupervised hierarchical clustering of these 176 samples
using these 819 genes shows that they cluster into the four
representative subtypes identified in the TCGA GBM dataset
(Figure 1B).
To examine if there were any differences in survival for the
entire set of 176 classified Rembrandt samples, we performed a
Kaplan-Meier analysis (Figure 1C). The 74 samples in the
Proneural class had significantly better outcome by the log rank
test (p = 1.16e-6). The other three gene expression classes did not
show appreciable survival differences. We next examined whether
gene expression class was predictive of outcome in grade II and
grade III gliomas, including both oligodendrogliomas and
astrocytomas in the analysis. We found that the proneural subtype
predicted significantly better outcome in lower grade gliomas
(p = 0.000265, Figure S1). To determine whether the prognostic
value of the Proneural subtype was independent of histology and
grade (GBM, oligodendroglioma, or astrocytoma), we performed a
similar analysis on each of these subsets. The Proneural subtype
demonstrated improved outcome for oligodendrogliomas (grades
II and III, p = 1.26e-4), but not astrocytomas (grades II and III,
p = 0.3) or GBM (grade IV, p = 0.588). Noushmehr et al [21] has
recently identified a specific subtype of diffuse glioma based on its
CpG methylation pattern that is enriched in the proneural
subtype, in oligodendrogliomas, and in all low-grade gliomas,
which has significantly improved outcome, corroborating our
findings.
We next performed a multivariate Cox Proportional Hazard
analysis of expression subtypes, adjusting for age and 1p/19q
deletion status (Table 3). Only 152 samples could be included in
this analysis, as 24 samples lacked SNP/copy number data for
assessing 1p/19q status. As expected, the classic (HR = 6.25;
p = 0.0100) and neural (HR = 23.28; p = 0.0001) subtypes of
oligodendrogliomas had significantly worse outcome compared
to the proneural subtype. For astrocytomas, the classic subtype
had significantly worse outcome than proneural cases (HR = 8.48;
p = 0.0059), but the mesenchymal and neural subtypes did not
exhibit significantly different outcome from proneural samples.
To make sure that none of the classic or neural oligodendro-
gliomas were misclassified small cell GBMs, we carefully examined
patient age, chromosome 10 loss, and EGFR amplification status.
Of the six classic and five neural oligodenrogliomas, none of the
neural and only two of the classic oligodendrogliomas had high
patient age (.70), chr10 loss, and/or EGFR amplification. We
reanalyzed the survival data excluding these two cases (HF1150
and HF0510), and none of the statistical findings were affected,
except that the p-value for HR comparing classic and proneural
oligodendrogliomas increased to 0.0595 (marginally significant)
from 0.0100 (significant), which is not too surprising given that the
sample size of the oligodendrogliomas was reduced by two.
Gene Expression Analysis
To investigate mechanisms underlying the difference in survival
associated with the Proneural subtype, we performed several
differential gene expression analyses. First we compared the set of
proneural lower grade gliomas (both oligodendrogliomas and
astrocytomas; PN-OA) to proneural GBMs (PN-GBM). In
addition, because of the markedly improved survival of proneural
oligodendrogliomas (PN-Oligo), we compared the gene expression
of PN- Oligo to PN-GBM, proneural astrocytomas (PN-Astro),
neural oligodendrogliomas (N-Oligo), and classic oligodendrogli-
Figure 1. The proneural subtype is enriched in oligodendrogli-
omas and has longer survival. (A) Uns upervised hierarchical
clustering of 176 Rembrandt samples using TCGA classification genes
identifies the four major subtypes: Proneural (cyan), Neural (blue),
Mesenchymal (magenta), and Classic (orange). (B) Kaplan-Meier survival
samples shows the Proneural subtype has significantly better survival for
all cases (n = 176, p = 1.16e-6), and oligodendrogliomas (n = 43, p = 1.26e-
4), but not for astrocytomas (n = 32, p = 0.3), or GBM cases (n = 101,
p = 0.588).
doi:10.1371/journal.pone.0012548.g001
Proneural Glioma Signature
PLoS ONE | www.plosone.org 3 September 2010 | Volume 5 | Issue 9 | e12548
omas (C-Oligo). We also compared proneural oligodendrogliomas
to the combined set of classic and neural oligodendrogliomas.
Using minimum fold-change cutoff of 1.8 and a Benjamini-
Hochberg corrected False Discovery Rate (FDR) ,0.05, we
identified 779 probes differentially regulated between PN-Oligo
and PN-GBM (Table S3), and 576 probes differentially regulated
between PN-OA and PN-GBM (Table S4). Venn diagram analysis
of these two gene sets determined that 508 probes were in
common, 68 were unique to the PN-OA vs PN-GBM comparison,
and 271 were unique to the PN-Oligo vs PN-GBM comparison.
Thus, the union of these comparisons resulted in 847 differentially
expressed probes. A hierarchical clustering of the PN-Oligo, PN-
Astro, and PN-GBM samples using these 847 differentially
expressed probes is shown in Figure 2A. With identical false
discovery and fold-change criteria, we found no significant probes
differentially expressed between PN-Oligo and PN-Astro subtypes.
However, we did observe 1508 probes differentially expressed
between PN-Oligo and N-Oligo (Table S5), 2118 probes
differentially expressed between PN-Oligo and C-Oligo (Table
S6), and 190 probes differentially expressed between PN-Oligo
and the combined set of N-Oligo and C-Oligo (Table S7).
Complete results for these comparisons providing data on all
probes is also available in Tables S3, S4, S5, S6.
Copy Number Analysis
DNA Copy number data for the Rembrandt samples was
calculated using GenePattern version 3.2.0. Segmentation of raw
copy numbers was calculated using the GLAD module (version 2).
Significance analysis of amplifications and deletions was per-
formed using the GISTIC method [22] (version 3), with
amplification/deletion threshold 0.1 and minimum segment size
of four markers.
We identified a number of regions with significant amplifica-
tions or deletions in the PN-GBM and PN-Oligo subtypes
(Figure 2B). It is worth noting that the PN-Oligo samples had
highly significant frequency of co-deletion of chromosomes 1p36
and 19q13. While there were relatively few neural (n = 3) and
classic (n = 5) oligodendrogliomas with SNP data, and no
mesenchymal oligodendrogliomas, we did note that they generally
contained either 1p deletion or 19q deletion, but not both
deletions in any given tumor sample.
Integrated Copy Number and Gene Expression Analysis
To gain insight into the progression of proneural samples, we
performed an integrated analysis of DNA copy number and
mRNA gene expression within the proneural subtype of the
Rembrandt samples, specifically examining those changes between
PN-OA and PN-GBM samples. We observed 66 probe sets
corresponding to 52 genes that showed significant amplification or
deletion, as well as significant differences in gene expression
between these subtypes. A comprehensive list of the mRNA probes
that intersect with GISTIC copy number analysis is given in Table
S8, and a summary of the loci with both expression and copy
number alterations between PN-GBM and PN-OA samples is
provided in Table 4. Among these genes, of particular interest,
were the Hedgehog pathway transcription factor GLIS3 (amplified
at 9p23 and increased in PN-GBM), Meningioma 1 (MN1), RAS-
like 10A (RASL10A), Src homology 2 containing transforming
protein D (SHD), and Dishevelled associated activator of
morphogenesis 2 (DAAM2), all amplified in PN-OA samples
and increased in PN-OA relative to PN-GBM. Genes deleted in
PN-OA samples and downregulated relative to PN-GBM included
Epithelial membrane protein 3 (EMP3), Secreted phosphoprotein
1/Osteopontin (SPP1), and integrin-binding sialoprotein (IBSP).
Also of note was the fact that multiple genes associated with the
complement cascade (C1S, C1R, C1RL, F13A1, and CFI) were
within the set of genes showing both copy number and expression
changes between PN-OA and PN-GBM samples. All of these
complement cascade genes showed higher expression in PN-GBM
samples relative to PN-OA samples.
Secondly, to gain insight into differences in histologic differen-
tiation and survival, we also performed an integrated analysis of
DNA copy number and mRNA gene expression between PN-
Oligo samples and PN-GBM samples in the Rembrandt dataset.
We observed 323 probe sets corresponding to 240 genes that
showed significant amplification or deletion, as well as significant
differences in gene expression between these subtypes. A
comprehensive list of the mRNA probes that intersect with
GISTIC copy number analysis is given in Table S9. Among these
genes, of particular interest, were CD44 (amplified at 11p13 and
increased in PN-GBM), CDKN2C and CDC42 (both amplified in
Table 1. Classification accuracy of subtype predictor on the training set of TCGA samples generated using Prediction Analysis of
Microarrays (PAM) software.
CV Confusion Matrix (Threshold = 2.66891)
True\Predicted Classic Mesenchymal Neural Proneural Class Error rate
Classic 43 4 0 0 0.085
Mesenchymal 2 40 1 0 0.070
Neural 0 1 28 1 0.067
Proneural 1 0 2 37 0.075
Overall prediction accuracy was 148/160 = 92.5% correct.
doi:10.1371/journal.pone.0012548.t001
Table 2. Classification of Rembrandt samples using the TCGA
molecular subtype gene signatures derived from the PAM
classifier.
Subtype GBM Oligodendroglioma Astrocytoma All
Classic 42 6 3 51
Mesenchymal 21 0 5 26
Neural 12 5 8 25
Proneural 26 32 16 74
Total 101 43 32 176
The Proneural subtype dominates oligodendriogliomas and there are no
mesenchymal subtype in the oligodenroglioma samples.
doi:10.1371/journal.pone.0012548.t002
Proneural Glioma Signature
PLoS ONE | www.plosone.org 4 September 2010 | Volume 5 | Issue 9 | e12548
PN-GBM and deleted in PN-Oligo subtypes at 1p36), TGFB2,
TNC, AURKA, and VEGFA, all amplified in PN-GBM, and
SOX8 and Noggin (NOG) amplified in PN-Oligo samples.
Pathway Analysis
We performed four separate types of pathway analysis using gene
set enrichment analysis (GSEA), the Database for Annotation,
Visualization and Integrated Discovery (DAVID), Literature Lab
Analysis (LLA), and Ingenuity Pathway Analysis (IPA). The
comparison of PN-GBM to PN-OA subtypes identified a number
of gene ontologies and pathways that were significantly enriched in
multiple analyses (Table 5). Pathways, ontologies, and biological
processes that were differential between the PN-GBM and PN-OA
subtypes included several annotations associated with increased
vasculature, including blood coagulation, the complement cascade,
vasculature development, wound healing, immune response, and the
IKK-NFkB pathway. In addition, IPA identified significant overrep-
resentation of genes associated with cancer, cell death, proliferation,
glioblastomas, and astrocytomas. An overview of gene expression
differences between PN-OA and PN-GBM samples is shown in a
network diagram (Figure 3A) in which genes increased in PN-GBM
relative to PN-OA are red, and genes decreased in PN-GBM relative
to PN-OA are green. LLA of this gene set identified the
‘Thrombospondin-1 Induced Apoptosis in Microvascular Endothe-
lial Cells’ pathway as the most strongly associated (p = 0.0009) with
the differences between PN-GBM and PN-OA samples.
In addition, to investigate molecular mechanisms underlying
improved survival of the PN-Oligo subtype relative to other types of
oligodendrogliomas, we performed pathway analysis of the 129 genes
corresponding to the 190 probe sets differentially expressed between
PN-Oligo and the Classic and Neural Oligodendrogliomas. As
expected, the differential genes were indicative of the proneural
subtype, with some of the most significant annotations including
Differentiation of Astrocytes, Neurogenesis, Cell Death, Develop-
ment of Blood Vessels, and Benign Tumor (Table S10). A Network
analysis of this set of 129 genes revealed several hubs including GLI1,
RUNX2, MYC, BMP2, and NOTCH1 (Figure 3B). These data
suggest that differences in signaling in the Hedgehog and Notch
pathways likely play an important role in the improved outcomes of
PN-Oligo cases. Moreover, Literature Lab analysis of the gene sets
derived from the integrated expression and copy number analysis
comparing PN-Oligo to PN-GBM identified both the Notch
(p = 0.0098) and Hedgehog (p = 0.0434) pathways as significantly
associated with progression of the proneural subtype.
Discussion
Here we demonstrate that the Proneural gene expression signature
as defined by TCGA is enriched in gliomas with oligodendrogliomal
differentiation. This signature also predicts improved outcome for
oligodendrogliomas. Analysis of the copy number data from the
Rembrandt dataset demonstrated a high frequency of large losses of
chromosomes 1p and 19q in oligodendrogliomas, but not in
astrocytomas or GBMs. Twelve of the 42 oligodendrogliomas
showed co-deletion of at least 85% of chromosomes 1p and 19q,
and eleven of those twelve samples were PN-oligos. The other sample
with co-deletion of 1p/19q was a classic oligodendroglioma.
Nevertheless, our findings regarding the improved outcome of
proneural subtype remained significant in multivariate analyses
controlling for 1p/19q status and age at diagnosis. Multivariate
analysis also showed that the proneural subtype of astroctyoma has
significantly better outcome than the classic subtype of astrocytoma,
suggesting that the proneural gene expression signature carries
prognostic significance across histologic types in the diffuse gliomas.
Our integrated analysis of expression patterns and DNA copy
number of low-grade and high-grade proneural gliomas in the
Rembrandt dataset identified genes and pathways associated with
progression of this disease. Genes increased in high-grade
proneural GBMs (PN-GBM) at both the DNA and RNA level
included GLIS3, TGFB2, TNC, AURKA, and VEGFA. Network
analysis of these gene expression changes identified several
regulatory hubs, including GLI1, RUNX2, MYC, BMP2, and
NOTCH1. The GLI transcription factors are effectors of the
Hedgehog pathway and have been strongly implicated as key
regulators of glioblastoma behavior since their discovery. GLI1
regulates stem cell renewal and tumorigenicity of gliomas [23],
and targeted inhibition of the Hedgehog pathway results in partial
tumor regression in animal models [24]. Tenascin-C (TNC) is
Table 3. Cox proportional hazards (PH) models were used to examine the association between patient survival and four subtypes
after adjusting for patient age at diagnosis as well as 1p/19q status whenever applicable.
All Cases GBM Astrocytomas Oligodendrogliomas
Number of cases 152 101 32 36
Subtype
Classic 1.96 (CI: 1.23, 3.13);
p = 0.0045
1.17 (CI: 0.69, 1.96); p = 0.563 8.48 (CI: 1.85, 38.85); p = 0.0059 6.25 (CI: 1.55, 25.19); p = 0.0100
Mesenchymal 2.68 (CI: 1.51, 4.76);
p = 0.0008
1.56 (CI: 0.85, 2.85); p = 0.150 1.04 (CI: 0.29, 3.76); p = 0.9537 N/A
Neural 2.38 (CI: 1.38, 4.11);
p = 0.0018
1.57 (CI: 0.78, 3.17); p = 0.207 1.91 (CI: 0.72, 5.07); p = 0.1963 23.28 (CI: 4.63, 116.98); p = 0.0001
Age ($5vs,55) 3.14 (CI: 2.12, 4.64);
p
,
0.0001
2.90 (CI: 1.84, 4.55); p
,
0.0001 6.79 (CI: 2.77, 16.66); p
,
0.0001 4.97 (CI: 1.58, 15.70); p = 0.0062
1p/19q (deletion vs. no deletion) 0.42 (CI: 0.16, 1.05);
p = 0.0634
N/A N/A 2.23 (CI: 0.59, 8.46); p = 0.2380
Shown are Hazard Ratios (HRs) plus 95% confidence intervals (CI) and associated p-values. Significant p-values (,0.05) are shown in bold font. For the Cox PH models,
Proneural was used as the reference group for the subtype variable, patient age was dichotomized as ,55 or $55 with ,55 as the reference group, and no deletion in
1p/19q status was used as the reference group. Cox PH multivariate analysis was performed for all cases, cases with Astrocytoma, cases with GBM and cases with
Oligodendrogliomas, respectively. We note that 1p/19q deletion is present only in Oligodendrogliomas; hence, 1p/19q deletion status was not adjusted for in cases with
Astrocytoma or GBM.
doi:10.1371/journal.pone.0012548.t003
Proneural Glioma Signature
PLoS ONE | www.plosone.org 5 September 2010 | Volume 5 | Issue 9 | e12548
induced by TGFb signaling [25,26], and promotes invasion of
glioma cells through matrix metalloproteinase 12 [27].
Both Ingenuity Pathway Analysis and Literature Lab Analysis
identified the Notch pathway as being differentially regulated in low
grade and high-grade proneural gliomas. Several component s of the
Notch pathway, including DLL3 and HEY2 are reduced in PN-
GBM, while NOV/CCN3, which is associated with Notch inhibition
[28,29] was increased. These data suggest that Notch pathway
activity decreases as proneural gliomas progress, which could account
for the abnormal angiogenesis observed in high-grade gliomas, since
Notch activity is essential for coordination with VEGF-stimulated
sprouting [30]. We also observed several genes associated with
angiogenesis and oxidative stress in the analysis of low and high-grade
proneural gliomas. These genes included thrombospondin-1
(THBS1), vascular endothelial growth factor A (VEGFA), angiopoie-
tin 2 (ANGPT2), and mitochondrial superoxide dismutase 2 (SOD2).
Finally, we observed reduced expression of the pro-apoptotic Bcl2
family member, BH3 interacting domain death agonist (BID) in high-
grade proneural gliomas, suggesting that loss of BID expression
promotes survival of these high-grade tumors.
As gliomas progress from lower grade (grades II and III) to
GBM, hypoxia and necrosis develop centrally, while angiogenesis
emerges peripherally. These processes are related, with hypoxia-
inducible factors (e.g. VEGFA) secreted by hypoxic, perinecrotic
tumor cells, resulting in the development of new vessels. It has
been suggested that vascular pathology, including vascular
endothelial apoptosis, vascular occlusion and thrombosis, initiates
the development of central hypoxia and necrosis. Angiopoetin 2
has been implicated in initiating endothelial apoptosis by Tie2
receptor in this setting [31,32,33]. THBS-1, a protein that inhibits
angiogenesis by inducing apoptosis via activation of CD36 in
microvascular endothelial cells, may be relevant to these
mechanisms as well, as we found this pathway to be markedly
upregulated in GBMs compared to lower grade gliomas. The non-
receptor tyrosine kinase FYN is activated by THBS-1 through
CD36, activating the apoptosis inducing proteases like caspase-3
Figure 2. RNA and DNA copy number analysis of low- and high-grade proneural gliomas. (A) Supervised hierarchical clustering of
Proneural GBM, Proneural Oligodendrogliomas, and Proneural Astrocytomas using 847 probes significantly different between these subtypes based
on differential marker analysis (FDR ,5%, 1.8 Fold change). (B) Amplification and Deletion GISTIC Analysis of Proneural GBM and Proneural
Oligodendrogliomas. Affymetrix SNP 6.0 data from the Rembrandt dataset was analyzed for copy number gain and loss using the GISTIC algorithm in
GenePattern 3.0. A total of 26 Proneural GBM and 32 Proneural Oligodendroglioma samples were included in the analysis. No significant copy
number changes were identified in the set of 14 Proneural Astrocytomas.
doi:10.1371/journal.pone.0012548.g002
Proneural Glioma Signature
PLoS ONE | www.plosone.org 6 September 2010 | Volume 5 | Issue 9 | e12548
Table 4. Integrated analysis of differences in gene expression and copy number between Proneural GBM and Proneural
Oligodendroglioma/Astrocytoma samples.
Alteration Locus GISTIC q value Gene Symbols
GBM-Amp 12p13.32 1.21E-03 C1R, C1RL, C1S
GBM-Amp 9p23 1.34E-02 GLIS3, HAUS6
GBM-Amp 9p23 1.34E-02 PLIN2
GBM-Amp 6p21.31 4.30E-02 CLIC1, F13A1
GBM-Amp 6p21.31 4.30E-02 HISTH1C, HISTH2BK, HLA-DQA1, HLA-DQB1, HLA -DRA, HLA-DRB1
OA-Amp 22q11.1 4.69E-04 BCR
OA-Amp 22q11.1 4.69E-04 MN1, RASL10A, SEZ6L
OA-Amp 22q11.1 4.69E-04 SLC25A18
OA-Amp 8q24.22 8.23E-04 FAM84B
OA-Amp 19p13.3 3.10E-03 SHD, SLC1A6
OA-Amp 6p21.2 8.78E-03 DAAM2
OA-Del 19q13.31 1.19E-06 EMP3
OA-Del 4q21.23 8.58E-03 CCDC109B, CFI
OA-Del 4q21.23 8.58E-03 ENPEP, HERC5, IBSP, MLF1IP, SEC24D, SPP1, TDO2
Loci with amplification or deletion and corresponding changes in gene expression in Proneural GBM or Proneura l Oligodendrogliomas/Astrocytomas are shown.
doi:10.1371/journal.pone.0012548.t004
Table 5. Significant Gene Ontology and Pathway Annotations identified for 576 probes corresponding to 395 unique genes
differentially expressed between Proneural Oligodendrogliomas/Astrocytomas and Proneural GBM.
DAVID DAVID GSEA GSEA IPA IPA
GO Term Count FDR Count FDR Count p-value
Response To Wounding 47 1.89E-13 11 5.97E-05
Collagen Fibril Organization 14 1.02E-11 10 1.88E-10
Extracellular Matrix Organization 19 8.33E-09 23 0.16288991 3 1.86E-03
Skeletal System Development 29 2.58E-07 30 1.59E-09
Inflammatory Response 28 1.90E-06 120 1.37E-06
Cell Adhesion 40 3.90E-05 43 1.11E-05
Vasculature Development 21 5.88E-04 16 7.30E-09
Regulation Of Cell Proliferation 38 0.00586091 109 6.41E-11
ECM_Receptor_Interaction 79 0.0247256
Cell_Communication 107 0.0645318 5
Blood_Coagulation 38 0.20890287
Regulation_Of_MAPKKK_Cascade 18 0.14662187
Cancer 166 1.78E-27
Apoptosis 61 2.54E-10
Development 30 1.59E-09
Invasion 37 1.75E-09
Brain Cancer 25 4.04E-08
Cell Cycle Progression 51 5.69E-08
Mitosis 29 1.12E-06
Glioma 13 3.05E-05
Astrocytoma 8 4.23E-04
Proneural GBM samples have increased expression of genes associated with enhanced vascularization, proliferation, invasion, and inflammation. Count = number of
genes identified with each annotation by the three analyses. FDR = false discovery rate. P-value for IPA is based on the hyperge ometric distribution.
doi:10.1371/journal.pone.0012548.t005
Proneural Glioma Signature
PLoS ONE | www.plosone.org 7 September 2010 | Volume 5 | Issue 9 | e12548
and p38 protein kinases. p38 is a mitogen-activated kinase that
also induces apoptosis in some conditions, perhaps through AP-1
activation and the activation of genes that lead to apoptosis.
In summary, our in silico analysis of available molecular datasets
for low and high-grade gliomas has provided new insights into the
survival and progression of the proneural subtype. These findings
demonstrate the power and importance of publicly available
molecular datasets and the potential for future discoveries from
The Cancer Genome Atlas project.
Supporting Information
Table S1 Complete clinical data downloaded from the Re-
mbrandt public data repository (https://caintegrator.nci.nih.gov/
rembrandt/) using data available on November 11, 2009. Clinical
data were derived from contributing center institutional diagnoses
at Henry Ford Hospital, UCSF, Lee Moffitt Cancer Center, Dana
Farber Cancer Center, University of Wisconsin, and NCI.
Found at: doi:10.1371/journal.pone.0012548.s001 (0.15 MB
XLS)
Table S2 Survival and PAM subtype classification data for
Rembrandt samples used in the survival analysis.
Found at: doi:10.1371/journal.pone.0012548.s002 (0.05 MB
PDF)
Table S3 Complete Comparative Marker Selection Results
generated from Rembrandt data comparing low- and high-grade
proneural samples. Data were generated using GenePattern 3.0.
Found at: doi:10.1371/journal.pone.0012548.s003 (7.50 MB
TXT)
Table S4 Complete Comparative Marker Selection Results
generated from Rembrandt data comparing proneural oligoden-
droglioma and proneural GBM samples. Data were generated
using GenePattern 3.0.
Found at: doi:10.1371/journal.pone.0012548.s004 (7.50 MB
TXT)
Table S5 Complete Comparative Marker Selection Results
generated from Rembrandt data comparing proneural oligoden-
droglioma and neural oligodendroglioma samples. Data were
generated using GenePattern 3.0.
Found at: doi:10.1371/journal.pone.0012548.s005 (7.50 MB
TXT)
Table S6 Complete Comparative Marker Selection Results
generated from Rembrandt data comparing proneural oligoden-
droglioma and classic oligodendroglioma samples. Data were
generated using GenePattern 3.0.
Found at: doi:10.1371/journal.pone.0012548.s006 (7.50 MB
TXT)
Table S7 Significant Comparative Marker Selection Results
generated from Rembrandt data comparing proneural oligoden-
droglioma and all other oligodendroglioma samples. Data were
generated using GenePattern 3.0.
Found at: doi:10.1371/journal.pone.0012548.s007 (0.15 MB
XLS)
Figure 3. Network analysis of genes differentially expressed between low- and high-grade proneural gliomas identifies regulatory
hubs. (A) Protein interaction network generated by Ingenuity Pathway Analysis (IPA) of the 576 probes differentially expressed between Proneural
GBM and Proneural Oligodendrogliomas/Astrocytomas. Proteins marked in red are increased in Proneural GBM samples, and those in green are
decreased, relative to Proneural low grade gliomas. Solid lines represent direct interactions, and dashed lines represent indirect interactions. All
interactions are derived on the Ingenuity Knowledgebase, which is extracted from the PubMed literature. (B ) Protein interaction network based on
IPA analysis of 129 genes differentially expressed between PN-Oligodendrogliomas and Classic and Neural Oligodendrogliomas. Network hubs are
readily discernible around the Notch receptor, Myc oncogene, and the GLI1 transcription factor.
doi:10.1371/journal.pone.0012548.g003
Proneural Glioma Signature
PLoS ONE | www.plosone.org 8 September 2010 | Volume 5 | Issue 9 | e12548
Table S8 Integrated RNA and DNA Copy number analysis
comparison of low- and high-grade proneural gliomas.
Found at: doi:10.1371/journal.pone.0012548.s008 (0.19 MB
XLS)
Table S9 Integrated RNA and DNA Copy number analysis
comparison of proneural oligodendrogliomas and proneural GBM
samples.
Found at: doi:10.1371/journal.pone.0012548.s009 (0.15 MB
XLS)
Table S10 Gene Ontology analysis of the 129 genes differen-
tially expressed between proneural oligodendrogliomas and the all
other oligodendrogliomas.
Found at: doi:10.1371/journal.pone.0012548.s010 (0.02 MB
XLS)
Figure S1 Kaplan-Meier Analysis of Low-grade gliomas.
Found at: doi:10.1371/journal.pone.0012548.s011 (0.13 MB TIF)
Acknowledgments
The authors recognize the support of the Emory In Silico Center of
Research Excellence (ISCRE) for performance of these studies.
Author Contributions
Conceived and designed the experiments: TK JHS DB CSM. Performed
the experiments: LC. Analyzed the data: LC DAG QL BAJ SRC DB
CSM. Contributed reagents/materials/analysis tools: JHS. Wrote the
paper: LC DB CSM.
References
1. CBTRUS (2010) CBTRUS Statistical Report: Primary Brain Tumors and
Central Nervous System Tumors Diagnosed in the United States in 2004–2006.
Hinsdale, IL.
2. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK (2007) WHO Classification of
Tumours of the Central Nervous System. 4th ed. Lyon: Intl. Agency for
Research, 309 p.
3. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, et al. (2005)
Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma.
N Engl J Med 352: 987–996.
4. Taveras JM, Thompson HG, Jr., Pool JL (1962) Should we treat glioblastoma
multiforme? A study of survival in 425 cases. Am J Roentgenol Radium Ther
Nucl Med 87: 473–479.
5. Li A, Walling J, Ahn S, Kotliarov Y, Su Q, et al. (2009) Unsupervised analysis of
transcriptomic profiles reveals six glioma subtypes. Cancer Res 69: 2091–2099.
6. Verhaak RGW, Hoadley KA, Purdom E, Wang V, Qi Y, et al. (2010) Integrated
Genomic Analysis Identifies Clinically Relevant Subtypes of Glioblastoma
Characterized by Abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer
Cell 17: 98–110.
7. Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, et al. (2006)
Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern
of disease progr ession, and resemble stages in neurogenesis. Cancer Cell 9:
157–173.
8. TCGA-ResearchN etwork (2008) Comprehensi ve genomic characterization
defines human glioblastoma genes and core pathways. Nature 455: 1061–1068.
9. Madhavan S, Zenklusen JC, Kotliarov Y, Sahni H, Fine HA, et al. (2009)
Rembrandt: helping personalized medicine become a reality through integrative
translational research. Mol Cancer Res 7: 157–167.
10. Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, et al. (2003)
Exploration, normaliz ation, and summaries of high density oligonucleotide array
probe level data. Biostatistics 4: 249–264.
11. Tibshirani R, Hastie T, Narasimhan B, Chu G (2002) Diagnosis of multiple
cancer types by shrunken centroids of gene expression. Proc Natl Acad Sci U S A
99: 6567–6572.
12. Mantel N (1966) Evaluation of survival data and two new rank order statistics
arising in its consideration. Cancer Chemother Rep 50: 163–170.
13. Reich M, Liefeld T, Gould J, Lerner J, Tamayo P, et al. (2006) GenePattern 2.0.
Nat Genet 38: 500–501.
14. Eisen MB, Spellman PT, Brown PO, Botstein D (1998) Cluster analysis and
display of genom e-wide expression patterns. Proc Natl Acad Sci U S A 95:
14863–14868.
15. Saldanha AJ (2004) Java Treeview–extensible visualization of microarray data.
Bioinformatics 20: 3246–3248.
16. Dennis G, Jr., Sherman BT, Hosack DA, Yang J, Gao W, et al. (2003) DAVID:
Database for Annotation, Visualization, and Integrated Discovery. Genome Biol
4: P3.
17. Ingenuity [http://www.ingenuity.com/] last accessed May 18, 2010.
18. Subramanian A, Kuehn H, Gould J, Tamayo P, Mesirov JP (2007) GSEA-P: a
desktop application for Gene Set Enrichment Analysis. Bioinformatics 23:
3251–3253.
19. Febbo PG, Mulligan MG, Slonina DA, Stegmaier K, Di Vizio D, et al. (2007)
Literature Lab: a meth od of automated literature interrogation to infer biology
from microarray analysis. BMC Genomics 8: 461.
20. Dabney AR (2006) ClaNC: point-and-click software for classifying microarrays
to nearest centroids. Bioinformatics 22: 122–123.
21. Noushmehr H, Weisenberger DJ, Diefes K, Phillips HS, Pujara K, et al. (2010)
Identification of a CpG island methylator phenotype that defines a distinct
subgroup of glioma. Cancer Cell 17: 510–522.
22. Beroukhim R, Getz G, Nghiemphu L, Barretina J, Hsueh T, et al. (2007)
Assessing the significance of chromosomal aberrations in cancer: methodology
and application to glioma. Proc Natl Acad Sci U S A 104: 20007–20012.
23. Clement V, Sanchez P, de Tribolet N, Radovanovic I, Ruiz i Altaba A (2007)
HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell
self-renewal, and tumorigenicity. Curr Biol 17: 165–172.
24. Sarangi A, Valadez JG, Rush S, Abel TW, Thompson RC, et al. (2009)
Targeted inhibition of the Hedgehog pathway in established malignant glioma
xenografts enhances survival. Oncogene 28: 3468–3476.
25. Zhao Y, Young SL (1995) TGF-beta regulates expression of tenascin alternative-
splicing isoforms in fetal rat lung. Am J Physiol 268: L173–180.
26. Pearson CA, Pearson D, Shibahara S, Hofsteenge J, Chiquet-Ehrismann R
(1988) Tenascin: cDNA cloning and induction by TGF-beta. Embo J 7:
2977–2982.
27. Sarkar S, Nuttall RK, Liu S, Edwards DR, Yong VW (2006) Tenascin-C
stimulates glioma cell invasion through matrix metalloproteinase-12. Cancer Res
66: 11771–11780.
28. Minamizato T, Sakamoto K, Liu T, Kokubo H, Katsube K, et al. (2007)
CCN3/NOV inhibits BMP-2-induced osteoblast differentiation by interacting
with BMP and Notch signaling pathways. Biochem Biophys Res Commun 354:
567–573.
29. Sakamoto K, Yamaguchi S, Ando R, Miyawaki A, Kabasawa Y, et al. (2002)
The nephroblastoma overexpressed gene (NOV/ccn3) protein associates with
Notch1 extracellular domain and inhibits myoblast differentiation via Notch
signaling pathway. J Biol Chem 277: 29399–29405.
30. Williams CK, Li JL, Murga M, Harris AL, Tosato G (2006) Up-regulation of the
Notch ligand Delta-like 4 inhibits VEGF-induced endothelial cell function.
Blood 107: 931–939.
31. Holash J, Maisonpierre PC, Compton D, Boland P, Alexander CR, et al. (1999)
Vessel cooption, regression, and growth in tumors mediated by angiopoietins
and VEGF. Science 284: 1994–1998.
32. Zagzag D, Amirnovin R, Greco MA, Yee H, Holash J, et al. (2000) Vascular
apoptosis and involution in gliomas precede neovascularization: a novel concept
for glioma growth and angiogenesis. Lab Invest 80: 837–849.
33. Rong Y, Durden DL, Van Meir EG, Brat DJ (2006) ‘Pseudopalisading’ necrosis
in glioblastoma: a familiar morphologic feature that links vascular pathology,
hypoxia, and angiogenesis. J Neuropathol Exp Neurol 65: 529–539.
Proneural Glioma Signature
PLoS ONE | www.plosone.org 9 September 2010 | Volume 5 | Issue 9 | e12548
... When removing IDH-mut gliomas from proneural GBMs, no survival benefit could be seen, instead these tumors had even worse survival than other groups [12,13]. Similarly, classification into proneural, classical and mesenchymal subtypes was shown to be relevant also for gliomas of lower grade, although its clinical implications not as thoroughly studied [8,14,15]. In dLGG, proneural was the most dominant and prognostically favorable subgroup compared to other groups [5,14,15]. ...
... Similarly, classification into proneural, classical and mesenchymal subtypes was shown to be relevant also for gliomas of lower grade, although its clinical implications not as thoroughly studied [8,14,15]. In dLGG, proneural was the most dominant and prognostically favorable subgroup compared to other groups [5,14,15]. ...
... As expected, the IDH-mut dLGGs were mainly of proneural subtype [5,8,14], while IDH-wt gliomas with molecular features of GBM more often were mesenchymal. In line with our findings, previous studies showed that the classical signature is frequent in GBMs, but less common in grade 2 and 3 tumors [14,15]. The proneural dominance in IDH-mut tumors was a confounding factor, explaining the significantly longer survival correlated with this subtype in the total cohort. ...
Article
Full-text available
Objectives Accumulating evidence shows that mesenchymal transition of glioblastomas is associated with a more aggressive course of disease and therapy resistance. In WHO2021-defined adult-type diffuse gliomas of lower grade (dLGG), the transition of the tumor phenotype over time, has not been studied. Most efforts to correlate proneural, classical or mesenchymal phenotype with outcome in dLGG were made prior to the WHO 2021 classification. Here, we set out to investigate if phenotype predicted survival and tumor recurrence in a clinical cohort of dLGGs, re-classified according to the 2021 WHO criteria.Methods Using a TMA-based approach with five immunohistochemical markers (EGFR, p53, MERTK, CD44 and OLIG2), we investigated 183 primary and 49 recurrent tumors derived from patients with previously diagnosed dLGG. Of the 49 relapses, nine tumors recurred a second time, and one a third time.ResultsIn total, 71.0% of all tumors could be subtyped. Proneural was most dominant in IDH-mut tumors (78.5%), mesenchymal more common among IDH-wt tumors (63.6%). There was a significant difference in survival between classical, proneural and mesenchymal phenotypes in the total cohort (p
... Glioblastoma (GBM), the most common and malignant primary brain tumor, is IDH-wild-type, whereas IDH-mutant gliomas are classified as oligodendroglioma or astrocytoma. The majority of IDH-mutant gliomas have a proneural transcriptional phenotype; the gene expression profiles of these cells resemble oligodendrocyte progenitor cells (OPCs) or neural progenitor cells (NPCs) [2][3][4]. Therefore, OPCs are considered a likely cell of origin for IDH-mutant gliomas [5][6][7][8]. While the cell of origin in gliomagenesis is still disputed, the proliferation ability, distribution, and abundance in the adult brain further implicate OPCs in this process [9][10][11][12][13][14]. ...
Article
Full-text available
Isocitrate Dehydrogenase-1 (IDH1) is commonly mutated in lower-grade diffuse gliomas. The IDH1R132H mutation is an important diagnostic tool for tumor diagnosis and prognosis; however, its role in glioma development, and its impact on response to therapy, is not fully understood. We developed a murine model of proneural IDH1R132H-mutated glioma that shows elevated production of 2-hydroxyglutarate (2-HG) and increased trimethylation of lysine residue K27 on histone H3 (H3K27me3) compared to IDH1 wild-type tumors. We found that using Tazemetostat to inhibit the methyltransferase for H3K27, Enhancer of Zeste 2 (EZH2), reduced H3K27me3 levels and increased acetylation on H3K27. We also found that, although the histone deacetylase inhibitor (HDACi) Panobinostat was less cytotoxic in IDH1R132H-mutated cells (either isolated from murine glioma or oligodendrocyte progenitor cells infected in vitro with a retrovirus expressing IDH1R132H) compared to IDH1-wild-type cells, combination treatment with Tazemetostat is synergistic in both mutant and wild-type models. These findings indicate a novel therapeutic strategy for IDH1-mutated gliomas that targets the specific epigenetic alteration in these tumors.
... On the other hand, gliomas are characterized by substantial heterogeneity among tumors, underscored by the identification of several molecular subtypes (classical, mesenchymal, neural, and proneural) [31]. Since proneural gliomas are always associated with better survival than other subtypes [32], [33], we compared sensitivity scores among molecular subtypes of LGG patients to examine whether the proneural patients possess potentially more backup resources for the IGF-1R inhibition. As LGG patients are all missing information on molecular subtypes in the TCGA database, the drug sensitivity exploration was hence focused on LGG patients from the CGGA database. ...
Article
Full-text available
Increasing evidence suggests that communication between tumor cells (TCs) and tumor-associated macrophages (TAMs) plays a substantial role in promoting progression of low-grade gliomas (LGG). Hence, it is becoming critical to model TAM-TC interplay and interrogate how the crosstalk affects prognosis of LGG patients. This paper proposed a translational research pipeline to construct the multicellular interaction gene network (MIGN) for identification of druggable targets to develop novel therapeutic strategies. Firstly, we selected immunotherapy-related feature genes (IFGs) for TAMs and TCs using RNA-seq data of glioma mice from preclinical trials. After translating the IFGs to human genome, we constructed TAM- and TC- associated networks separately, using a training set of 524 human LGGs. Subsequently, clustering analysis was performed within each network, and the concordance measure K-index was adopted to correlate gene clusters with patient survival. The MIGN was built by combining the clusters highly associated with survival in TAM- and TC-associated networks. We then developed a MIGN-based survival model to identify prognostic signatures comprised of ligands, receptors and hub genes. An independent cohort of 172 human LGG samples was leveraged to validate predictive accuracy of the signature. The areas under time-dependent ROC curves were 0.881, 0.867, and 0.839 with respect to 1-year, 3-year, and 5-year survival rates respectively in the validation set. Furthermore, literature survey was conducted on the signature genes, and potential clinical responses to targeted drugs were evaluated for LGG patients, further highlighting potential utilities of the MIGN signature to develop novel immunotherapies to extend survival of LGG patients.
... 75 Purvalanol A has shown synergism with chemotherapeutics such as paclitaxel to suppress the growth of Hela cells in part through reducing the expression of Bcl-2 anti-apoptotic proteins. [76][77][78] The anti-cancer function of this CDK inhibitor is demonstrated in vitro by inducing apoptosis in MDA-MB-231 and MCF-7 BC cell lines. 75 SNS-032 SNS-032, also known as BMS 387032 and developed by Sunesis, has a thiazole unit and selectively inhibits CDK2, 7, and 9. 79 The comparison of this CDK inhibitor with flavopiridol represented its lower toxicity, higher suppression of transcription, and higher apoptosis induction. ...
Article
Background: Despite extensive attempts for the treatment of breast cancer (BC), it is the most prevalent cancer type among women, and its treatment remains elusive, particularly in patients with advanced disease. Although there are several therapeutic options, none of them is effective for complete relief, especially in metastatic patients. Cancer cells exhibit a high proliferation rate, which is usually associated with the dysregulation of cell cycle progress. Various proteins such as cyclin-dependent kinases (CDKs) and cyclins are involved in cell cycle modulation. Methods: Databases including PubMed, Scopus, WebofScience and Google Scholar were used to extract information. Articles published in English until 2022 were used. Results: Regarding the dysregulation of various CDKs in several cancer types, the pharmacologicalinhibitors of CDKs have extensively been evaluated to treat several cancer types such as BC. The blockade of CDKs strongly suppresses tumor growth through cell cycle arrest. Moreover, the combination of CDK inhibitors and other anti-cancer therapeutics has demonstrated potent synergistic effects on the treatment of various cancers. Conclusion: Therefore, various CDK inhibitors have been designed and evaluated as antiproliferative therapeutics to suppress the proliferation of cancer cells. Pan CDK inhibitors, including flavopiridol, dinaciclib, purvalanol A, SNS-032, and roscovitine, are the most effective CDK inhibitors investigated in several studies. They inhibit various CDKs such as the CDK1, 2, 4, 5, 6, 7, and 9. In this review, it is attempted to discuss the efficacy of Pan CDK inhibitors as an anti-cancer therapy in BC.
... The expression pattern of GLIS3 varies significantly in different types of cancers. GLIS3 is detected in the highly proliferative group of central neurological tumors such as ventricular meningioma and cerebral glioblastoma (19,20). In contrast, reduced GLIS3 expression was observed in chromophobe renal cell carcinoma (21). ...
Article
Full-text available
Background: Gastric cancer is the most prevalent solid tumor form. Even after standard treatment, recurrence and malignant progression are nearly unavoidable in some cases of stomach cancer. GLIS Family Zinc Finger 3 (GLIS3) has received scant attention in gastric cancer research. Therefore, we sought to examine the prognostic significance of GLIS3 and its association with immune infiltration in gastric cancer. Method: Using public data from The Cancer Genome Atlas (TCGA), we investigated whether GLIS3 gene expression was linked with prognosis in patients with stomach cancer (STAD). The following analyses were performed: functional enrichment analysis (GSEA), quantitative real-time PCR, immune infiltration analysis, immunological checkpoint analysis, and clinicopathological analysis. We performed functional validation of GLIS3 in vitro by plate cloning and CCK8 assay. Using univariate and multivariate Cox regression analyses, independent prognostic variables were identified. Additionally, a nomogram model was built. The link between OS and subgroup with GLIS3 expression was estimated using Kaplan-Meier survival analysis. Gene set enrichment analysis utilized the TCGA dataset. Result: GLIS3 was significantly upregulated in STAD. An examination of functional enrichment revealed that GLIS3 is related to immunological responses. The majority of immune cells and immunological checkpoints had a positive correlation with GLIS3 expression. According to a Kaplan-Meier analysis, greater GLIS3 expression was related to adverse outcomes in STAD. GLIS3 was an independent predictive factor in STAD patients, as determined by Cox regression (HR = 1.478, 95%CI = 1.478 (1.062-2.055), P=0.02). Conclusion: GLIS3 is considered a novel STAD patient predictive biomarker. In addition, our research identifies possible genetic regulatory loci in the therapy of STAD.
... 25,26) Some research findings revealed that GLIS3 is highly expressed and may involve in the carcinogenesis and progression of a few cancers. [31][32][33][34] Besides that, one previous study revealed that their gene-set enrichment analysis (GSEA) displayed an association of GLIS3 with BC. 35) Accordingly, the relationship between GLIS3 and its partners may bring TNBC a new probability in the progression of therapeutic strategies. ...
Article
Triple-negative breast cancer (TNBC) puts a great threat to women’s health. GLIS family zinc finger 3 (GLIS3) belongs to the GLI transcription factor family and acts as a critical factor in cancer progression. Nevertheless, the part of GLIS3 played in TNBC is not known. Immunohistochemical (IHC) staining analysis displayed that GLIS3 was highly expressed in TNBC tissues. The effect of GLIS3 on the malignant phenotype of TNBC was tested in two different cell lines according to GLIS3 regulation. Upregulation of GLIS3 promoted the proliferation, migration, and invasion of TNBC cell lines, whereas the knockdown of GLIS3 suppressed these tumor activities. Inhibition of GLIS3 induced TNBC cell apoptosis. Furthermore, study as immunofluorescence and electrophoretic mobility shift assay confirmed that the nuclear factor-κB (NF-κB) signaling pathway activated by GLIS3 played an important role in TNBC cells’ malignant phenotype. In conclusion, the present work demonstrated that GLIS3 acts as a crucial element in TNBC progression via activating the NF-κB signaling pathway. Accordingly, above mentioned findings indicated that modulation of GLIS3 expression is a potential tactic to interfere with the progression of TNBC. Fullsize Image
... Qi et al. discovered that CAPG was the candidate biomarker of GBM [38]. CAPG was identified to be up-regulated in glioblastoma cell lines in previous study [26] and GLIS3 was found to be an overexpressed gene in GBM tissues [39]. GEPIA data also indicated that CAPG and GLIS3 were up-regulated in GBM tissues relative to deceased normal tissues ( Fig. 2A). ...
Article
Full-text available
Background Glioblastoma (GBM) is an aggressive and malignant brain tumor with extremely poor prognosis. Despite advances in treatment, the pathogenesis of GBM remains elusive. Mounting studies have revealed the critical role of circular RNAs (circRNAs) in the development and progression of human cancers including GBM, but the comprehension of their functions is still insufficient. In this study, we investigated the expression profile of a circRNA derived from GLIS family zinc finger 3 (GLIS3) in GBM and normal astrocytes. CircGLIS3 expression was detected through quantitative real-time polymerase chain reaction (qRT-PCR) analysis. Functional experiments were performed to analyze the influence of circGLIS3 on GBM cell proliferation and apoptosis. In addition, mechanism assays were to uncover the potential regulatory mechanism of circGLIS3. Results CircGLIS3 was up-regulated in GBM cells and knockdown of circGLIS3 significantly hampered proliferation and promoted apoptosis of GBM cells. Furthermore, circGLIS3 positively regulated CAPG and GLIS3 by sponging miR-449c-5p to affect GBM cell proliferation and apoptosis. Conclusions In summary, our study identified that circGLIS3 could promote proliferation and inhibit apoptosis of GBM cells via targeting miR-449c-5p/GLIS3/CAPG axis in vitro. This study could offer a novel molecular perspective for further investigation into mechanisms essential to GBM progression.
... Primary and secondary GBMs also have significantly different histories, and dissimilar age and gender distribution (41). Low-grade gliomas such as diffuse astrocytoma, oligodendroglioma and oligoastrocytoma display proneural signatures, further establishing that secondary GBM tumours likely arise from a common precursor cell (41,70). The parallels in phenotype, histologic appearance, and mortality rates of primary and secondary GBM suggest that their malignancy is better described by similarities in gene expression rather than by differences. ...
Thesis
Full-text available
Glioblastoma multiforme (GBM) represents the most malignant incarnation of glial tumours – a World Health Organisation (WHO) grade IV brain malignancy. GBM is the most common primary brain tumour in adults, accounting for 78% of all malignant central nervous system (CNS) tumours, and affecting 2-3 people per 100,000 in Europe and North America, with an average survival of only 14.6 months. Despite continued research and incremental advances in imaging, surgery, and chemoradiotherapy, patient survival has stagnated in the past decade, with several promising lines of investigation failing to fully deliver on their anticipated translational outcomes. Recent advances in genetic sequencing and computational biology have allowed the simultaneous comparison of large numbers of patient cancer cell genomes and identified several GBM subtypes. It is hoped that such stratification will one day allow clinicians to tailor treatments specific to each GBM subtype as has already happened in cancers like medulloblastoma. However, despite best efforts, GBM remains highly aggressive, infiltrative, and treatment-resistant, rendering it incurable by current treatment modalities. Invasion of tumour cells into normal brain prohibits a surgical cure, while a high cancer stem cell (CSC) component resists treatment with radiation and temozolomide (TMZ) – both of which are more effective against rapidly dividing cells – and relapse remains the rule. Molecular mechanisms underlie GBM’s treatment resistance, and elucidating the key drivers that garner inherent resistance to the quiescent, stem-like fraction of cells that lead to treatment failure therefore presents as an exciting area of research that may uncover new potential drug targets that improve patient outcomes. This study has shown that the proliferation rate of GBM cells is spectral, approximating a positively skewed normal distribution, with highly proliferative cells at one end and quiescent cells at the other. The quiescent cell fraction was subsequently shown to be inherently more resistant to chemoradiotherapy than the proliferative fraction. The quiescent fraction also displayed increased size, complexity, rates of migration and invasion, secretion of extracellular matrix-degrading enzymes, and invadopodia activity than their proliferative counterparts. Similarly, quiescent cells proliferated slower as intracranial tumours but displayed significantly greater invasive properties than a subset of proliferative cells grown in vivo. mRNA expression analysis revealed the genetic signature that underpins the disparity in proliferation rate between quiescent and proliferative cells, and the putative genes which are responsible for the malignant properties identified in both populations. This body of work has uncovered the inherently dichotomous treatment response of quiescent and rapidly dividing GBM cells, as well as the difference in their abilities to migrate and invade. This study has also shed light on the fundamental molecular mechanisms that are at the root of treatment resistance and malignancy in this disease. It is hoped that this expression signature will help to inform future studies and treatments that target these differences and make GBM less of a death sentence and more of manageable, chronic disease.
... CircGLIS3 is originated from gene GLIS3, which encodes a nuclear protein with dual transcriptional influence. A few studies have referred to the correlation of the GLIS3 transcriptional level with higher grade of gliomas and with poor outcome of ependymoma (Lukashova-v et al., 2007;Cooper et al., 2010), but the function and mechanism have not been discovered. An abnormal expression of circGLIS3 in HGG was only mentioned by Song et al. (2016). ...
Article
Full-text available
High-grade glioma is highly invasive and malignant, resistant to combined therapies, and easy to relapse. A better understanding of circular RNA (circRNA) biological function in high-grade glioma might contribute to the therapeutic efficacy. Here, a circRNA merely upregulated in high-grade glioma, circGLIS3 (hsa_circ_0002874, originating from exon 2 of GLIS3 ), was validated by microarray and Real-time quantitative reverse transcription PCR (qRT-PCR). The role of circGLIS3 in glioma was assessed by functional experiments both in vitro and in vivo . Fluorescence in situ hybridization (FISH), RNA pull-down, RNA immunoprecipitation (RIP), and immunohistochemical staining were performed for mechanistic study. Cocultured brain endothelial cells with glioma explored the role of exosome-derived circGLIS3 in the glioma microenvironment. We found that upregulation of circGLIS3 promoted glioma cell migration and invasion and showed aggressive characteristics in tumor-bearing mice. Mechanistically, we found that circGLIS3 could promote the Ezrin T567 phosphorylation level. Moreover, circGLIS3 could be excreted by glioma through exosomes and induced endothelial cell angiogenesis. Our findings indicate that circGLIS3 is upregulated in high-grade glioma and contributes to the invasion and angiogenesis of glioma via modulating Ezrin T567 phosphorylation.
Article
GLIS3 is highly expressed in multiple cancers, but it has not been studied in gastric adenocarcinoma (GAC). Based on bioinformatics analysis, the prognostic significance of GLIS3 in GAC was analyzed. GAC cells were transfected with small interfering (si)-GLIS3 and GLIS3 overexpression plasmid as well as treated with SB505124 [an inhibitor for transforming growth factor beta receptor 1 (TGFβR1)] and dorsomorphin [an inhibitor for bone morphogenetic protein receptor 1 (BMPR1)]. The GLIS3 expression was detected using qRT-PCR. The impacts of GLIS3 on the proliferation, invasion and migration of GAC cells were measured using cell function assays. The activation of phosphor (p)-Smad1/5 was tested by immunofluorescence. Western blot was utilized to measure the level of transforming growth factor (TGF)-β1/Smad1/5 signaling pathway-related proteins (TGF-β1, p-Smad1, Smad1, p-Smad5, Smad5). GLIS3 was expressed at high levels in GAC tissues and cell lines and its high expression could indicate the poor prognosis of GAC patients. GLIS3 inhibition declined the proliferative, invasive and migratory capabilities as well as TGF-β1 expression and phosphorylation of Smad1/5 in GAC cells. Overexpressed GLIS3 promoted proliferation, migration, invasion, TGF-β1 expression and Smad1/5 phosphorylation in GAC cells, with SB505124 reversing the effects of overexpressed GLIS3 on proliferation, migration, invasion and Smad1/5 phosphorylation whereas dorsomorphin exhibiting no influence on GLIS3-induced effects. GLIS3 facilitated the malignant phenotype of GAC cells via regulating TGF-β1/TGFβR1/Smad1/5 pathway, which may be a novel prognostic indicator of GAC and provided a target for GAC treatment.
Article
Full-text available
Background: Functional annotation of differentially expressed genes is a necessary and critical step in the analysis of microarray data. The distributed nature of biological knowledge frequently requires researchers to navigate through numerous web-accessible databases gathering information one gene at a time. A more judicious approach is to provide query-based access to an integrated database that disseminates biologically rich information across large datasets and displays graphic summaries of functional information. Results: Database for Annotation, Visualization, and Integrated Discovery (DAVID; http://www.david.niaid.nih.gov) addresses this need via four web-based analysis modules: 1) Annotation Tool - rapidly appends descriptive data from several public databases to lists of genes; 2) GoCharts - assigns genes to Gene Ontology functional categories based on user selected classifications and term specificity level; 3) KeggCharts - assigns genes to KEGG metabolic processes and enables users to view genes in the context of biochemical pathway maps; and 4) DomainCharts - groups genes according to PFAM conserved protein domains. Conclusions: Analysis results and graphical displays remain dynamically linked to primary data and external data repositories, thereby furnishing in-depth as well as broad-based data coverage. The functionality provided by DAVID accelerates the analysis of genome-scale datasets by facilitating the transition from data collection to biological meaning.
Article
The fourth edition of the World Health Organization (WHO) classification of tumours of the central nervous system, published in 2007, lists several new entities, including angiocentric glioma, papillary glioneuronal tumour, rosette-forming glioneuronal tumour of the fourth ventricle, papillary tumour of the pineal region, pituicytoma and spindle cell oncocytoma of the adenohypophysis. Histological variants were added if there was evidence of a different age distribution, location, genetic profile or clinical behaviour; these included pilomyxoid astrocytoma, anaplastic medulloblastoma and medulloblastoma with extensive nodularity. The WHO grading scheme and the sections on genetic profiles were updated and the rhabdoid tumour predisposition syndrome was added to the list of familial tumour syndromes typically involving the nervous system. As in the previous, 2000 edition of the WHO ‘Blue Book', the classification is accompanied by a concise commentary on clinico-pathological characteristics of each tumour type. The 2007 WHO classification is based on the consensus of an international Working Group of 25 pathologists and geneticists, as well as contributions from more than 70 international experts overall, and is presented as the standard for the definition of brain tumours to the clinical oncology and cancer research communities world-wide
Article
Glioblastoma (GBM) is a highly malignant, rapidly progressive astrocytoma that is distinguished pathologically from lower grade tumors by necrosis and microvascular hyperplasia. Necrotic foci are typically surrounded by "pseudopalisading" cells-a configuration that is relatively unique to malignant gliomas and has long been recognized as an ominous prognostic feature. Precise mechanisms that relate morphology to biologic behavior have not been described. Recent investigations have demonstrated that pseudopalisades are severely hypoxic, overexpress hypoxia-inducible factor (HIF-1), and secrete proangiogenic factors such as VEGF and IL-8. Thus, the microvascular hyperplasia in GBM that provides a new vasculature and promotes peripheral tumor expansion is tightly linked with the emergence of pseudopalisades. Both pathologic observations and experimental evidence have indicated that the development of hypoxia and necrosis within astrocytomas could arise secondary to vaso-occlusion and intravascular thrombosis. This emerging model suggests that pseudopalisades represent a wave of tumor cells actively migrating away from central hypoxia that arises after a vascular insult. Experimental glioma models have shown that endothelial apoptosis, perhaps resulting from angiopoetin-2, initiates vascular pathology, whereas observations in human tumors have clearly demonstrated that intravascular thrombosis develops with high frequency in the transition to GBM. Tissue factor, the main cellular initiator of thrombosis, is dramatically upregulated in response to PTEN loss and hypoxia in human GBM and could promote a prothrombotic environment that precipitates these events. A prothrombotic environment also activates the family of protease activated receptors (PARs) on tumor cells, which are G-protein-coupled and enhance invasive and proangiogenic properties. Vaso-occlusive and prothrombotic mechanisms in GBM could readily explain the presence of pseudopalisading necrosis in tissue sections, the rapid peripheral expansion on neuroimaging, and the dramatic shift to an accelerated rate of clinical progression resulting from hypoxia-induced angiogenesis.