ArticlePDF Available

Evaluation of the Psoriasis Transcriptome across Different Studies by Gene Set Enrichment Analysis (GSEA)

PLOS
PLOS ONE
Authors:

Abstract and Figures

Our objective was to develop a consistent molecular definition of psoriasis. There have been several published microarray studies of psoriasis, and we compared disease-related genes identified across these different studies of psoriasis with our own in order to establish a consensus. We present a psoriasis transcriptome from a group of 15 patients enrolled in a clinical study, and assessed its biological validity using a set of important pathways known to be involved in psoriasis. We also identified a key set of cytokines that are now strongly implicated in driving disease-related pathology, but which are not detected well on gene array platforms and require more sensitive methods to measure mRNA levels in skin tissues. Comparison of our transcriptome with three other published lists of psoriasis genes showed apparent inconsistencies based on the number of overlapping genes. We extended the well-established approach of Gene Set Enrichment Analysis (GSEA) to compare a new study with these other published list of differentially expressed genes (DEG) in a more comprehensive manner. We applied our method to these three published psoriasis transcriptomes and found them to be in good agreement with our study. Due to wide variability in clinical protocols, platform and sample handling, and subtle disease-related signals, intersection of published DEG lists was unable to establish consensus between studies. In order to leverage the power of multiple transcriptomes reported by several laboratories using different patients and protocols, more sophisticated methods like the extension of GSEA presented here, should be used in order to overcome the shortcomings of overlapping individual DEG approach.
Content may be subject to copyright.
Evaluation of the Psoriasis Transcriptome across
Different Studies by Gene Set Enrichment Analysis
(GSEA)
Mayte Sua
´rez-Farin
˜as
1,2
, Michelle A. Lowes
1
, Lisa C. Zaba
1
, James G. Krueger
1
*
1Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, United States of America, 2Center for Clinical and Translational Science, The
Rockefeller University, New York, New York, United States of America
Abstract
Background:
Our objective was to develop a consistent molecular definition of psoriasis. There have been several published
microarray studies of psoriasis, and we compared disease-related genes identified across these different studies of psoriasis
with our own in order to establish a consensus.
Methodology/Principal Findings:
We present a psoriasis transcriptome from a group of 15 patients enrolled in a clinical
study, and assessed its biological validity using a set of important pathways known to be involved in psoriasis. We also
identified a key set of cytokines that are now strongly implicated in driving disease-related pathology, but which are not
detected well on gene array platforms and require more sensitive methods to measure mRNA levels in skin tissues.
Comparison of our transcriptome with three other published lists of psoriasis genes showed apparent inconsistencies based
on the number of overlapping genes. We extended the well-established approach of Gene Set Enrichment Analysis (GSEA)
to compare a new study with these other published list of differentially expressed genes (DEG) in a more comprehensive
manner. We applied our method to these three published psoriasis transcriptomes and found them to be in good
agreement with our study.
Conclusions/Significance:
Due to wide variability in clinical protocols, platform and sample handling, and subtle disease-
related signals, intersection of published DEG lists was unable to establish consensus between studies. In order to leverage
the power of multiple transcriptomes reported by several laboratories using different patients and protocols, more
sophisticated methods like the extension of GSEA presented here, should be used in order to overcome the shortcomings of
overlapping individual DEG approach.
Citation: Sua
´rez-Farin
˜as M, Lowes MA, Zaba LC, Krueger JG (2010) Evaluation of the Psoriasis Transcriptome across Different Studies by Gene Set Enrichment
Analysis (GSEA). PLoS ONE 5(4): e10247. doi:10.1371/journal.pone.0010247
Editor: H. Peter Soyer, The University of Queensland, Australia
Received January 8, 2010; Accepted March 27, 2010; Published April 20, 2010
Copyright: ß2010 Sua
´rez-Farin
˜as 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, provided the original author and source are credited.
Funding: This research was supported by a Clinical and Translational Science Award grant UL1RR024143; MSF is supported by the Milstein Program in Medical
Research; LZ is supported by National Institutes of Health (NIH) Medical Scientist Training Program grant GM07739; and ML by K23 AR052404-01A1 and the Doris
Duke Foundation. 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: jgk@rockefeller.edu
Introduction
The study of human diseases such as psoriasis has benefited
significantly from analysis of the transcriptome, the global gene
expression of a diseased tissue compared to its healthy counterpart.
However, as more studies are carried out independently in
multiple laboratories, effective methodology to leverage these
multiple studies becomes necessary. Such methodologies have
significant hurdles to overcome: first, multiple studies are likely to
use different platforms, different sample dissection, handling and
preparation, and, especially, different definition of the non-
diseased counterpart, resulting in different physical samples being
hybridized against different platforms [1,2,3]. Second, computa-
tional analysis and statistical treatment required to assess the
transcriptome are just as likely to be considerably different.
In many instances, all that is available from published studies
are lists of differentially expressed genes (DEG). It is tempting to
evaluate the agreement between studies simply by evaluating the
intersection between the published lists, the ‘‘Venn diagram
approach’’. However, such an approach suffers serious method-
ological shortcomings [4,5,6]. Use of the original raw data of the
studies has shown that studies which are apparently discordant in
terms of their overlapping individual DEG lists are, in fact, both
concordant and predictive [4,5]. However, most of the time the
original raw data is unavailable, and furthermore a complete
reanalysis of all data is needlessly laborious. In such cases use of
the published lists of DEG is a necessity. Here we present an
extension to the widely used Gene Set Enrichment Analysis
(GSEA) method, where it suffices to have full access to the
complete list of gene expression values for a single study, while the
remaining studies only require the DEG list.
In the last few years, the use of Gene-Sets approach had
emerged as a powerful tool to identify sets of functionally related
genes or pathways that are associated with a disease phenotype
[7,8]. Gene-Sets based methods were designed to address
limitations of conventional single gene methods [6] by evaluating
PLoS ONE | www.plosone.org 1 April 2010 | Volume 5 | Issue 4 | e10247
differential expression patterns of gene groups instead of individual
genes. GSEA, introduced by Mootha et al [9] and further
developed by Subramanian et al [10], was one of the first method
using the Gene-Sets approach, and is arguably the most widely
used of such methods. Here we use GSEA as a conventional
approach to identify pathways related to the psoriatic phenotype.
Furthermore, we propose to extend the use of GSEA as a tool to
easily cross-compare prior lists of DEG genes.
We developed this method specifically to compare several high-
quality studies that defined the psoriasis transcriptome by
identifying DEG between psoriatic lesions and non-lesional tissue
from the same patients [11,12,13,14,15]. Those studies had
identified key genes involved in psoriasis pathogenesis, using a
non-biased approach. Because the genomic data for more recent
studies is more comprehensive than in the earlier studies due to the
larger number of genes represented in the latest Affymetrix chips,
we chose to compare the transcriptomes for studies published since
2003 [12,14,15].
We recently conducted a clinical trial of 15 psoriasis patients
with the TNF inhibitor etanercept [16], and performed a time-
course experiment using HGU 133 2.0 microarray chips [17]. By
analyzing the baseline data from this experiment, we generated
our psoriasis transcriptome comparing baseline-paired values of
lesional versus non-lesional skin. We have compared our
transcriptome with the three others described above, and
introduce the concept of using GSEA as a more robust way of
comparing genomic data.
Results
Disease-modulated genes
The analysis of our data identified a psoriasis transcriptome
composed of 732 up-regulated probesets (representing 579 genes
with unique ENTREZ identifier) and 890 down-regulated
probesets (703 genes) with fold change (FCH) greater than 2 and
false discovery rate (FDR) less than 0.05 (Table 1, and Table S1).
Certain genes with low expression on the Affymetrix chip were
confirmed by RT-PCR, and will be discussed in the next section.
To further consider the biological significance of our data, we
used GSEA in the classical manner, to identify pathways that
correlate with the psoriatic phenotype [10,18]. GSEA evaluates
how genes in queried pathways are distributed in the fold change
(lesional versus non-lesional) ordered list generated by our data (all
probesets included). This is quantified by using the Enrichment
Score (ES), a weighted Kolmogorov-Smirnov-like statistic that
evaluates if the members of the pathway are randomly distributed
or found at the extremes (top or bottom) of the list. If genes in a
pathway rank at the top of the new fold change list, ie. they are
overrepresented at the top, then the enrichment score (ES) will be
close to 1. Conversely if the ES = 21, then genes are overrepre-
sented at the bottom of our fold change data. A perfect agreement
is reached if ES = 1 for the up-regulated genes and ES = 21 for the
down-regulated genes. A normalized enrichment score (NES) takes
into account the number of genes in the pathway. A positive NES
indicates that the list of genes is enriched at the ‘‘top’’ of the
ordered fold change list, and a negative NES indicates that the list
in question is enriched at the ‘‘bottom’’ of the list.
GSEA may be used with well known ‘‘canonical’’ pathways and
Gene ontology categories, but also with sets that contain genes
sharing the same transcription factor binding site, the same
microRNA binding motif or the same cis-regulatory motif. It can
also be used with curated collections such as GeneSigDb (http://
compbio.dfci.harvard.edu/genesigdb), which contain gene signa-
tures of cancer, viral and stem cell biology, and the CGP collection
of the MSigDb (http://www.broadinstitute.org/gsea/msigdb/)
that contains gene expression signatures of genetic and chemical
perturbations, or computational derived sets such as cancer
modules (presented in [19]).
We queried our psoriasis transcriptome with a set of cytokine-
treated keratinocyte pathways for IL-17, TNF, IL-17+TNF, IFNc,
and IL-22, reported in [17,20] and IL-1a[21]. GSEA showed that
those pathways were enriched in lesional skin of psoriasis patients
(Table 2). We used a list (‘‘IL-17 Gaffen’’) considered to be IL-17-
gene targets defined by Shen et al [22]. This was also significantly
enriched in our psoriasis transcriptome. An IFNa-keratinocyte
pathway [14] was also significantly enriched in lesional skin,
supporting the potential role for IFNadiscussed by Yao et al [14].
A list of genes representing the transcriptome of inflammatory
myeloid DCs [23] was also significantly enriched in lesional skin.
In addition, the cell cycle and TLR signaling from the collection of
canonical pathways (C2 CP) available at the Molecular Signature
Database (MSigDb) were enriched in psoriasis lesional skin.
Gudjonsson et al reported lack of evidence of enrichment of the
Hedghog Signalling Pathways in psoriasis [24]. GSEA analysis did
not detect any significant enrichment of this pathway with the
psoriasis phenotype. (ES = 0.37, NES = 1.05 and p = 0.39).
Comparison with other published studies
We next performed a comparative analysis of the DEG lists of
these three studies [12,14,15] with our transcriptome. Table 1
summarizes the main characteristics of the four studies. Zhou’s
study which uses early hgu95 (a,b,c,d,e) chips, reported 397 up-
regulated and 613 down-regulated probesets representing 270 and
397 unique genes respectively [15]. Yao et al conducted a study on
Table 1. Description of studies.
Zhou Yao Gudhjonsson Sua
´rez-Farin
˜as
Platform/chips hgu95 a,b,c,d,e hgu133plus2 hgu133plus2 hgu133a2
Sample size 16 26 58 15
Expression Algorithm dChip GCRMA RMA GCRMA
Statistical test t-test Sam (paired) t-test Paired t-test Moderated paired t-test
Multiple Hypothesis correction none FDR through permutations FDR through permutations FDR Benjamini-Hochberg
Cut-off FCH.2, p,0.05 FCH.2, q-value,0.05 FCH.2, FDR,0.05 FCH.2, FDR,0.05
#Up-regulated probesets (genes) 397 ps (270 genes) 1408 ps. (974 genes) 721 ps (508 genes) 732 ps (579 genes)
#of Down-regulated probesets (genes) 613 ps (397 genes) 1465 ps (853 genes) 364 ps (248 genes) 890 ps (703 genes)
doi:10.1371/journal.pone.0010247.t001
Psoriasis Transcriptomes
PLoS ONE | www.plosone.org 2 April 2010 | Volume 5 | Issue 4 | e10247
hgu133plus2 chips and reported 1408 up-regulated and 1465
down-regulated probesets (974 and 853 genes respectively) [14].
More recently, Gudjonsson et al also using hgu133plus2 chips with
a large sample size [12] reported a set of 721 up-regulated and 364
down-regulated probesets (508 genes and 248 genes).
Figure 1 shows a Venn diagram illustrating the intersection
between the four studies. There were approximately 11,000
ENTREZ identifiers common to the 4 chip series, but only 126
genes were up-regulated (Figure 1A) and 38 down-regulated
(Figure 1B) in all four studies. The numbers of upregulated genes
in our transcriptome were similar to the Gudhjonsson group, but
less than Yao’s. The number of down-regulated genes were greater
than the Gudhjonsson group, and again, less than Yao’s.
The list of the DEG in common in the 4 studies (Table S2)
contains many genes known to be upregulated in psoriasis, such as
IFNc-regulated genes STAT-1, STAT-3 and MX1; antimicrobial
peptides (beta defensin 4, lipocalin 2, S100A7); and pro-
inflammatory proteins such as IL-8, CXCL1, IL-1F9,
TNFSF10/TRAIL. However, some genes known to be upregu-
lated in psoriasis were only identified in one study. For example
STAT2 was only identified in our study, JAK3 only in Yao’s, and
IL-12RB1 only in Zhou’s study. The set of genes that were down-
regulated in all 4 studies include CCL27, also called Cutaneous T
cell –attracting chemokine (CTACK), which has a role in memory
T cell homing to the skin [25], and Aquaporin 9, a member of a
family of proteins that form water channels across membranes
[26].
Some well-recognized inflammatory genes involved in psoriasis
were not detected by most of the four studies, for example IFNc,
IL-17, iNOS. This is due to the fact that the expression of these
genes is usually low on the Affymetrix gene array platform (0–4
range of expression in log
2
-scale) and hence fold change is not
accurately measured. Most analysis pipelines filter out low
abundance genes so they may be excluded from the statistical
analysis, or the resultant fold change is very low, albeit significant.
This is a major limitation of the use of these arrays for the study of
these genes.
To confirm the role of these genes in psoriasis, we analyzed RT-
PCR data of many of these inflammatory genes. We used data
from the same clinical trial of etanercept treatment of psoriasis
[16], however, we compared only baseline lesional skin and non-
lesional skin. The fold change and p-value for each gene by RT-
PCR is presented in Table 3, and all of these genes except LTA
and p35 were significantly different in lesional skin (p,0.05). We
also present the fold change of any of these genes that were
detectable in any of the microarray studies. It can be seen that IL-
23p19, IL-12/23p40, IL-22, IFNc, IL-6 were not found to be
differentially expressed by any of the four studies, but the fold
change by RT-PCR was greater than 4. Furthermore, IL-17, IL-
20, CCL4, iNOS and CCL3 were detected in only one study, and
with a lower fold change than detected by RT-PCR (which was
greater than 5.5). We also included the percentage of samples in
our study with low intensity, as defined as expression values less
than 4. For example, p19 has expression of less than 4. It can be
seen that the genes that were not detected by any or only one
microarray study had low abundance in more than 87% of the
samples.
Correlation of other published psoriasis transcriptomes
compared to our transcriptome using Gene Set
Enrichment Analysis (GSEA)
A more robust approach beyond a Venn diagram was required
to overcome these limitations. We propose to use a Gene-Sets
approach to analyze how published DEG rank in our fold change
data. This reduces the bias due to preprocessing steps, statistical
protocols and stringency of cut-offs. This approach is successfully
used in the Connectivity Map [27], a pattern recognition instance
that correlates disease signatures (based on gene expression of any
platform) with drugs. The idea is to use the GSEA framework [10]
by considering the published DEG as a pathway or gene set, and
quantify how well the up (and down) regulated genes rank in the
ordered fold change for all genes in our data. This will generate an
ES for up-regulated genes and one for the down-regulated genes.
The connectivity score (CS) can be used to give a measure of
agreement between studies, by combining the two ES into one
final value, as used in the connectivity map to rank drugs that
better correlate with disease. A value of CS near 1 would indicate
perfect agreement between a study list and our analysis, whereas 0
would indicate no agreement, and 21 a negative correlation.
GSEA showed that there was highly significant enrichment of
psoriasis DEGs, both up- and down-regulated genes, from the
three studies compared to our data (Table 4). The GSEA plot for
Table 2. Pathways enriched in Psoriasis lesions by using GSEA.
PATHWAYS No. of genes in pathway ES NES FDR
IFNaUp in KC (Yao) 28 0.91 2.74 ,10
24
IL17 and TNF Up in KC 30 0.89 2.69 ,10
24
IL17 Up in KC 46 0.87 2.86 ,10
24
IL1 Up in KC 34 0.85 2.66 ,10
24
IL17 GAFFEN 27 0.81 2.43 ,10
24
IFNcUp in KC 872 0.47 2.31 ,10
24
TNF Up in KC 472 0.46 2.14 ,10
24
IL22 Up in KC 10 0.89 2.07 ,10
24
Terminal Differentiation 33 0.69 2.10 ,10
24
Inflammatory myeloid DCs (psoriasis) 121 0.42 1.68 ,10
24
Cell Cycle (KEGG) 64 0.58 2.06 ,10
24
TLR signaling Pathway (KEGG) 58 0.59 2.05 ,10
24
Cytokine-Cytokine receptor interaction (KEGG) 111 0.44 1.71 0.02
doi:10.1371/journal.pone.0010247.t002
Psoriasis Transcriptomes
PLoS ONE | www.plosone.org 3 April 2010 | Volume 5 | Issue 4 | e10247
up and down-regulated genes in Zhou’s, Yao’s and Gudjonsson’s
lists is shown in Figure 2. P-values for ES and CS were calculated
using 10,000 simulations. For Zhou’s list, the ES for the up-
regulated genes was 0.86 (p,0.0001) and 20.63 (p,0.0001) for
the down-regulated genes (Figure 2A). The CS = 0.75 (p,0.0001)
indicates a positive significant agreement between Zhou’s
signature and our data. For Yao’s transcriptome (Figure 2B), the
ES = 0.90 (p,0.0001) for the up-regulated genes and ES = 20.86
(p,0.0001) for the down–regulated genes, for CS = 0.88
(p,0.0001), which also indicates a positive significant agreement.
For Gudjonsson’s transcriptome (Figure 2C), the ES = 0.93
(p,0.0001) for the up-regulated genes and ES = 20.91
(p,0.0001) for the down–regulated genes, for CS = 0.92
(p,0.0001), which also indicates a positive significant agreement.
In general, a better agreement was observed among up-regulated
genes for all studies. In addition, Yao’s transcriptome correlated
better with our study than Zhou’s, which is not surprising since the
array series and the statistical protocols used in Yao’s and ours
were more similar than those in Zhou’s.
We used the same approach to compare two published DEGs of
other skin diseases produced by own group: squamous cell
carcinoma (SCC) [28] and basal cell carcinoma (BCC) [29]. The
CS for SCC was 0.69 and the CS of BCC was 0.42, considerably
lower than in psoriasis (Table 4). This degree of enrichment is
most likely reflects the origin of this data from our own lab, and
the cutaneous nature of the specimens, as well as epidermal
hyperproliferation and inflammation in all these three diseases.
Discussion
Investigators might be surprised at the lack of overlap between
DEG lists, as shown in the Venn diagram (Figure 1). However, if
one considers all the variables involved in the four studies,
summarized in Table 1, it is not that surprising. Although all the
studies were conducted using Affymetrix platform, they used
different array series, which may contribute to variability in results.
Besides the obvious laboratory effect due to sample preparation,
technician experience, equipment calibration, and the use of
different preprocessing algorithms [30], alternative statistical tests
and stringent cut-offs also contribute to different results [3,31].
Measuring agreement of microarray studies by overlap of DEG
lists generated by individual studies has been largely criticized [5]
because it is highly inconsistent, even in the presence of small
variation in the data as in the case of technical replicates [32,33].
A low overlap between DEG does not directly indicate low
agreement between studies [3,31,33].
Here we present our new data on the psoriasis transcriptome
from our patients, as well as a comparison of our data with three
published DEG lists for psoriasis. We find only 164 genes in
common for the four lists. However by changing the focus of the
single-gene approach behind the intersection of DEG involved in
psoriasis and using a gene set approach, a closer biological
similarity between the studies is revealed. The extension of GSEA
presented here enables us to see that cellular processes and
molecular signature involved in psoriasis is very robust across the
studies. In this paper, we extended the use of GSEA to compare
new expression data with previously published DEG lists in order
to validate psoriasis disease-related gene profiles. It is worth
noting that this approach is applicable to expression data
obtained through deep sequencing (potentially improving sensi-
tivity for low abundance genes and cross-hybridization problems
of current microarray technology). Moreover, this approach is
easily extendable to other omics applications and more complex
phenotypes. Since the method is based on ranking a list according
to a phenotype, the ranked list can be derived from other
measures besides gene expression fold changes from microarray
chips or deep sequencing; such phenotype measures may include
odd ratios of single nucleotide polymorphisms (SNPs), or a
microRNA profile assay derived from microarray technologies or
deep sequencing.
The use of GSEA as a gene set approach is not unique:
extensions to GSEA [34] and other Gene Set methods and
Figure 1. Comparison of four psoriasis transcriptomes. A. Venn
diagram showing the comparison of the number of up-regulated genes
in common and distinct for the four studies. B. Venn diagram showing
the comparison of the number of down-regulated genes in common
and distinct for the four studies.
doi:10.1371/journal.pone.0010247.g001
Psoriasis Transcriptomes
PLoS ONE | www.plosone.org 4 April 2010 | Volume 5 | Issue 4 | e10247
statistics have also been proposed, and could also be used to
compare transcriptomes. Efron and Tibshirani [35] proposed the
MaxMean statistic instead of the weighted Kolmogorov Smirnov
statistics used in the classical GSEA. Dinu et al [36] extended the
single gene analysis SAM and proposed SAM-GS. See [7,37] for a
comparative study of different gene set enrichment methods.
Extensions other than the classical difference between two
phenotypes have also been reported. For example, we used the
time slope of a mixed effect model as a phenotype to evaluate the
time-response of cytokines pathways to psoriasis treatment with
TNF inhibitor [17].
In this report, we show an excellent and simple method for
researchers seeking validation of their own expression data with
published lists from different studies.
Materials and Methods
Patients
Twenty adult patients with moderate to severe psoriasis were
treated with etanercept 50 mg subcutaneously twice weekly for 12
weeks (clinical trial no. NCT00116181). The clinical and
histological response of patients in this trial was previously
published [16]. The gene array was performed on samples from
15 sequential patients [17]. RT-PCR was performed on samples
from all 20 patients.
Ethics Statement
The clinical trial (no. NCT00116181) was conducted according
to the principles expressed in the Declaration of Helsinki and
informed consent for their information to be stored in the hospital
database and used for research was obtained from all patients in
written form. This research was conducted under protocol JKR-
0542 approved by the Rockefeller University Institutional Review
Board.
Table 3. RT-PCR validation.
RT-PCR Microarray FCH (log2)
1
Gene Symbol FCH (log2) FCH p.value Suarez-Farinas Zou Yao Gudjonsson % Low Int
2
p19 IL23A 2.66 6.34 0.011 100
p40 IL12B 4.03 16.32 1.8610
205
97
LTA1 LTA 0.83 1.77 0.302 100
IL22 IL22 3.96 15.53 1.5610
24
100
IFNcIFNg 2.28 4.85 2.8610
24
100
IL4 IL4 21.45 0.36 0.034 100
IL6 IL6 2.66 6.33 7.1610
24
100
IL17 IL17A 6.17 71.87 3.8610
25
1.15 93
IL20 IL20 3.95 15.48 1.4610
25
1.03
CCL4 CCL4 2.83 7.12 9.2610
25
1.38 87
iNOS NOS2 6.37 82.91 1.7610
29
1.11 100
p35 IL12A 21.58 0.34 0.186 100
CCL3 CCL3 2.46 5.52 6.2610
23
1.07
AREG AREG 2.05 4.15 1.2610
23
2.54 2.15 1.35 7
CCL20 CCL20 3.08 8.45 4.7610
25
2.79 3.64 2.90 67
IL19 IL19 5.45 43.57 2.0610
25
1.43 2.21 1.88 8
IL1bIL1B 3.66 12.66 6.8610
26
2.61 1.97 1.11 83
K16 KRT16 5.10 34.34 8.3610
29
3.68 1.99 4.57 4.11 0
MX1 MX1 3.56 11.83 4.1610
25
3.23 3.24 3.24 2.32 0
IL8 IL8 6.19 72.82 1.0610
27
5.85 2.96 4.67 4.03 47
bDefensin DEFB4 4.12 17.42 6.5610
28
5.96 1.96 7.34 7.07 1
1
The largest fold change (FCH) was reported when there were several probesets representing the same gene.
2
Percentage of samples with low intensity as defined by having expression smaller that 4.
doi:10.1371/journal.pone.0010247.t003
Table 4. GESA analysis of published transcriptomes with our
data.
Gene Set
No. of genes
in pathway ES NES FDR CS
Gudjonsson - UP 386 0.93 4.25 ,10
24
0.92
Gudjonsson - Down 153 20.91 23.77 ,10
24
Yao - UP 670 0.90 4.31 ,10
24
0.88
Yao – Down 487 20.86 24.06 ,10
24
Zhou - UP 199 0.86 3.69 ,10
24
0.75
Zhou - Down 227 20.63 22.71 ,10
24
SCC LSvsNL UP 859 0.69 3.37 ,10
24
0.69
SCC LSvsNL Down 655 20.70 23.41 ,10
24
BCC LS vs Normal UP 191 0.38 1.61 ,10
24
0.42
BCC LS vs Normal Down 326 20.46 22.09 ,10
24
ES: Enrichment Score; NES: Normalized Enrichment Score; FDR: False Discovery
Rate.
doi:10.1371/journal.pone.0010247.t004
Psoriasis Transcriptomes
PLoS ONE | www.plosone.org 5 April 2010 | Volume 5 | Issue 4 | e10247
Psoriasis Transcriptomes
PLoS ONE | www.plosone.org 6 April 2010 | Volume 5 | Issue 4 | e10247
Sample Preparation and Hybridization
RNA was extracted from skin biopsies and hybridized to
Affymetrix hgu133a2 chips as described in [17].
RTPCR
Primers and Probes for TaqMan RT-PCR assays had been
previously described [16]. New primers used in this study were
CCL3 (Hs00234142_m1), CCL4 (Hs99999148_m1), AREG
(Hs00950669_m1), IL-19 (Hs00604657_m1). All assays were
obtained from Applied Biosystems. Data was normalized using
human acidic ribosomal protein as a housekeeping gene [16].
Statistical Analysis
Expression values were obtained using gcrma algorithm. Samples
were filtered for unreliable low expression value and low variability.
To compare lesional with non-lesional values, the moderated t-test
available at limma package was used. P-values were adjusted for
multiple hypothesis correction using the Benjamini-Hochberg
approach, which controls the false discovery rate (FDR). Probesets
with FDR,0.05 and more than 2 fold change (FCH) were
considered differentially expressed. All analysis was carried out
using R programming language (www.R-project.org) and Biocon-
ductor packages (www.bioconductor.org).
Annotations were obtained by using Bioconductor hgu133a2.db
and hgu133plus2.db packages version 2.3.5. Mappings were based
on ENTREZ identifiers, provided by Entrez Gene fftp://
ftp.ncbi.nlm.nih.gov/gene/DATA with a data stamp of Sept 1,
2009.
Gene Set Enrichment Analysis (GSEA) was conducted using
GSEA software [18].
Data Repository
This data has been deposited at the public repository Gene
Expression Omnibus (GEO) (http://www.ncbi.nlm.nih.gov/geo/)
with accession number GSE11903.
Supporting Information
Table S1 DEG genes identified in this study (FDR,0.05,
FCH.2).
Found at: doi:10.1371/journal.pone.0010247.s001 (0.52 MB
PDF)
Table S2 Genes consistently identified by the four studies.
Found at: doi:10.1371/journal.pone.0010247.s002 (0.08 MB
PDF)
Acknowledgments
We thank Dr J. Mee for sharing his genomic data with our group. We
thank Irma Cardinale and RU Genomic facility for the hybridization of
Affymetrix chips. We thank Dr. Marcelo O. Magnasco and Dr. Leanne M.
Huang-Johnson for valuable comments and suggestions.
Author Contributions
Conceived and designed the experiments: MSF JGK. Performed the
experiments: LCZ. Analyzed the data: MSF MAL JGK. Contributed
reagents/materials/analysis tools: JGK. Wrote the paper: MSF MAL JGK.
Interpreted data analysis: JGK. Interpreted results: MAL LCZ.
References
1. Irizarry RA, Warren D, Spencer F, Kim IF, Biswal S, et al. (2005) Multiple-
laboratory comparison of microarray platforms. Nat Methods 2: 345–350.
2. Larkin JE, Frank BC, Gavras H, Sultana R, Quackenbush J (2005)
Independence and reproducibility across microarray platforms. Nat Methods
2: 337–343.
3. Shi LM, Perkins RG, Fang H, Tong WD (2008) Reproducible and reliable
microarray results through quality control: good laboratory proficiency and
appropriate data analysis practices are essential. Current Opinion in
Biotechnology 19: 10–18.
4. Suarez-Farinas M, Magnasco MO (2007) Comparing microarray studies.
Methods Mol Biol 377: 139–152.
5. Suarez-Farinas M, Noggle S, Heke M, Hemmati-Brivanlou A, Magnasco MO
(2005) Comparing independent microarray studies: the case of human
embryonic stem cells. BMC Genomics 6: 99.
6. Shi J, Walker MG (2007) Gene set enrichment analysis (GSEA) for interpreting
gene expression profiles. Current Bioinformatics 2: 133–137.
7. Abatangelo L, Maglietta R, Distaso A, D’Addabbo A, Creanza TM, et al. (2009)
Comparative study of gene set enrichment methods. BMC Bioinformatics 10:
275.
8. Nam D, Kim SY (2008) Gene-set approach for expression pattern analysis (vol 9,
pg 189, 2008). Briefings in Bioinformatics 9: 450–450.
9. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, et al. (2003)
PGC-1alpha-responsive genes involved in oxidative phosphorylation are
coordinately downregulated in human diabetes. Nat Genet 34: 267–273.
10. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, et al. (2005)
Gene set enrichment analysis: a knowledge-based approach for interpreting
genome-wide expression profiles. Proc Natl Acad Sci U S A 102: 15545–15550.
11. Bowcock AM, Shannon W, Du F, Duncan J, Cao K, et al. (2001) Insights into
psoriasis and other inflammatory diseases from large-scale gene expression
studies. Hum Mol Genet 10: 1793–1805.
12. Gudjonsson JE, Ding J, Li X, Nair RP, Tejasvi T, et al. (2009) Global gene
expression analysis reveals evi dence for decreased lipid biosyn thesis and
increased innate immunity in uninvolved psoriatic skin. J Invest Dermatol
129: 2795–2804.
13. Oestreicher JL, Walters IB, Kikuchi T, Gilleaudeau P, Surette J, et al. (2001)
Molecular classification of psoriasis disease-associated genes through pharma-
cogenomic expression profiling. Pharmacogenomics J 1: 272–287.
14. Yao Y, Richman L, Morehouse C, de los Reyes M, Higgs BW, et al. (2008) Type
I interferon: potential therapeutic target for psoriasis? PLoS One 3: e2737.
15. Zhou X, Krueger JG, Kao MC, Lee E, Du F, et al. (2003) Novel mechanisms of
T-cell and dendritic cell activation revealed by profiling of psoriasis on the
63,100-element oligonucleotide array. Physiol Genomics 13: 69–78.
16. Zaba LC, Cardinale I, Gilleaudeau P, Sullivan-Whalen M, Farinas MS, et al.
(2007) Amelioration of epidermal hyperplasia by TNF inhibition is associated
with reduced Th17 responses. J Exp Med 204: 3183–3194.
17. Zaba LC, Suarez-Farinas M, Fuentes-Duculan J, Nograles KE, Guttman-
Yassky E, et al. (2009) Effective treatment of psoriasis with etanercept is linked to
suppression of IL-17 signaling, not immediate response TNF genes. J Allergy
Clin Immunol 124: 1022-1010 e1021–1395.
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. Segal E, Shapira M, Regev A, Pe’er D, Botstein D, et al. (2003) Module
networks: identifying regulatory modules and their condition-specific regulators
from gene expression data. Nat Genet 34: 166–176.
20. Nograles KE, Zaba LC, Guttman-Yassky E, Fuentes-Duculan J, Suarez-
Farinas M, et al. (2008) Th17 cytokines interleukin (IL)-17 and IL-22 modulate
distinct inflammatory and keratinocyte-response pathways. Br J Dermatol 159:
1086–1091.
21. Mee JB, Johnson CM, Morar N, Burslem F, Groves RW (2007) The psoriatic
transcriptome closely resembles that induced by interleukin-1 in cultured
keratinocytes: dominance of innate immune responses in psoriasis. Am J Pathol
171: 32–42.
22. Shen F, Gaffen SL (2008) Structure-function relationships in the IL-17 receptor:
implications for signal transduction and therapy. Cytokine 41: 92–104.
23. Zaba LC, Fuentes-Duculan J, Eungdamrong NJ, Johnson-Huang LM,
Nograles KE, et al. (2010) Identification of TRAIL and other molecules that
distinguish inflammatory DCs from resident DCs in psoriasis. J Allergy Clin
Immunol, (Accepted).
Figure 2. Rank of the published DEG lists in our fold change data. A. Rank of Zhou’s genes in our data set ordered by fold change expression,
with Enrichment plots for the up-regulated and downregulated genes data below. B. As A but for Yao’s study. C. As A but for Gudjonsson’s study. A
detailed explanation of Enrichment plots can be found in Figure 1 of original Subramanian et al [10].
doi:10.1371/journal.pone.0010247.g002
Psoriasis Transcriptomes
PLoS ONE | www.plosone.org 7 April 2010 | Volume 5 | Issue 4 | e10247
24. Gudjonsson JE, Aphale A, Grachtchouk M, Ding J, Nair RP, et al. (2009) Lack
of evidence for activation of the hedgehog pathway in psoriasis. J Invest
Dermatol 129: 635–640.
25. Homey B, Alenius H, Muller A, Soto H, Bowman EP, et al. (2002) CCL27-
CCR10 interactions regulate T cell-medi ated skin inflammation. Nature
Medicine 8: 157–165.
26. Boury-Jamot M, Sougrat R, Tailhardat M, Le Varlet B, Bonte F, et al. (2006)
Expression and function of aquaporins in human skin: Is aquaporin-3 just a
glycerol transporter? Biochim Biophys Acta 1758: 1034–1042.
27. Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, et al. (2006) The
Connectivity Map: using gene-expression signatures to connect small molecules,
genes, and disease. Science 313: 1929–1935.
28. Haider AS, Peters SB, Kaporis H, Cardinale I, Fei J, et al. (2006) Genomic
analysis defines a cancer-specific gene expression signature for human squamous
cell carcinoma and distinguishes malignant hyperproliferation from benign
hyperplasia. J Invest Dermatol 126: 869–881.
29. Kaporis HG, Guttman-Yassky E, Lowes MA, Haider AS, Fuentes-Duculan J,
et al. (2007) Human basal cell carcinoma is associated with Foxp3+T cells in a
Th2 dominant microenvironment. J Invest Dermatol 127: 2391–2398.
30. Irizarry RA, Wu Z, Jaffee HA (2006) Comparison of Affymetrix GeneChip
expression measures. Bioinformatics 22: 789–794.
31. Shi L, Jones WD, Jensen RV, Harris SC, Perkins RG, et al. (2008) The balance
of reproducibility, sensitivity, and specificity of lists of differentially expressed
genes in microarray studies. BMC Bioinformatics 9 Suppl 9: S10.
32. Tan PK, Downey TJ, Spitznagel EL, Jr., Xu P, Fu D, et al. (2003) Evaluation of
gene expression measurements from commercial microarray platforms. Nucleic
Acids Res 31: 5676–5684.
33. Zhang M, Yao C, Guo Z, Zou J, Zhang L, et al. (2008) Apparently low
reproducibility of true differential expression discoveries in microarray studies.
Bioinformatics 24: 2057–2063.
34. Jiang Z, Gentleman R (2007) Extensions to gene set enrichment. Bioinformatics
23: 306–313.
35. Efron B, Tibshirani R (2007) On testing the significance of sets of genes. Annals
of Applied Statistics 1: 107–129.
36. Dinu I, Potter JD, Mueller T, Liu Q, Adewale AJ, et al. (2009) Gene-set analysis
and reduction. Brief Bioinform 10: 24–34.
37. Ackermann M, Strimmer K (2009) A general modular framework for gene set
enrichment analysis. BMC Bioinformatics 10: 47.
Psoriasis Transcriptomes
PLoS ONE | www.plosone.org 8 April 2010 | Volume 5 | Issue 4 | e10247

Supplementary resources (2)

... o Find the minimizer (�, �) to the following optimization problem using coordinate descent. The solutions are equations [11] and [12]. ...
... Setting the gradient with respect to to 0, the coordinate-wise update for has the form: [11] where ( ) = ′ − ∑ � ≠ − � ( ) , is the partial residual excluding the contribution of . As for , if � > 0, then the gradient of (9) with respect to is ...
Preprint
Full-text available
Background Associated with high-dimensional omics data there are often “meta-features” such as pathways and functional annotations that can be informative for predicting an outcome of interest. We extend to Cox regression the regularized hierarchical framework of Kawaguchi et al. (2022) (1) for integrating meta-features, with the goal of improving prediction and feature selection performance with time-to-event outcomes. Methods A hierarchical framework is deployed to incorporate meta-features. Regularization is applied to the omic features as well as the meta-features so that high-dimensional data can be handled at both levels. The proposed hierarchical Cox model can be efficiently fitted by a combination of iterative reweighted least squares and cyclic coordinate descent. Results In a simulation study we show that when the external meta-features are informative, the regularized hierarchical model can substantially improve prediction performance over standard regularized Cox regression. We illustrate the proposed model with applications to breast cancer and melanoma survival based on gene expression profiles, which show the improvement in prediction performance by applying meta-features, as well as the discovery of important omic feature sets with sparse regularization at meta-feature level. Conclusions The proposed hierarchical regularized regression model enables integration of external meta-feature information directly into the modeling process for time-to-event outcomes, improves prediction performance when the external meta-feature data is informative. Importantly, when the external meta-features are uninformative, the prediction performance based on the regularized hierarchical model is on par with standard regularized Cox regression, indicating robustness of the framework. In addition to developing predictive signatures, the model can also be deployed in discovery applications where the main goal is to identify important features associated with the outcome rather than developing a predictive model.
... Some DEGs, however, are known to be involved in the pathogenesis of psoriasis (e.g., SERPINB3 and SERPINB4) [37,38]. To better illustrate the differences between the effect of DSC and SEC treatments, we performed semi-supervised hierarchical clustering of samples based on a panel consisting of 48 psoriasis-defining genes selected from a range of studies that compared the bulk transcriptome in NL skin with LS skin, in addition to some involved in TRM cell biology [39][40][41][42][43][44][45] (Table S5) (Figure 4c,d). The role of some of these genes in psoriasis has not been fully elucidated. ...
Article
Full-text available
Secukinumab and Dead Sea treatment result in clear skin for many psoriasis patients, through distinct mechanisms. However, recurrence in the same areas after treatments suggests the existence of a molecular scar. We aimed to compare the molecular and genetic differences in psoriasis patients who achieved complete response from secukinumab and Dead Sea climatotherapy treatments. We performed quantitative immunohistochemical and transcriptomic analysis, in addition to digital spatial profiling of skin punch biopsies. Histologically, both treatments resulted in a normalization of the lesional skin to a level resembling nonlesional skin. Interestingly, the transcriptome was not normalized by either treatments. We revealed 479 differentially expressed genes between secukinumab and Dead Sea climatotherapy at the end of treatment, with a psoriasis panel identifying SERPINB4, SERPINB13, IL36G, IL36RN, and AKR1B10 as upregulated in Dead Sea climatotherapy compared with secukinumab. Using digital spatial profiling, pan-RAS was observed to be differentially expressed in the microenvironment surrounding CD103+ cells, and IDO1 was differentially expressed in the dermis when comparing the two treatments. The differences observed between secukinumab and Dead Sea climatotherapy suggest the presence of a molecular scar, which may stem from mechanistically different pathways and potentially contribute to disease recurrence. This may be important for determining treatment response duration and disease memory.
... GSEA (v 4.0.3) [27] was used to acquire biomarkers related to dramatically enriched pathways based on the Hallmark background gene set. The single-sample GSEA (ssGSEA) algorithm was used to assess the abundance of immune cell infiltration in all samples from the two risk groups. ...
Article
Full-text available
Background Natural killer cells (NKs) may be involved in multiple myeloma (MM) progression. The present study elucidated the correlation between NKs and the progression of MM using single-cell binding transcriptome probes to identify NK cell-related biomarkers. Methods Single-cell analysis was performed including cell and subtype annotation, cell communication, and pseudotime analysis. Hallmark pathway enrichment analysis of NKs and NKs-related differentially expressed genes (DEGs) were conducted using Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and protein–protein interaction (PPI) networks. Then, a risk model was structured based on biomarkers identified through univariate Cox regression analysis and least absolute shrinkage and selection operator regression analysis and subsequently validated. Additionally, correlation of clinical characteristics, gene set enrichment analysis, immune analysis, regulatory network, and drug forecasting were explored. Results A total of 13 cell clusters were obtained and annotated, including 8 cell populations that consisted of NKs. Utilizing 123 PPI network node genes, 8 NK-related DEGs were selected to construct a prognostic model. Immune cell infiltration results suggested that 11 immune cells exhibited marked differences in the high and low-risk groups. Finally, the model was used to screen potential drug targets to enhance immunotherapy efficacy. Conclusion A new prognostic model for MM associated with NKs was constructed and validated. This model provides a fresh perspective for predicting patient outcomes, immunotherapeutic response, and candidate drugs.
... Recent advances in gene expression profiling have illuminated the pathogenesis of psoriasis [16,17]. Weighted gene co-expression network analysis (WGCNA), a robust method for associating genes with phenotypic traits, played a key role in revealing gene networks pertinent to psoriasis [18][19][20]. ...
Article
Full-text available
Psoriasis is a globally prevalent chronic inflammatory skin disease. This study aimed to scrutinize the hub genes related to inflammation and potential molecular mechanisms in psoriasis. Utilizing mRNA expression profiles from public datasets GSE13355, GSE78097, and GSE14905, we set up a comprehensive analysis. Initially, we selected differentially expressed genes (DEGs) from psoriasis and control samples in GSE13355, followed by calculating inflammatory indices using genomic set variation analysis (GSVA). Weighted gene co-expression network analysis (WGCNA) was then applied to link significant modules with the inflammatory index. This process helped us identify differentially expressed inflammation-related genes (DE-IRGs). A protein-protein interaction (PPI) network was established, with the molecular complex detection (MCODE) plug-in pinpointing six chemokine genes (CCR7, CCL2, CCL19, CXCL8, CXCL1, and CXCL2) as central hub genes. These genes demonstrated pronounced immunohistochemical staining in psoriatic tissues compared to normal skin. Notably, the CCR7 gene exhibited the highest potential for m6A modification sites. Furthermore, we constructed transcription factor-microRNA-mRNA networks, identifying 139 microRNAs and 52 transcription factors associated with the hub genes. For the LASSO logistic regression model, the area under the curve (AUC) in the training set was 1, and in the two validation cohorts GSE78097 and GSE14905 were 1 and 0.872, respectively. In conclusion, our study highlights six chemokine genes (CCR7, CCL2, CCL19, CXCL8, CXCL1, and CXCL2) as potential biomarkers in psoriasis, providing insights into the immune and inflammatory responses as pivotal instances in disease pathogenesis. These findings pave the way for exploring new therapeutic targets, particularly focusing on chemokine-associated pathways in psoriasis treatment.
... Anti-TNF-α therapies block the free soluble fraction and the membrane fraction of this cytokine. Anti-TNF has been shown to act more rapidly on Th17 populations than on Th1 populations, and the inhibition of the Th1 population appears to have a stronger correlation with clinical improvement [15][16]. TNF can be produced by various cells, including keratinocytes and Th1 and Th17 cells. ...
... In the GSE56815 dataset, the expression matrices of 34 MRG were extracted, and the MRG scores between OP and control samples were calculated using the gene set variation analysis (GSVA) package (version 1.42.0) (Suarez-Farinas et al., 2010). ...
Article
Full-text available
Background: Osteoporosis (OP) is a chronic bone metabolic disease and a serious global public health problem. Several studies have shown that mitophagy plays an important role in bone metabolism disorders; however, its role in osteoporosis remains unclear. Methods: The Gene Expression Omnibus (GEO) database was used to download GSE56815, a dataset containing low and high BMD, and differentially expressed genes (DEGs) were analyzed. Mitochondrial autophagy-related genes (MRG) were downloaded from the existing literature, and highly correlated MRG were screened by bioinformatics methods. The results from both were taken as differentially expressed (DE)-MRG, and Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed. Protein-protein interaction network (PPI) analysis, support vector machine recursive feature elimination (SVM-RFE), and Boruta method were used to identify DE-MRG. A receiver operating characteristic curve (ROC) was drawn, a nomogram model was constructed to determine its diagnostic value, and a variety of bioinformatics methods were used to verify the relationship between these related genes and OP, including GO and KEGG analysis, IP pathway analysis, and single-sample Gene Set Enrichment Analysis (ssGSEA). In addition, a hub gene-related network was constructed and potential drugs for the treatment of OP were predicted. Finally, the specific genes were verified by real-time quantitative polymerase chain reaction (RT-qPCR). Results: In total, 548 DEGs were identified in the GSE56815 dataset. The weighted gene co-expression network analysis(WGCNA) identified 2291 key module genes, and 91 DE-MRG were obtained by combining the two. The PPI network revealed that the target gene for AKT1 interacted with most proteins. Three MRG (NELFB, SFSWAP, and MAP3K3) were identified as hub genes, with areas under the curve (AUC) 0.75, 0.71, and 0.70, respectively. The nomogram model has high diagnostic value. GO and KEGG analysis showed that ribosome pathway and cellular ribosome pathway may be the pathways regulating the progression of OP. IPA showed that MAP3K3 was associated with six pathways, including GNRH Signaling. The ssGSEA indicated that NELFB was highly correlated with iDCs (cor = −0.390, p < 0.001). The regulatory network showed a complex relationship between miRNA, transcription factor(TF) and hub genes. In addition, 4 drugs such as vinclozolin were predicted to be potential therapeutic drugs for OP. In RT-qPCR verification, the hub gene NELFB was consistent with the results of bioinformatics analysis. Conclusion: Mitophagy plays an important role in the development of osteoporosis. The identification of three mitophagy-related genes may contribute to the early diagnosis, mechanism research and treatment of OP.
... Subsequently, different biologics such as Alefacept [31], Fontolizumab (anti-IFN-γ) and Infliximab [32], have been evaluated to further investigate the immune mechanism of psoriasis. With the emergence of new technologies, they have also introduced gene set enrichment analysis (GSEA) [33], RNA-seq [34], and Single-cell RNA seq [35] to study the mechanisms underlying psoriasis. In 2022, they noted that the small-molecule targeted drug Janus kinase (JAK) 1-3 inhibitor was effective against moderate to severe psoriasis, but safety concerns remain, and no JAK inhibitor has received regulatory approval for psoriasis. ...
Article
Full-text available
Background It is now understood that T cells play a key role in the occurrence and development of psoriasis. Herein, a bibliometric analysis was conducted to summarize the content and trends of T cell-related research in psoriasis. Methods A bibliometric analysis was conducted on publications pertaining to T cells in psoriasis between 2003 and 2022 retrieved from the Web of Science Core Collection (WoSCC) database using tools such as CiteSpace, the Bibliometrix R package, and VOSviewer. Results The study included a total of 3595 articles authored by 14,188 individuals, including all coauthors in article bylines. The Laboratory for Investigative Dermatology at Rockefeller University, led by James G Krueger, has made significant contributions to this field through focusing on the pathogenesis of psoriasis and exploring the potential of using biological agents to treat psoriasis. Furthermore, targeted inhibitors have significantly impacted the treatment of psoriasis, with researchers focusing on small-molecule targeted drugs as a new area of research that could potentially replace biological agents. Conclusions Research has established the efficacy and long-term safety of targeted inhibition of T cell-related targets. Deucravacitinib, a psoriasis treatment drug targeting TYK2 as an allosteric inhibitor, has attracted significant attention and raised high expectations.
... Serine proteases are pivotal for maintaining skin barrier homeostasis. SERPINB3, a serine protease inhibitor, is highly expressed in the skin and serum of patients with psoriasis and is positively correlated with disease severity [32][33][34]. In an in vitro study of psoriasis, the expression of in ammatory chemokines such as CXCL1, CXCL5, and CXCL8 in keratinocytes was suppressed when SERPINB3 was knocked out [34]. ...
Article
Background: Psoriasis is a chronic immune-mediated skin disease. However, the pathogenesis is not yet well established. Objectives: This study aimed to screen psoriasis biomarker genes and analyse their significance in immune cell infiltration. Materials & methods: GSE13355 and GSE14905 datasets were downloaded from Gene Expression Omnibus (GEO) as training groups to establish the model. GSE30999 obtained from GEO was used to validate the model. Differential expression and multiple enrichment analyses were performed on 91 psoriasis samples and 171 control samples from the training group. The "LASSO" regression model and support vector machine model were used to screen and verify genes implicated in psoriasis. Genes with an area under the ROC curve >0.9 were selected as candidate biomarkers and verified in the validation group. Differential analysis of immune cell infiltration was performed on psoriasis and control samples using the "CIBERSORT" algorithm. Correlation analyses between the screened psoriasis biomarkers and 22 types of immune cell infiltration were performed. Results: In total, 101 differentially expressed genes were identified, which were mainly shown to be involved in regulating cell proliferation and immune functions. Three psoriasis biomarkers, BTC, IGFL1, and SERPINB3, were identified using two machine learning algorithms. These genes showed high diagnostic value in training and validation groups. The proportion of immune cells during immune infiltration differed between psoriasis and control samples, which was associated with the three biomarkers. Conclusion: BTC, IGFL1, and SERPINB3 are associated with the infiltration of multiple immune cells, and may therefore be used as biomarkers for psoriasis.
Article
Full-text available
Pancreatic cancer shows malignancy around the world standing in 4th position for causing death globally. This cancer is majorly divided into exocrine and neuroendocrine where exocrine pancreatic ductal adenocarcinoma is observed to be nearly 85% of cases. The lack of diagnosis of pancreatic cancer is considered to be one of the major drawbacks to the prognosis and treatment of pancreatic cancer patients. The survival rate after diagnosis is very low, due to the higher incidence of drug resistance to cancer which leads to an increase in the mortality rate. The transcriptome analysis for pancreatic cancer involves dataset collection from the ENA database, incorporating them into quality control analysis to the quantification process to get the summarized read counts present in collected samples and used for further differential gene expression analysis using the DESeq2 package. Additionally, explore the enriched pathways using GSEA software and represented them by utilizing the enrichment map finally, the gene network has been constructed by Cytoscape software. Furthermore, explored the hub genes that are present in the particular pathways and how they are interconnected from one pathway to another has been analyzed. Finally, we identified the CDKN1A, IL6, and MYC genes and their associated pathways can be better biomarker for the clinical processes to increase the survival rate of of pancreatic cancer.
Article
Full-text available
Fat intake is among the most significant triggers for symptom development in patients with irritable bowel syndrome (IBS). Nevertheless, long-term restriction in fatty foods ingestion may lead to nutritional inadequacies. This study aimed to identify the crucial genes involved in lipid-induced gastrointestinal symptoms, contributing to helping IBS patients regulate fat. The clinical characteristics of the subjects were collected by questionnaire investigation and analyzed using multivariate logistic regression. Differentially expressed genes (DEG) and signaling pathways were analyzed by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. ImmuInfiltration and CIBERSORT packages evaluated small intestine immune cell infiltration. Random forest and SVM-RFE algorithms were used to select hub genes. A receiver operating characteristic curve was used to access the diagnostic significance of each hub gene. Gene Set Enrichment Analysis (GSEA) was performed to identify hub genes’ molecular processes in IBS development after lipid infusion. IBS patients’ risk, severity, and quality of life increased with fat intake. In total, 116 robust DEGs were identified in IBS patients after lipid infusion using the GSE166869 dataset and were mainly clustered in the immune and inflammatory pathways. IBS patients had greater Neutrophils, CD4⁺ T cells, and M1 Macrophages than healthy controls. Furthermore, infiltration levels of Neutrophils and resting memory CD4⁺ T cells were inversely related to the expression of hub genes (IGKV1D-43, IGKV1-12, APOD, FCGR2A and IGKV2-29). After lipid infusion, GSEA results of each hub gene indicated the relevance of proinflammatory pathways in IBS pathogenesis. After verification, only APOD and FCGR2A were stably downregulated in small intestinal mucosa and plasma of IBS patients. The area under the curve of APOD combined with FCGR2A expression was 0.9. APOD and FCGR2A may be promising biomarkers for IBS diagnosis and lipid-sensitive IBS patients. Their potential roles in the immune microenvironment of the small intestinal mucosa may provide a vital clue to IBS precision therapy.
Article
Full-text available
Gene set enrichment analysis (GSEA) is a statistical method to determine if predefined sets of genes are differentially expressed in different phenotypes. Predefined gene sets may be genes in a known metabolic pathway, located in the same cytogenetic band, sharing the same Gene Ontology category, or any user-defined set. In microarray experiments where no single gene shows statistically significant differential expression between phenotypes, GSEA has identified significant differentially expressed sets of genes, even where the average difference in expression between two phenotypes is only 20% for genes in the gene set. The gene set identified in the first GSEA analysis (oxidative phosphorylation genes differentially expressed in diabetic versus non-diabetic patients) was subsequently confirmed by independent laboratory studies published in the New England Journal of Medicine. Since the first paper on GSEA was published, many extensions and alternative methods have been described in the literature. In this paper, we describe the original GSEA algorithm, subsequent extensions and alternatives, results of some of the applications, some limitations of the methods and caveats for users, and possible future research directions. GSEA and related methods are complementary to conventional single-gene methods. Single gene methods work best when individual genes have large effects and there is small variance within the phenotype. GSEA is likely to be more powerful than conventional single-gene methods for studying the large number of common diseases in which many genes each make subtle contributions. It is a tool that deserves to be in the toolbox of bioinformatics practitioners.
Article
Full-text available
Reproducibility is a fundamental requirement in scientific experiments. Some recent publications have claimed that microarrays are unreliable because lists of differentially expressed genes (DEGs) are not reproducible in similar experiments. Meanwhile, new statistical methods for identifying DEGs continue to appear in the scientific literature. The resultant variety of existing and emerging methods exacerbates confusion and continuing debate in the microarray community on the appropriate choice of methods for identifying reliable DEG lists. Using the data sets generated by the MicroArray Quality Control (MAQC) project, we investigated the impact on the reproducibility of DEG lists of a few widely used gene selection procedures. We present comprehensive results from inter-site comparisons using the same microarray platform, cross-platform comparisons using multiple microarray platforms, and comparisons between microarray results and those from TaqMan - the widely regarded "standard" gene expression platform. Our results demonstrate that (1) previously reported discordance between DEG lists could simply result from ranking and selecting DEGs solely by statistical significance (P) derived from widely used simple t-tests; (2) when fold change (FC) is used as the ranking criterion with a non-stringent P-value cutoff filtering, the DEG lists become much more reproducible, especially when fewer genes are selected as differentially expressed, as is the case in most microarray studies; and (3) the instability of short DEG lists solely based on P-value ranking is an expected mathematical consequence of the high variability of the t-values; the more stringent the P-value threshold, the less reproducible the DEG list is. These observations are also consistent with results from extensive simulation calculations. We recommend the use of FC-ranking plus a non-stringent P cutoff as a straightforward and baseline practice in order to generate more reproducible DEG lists. Specifically, the P-value cutoff should not be stringent (too small) and FC should be as large as possible. Our results provide practical guidance to choose the appropriate FC and P-value cutoffs when selecting a given number of DEGs. The FC criterion enhances reproducibility, whereas the P criterion balances sensitivity and specificity.
Article
Full-text available
Psoriasis is an immune-mediated disease characterized by aberrant epidermal differentiation, surface scale formation, and marked cutaneous inflammation. To better understand the pathogenesis of this disease and identify potential mediators, we used whole genome array analysis to profile paired lesional and nonlesional psoriatic skin and skin from healthy donors. We observed robust overexpression of type I interferon (IFN)-inducible genes and genomic signatures that indicate T cell and dendritic cell infiltration in lesional skin. Up-regulation of mRNAs for IFN-alpha subtypes was observed in lesional skin compared with nonlesional skin. Enrichment of mature dendritic cells and 2 type I IFN-inducible proteins, STAT1 and ISG15, were observed in the majority of lesional skin biopsies. Concordant overexpression of IFN-gamma and TNF-alpha-inducible gene signatures occurred at the same disease sites. Up-regulation of TNF-alpha and elevation of the TNF-alpha-inducible gene signature in lesional skin underscore the importance of this cytokine in psoriasis; these data describe a molecular basis for the therapeutic activity of anti-TNF-alpha agents. Furthermore, these findings implicate type I IFNs in the pathogenesis of psoriasis. Consistent and significant up-regulation of type I IFNs and their associated gene signatures in psoriatic skin suggest that type I IFNs may be potential therapeutic targets in psoriasis treatment.
Article
Full-text available
Motivation: Differentially expressed gene (DEG) lists detected from different microarray studies for a same disease are often highly inconsistent. Even in technical replicate tests using identical samples, DEG detection still shows very low reproducibility. It is often believed that current small microarray studies will largely introduce false discoveries. Results: Based on a statistical model, we show that even in technical replicate tests using identical samples, it is highly likely that the selected DEG lists will be very inconsistent in the presence of small measurement variations. Therefore, the apparently low reproducibility of DEG detection from current technical replicate tests does not indicate low quality of microarray technology. We also demonstrate that heterogeneous biological variations existing in real cancer data will further reduce the overall reproducibility of DEG detection. Nevertheless, in small subsamples from both simulated and real data, the actual false discovery rate (FDR) for each DEG list tends to be low, suggesting that each separately determined list may comprise mostly true DEGs. Rather than simply counting the overlaps of the discovery lists from different studies for a complex disease, novel metrics are needed for evaluating the reproducibility of discoveries characterized with correlated molecular changes. Supplementaty information: Supplementary data are available at Bioinformatics online.
Article
Approximately 2% of the Caucasian population is affected by psoriasis (PS); a chronic inflammatory skin disease triggered by both genetic and environmental risk factors. In addition to a major contribution from the HLA class I region, PS susceptibility loci have been mapped to a number of regions including 1q21, 3q21, 4qter, 14q31-q32, 17q24-q25, 19p13.3 and 20p. Some of these overlap with loci implicated in other autoimmune/inflammatory diseases. Global gene expression studies are beginning to provide insights into the etiology of these and other complex diseases. We used Affymetrix oligonucleotide arrays comprising approximately 12 000 known genes to initiate a more comprehensive analysis of the transcriptional changes that occur in involved and uninvolved skin of 15 psoriatic patients versus six normal controls. Expression levels of the transcripts detected on the arrays were first used to determine the relationship of samples to each other using hierarchical clustering. This analysis clearly differentiated involved psoriatic skin from uninvolved and normal skin. Clusters of differentially expressed genes with similar expression patterns in the same samples were then identified. Six out of 32 clusters contained a total of 177 transcripts that were differentially expressed in involved psoriatic skin versus normal skin. These differences were independent of the gender, age, skin site and HLA class I status of the patient. Ten of the 177 genes were also differentially expressed in uninvolved skin, and several mapped to regions previously shown to harbor psoriasis susceptibility loci.