Sei sulla pagina 1di 8

Chen and Wang Journal of Clinical Bioinformatics 2011, 1:35 JOURNAL OF

http://www.jclinbioinformatics.com/content/1/1/35 CLINICAL BIOINFORMATICS

REVIEW Open Access

Significance of bioinformatics in research of


chronic obstructive pulmonary disease
Hong Chen1 and Xiangdong Wang1,2*

Abstract
Chronic obstructive pulmonary disease (COPD) is an inflammatory disease characterized by the progressive
deterioration of pulmonary function and increasing airway obstruction, with high morality all over the world. The
advent of high-throughput omics techniques provided an opportunity to gain insights into disease pathogenesis
and process which contribute to the heterogeneity, and find target-specific and disease-specific therapies. As an
interdispline, bioinformatics supplied vital information on integrative understanding of COPD. This review focused
on application of bioinformatics in COPD study, including biomarkers searching and systems biology. We also
presented the requirements and challenges in implementing bioinformatics to COPD research and interpreted
these results as clinical physicians.
Keywords: bioinformatics, genomics, proteomics, chronic obstructive pulmonary disease, biomarkers

Introduction methodologies, such as genomics and proteomics, are


Chronic obstructive pulmonary disease (COPD) is an commonly used. The variety of data from biology,
inflammatory disease characterized by the progressive mainly in the form of DNA, RNA and protein sequences
deterioration of pulmonary function and increasing air- is putting heavy demand in computer sciences and com-
way obstruction [1,2]. It can be caused by inflammatory putational biology. Bioinformatics, including many sub-
responses triggered by noxious particles or gas, most disciplines, such as genomics, proteomics and system
commonly from tobacco smoking and is accompanied biology, is an integration of mathematical, statistical,
by chronic bronchitis and emphysema [3,4]. Some and computational methods to analyze biological, bio-
patients go on to require long-term oxygen therapy or chemical, and biophysical data. Compared to wet-lab
even lung transplantation [3]. COPD was ranked as method, bioinformatics focused on data mining via com-
fourth leading cause of death worldwide and is esti- putational means. Sophisticated bioinformatics techni-
mated to become the top third cause of mortality by ques are developed to analyze the vast amount of data
2020 [5]. According to the data in China, COPD ranks generated from genomics and proteomics studies, such
as the fourth leading cause of death in urban areas and as gene and protein function, interactions and metabolic
third in rural areas[6]. The high mortality and morbidity and regulatory pathways. However, there is still a great
with COPD, and its chronic progressive nature, have challenge to combine the computer figures with clinical
promoted the need to investigate the underlying data for both bench-scientists and bedside-physicians.
mechanisms and identify biomarkers for diagnosis, prog- In COPD studies, there are usually three ways to ana-
nosis and drug target. lyze ‘omics’ data: 1) search correlation between single
The understanding of COPD increased by advanced gene or protein and some clinical features in order to
molecular biology approaches, genetically modified ani- find diagnostic or prognostic biomarkers; 2) integrate
mals, virally administered genes, and high-throughput clinical and wet-lab information, or omics data from dif-
transcriptional profiling approaches. High-throughput ferent levels for database establishment and computa-
tional models. In this current review, we discussed
application of bioinformatics in COPD study. We also
* Correspondence: xiangdong.wang@telia.com
1
Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University,
presented the requirements and challenges in imple-
Shanghai, China menting bioinformatics to COPD research, and gave
Full list of author information is available at the end of the article

© 2011 Chen and Wang; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative
Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly cited.
Chen and Wang Journal of Clinical Bioinformatics 2011, 1:35 Page 2 of 8
http://www.jclinbioinformatics.com/content/1/1/35

some suggestions on how to interprete these results as case-control genomics studies focused on the association
clinical physicians. of several gene alleles with lung function. 105V/114V
alleles of GSTP1 and 113H/139H alleles of mEPHX and
Application of bioinformatics in biomarker the combination of genotypes with same alleles were
searching associated with imbalanced oxidative stress and lung
The diagnosis of COPD is based on the presence of function in patients [19]. Polymorphisms in ADAM33
typical symptoms of cough and shortness of breath, were associated with COPD and lung function decline
together with the presence of risk factors, and is con- in long-term smokers [20] and general population [21].
firmed by spirometry. Therefore, searching for better The variants and their combinations of eNOS -786C,
biomarkers with high specificity and sensitivity indicat- -922G, and 4A alleles in endothelial cells contribute to
ing the staging and severity of COPD remain as major disturbed pulmonary function and oxidative stress in
concerns for clinical physicians. The main value of bio- COPD[22].
markers in COPD would be in early diagnosis and to An interleukin13 polymorphism in the promoter
provide the early proof of drug efficacy during the treat- region may modulate the adverse effects of cigarette
ment [7]. As a biomarker for COPD, it is expected to be smoking on pulmonary function in long-term cigarette
detected in human lung fluids or tissues, sensitive to the smokers [23]. These genomics findings suggested that
progress of COPD, disease-specific to COPD and asso- environmental influences were important in COPD.
ciated with the status of patients [8]. In these researches, Genetic polymorphisms contributed to the development
selected genes or proteins usually combined with several of COPD, especially to the declined lung function. The
clinical features, such as disease susceptibility, lung diversity in human genes could help us to understand
function, via statistical methods, i.e. logistic regression. the susceptibility among different ethics and different
populations. The dissection of the genetic basis of com-
Genomics plex diseases and the development of highly individua-
It is believed that many genetic factors increase a per- lized therapies remain lofty but achievable goals [24].
son’s risk of developing COPD[9]. The high mortality
and morbidity associated with COPD, and its chronic Proteomics
and progressive nature, has prompted the use of mole- Proteomics is the systematic study of the many and
cular genetic studies in an attempt to identify suscept- diverse properties of protein profiles in a parallel man-
ibility factors for the disease. The advent of high- ner with the aim of providing detailed descriptions of
throughput methodologies to study genetic background the structure, function and control of biological systems
variability, epigenetic regulation allowed us to explain in health and disease [25]. A major research objective is
the individual variability in the susceptibility of human to search for biomarkers in complex biological fluids.
diseases. The proteomic analysis highlights the avenues to inves-
Single nucleotide polymorphism (SNP) was a common tigate protein profiles of cells, biopsies and fluids,
method in COPD study. These analyses usually study a explore protein-based mechanisms of human diseases,
group of candidate genes, and then perform statistical define subgroups of disease, and identify novel biomar-
test in different populations. The gene-related suscept- kers for diagnosis, therapy and prognosis of multiple
ibility can be approached by testing or unbiased study diseases and discover new targets for drug development.
designs [10]. By SNP microarray, many genetic factors In particular, the application of complementary
were found to be related with the individual risk of approaches, including gel- and liquid chromatography
developing COPD [9]. Apart from recognized deficiency mass spectrometry-based proteomic techniques on spu-
of alpha1antitrypsin [9], genomics in COPD found that tum and/or bronchoalveolar lavage may provide a better
other gene alleles, such as IREB2[11], CYP2E1 and understanding of the proteome differentially expressed
NAT2[12], CYP1A1, CYP1A2 and CYBA[13], TNF-a among the courses, severities and populations of COPD
[14] were associated with COPD susceptibility. PIM3 [7]. We have previously reviewed the clinical studies on
allele of the alpha1antitrypsin gene had an association COPD proteomics, highlighted the proteomic-oriented
with the pathogenesis of COPD in the Indian population methods applied and evaluated the diagnostic or prog-
[15]. The polymorphisms in SP-A1 and SP-A2[16], nostic values of potential biomarkers [8]. Those studies
COX2 and p53 risk-alleles [17]might be genetic factors mainly focused on disease classification, biomarker
contributing to the susceptibility to COPD. On the detection, or identification of mechanism, while the
other hand, COPD could influence single gene expres- three components are related with each other in COPD
sion as well, such as cathepsin inhibitory cystatin A[18]. (Figure 1).
COPD is featured by decline in lung function in dis- An important goal of proteomic studies is to under-
ease progression. Therefore, except susceptibility, other stand biological roles of specific proteins and develop
Chen and Wang Journal of Clinical Bioinformatics 2011, 1:35 Page 3 of 8
http://www.jclinbioinformatics.com/content/1/1/35

Metabolomics
Metabolomics is a global way to understanding regula-
tion of metabolic pathways and metabolic networks of a
biologic system[36]. Metabolites trigonelline, hippurate
and formate in urinary were identified to be associated
with baseline lung function of COPD patients and con-
sidered to reflect lifestyle differences affecting overall
health [37]. Another way to analyze omics data is clus-
tering data to form a specific pattern for different
groups by principal component analysis (PCA). Combi-
nation of PCA and metabolomics identified the meta-
bolic fingerprint of exhaled breath condensate of COPD
patients [38].

Multiplexed ELISA
A recent advancement in ELISA is the multiplexed
ELISA which could determine multiple proteins within
a single tissue sample. It has recently been shown to be
more sensitive than standard ELISA once optimized for
a particular cytokine [39-41] and could be a promising
Figure 1 Overview of the utility of bioinformatics in chronic diagnostic assay in lung diseases [42]. Moreover, the
obstructive pulmonary disease. Both genomics and proteomics
measurement of multiple cytokines is required for many
provide information on candidates. By searching in various datasets
and combining with clinical profiles, ‘omis’ studies may help to diseases, particularly those like COPD that arise from a
explain questions on disease classification, biomarker detection, and complex process of initiation and progression of inflam-
identification of mechanism. mation network. Simultaneous detection of multiple
cytokines will undoubtedly provide a more powerful
tool to quantifiably measure cytokines in different stages
new therapeutic targets [26]. Although there are few of COPD. With help of this high-throughput measure-
proteomics studies performed in COPD patients, several ment platform, we could integrate both biologic and
potential proteins have been regarded as biomarkers. clinical data to inform predictive multiscale models ran-
For example, matrix metalloproteinase (MMP)-13 and ging from the molecular to the organ levels, as shown in
thioredoxin-like 2 in lungs increased in patients with Figure 2. For example, the integration of IL-9 pathway
COPD [27]. The serine and MMP proteinase network and CCR3 pathway between biological function and
was considered as an important feature in predicting pathology demonstrated cell proliferation-related remo-
clinical worsening of airway obstruction [28]. Pulmonary deling, intracellular signal-associated inflammatory
surfactant A was found to link to the pathogenesis of responses and over-activation of kinases-correlated
COPD and could be considered as a potential COPD emphysema in the pathogenesis of COPD. We propose
biomarker [29]. Proteomic screening of sputum yields this new way will increase our insight into disease pro-
potential biomarkers of inflammation [30]. Airway and cess and have great potential to identify new biomarkers
parenchymal phenotypes of COPD were suggested to be for disease diagnosis as well as novel therapeutic targets.
associated with unique systemic serum biomarker pro-
files [31]. The utility of proteomic profiling would Systems biology and database establishment
improve the understanding of molecular mechanisms The difficulties encountered whilst exploring pathogen-
involved in cigarette smoking-related COPD by identify- esis and searching for biomarkers may be due in part to
ing plasma proteins that correlate with declined lung the complex nature of COPD, which comprises a broad
function [32]. The concentrations of neutrophil defen- spectrum of histopathological findings and respiratory
sins 1 and 2, calgranulin A, and calgranulin B were ele- symptoms [43]. All genetic information and molecular
vated in smokers with COPD when compared to knowledge need to be semantically incorporated and
asymptomatic smokers [33]. Other candidates, like associated with clinical and experimental data. System
serum amyloid A [34], plasma retinal-binding protein, biology in COPD (as reviewed before[44]) presented a
apolipoprotein E, inter-alpha-trypsininhibitor heavy manifold understanding of the complexity of COPD,
chain H4, and glutathione peroxidase[35], were also therefore advanced biomedical research and drug devel-
been detected in plasma in COPD patients by proteo- opment. This approach relies on global genome, tran-
mics approaches. scriptome, proteome, and metabolome data sets
Chen and Wang Journal of Clinical Bioinformatics 2011, 1:35 Page 4 of 8
http://www.jclinbioinformatics.com/content/1/1/35

Figure 2 The integration of IL-9 pathway and CCR3 pathway between biological function and pathology demonstrated cell
proliferation-related remodeling, intracellular signal-associated inflammatory responses and over-activation of kinases-correlated
emphysema in the pathogenesis of COPD.

collected in cross-sectional patient cohorts with high- methods (KHM) to study a large dataset, acquired from
throughput measurement platforms and integrated with 415 COPD patients, to assess the presence of hidden
biologic and clinical data to inform predictive multidi- structures in data corresponding to the different COPD
mensional models ranging from the molecular to the phenotypes observed in clinical practice. This study may
organ levels[45]. be considered as a methodological example showing
Comandini etc. [46] assayed a number of published possible applications of intelligent data analysis and
studies by creating a smoker datasets on which to per- visual exploratory techniques to investigate clinical
form data-mining analysis. They utilized Ingenuity Path- aspects of chronic pathologies where a mathematical
ways Analysis, a web-based application that enables referring model is generally missing[48].
identifying relationships, biological mechanisms, func-
tions, and pathways of relevance associated with the How to understand bioinformatics as clinical
molecules under study. Their findings supported the physicians
central role of anti-oxidant genes in smoking population COPD has been approached by genomic and proteomic
and suggested Nrf2 may be a COPD risk biomarker. A technologies to allow us to identify patterns of gene/
brand new knowledge base was generated from clinical protein expression that track with clinical disease or to
and experimental data for COPD based on BioXM soft- identify new pathways involved in disease pathogenesis.
ware platform [47]. This integrated database reduced The results from these initial studies highlight the
implementation time and effort for the knowledge base potential for these omics approaches to reveal novel
compared to similar systems and provided a free, com- insights into the pathogenesis of COPD and provide
prehensive, easy to use resource for all COPD related new tools to improve diagnosis, clinical classification,
clinical research. course prediction, and response to therapy. Existing
Clinical profiles could also be considered as a form of knowledge such as genotype-phenotype relations or sig-
omics data since it provided a large quantity of patients’ nal transduction pathways must be semantically inte-
information in a direct conservative way. An in-silico grated and dynamically organized into structured
research applied various explorative analysis techniques networks that are connected with clinical and experi-
(PCA, MCA, MDS) and unsupervised clustering mental data. This will require collaboration among
Chen and Wang Journal of Clinical Bioinformatics 2011, 1:35 Page 5 of 8
http://www.jclinbioinformatics.com/content/1/1/35

multidisciplinary groups with expertise in the respective


technologies, bioinformatics, and clinical medicine for
the disease. More and more clinical physicians began to
realize the promise of these studies and the potential to
revolutionize the diagnosis and treatment of COPD,
while obstacles still existed between these laboratory
findings and their applications in clinical practice.
Omics results need to be interpreted by translational
medicine and systems biology. Since the vocabulary of
systems biology is different than that of molecular biol-
ogy or clinical research, the biggest challenge is the shift
in thinking[49].
Instead of a single-factor approach, which is highly
effective in the lab, we need to think globally as clinical
physicians. We need to shift from an approach that tries
Figure 3 Bioinformatics begin to take part in COPD
to explain lung pathogenesis by “one molecule, one cell investigation. The collaboration of epidemiologists, clinicians,
type” to approach that looks at the network of interac- geneticists and specialists in bioinformatics, in addition to specialists
tions between multiple molecules, pathways, and cells. in disciplines less familiar to epidemiologists, is critical for further
Given that all the samples were collected from human, study. More co-operations are still needed.
it would be of great significance to standardize patient
groups. Criteria of clinical informatics and medical
informatics, including age, gender, smoking history, sta- need to explore the COPD-specific and/or related tran-
ging, complications and clinical signs as well as exami- scriptional factors and regulation networks generated
nations, should be fully considered before and after any from omics and bioinformatics like in other diseases
omics investigation. We also need to pay attention to [41].
the relation between clinical data and laboratory find-
ings. For this to occur, a well-done history and physical Conclusions
examination would be helpful to supplement these The use of high-throughput techniques for gene and
laboratory figures by providing multiple features of protein expression profiling and of computerized data-
human COPD. bases has become a mainstay of biomedical research.
Although all of these exciting technological advances There is a need to perform omics studies on patients
that exponentially increase the levels of knowledge with COPD, describing the association with the disease
about every disease and model serve as facilitators of in terms of specificity, severity, progress and prognosis
integration, they do not inherently provide integrative and monitoring the efficacy of therapies. These omics
models of disease. Therefore, we proposed that digitaliz- analysis highlight the ways to investigate protein profiles
ing essential clinical profiles, such as symptoms and of cells, biopsies and fluids, explore protein-based
signs, by questionnaires and/or scores, would provide mechanisms of human diseases, identify novel biomar-
direct vision for physicians and shrink the distance kers for diagnosis, therapy and prognosis of multiple
between lab discovery and clinical condition. The com- diseases and discover new targets for drug development,
bination of epidemiologists, clinicians, geneticists and as shown in Figure 3. Although the number of clinical
specialists in bioinformatics, in addition to specialists in studies on COPD is limited, they still serve as the out-
disciplines less familiar to epidemiologists, is critical to standing initiation for proteomic research in such a
be prepared for new phenotypic characterizations based complex disease. The analysis of protein profiles that
on transcriptome and proteome [50]. Even though we are up- or down-regulated, modified, secreted in the air-
have spotted considerable advancement in bioinfor- ways during the disease may yield vital evidences to
matics, it still calls for more collaboration to fulfill its understand the pathogenesis and discover new therapeu-
potential (Figure 3). Interdisciplinary teams should allow tic targets for the disease (Figure 4) [52]. With many
us to access omics datasets integratively and generate a guidelines now in place and model studies on which to
global model of COPD. It is important to have a special design future experiments, there is reason to be optimis-
attention from proteomic scientists to explore the com- tic that candidate protein biomarkers will be discovered
bination between advanced proteomic biotechnology, using proteomics and translated into clinical assays[53].
clinical proteomics, tissue imaging and profiling, and With better study design standardization and the imple-
organ dysfunction score systems, to improve the clinical mentation of novel technologies to reach the optimal
outcomes of these patients [51]. There is still a great research standard, there is enough reason be optimistic
Chen and Wang Journal of Clinical Bioinformatics 2011, 1:35 Page 6 of 8
http://www.jclinbioinformatics.com/content/1/1/35

Lungs Laser Capture Microdissection:


Tissue frozen section Isolation of Specific Cells Selected cells

Statistical analysis
Resources
Probe Set Gene
100057_at Cluster Incl AI838320:UI-M-AO0-aby-f-05-0-UI.s1 Mus musculus cDNA, 3 end /clone=UI-M-AO0-aby-f-05-0-UI /clone_end=3 /gb=AI838320 /gi=5472533 /ug=Mm.4467 /len=529
100059_at cytochrome b-245, alpha polypeptide
100069_at cytochrome P450, 2f2
100311_f_at eosinophil-associated ribonuclease 1
100331_g_at thioredoxin peroxidase, pseudogene 1
100380_at H3 histone, family 3A
100397_at TYRO protein tyrosine kinase binding protein
100569_at annexin A2
100583_at Cluster Incl J00475:Mouse germline IgH chain gene, DJC region- segment D-FL16.1 /cds=(0,1033) /gb=J00475 /gi=194417 /ug=Mm.6160 /len=1034
100597_at glycogenin 1
100711_at ribosomal protein L10A

Criteria for genes


100944_at Cluster Incl AA958903:ua19f08.r1 Mus musculus cDNA, 5 end /clone=IMAGE-1347207 /clone_end=5 /gb=AA958903 /gi=3125133 /ug=Mm.6381 /len=384
100946_at neuraminidase 1
101019_at cathepsin C
101045_at hydroxyacyl-Coenzyme A dehydrogenase, type II
101048_at protein tyrosine phosphatase, receptor type, C
101137_at ribosomal protein S3
101212_at ribosomal protein S7
101482_at protein phosphatase 1, catalytic subunit, gamma isoform
101656_f_at Cluster Incl U60442:Mus musculus Ig kappa light chain mRNA, partial cds /cds=(0,363) /gb=U60442 /gi=1421746 /ug=Mm.30406 /len=391

Differentially
101753_s_at P lysozyme structural
101781_f_at Cluster Incl V00754:Mouse gene coding for embryonic H3 histone /cds=(0,407) /gb=V00754 /gi=51297 /ug=Mm.57118 /len=408
101876_s_at histocompatibility 2, T region locus 10
101881_g_at procollagen, type XVIII, alpha 1
101908_s_at CEA-related cell adhesion molecule 2
101955_at heat shock 70kD protein 5 (glucose-regulated protein, 78kD)
101973_at Cbp/p300-interacting transactivator, w ith Glu/Asp-rich carboxy-terminal domain, 2
102029_at interleukin 16
102104_f_at Cluster Incl AI504305:vl04g11.x1 Mus musculus cDNA, 3 end /clone=IMAGE-963236 /clone_end=3 /gb=AI504305 /gi=4402156 /ug=Mm.42485 /len=580

cDNA Microarray:
102156_f_at Cluster Incl M80423:Mus castaneus IgK chain gene, C-region, 3 end /cds=(0,322) /gb=M80423 /gi=196865 /ug=Mm.46804 /len=323

expressed
102196_at Cluster Incl U37413:Guanine nucleotide binding protein, alpha 11 /cds=(57,1136) /gb=U37413 /gi=1051237 /ug=Mm.989 /len=1342
102207_at delta-like homolog (Drosophila)
102208_at sialyltransferase 10 (alpha-2,3-sialyltransferase VI)
102217_at G protein-coupled receptor kinase 5
102372_at Cluster Incl M90766:Mouse Ig active joining chain (J chain) mRNA of the b allele, complete cds /cds=(21,500) /gb=M90766 /gi=194473 /ug=Mm.1192 /len=501
102585_f_at Cluster Incl AB017349:Mus musculus mRNA for immunoglobulin light chain V region, partial cds /cds=(0,353) /gb=AB017349 /gi=4519566 /ug=Mm.57251 /len=360
102641_at SFFV proviral integration 1
102767_at EST AA536815

Key Genes
102806_g_at CEA-related cell adhesion molecule 1
102906_at T-cell specific GTPase
102920_at Cluster Incl AW215585:up06c04.x1 Mus musculus cDNA, 3 end /clone=IMAGE-2651238 /clone_end=3 /gb=AW215585 /gi=6526261 /ug=Mm.1622 /len=329
102995_s_at granzyme A
103035_at ATP-binding cassette, sub-family B (MDR/TAP), member 2
103089_at CD48 antigen
103200_at Cluster Incl AA711773:vu58g05.r1 Mus musculus cDNA, 5 end /clone=IMAGE-1195640 /clone_end=5 /gb=AA711773 /gi=2721691 /ug=Mm.1902 /len=473
103240_f_at eosinophil-associated ribonuclease 3
103448_at S100 calcium binding protein A8 (calgranulin A)
103462_at Cluster Incl AW122239:UI-M-BH2.2-aov-f-02-0-UI.s1 Mus musculus cDNA, 3 end /clone=UI-M-BH2.2-aov-f-02-0-UI /clone_end=3 /gb=AW122239 /gi=6097702 /ug=Mm.2173 /len=549

Identification
103560_at Cluster Incl AW124401:UI-M-BH2.1-ape-d-04-0-UI.s1 Mus musculus cDNA, 3 end /clone=UI-M-BH2.1-ape-d-04-0-UI /clone_end=3 /gb=AW124401 /gi=6099931 /ug=Mm.19119 /len=217
103887_at S100 calcium-binding protein A9 (calgranulin B)
104014_at hemochromatosis
104071_at Cluster Incl AI852433:UI-M-BH0-ajo-g-04-0-UI.s1 Mus musculus cDNA, 3 end /clone=UI-M-BH0-ajo-g-04-0-UI /clone_end=3 /gb=AI852433 /gi=5496304 /ug=Mm.21639 /len=544
104093_at lymphocyte specific 1
104173_at Cluster Incl AA797989:ub60d04.r1 Mus musculus cDNA, 5 end /clone=IMAGE-1382119 /clone_end=5 /gb=AA797989 /gi=2860944 /ug=Mm.27253 /len=413
104267_at Cluster Incl AI844736:UI-M-AL1-ahq-c-10-0-UI.s1 Mus musculus cDNA, 3 end /clone=UI-M-AL1-ahq-c-10-0-UI /clone_end=3 /gb=AI844736 /gi=5488642 /ug=Mm.28535 /len=406
104298_at Cluster Incl AI842544:UI-M-AQ1-aea-e-05-0-UI.s1 Mus musculus cDNA, 3 end /clone=UI-M-AQ1-aea-e-05-0-UI /clone_end=3 /gb=AI842544 /gi=5476757 /ug=Mm.22337 /len=457
104415_at EST AI461938
104606_at CD52 antigen
104735_at Cluster Incl AI842065:UI-M-AN1-afg-a-10-0-UI.s1 Mus musculus cDNA, 3 end /clone=UI-M-AN1-afg-a-10-0-UI /clone_end=3 /gb=AI842065 /gi=5476278 /ug=Mm.24647 /len=444
160108_at p8 protein
160117_at Cluster Incl AI850638:UI-M-BG1-aik-e-06-0-UI.s1 Mus musculus cDNA, 3 end /clone=UI-M-BG1-aik-e-06-0-UI /clone_end=3 /gb=AI850638 /gi=5494544 /ug=Mm.19258 /len=381 /NOTE=rep
160156_at Cluster Incl AA981015:vx55c11.r1 Mus musculus cDNA, 5 end /clone=IMAGE-1279124 /clone_end=5 /gb=AA981015 /gi=3159551 /ug=Mm.22383 /len=343 /NOTE=replacement for probe
160386_at Cluster Incl AI835981:UI-M-AQ0-aah-d-09-0-UI.s2 Mus musculus cDNA, 3 end /clone=UI-M-AQ0-aah-d-09-0-UI /clone_end=3 /gb=AI835981 /gi=5470234 /ug=Mm.30099 /len=318 /NOTE=r
160388_at Cluster Incl AI848668:UI-M-AH1-agv-a-10-0-UI.s1 Mus musculus cDNA, 3 end /clone=UI-M-AH1-agv-a-10-0-UI /clone_end=3 /gb=AI848668 /gi=5492539 /ug=Mm.30119 /len=421 /NOTE=re
160402_at ribosomal protein L13
160493_at Cd63 antigen
160495_at aryl-hydrocarbon receptor
160502_at cellular repressor of E1A-stimulated genes

Hierarchical Clustering Lists of differentially


expressed genes
Targets
Real Time Quantitative PCR:
Assess Gene Expression
Pathophysiologies

Figure 4 Clinical bioinformatics can be generated from the analysis of COPD-specific pathological alterations. Patients with COPD can
be selected by clinical informatics and criteria and the specific area is selected by micro-dissection. The tissue can be used for genomic (or
proteomic) analysis and identified biomarkers are validated for the understanding of the pathogenesis.

about the future of omics research and its clinical impli- References
1. Celli BR, MacNee W: Standards for the diagnosis and treatment of
cations [54]. Clinical bioinformatics on COPD could be patients with COPD: a summary of the ATS/ERS position paper. Eur Respir
achieved from the combination of clinical informatics, J 2004, 23(6):932-946.
medical informatics, bioinformatics and informatics by 2. Pauwels RA, Rabe KF: Burden and clinical features of chronic obstructive
pulmonary disease (COPD). Lancet 2004, 364(9434):613-620.
collaborations among clinicians, bioinformaticians, com- 3. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L,
puter scientists, biologists, and mathematicians. Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, et al: The nature of
small-airway obstruction in chronic obstructive pulmonary disease. N
Engl J Med 2004, 350(26):2645-2653.
Author details 4. Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P, Fukuchi Y,
1 Jenkins C, Rodriguez-Roisin R, van Weel C, et al: Global strategy for the
Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University,
Shanghai, China. 2Biomedical Research Center, Zhongshan Hospital, Fudan diagnosis, management, and prevention of chronic obstructive
University, Shanghai, China. pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med
2007, 176(6):532-555.
Authors’ contributions 5. Lopez AD, Murray CC: The global burden of disease, 1990-2020. Nat Med
HC mainly wrote the manuscript, as well as the revision. XDW conceived of 1998, 4(11):1241-1243.
the review and edited and wrote part of the manuscript. All authors read 6. Fang X, Wang X, Bai C: COPD in China: the burden and importance of
and approved the final manuscript. proper management. Chest 2011, 139(4):920-929.
7. Casado B, Iadarola P, Luisetti M, Kussmann M: Proteomics-based diagnosis
Competing interests of chronic obstructive pulmonary disease: the hunt for new markers.
The authors declare that they have no competing interests. Expert Rev Proteomics 2008, 5(5):693-704.
8. Chen H, Wang D, Bai C, Wang X: Proteomics-based biomarkers in chronic
Received: 3 October 2010 Accepted: 20 December 2011 obstructive pulmonary disease. J Proteome Res 2010, 9(6):2798-2808.
Published: 20 December 2011 9. Pauwels RA, Buist AS, Calverley PM, Jenkins CR, Hurd SS: Global strategy for
the diagnosis, management, and prevention of chronic obstructive
Chen and Wang Journal of Clinical Bioinformatics 2011, 1:35 Page 7 of 8
http://www.jclinbioinformatics.com/content/1/1/35

pulmonary disease. NHLBI/WHO Global Initiative for Chronic Obstructive radiographic phenotype and smoking status on peripheral blood
Lung Disease (GOLD) Workshop summary. Am J Respir Crit Care Med 2001, biomarker patterns in chronic obstructive pulmonary disease. PLoS One
163(5):1256-1276. 2009, 4(8):e6865.
10. Silverman EK, Spira A, Pare PD: Genetics and genomics of chronic 32. York TP, van den Oord EJ, Langston TB, Edmiston JS, McKinney W,
obstructive pulmonary disease. Proc Am Thorac Soc 2009, 6(6):539-542. Webb BT, Murrelle EL, Zedler BK, Flora JW: High-resolution mass
11. DeMeo DL, Mariani T, Bhattacharya S, Srisuma S, Lange C, Litonjua A, spectrometry proteomics for the identification of candidate plasma
Bueno R, Pillai SG, Lomas DA, Sparrow D, et al: Integration of genomic and protein biomarkers for chronic obstructive pulmonary disease.
genetic approaches implicates IREB2 as a COPD susceptibility gene. Am Biomarkers 2010, 15(4):367-377.
J Hum Genet 2009, 85(4):493-502. 33. Merkel D, Rist W, Seither P, Weith A, Lenter MC: Proteomic study of
12. Arif E, Vibhuti A, Alam P, Deepak D, Singh B, Athar M, Pasha MA: human bronchoalveolar lavage fluids from smokers with chronic
Association of CYP2E1 and NAT2 gene polymorphisms with chronic obstructive pulmonary disease by combining surface-enhanced laser
obstructive pulmonary disease. Clin Chim Acta 2007, 382(1-2):37-42. desorption/ionization-mass spectrometry profiling with mass
13. Vibhuti A, Arif E, Mishra A, Deepak D, Singh B, Rahman I, Mohammad G, spectrometric protein identification. Proteomics 2005, 5(11):2972-2980.
Pasha MA: CYP1A1, CYP1A2 and CYBA gene polymorphisms associated 34. Bozinovski S, Hutchinson A, Thompson M, Macgregor L, Black J, Giannakis E,
with oxidative stress in COPD. Clin Chim Acta 2010, 411(7-8):474-480. Karlsson AS, Silvestrini R, Smallwood D, Vlahos R, et al: Serum amyloid a is
14. Zhan P, Wang J, Wei SZ, Qian Q, Qiu LX, Yu LK, Song Y: TNF-308 gene a biomarker of acute exacerbations of chronic obstructive pulmonary
polymorphism is associated with COPD risk among Asians: meta-analysis disease. Am J Respir Crit Care Med 2008, 177(3):269-278.
of data for 6,118 subjects. Mol Biol Rep 2011, 38(1):219-227. 35. Bandow JE, Baker JD, Berth M, Painter C, Sepulveda OJ, Clark KA, Kilty I,
15. Gupta J, Bhadoria DP, Lal MK, Kukreti R, Chattopadhaya D, Gupta VK, VanBogelen RA: Improved image analysis workflow for 2-D gels enables
Dabur R, Yadav V, Chhillar AK, Sharma GL: Association of the PIM3 allele of large-scale 2-D gel-based proteomics studies–COPD biomarker discovery
the alpha-1-antitrypsin gene with chronic obstructive pulmonary study. Proteomics 2008, 8(15):3030-3041.
disease. Clin Biochem 2005, 38(5):489-491. 36. Nicholson JK, Connelly J, Lindon JC, Holmes E: Metabonomics: a platform
16. Saxena S, Kumar R, Madan T, Gupta V, Muralidhar K, Sarma PU: Association for studying drug toxicity and gene function. Nat Rev Drug Discov 2002,
of polymorphisms in pulmonary surfactant protein A1 and A2 genes 1(2):153-161.
with high-altitude pulmonary edema. Chest 2005, 128(3):1611-1619. 37. McClay JL, Adkins DE, Isern NG, O’Connell TM, Wooten JB, Zedler BK,
17. Arif E, Vibhuti A, Deepak D, Singh B, Siddiqui MS, Pasha MA: COX2 and p53 Dasika MS, Webb BT, Webb-Robertson BJ, Pounds JG, et al: (1)H nuclear
risk-alleles coexist in COPD. Clin Chim Acta 2008, 397(1-2):48-50. magnetic resonance metabolomics analysis identifies novel urinary
18. Butler MW, Fukui T, Salit J, Shaykhiev R, Mezey JG, Hackett NR, Crystal RG: biomarkers for lung function. J Proteome Res 2010, 9(6):3083-3090.
Modulation of Cystatin A Expression in Human Airway Epithelium 38. de Laurentiis G, Paris D, Melck D, Maniscalco M, Marsico S, Corso G,
Related to Genotype, Smoking, COPD, and Lung Cancer. Cancer Res 2011, Motta A, Sofia M: Metabonomic analysis of exhaled breath condensate in
71(7):2572-2581. adults by nuclear magnetic resonance spectroscopy. Eur Respir J 2008,
19. Vibhuti A, Arif E, Deepak D, Singh B, Qadar Pasha MA: Genetic 32(5):1175-1183.
polymorphisms of GSTP1 and mEPHX correlate with oxidative stress 39. Trune DR, Larrain BE, Hausman FA, Kempton JB, Macarthur CJ:
markers and lung function in COPD. Biochem Biophys Res Commun 2007, Simultaneous measurement of multiple ear proteins with multiplex
359(1):136-142. ELISA assays. Hear Res 2010.
20. Sadeghnejad A, Ohar JA, Zheng SL, Sterling DA, Hawkins GA, Meyers DA, 40. Elshal MF, McCoy JP: Multiplex bead array assays: performance evaluation
Bleecker ER: Adam33 polymorphisms are associated with COPD and lung and comparison of sensitivity to ELISA. Methods 2006, 38(4):317-323.
function in long-term tobacco smokers. Respir Res 2009, 10:21. 41. Ray CA, Bowsher RR, Smith WC, Devanarayan V, Willey MB, Brandt JT,
21. van Diemen CC, Postma DS, Vonk JM, Bruinenberg M, Schouten JP, Dean RA: Development, validation, and implementation of a multiplex
Boezen HM: A disintegrin and metalloprotease 33 polymorphisms and immunoassay for the simultaneous determination of five cytokines in
lung function decline in the general population. Am J Respir Crit Care human serum. J Pharm Biomed Anal 2005, 36(5):1037-1044.
Med 2005, 172(3):329-333. 42. Farlow EC, Patel K, Basu S, Lee BS, Kim AW, Coon JS, Faber LP, Bonomi P,
22. Arif E, Ahsan A, Vibhuti A, Rajput C, Deepak D, Athar M, Singh B, Pasha MA: Liptay MJ, Borgia JA: Development of a multiplexed tumor-associated
Endothelial nitric oxide synthase gene variants contribute to oxidative autoantibody-based blood test for the detection of non-small cell lung
stress in COPD. Biochem Biophys Res Commun 2007, 361(1):182-188. cancer. Clin Cancer Res 2010, 16(13):3452-3462.
23. Sadeghnejad A, Meyers DA, Bottai M, Sterling DA, Bleecker ER, Ohar JA: 43. Bhattacharya S, Srisuma S, Demeo DL, Shapiro SD, Bueno R, Silverman EK,
IL13 promoter polymorphism 1112C/T modulates the adverse effect of Reilly JJ, Mariani TJ: Molecular biomarkers for quantitative and discrete
tobacco smoking on lung function. Am J Respir Crit Care Med 2007, COPD phenotypes. Am J Respir Cell Mol Biol 2009, 40(3):359-367.
176(8):748-752. 44. Agusti A, Sobradillo P, Celli B: Addressing the complexity of chronic
24. Desai AA, Hysi P, Garcia JG: Integrating genomic and clinical medicine: obstructive pulmonary disease: from phenotypes and biomarkers to
searching for susceptibility genes in complex lung diseases. Transl Res scale-free networks, systems biology, and P4 medicine. Am J Respir Crit
2008, 151(4):181-193. Care Med 2011, 183(9):1129-1137.
25. Patterson SD, Aebersold RH: Proteomics: the first decade and beyond. Nat 45. Auffray C, Adcock IM, Chung KF, Djukanovic R, Pison C, Sterk PJ: An
Genet 2003, 33(Suppl):311-323. integrative systems biology approach to understanding pulmonary
26. Marko-Varga G, Fehniger TE: Proteomics and disease–the challenges for diseases. Chest 2010, 137(6):1410-1416.
technology and discovery. J Proteome Res 2004, 3(2):167-178. 46. Comandini A, Marzano V, Curradi G, Federici G, Urbani A, Saltini C: Markers
27. Lee EJ, In KH, Kim JH, Lee SY, Shin C, Shim JJ, Kang KH, Yoo SH, Kim CH, of anti-oxidant response in tobacco smoke exposed subjects: a data-
Kim HK, et al: Proteomic analysis in lung tissue of smokers and COPD mining review. Pulm Pharmacol Ther 2010, 23(6):482-492.
patients. Chest 2009, 135(2):344-352. 47. Maier D, Kalus W, Wolff M, Kalko SG, Roca J, Marin de Mas I, Turan N,
28. Sepper R, Prikk K: Proteomics: is it an approach to understand the Cascante M, Falciani F, Hernandez M, et al: Knowledge management for
progression of chronic lung disorders? J Proteome Res 2004, 3(2):277-281. systems biology a general and visually driven framework applied to
29. Ohlmeier S, Vuolanto M, Toljamo T, Vuopala K, Salmenkivi K, Myllarniemi M, translational medicine. BMC Syst Biol 2011, 5:38.
Kinnula VL: Proteomics of human lung tissue identifies surfactant protein 48. Paoletti M, Camiciottoli G, Meoni E, Bigazzi F, Cestelli L, Pistolesi M,
A as a marker of chronic obstructive pulmonary disease. J Proteome Res Marchesi C: Explorative data analysis techniques and unsupervised
2008, 7(12):5125-5132. clustering methods to support clinical assessment of Chronic
30. Gray RD, MacGregor G, Noble D, Imrie M, Dewar M, Boyd AC, Innes JA, Obstructive Pulmonary Disease (COPD) phenotypes. J Biomed Inform
Porteous DJ, Greening AP: Sputum proteomics in inflammatory and 2009, 42(6):1013-1021.
suppurative respiratory diseases. Am J Respir Crit Care Med 2008, 49. Studer SM, Kaminski N: Towards systems biology of human pulmonary
178(5):444-452. fibrosis. Proc Am Thorac Soc 2007, 4(1):85-91.
31. Bon JM, Leader JK, Weissfeld JL, Coxson HO, Zheng B, Branch RA, 50. Kauffmann F: Post-genome respiratory epidemiology: a multidisciplinary
Kondragunta V, Lee JS, Zhang Y, Choi AM, et al: The influence of challenge. Eur Respir J 2004, 24(3):471-480.
Chen and Wang Journal of Clinical Bioinformatics 2011, 1:35 Page 8 of 8
http://www.jclinbioinformatics.com/content/1/1/35

51. Wang X, Adler KB, Chaudry IH, Ward PA: Better understanding of organ
dysfunction requires proteomic involvement. J Proteome Res 2006,
5(5):1060-1062.
52. Plymoth A, Yang Z, Lofdahl CG, Ekberg-Jansson A, Dahlback M, Fehniger TE,
Marko-Varga G, Hancock WS: Rapid proteome analysis of bronchoalveolar
lavage samples of lifelong smokers and never-smokers by micro-scale
liquid chromatography and mass spectrometry. Clin Chem 2006,
52(4):671-679.
53. Reisdorph NA, Reisdorph R, Bowler R, Broccardo C: Proteomics methods
and applications for the practicing clinician. Ann Allergy Asthma Immunol
2009, 102(6):523-529.
54. Lin JL, Bonnichsen MH, Nogeh EU, Raftery MJ, Thomas PS: Proteomics in
detection and monitoring of asthma and smoking-related lung diseases.
Expert Rev Proteomics 2010, 7(3):361-372.

doi:10.1186/2043-9113-1-35
Cite this article as: Chen and Wang: Significance of bioinformatics in
research of chronic obstructive pulmonary disease. Journal of Clinical
Bioinformatics 2011 1:35.

Submit your next manuscript to BioMed Central


and take full advantage of:

• Convenient online submission


• Thorough peer review
• No space constraints or color figure charges
• Immediate publication on acceptance
• Inclusion in PubMed, CAS, Scopus and Google Scholar
• Research which is freely available for redistribution

Submit your manuscript at


www.biomedcentral.com/submit

Potrebbero piacerti anche