Está en la página 1de 12

J BIOCHEM MOLECULAR TOXICOLOGY

Volume 15, Number 5, 2001

Discovery in Toxicology: Mediation by Gene Expression


Array Technology
Hisham K. Hamadeh, Pierre Bushel, Richard Paules, and Cynthia A. Afshari
National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; E-mail: afshari@niehs.nih.gov

Received 24 April 2001; accepted 27 April 2001

ABSTRACT: Toxicogenomics is a term that represents the analysis of histopathological or biochemical mark-
the merging of toxicology with novel genomics tech- ers often does not provide information about a toxi-
niques. Data generated in the new-age era of toxico- cant’s mechanism of action. In addition, gross patho-
logy is relatively complex, requires new bioinformat- logical changes are insensitive for detecting toxicity at
ics tools for adequate interpretation, and allows for the low doses or early time points leading to incorrect as-
rapid generation of testable hypotheses. Hazard iden-
sessment of potential toxicity or safety. In order to better
tification and risk assessment processes will advance
from the use of genomics techniques, which will lead
evaluate the hazard associated with exposure to chem-
to greater understanding of mechanism(s) of action of icals at varying doses and exposure, one needs to un-
toxicants, development of novel biomarkers of expo- derstand the specific mechanism of action of a toxicant.
sure and effect, and better identification of sensitive Gene expression changes associated with signal
subpopulations. ° C 2001 John Wiley & Sons, Inc.∗ J pathway activation can provide chemical-specific in-
Biochem Mol Toxicol 15:231–242, 2001 formation on the mechanistic and biological effects
of a chemical. A standard method used to study the
KEY WORDS: Toxicogenomics, Genomics, Toxicology, changes in gene expression is the Northern blot. This
Microarrays, cDNA, Single Nucleotide Polymor- method, however, is relatively labor intensive and is
phisms, Pattern Recognition, Bioinformatics, Gene only practical for examining expression changes for a
Expression, Biomarkers
limited number of genes. However, the advancements
in genome sequencing present researchers with the
INTRODUCTION opportunity to study genome-wide expression alter-
ations. New technologies, including DNA microarrays,
The field of toxicology has traditionally focused on will facilitate studies that glean insight into the cellu-
assessing toxicity or investigating the mechanisms of lar response to chemical exposure. While traditional
action of potentially hazardous agents. The evolution of assays, like Northern blots, measure the expression of
new, innovative technologies coupled with the advance only one gene at a time, DNA microarrays measure the
of genomic information is revolutionizing the future of expression of tens of thousands of genes in an equiva-
toxicological analyses. This review will describe the ap- lent amount of time. In addition, array technology al-
plication of microarray technology to facilitate discov- lows newly discovered or unidentified genes to be in-
ery of mechanistic pathways associated with toxicant cluded in the analysis, providing a remarkable potential
action as well as identification of novel targets for de- to accelerate the discovery of toxicant targets.
velopment of susceptibility models. DNA microarrays [1] provide a revolutionary plat-
form to perform genome-wide gene expression analy-
DNA MICROARRAYS ses through comparison of virtually any two biological
samples. There are two basic types of microarrays used
Classically, toxicologists assess potential adverse in gene expression analyses: oligonucleotide-based ar-
health outcomes resulting from chemical exposure by rays and cDNA arrays. Both types yield compara-
using gross endpoints such as body and organ weight ble results, though they differ in their methodology.
changes and histopathological observations. However, Oligonucleotide arrays are made using specific chemi-
cal synthesis steps by means of a series of photolitho-
Correspondence to: Cynthia A. Afshari.
graphic mask [2], light [3], or other methods [4] to
c 2001 John Wiley & Sons, Inc. ∗ This article is a US Government work
° generate the sequence order in the synthesis. The re-
and, as such, is in the public domain in the United States of America. sult of these processes is the generation of high density

231
232 HAMADEH ET AL. Volume 15, Number 5, 2001

arrays of short oligonucleotides (∼20 bases) probes that of the photomultiplier tubes associated with each laser.
are synthesized in predefined positions [2]. Oligonu- A simplified overview of the protocol for generating
cleotide arrays may be used for expression studies as gene expression data using microarrays is illustrated
well as sequence analysis. in Figure 1.
cDNA microarrays can be used to study differences Data acquired from scanning the microarray chips
in gene expression in different cells or tissues. Partial are converted to 16-bit TIFF images corresponding to
cDNAs (0.5–2 kb in length) that correspond to unique each of the individual fluors. Specialized image pro-
gene sequences are usually spotted onto the surface of cessing software overlays the two images and assigns a
treated glass slides using high speed robotic printers [1] color intensity to each spot representing the amount of
that allow the user to configure the placement of cD- fluorescence. The result is a red–green pseudo-color ra-
NAs on a glass substrate or chip. Spotted cDNAs can tio image with colors for each cDNA signal ranging be-
represent either sequenced genes of known function or tween green and red. For example, if a gene is expressed
collections of partially sequenced cDNA derived from in equal amounts in both the control and treated sam-
expressed sequence tags (ESTs) corresponding to ples, the cDNA spot is yellow, indicating an equal ratio
messenger RNAs of genes of unknown function. It is of both fluors hybridizing with that gene. If a gene were
possible to print greater than 25,000 spots of cDNAs highly expressed in one of the samples, then the spot
on a single chip. To adhere to the surface of a glass would be predominantly red or green (Figure 2).
slide, the cDNA can be covalently linked or bound The sensitivity of microarray data is dynamic and
to a coated surface by electrostatic interaction. After depends on the confidence level associated with the tar-
the DNA is spotted on the chip, it is dried, UV cross- get definition and image analysis processes. Statistical
linked, and blocked. While UV cross-linking attaches analysis at low confidence levels yields an increased
the spotted DNA more firmly to the slide, blocking sensitivity for detecting subtle changes in gene expres-
prevents fluorescently labeled target cDNA from non- sion levels but inherently results in the inclusion of false
specifically binding to the slide coating during hy- positives as significantly regulated genes. Conversely,
bridization. Blocking is important for minimizing the high confidence levels decrease the sensitivity of detec-
background around the spotted DNA in order to op- tion and might omit genes with significant expression
timize the signal-to-noise ratio. A low noise-to-signal changes (false negatives). A successful strategy to de-
ratio maximizes the sensitivity of detection. Optimal termine significant gene expression responses utilizes
microarray measurements can detect changes as small lower confidence levels in combination with biological
as 1.2-fold increase or decrease in gene expression. and hybridization replicates. Replicate data sets are an-
Any biological sample from which high quality alyzed using a binomial distribution [5]. Table 1 demon-
RNA can be isolated may be used for microarray anal- strates the correlation between confidence levels and
ysis. For example, one may compare tissue types such false negative genes.
as normal versus diseased tissue, or untreated versus
treated tissue. Gene expression analysis using cDNA
microarrays is conducted by the simultaneous com- ANALYSIS TOOLS
parison of at least two biological samples. Differential FOR DNA MICROARRAYS
gene expression measurements are achieved by a com-
petitive, simultaneous hybridization using a two-color The amount of data derived from microarray anal-
fluorescence labeling approach. Isolated RNA is con- ysis is relatively difficult to analyze without the utiliza-
verted to fluorescently labeled “targets” by a reverse tion of computational tools that can efficiently handle
transcriptase reaction using a modified nucleotide, typ- large volumes of data. Thus the field of bioinformatics
ically dUTP or dCTP conjugated with a chromophore. has emerged. Bioinformatics in gene expression analy-
The two RNAs being compared are labeled with differ- sis deals with tasks including spot location, definition,
ent fluorescent tags, traditionally either Cy3 or Cy5, so and intensity calculations from raw scanned images,
that each RNA has a different energy emission wave- transformation of data sets into more easily quantifi-
length or color when excited by dual lasers. The fluo- able forms, application of analysis tools to extract asso-
rescently labeled targets are mixed and hybridized on ciations between gene expression levels, and extraction
a microarray chip. The array is scanned at two wave- of information from analyses that facilitate the devel-
lengths using independent laser excitation of the two opment of new testable hypotheses.
fluors at 632 and 532 nm wavelengths for the red (Cy5) Changes in gene expression are quantified from
and green (Cy3) labels, respectively. Thus, one RNA can raw images using image analysis algorithms and data
be regarded as labeled in “red” and the other in “green.” acquisition procedures to detect target regions, com-
The intensity of the signals relative to each other and pute feature pixel intensity values, subtract background
the background can be optimized by adjusting the gains intensity, and compare the pixel intensity values of a
Volume 15, Number 5, 2001 GENE ARRAY TECHNOLOGY IN TOXICOLOGY 233

FIGURE 1. High-density gene expression analysis using cDNA microarrays. This illustration shows mRNA samples derived from in vivo
TCDD-treated rats or in vitro TCDD-treated liver cells used to prepare fluorescently labeled cDNA by reverse transcription in the presence of
fluorescently labeled (Cy3 or Cy5) nucleotide precursors. Competitive hybridization of the untreated and treated labeled RNA samples allows
for measuring the relative abundance of the transcripts from each gene as reflected by the ratio of “red” to “green” fluorescence from each spot
on the array. The abundance of mRNA copies corresponding to CYP1A1, 1A2, and 1B1 is greater in the TCDD-treated biological models than in
the unexposed counterparts. This is evidenced by the mostly “red” fluorescence measured at the spots representing those genes on the array.

cDNA between two samples. Normalization is regu- calculated for each gene target and processed into a
larly performed during data analysis to adjust and cor- standard 24-bit pseudo-color composite image for vi-
rect the variability in the intensity values obtained from sualization of the gene expression profile.
the scanned image. This is achieved by using the inten- Pixel intensity values, calculated ratio, and statis-
sity values for either a standard set of control genes tics for each gene are stored as records in files for subse-
or for the entire collection of the targets represented quent data management and analysis purposes. For in-
on the DNA chip. A final ratio pixel intensity value is stance, the microarray database ArrayDB was designed
234 HAMADEH ET AL. Volume 15, Number 5, 2001

FIGURE 2. Color gradation of microarray spots resulting from competitive hybridization of two fluorescently tagged cDNA populations on a
DNA microarray. This colorimetric representation of the ratio of the “red” signal to the “green” signal indicates that if the transcript copies of a
gene were found exclusively in one of the samples of the comparison pair, the color of the respective spot would be at one end of the spectrum.
A gene whose transcripts are found in approximately equal amounts in both samples would be indicated as yellow.

as a relational database management system to store, At the onset of the microarray era, simple statistical
analyze, and associate gene expression data with infor- methods facilitated the interpretation of complex and
mation from remote biological resources [6]. A more large-scale microarray gene expression results. For ex-
comprehensive database has recently been developed ample, a set number of + and − standard deviations
to correlate gene expression patterns with information from the mean intensity values were typically used to
about pharmacological compounds [7]. identify changes in biologically relevant genes in most
Inherent with microarray gene expression data is published microarray-related studies. However, more
the nonlinearity nature of the data and the low qual- sophisticated methods have been developed in which
ity or unreliability of data points that are generated computed confidence intervals are used to determine
from below threshold detection levels, anomalies on significantly changed genes from a ratio distribution of
the microarray chip, or essentially missing cDNA fea- gene expression data [8]. More importantly, differen-
tures. Typical microarray data analysis software in- tially expressed genes can be validated by comparing
cludes functions to log transform gene expression data replicate measurements of microarrays and perform-
into a more linear state and to essentially stabilize the ing biological assays to confirm the significance of al-
variance in the data. In addition, cut-off values may be tered expression [9,10]. Hence, for a given confidence
applied to the pixel size of cDNA features as well as the level, the probability of chance occurrences in replicate
intensity range of the values to refine the data set for experiments can be determined using a binomial dis-
further analysis. The use of these processing features is tribution model [11].
a form of “filtering” to microarray gene expression data At this time, these methods do not control for
and improves the reliability of results. false positives and false negatives, low fold changes,

TABLE 1. Number of Genes Altered in a Statistically Significant Manner for an Experiment Replicated Three Times
Times Gene Flagged # of Genes Expected to be
# of Outliers at 99% Confidencea by Chance Flagged by Chanceb Probabilityc

1650 0 1650 0.5


215 1 50 0.25
88 2 1 0.01
48 3 0 0.001
a
# of genes that were altered in a statistically significant manner in replicate experiments.
b
Probability of genes flagged by chance based on a 1650 gene chip.
c
Probability calculations using a binomial distribution (5).
Volume 15, Number 5, 2001 GENE ARRAY TECHNOLOGY IN TOXICOLOGY 235

variation of data across independent replicate experi- Self-organizing maps (SOMs) is another useful
ments, nor do they account for genes exhibiting a bias algorithm used to interpret and visualize large high-
for incorporation of a particular fluor, as detected from dimensional data sets in two-dimensional space. Basi-
microarray fluor-reverse experiments. Fortunately, cally, SOMs are an array of interconnected cells that be-
microarray data has attracted significant attention come refined to various input signal patterns or classes
and gained concerted interest from practitioners of patterns in an orderly fashion. These artificial neu-
in the statistics, mathematics, and applied physics ral networks undergo a competitive and unsupervised
communities as well as expert bioinformatics compa- iterative learning process. When data is used to train
nies committed to applying rigorous and innovative SOMs, structure is imposed on the data with neigh-
computational error models to measurements of gene boring nodes defining related clusters. In essence, the
expression data. For instance, a pioneering approach SOM constitutes a new paradigm in artificial intelli-
to analysis of microarray data is currently underway gence and cognitive learning and only recently has been
using various mixed models and general analysis implemented into a computer program that can be uti-
of variance models to assess and increase statistical lized as a genetic neural network model for interpreting
significance in the microarray-derived gene expression and displaying various patterns of gene expression data
data [12]. Similarly, a combination of robust additive [15].
and multiplicative error models are being used to Finally, discriminant analysis is commonly used in
account for the uncertainty and variation in measuring multivariate analysis to determine which variables best
gene expression data as well as to assign confidence discriminate between two or more groups. The general
and probability values, weight, and error bars to concept underlying the use of discriminant analysis to
define statistical significance of individual genes analyze microarray data is to determine whether classes
[13]. of gene expression measurements significantly differ
The final stage in microarray gene expression anal- with regard to distinct and characteristic gene expres-
ysis is multivariate analysis, in which higher-order sion profiles. Microarray data is used as a set of training
computational tools and procedures are utilized to as- observations to assess and rank the importance of par-
sist with the interpretation and visualization of com- ticular genes for discrete sample classification. When
plex multivariate microarray gene expression data. Al- identified, hallmark gene markers are used to predict
though there is no definitive answer as to which form the classifications of a collection of test microarray treat-
of multivariate analysis to use on microarray data, it ments as well as apply robust pattern recognition pro-
is critical to understand the implicit characteristics of cedures to large volumes of complex gene expression
each algorithm with the intention of prudently exploit- data. Computationally, discriminant analysis is very
ing inherent advantages of each to ultimately assemble similar to the analysis of variance; however, a novel
the best insight and interpretation of analyzed results. approach that combines a genetic algorithm and the
For example, hierarchical clustering methods are es- k-nearest neighbor method has been described to posi-
sentially an iterative process to organize objects into tively identify genes that discriminate between classes
groups with similar attributes based on resemblance, of tumor cells [16] and toxicant treatments [17].
proximity, similarity, or dissimilarity measurements. Data derived from microarray analysis may be ap-
The four basic steps in conducting a cluster analysis plied to the identification of complex genetic diseases,
on microarray data are 1) data collection and selection drug targets, toxicant and pathogen analysis, detection
of variables (genes and experiments) for analysis, 2) of differences over time between normal and diseased
generation of a resemblance matrix using a particular states of tissues, and mutation/polymorphism analy-
mathematical formula to measure the extent of similar- sis. The field of genomics that deals with generating
ity, 3) selection and execution of an iterative clustering high-output gene expression data will have a great im-
method to produce a phylogenetic tree diagram (den- pact on preventive medicine where diseases will be
drogram) with the branches representing the amount sub-typed prior to the onset of physiological symp-
of similarity between clusters, and 4) determination of toms or progressed stages and drugs will be designed
where to “cut” the tree into a select number of nodes. to target causes of disease rather than symptoms. There
Recently, robust computational tools and an interactive has already been some interesting studies published
graphical interface have been developed to facilitate in these areas. For example, gene expression analysis
the compilation and visualization of clustered gene ex- of serum-exposed human fibroblasts highlighted the
pression data [14] (Figure 3). Although cluster analysis physiological role of these cells in wound repair [18].
is a classic and rather simple partitioning technique, its This type of analysis was also used to monitor signal
application is relatively new to the field of microarray transduction during yeast pheromone response [19],
data analysis and is not supported by any comprehen- define regulatory pathways in stem cells as well as
sive body of statistical literature. their global genetic program [20], and validate drug
236 HAMADEH ET AL. Volume 15, Number 5, 2001

FIGURE 3. Two-dimensional hierarchical clustering analysis of 28 animals and 457 genes that were altered in a statistically significant manner at
95% confidence level in at least two replicate hybridizations at 2 time points [13]. Clustering analysis was performed across genes but not animals.
Genes are represented on the vertical axis while animals are plotted on the horizontal axis. Red or green indicates upregulation or suppression
respectively while black indicates no change in levels of mRNA as a result of exposure. Amplified node II shows an example of genes that were
induced by the peroxisome proliferators (PP) collectively but suppressed with phenobarbital treatment while node I is an example of genes that
were upregulated by phenobarbital (PB) treatment at the 24 h and 2 week time points while levels mostly decreased or remained unchanged
with the pp exposures.

target and identify secondary drug target effects [21]. toxicology [32–39]. However, researchers are just start-
The technology has been used to discover potential hu- ing to test whether the application of microarrays can
man biomarkers of ionizing radiation by ex vivo irra- help answer key toxicological questions. The under-
diation of human peripheral blood lymphocytes [22]. lying basis of chemical effects stems from the com-
This study suggests that monitoring the gene expres- plex interaction between the environment and different
sion of these biomarkers may provide estimates of genes. By understanding the molecular details of this
environmental radiation exposures. The collective re- interaction, we can identify toxicity-associated genes
sults of these studies provide tangible evidence that and cellular response mechanisms. This would allow
this technology can be utilized to confront a variety of for the characterization of these “response” genes and
important medical problems that have been intractable the elucidation of the mechanisms by which they are
with past technology. regulated.
Microarray technology promises to speed the an-
notation of uncharacterized or poorly characterized
cDNA MICROARRAYS AND DISCOVERY genes. The belief is based on the premise that shared
IN TOXICOLOGY expression often implies shared function. Thus, when
a gene with an unknown function is included in a
Although the field of toxicology has yet to benefit cluster of known genes sharing similar gene expres-
from this revolutionary technology, other disciplines, sion patterns, it is implied that the function of the un-
including cancer biology, cell biology, physiology, and known gene bears similarity to its neighbors in that
pharmacology, have made great advances using DNA cluster. This hypothesis was first tested by Brown and
microarrays [13,23–31]. colleagues in 1998 in a study of global gene expres-
Many reports describe the theoretical application of sion in Saccharomyces cerevisiae, that demonstrated co-
high-density gene expression analysis to challenges in regulation of genes that participated in shared cellular
Volume 15, Number 5, 2001 GENE ARRAY TECHNOLOGY IN TOXICOLOGY 237

function [14]. Theoretically, clustering will aid in de- were mapped into functional pathways present in the
termining the function of a substantial number of un- mitochondria. This demonstrates the potential of dis-
known genes, even when sequence similarity has not covering novel alterations in the expression levels of
been established with characterized genes [40–42]. genes associated with toxicant-specific molecular path-
A demonstration of the assignment of putative ways [17]. Other studies that demonstrated the ability
function to a number of unknown genes is derived of microarrays to discover new genes associated with
by studying gene expression patterns generated toxicant responses have been reviewed [44].
in the livers of rats that were treated in vivo with
phenobarbital or one of three peroxisome proliferators
(clofibrate, Wyeth 14,643, gemfibrozil) [17]. Male MICROARRAYS AND THE
Sprague-Dawley rats were exposed to one of the EXPLORATION OF POLYMORPHISMS
compounds, then livers were harvested 24 h post
single-dose or 2 weeks post daily-dosage. In this study, DNA sequence polymorphisms are discovered
clustering illustrated upregulation of genes in response by comparing the base sequence of genes across a
to phenobarbital treatment but not with peroxisome population and observing interindividual variation.
proliferator exposures (Figure 3). Not only did these In addition to traditional sequencing methodologies,
genes have a high correlation (R > + 0.8) in their expres- oligonucleotide-based arrays are also useful in geno-
sion levels, but were also related biologically. Genes in typing. Once a target gene has been identified, oligo
node II, which were upregulated by peroxisome prolif- arrays can be used for conducting mutation and poly-
erators but suppressed by phenobarbital, have reported morphism screening [2,45]. However, limitations of
involvement in mitochondrial b-oxidation. This corrob- oligonucleotide-based microarrays include the inabil-
orates previously published findings demonstrating ity to detect length polymorphisms or large deletions
the induction of b-oxidation by peroxisome prolifera- and insertions [46,47].
tors [43]. Node I contains genes that are mostly involved DNA base (deletion or substitution) changes that
in the detoxification and metabolism of phenobarbital. occur at a population frequency greater than 1% are
Consequently these genes were upregulated with common variations that affect host susceptibility to
phenobarbital treatment, but not with peroxisome a variety of factors [48]. Single nucleotide polymor-
proliferators. Interestingly, an EST is included in node I, phisms (SNPs), which are a form of genetic variation,
indicating similarity in the expression pattern between occur approximately once every 1000 bases [49]. Al-
this unknown gene and the rest of the genes in this node. though interindividual similarity is more than 99.9% in
The classification of this EST in node I raises the ques- the human genetic makeup, it is subtle variation such
tion of whether we can infer that this gene is involved as polymorphisms that are responsible for differences
in the detoxification and metabolism of phenobarbital. in resistance or sensitivity to certain drugs or toxicants
This study illustrates the feasibility of discovering the [50–54]. Polymorphisms in enzymes have been shown
function of unknown genes, that may lead to a better to affect their inducibility, expression level, and activity
understanding of mechanisms of action by using cDNA [55]. DNA microarrays offer a novel method of study-
microarrays containing significant numbers of ESTs to ing variations in the response to toxicants or drugs by
study toxicant-exposed biological samples in vivo. monitoring genome-wide differences in gene expres-
The study of mechanisms of toxicant action is vi- sion of key metabolizing or detoxification enzymes in
tal to understanding their adverse effects on human a target cell type.
health. Mechanistic information is crucial to making Under the premise that some polymorphisms lead
more informed regulatory decisions regarding expo- to wide fluctuations in gene expression across indi-
sure levels. Because of the ability to monitor expression viduals, one could screen individuals and potentially
of thousands of genes on a cDNA chip, researchers can identify new polymorphisms. Changes in gene ex-
now study the effects of a chemical insult on multi- pression between resistant versus susceptible individ-
ple molecular pathways. This investigative approach uals may indicate alterations in a signaling/metabolic
allows for the accumulation of mechanistic data that pathway, allowing further definition of the exact gene
will provide an insight into vital cellular pathways that that contains the critical polymorphism. The inclu-
are affected by the exposure to a chemical agent. sion of ESTs on cDNA chips generates the potential
Given the large number of genes on a chip, high- to discover new members of gene families involved
density gene expression data theoretically should sum- in interindividual susceptibility to human disease. Un-
marize the findings from previous reports as well as fortunately, polymorphisms do not always affect the
identify unreported alterations of genes. Figure 4 de- levels of mRNA expression, thus rendering them un-
picts a portion of a toxicant effector pathway map where detectable if cDNA arrays were the only tool used for
gene expression data from phenobarbital-treated livers this purpose. Other challenges to using microarrays for
238 HAMADEH ET AL. Volume 15, Number 5, 2001

FIGURE 4. Map of Effector Pathways of Toxicant (MEPT) for the action of phenobarbital in the mitochondria. mRNA levels corresponding to
all enzymes on the diagram were found to be statistically altered by phenobarbital treatment of Sprague-Dawley rats when gene expression
data was analyzed at a 95% confidence level using Array Suite software. The diagram shows data from two time points: 24 h post single-dose
and two week post daily-dosage. Enzymes enclosed in blue boxes were altered in this study and were previously reported in the literature
to be modulated by phenobarbital treatment while those in green were not previously associated with phenobarbital exposure and are novel
observations. (o/o) denotes modulation of mRNA level at the two time points where the spots before and after the slash correspond to the 24 h
and 2 week time points respectively. Up- or downregulation is denoted by a red or green spot respectively, and a gray spot denotes no change.

FIGURE 5. Clustering output highlighting interanimal differences in gene alteration in response to chemical exposure. Validated gene expression
data from livers of phenobarbital treated animals (24 h post single-dose or 2 week post daily-dosage) were clustered and the output displayed
as a dendrogram. Three rats were represented from each time point. Nodes on the hierarchical tree, highlighting differences in gene expression
patterns between animals receiving similar exposures were magnified. Deviant animals are marked as indicated ∗.
Volume 15, Number 5, 2001 GENE ARRAY TECHNOLOGY IN TOXICOLOGY 239

this purpose include tissue specificity of gene expres- the automated hybridization stations and autoloaded
sion, limited availability of human target tissue, and scanners are starting to emerge. Because techniques in-
definition of reference samples. volved in microarray studies are not yet standardized,
In addition to comparison of normal versus dis- researchers face technical difficulties at different stages
eased, or sensitive versus resistant organisms or indi- when performing the assay. The success of hybridiza-
viduals, the identification of candidate genes that might tion is highly correlated with superior RNA quality,
play a role in susceptibility to toxic agents can be con- good signal-to-noise ratio, low cross-hybridization, and
ducted using microarray analysis of samples from hu- exclusion of fluor incorporation bias during labeling re-
man derived cell lines. Hypotheses generated from in actions. Microarray technology monitors fluctuations
vitro models can then be tested in the in vivo envi- in mRNA levels, but it does not verify if the message
ronment. For example, interindividual variation in sus- was translated into functional protein. Proteomics, a
ceptibility to carcinogenic effects of arsenic has been field that deals with global protein expression and func-
reported [56–59]. Since the mechanism of arsenic toxic- tion, will complement and extend data obtained us-
ity is not fully understood, inclusion of ESTs on cDNA ing genomic approaches [67–69]. Studies using paral-
microarrays may aid in the discovery of novel genes lel analysis of samples with both high density gene
that play a central role in sensitivity. Fresh or immortal- and protein expression tools will lead to verification
ized cell populations (ex. keratinocytes) derived from of the relevance and importance of gene expression
healthy human donors could be screened by biological data. Challenges facing proteomics efforts include over-
assays such as viability in order to measure the rela- coming labor intensive steps (i.e. protein fractiona-
tive resistance or susceptibility of different cell pop- tion, purification, separation) and the inability to match
ulations. This would identify cells sensitive and re- the high output of data generated by genomics at the
sistant to the toxic effects of arsenic. Comparison of present time.
the gene expression profiles of arsenic-resistant cells to Identification of environmental carcinogens and
sensitive cells should indicate mechanistic differences other hazards constitutes a major challenge in the field
between the two responses to arsenic exposure. Addi- of toxicology. The risk assessment process associated
tionally this analysis would potentially identify candi- with these chemicals is limited because of the use of
date genes that play a role in the increased sensitivity or a variety of assumptions, particularly relating to cross-
resistance to arsenic toxicity [60]. The hypotheses gen- species extrapolation, dose response, and estimated ex-
erated by these studies could be further investigated by posure levels of humans [70–73]. Thus, the need has
construction of cell or animal models that either over- arisen for elucidation of transspecies, chemical-specific
express or underexpress for those genes. biomarkers resulting from quantified exposures to
Although the field of toxicology is just beginning to drugs or toxic chemicals. This will permit the extrapola-
embrace the potential of DNA microarrays, investiga- tion of effects observed in surrogate systems to potential
tors in other scientific disciplines have benefited from effects in humans. At this time, there is a limited inven-
the recent advancements in the field of pharmacoge- tory of gene expression patterns that reflect responses
nomics [61–63]. Pharmacogenomics is a recent term re- to chemical effects in biological models. Other impor-
ferring to the identification and elucidation of genetic tant issues such as variations in gene expression due
variations that could serve as targets for therapeutic to the animals’ status, diet, circadian rhythm, or meth-
modalities or impact the efficacy of drugs [64–66]. The ods of tissue collection are not well understood. Cells
field of pharmacogenomics has realized the benefits of in culture also undergo remarkable changes in gene ex-
DNA microarrays and researchers in the field have al- pression solely due to culture conditions. Such informa-
ready used this technology to improve their drug dis- tion would be equally beneficial to the toxicology and
covery efforts. The future use of microarrays in the field pharmaceutical communities. Gene expression profiles
of toxicology will allow for a versatile approach to in- for compounds where toxicological, pharmacological,
vestigate interindividual susceptibility to toxicants due and pathological information is abundant should be
to genetic factors. generated in several biological models of interest. Dif-
ferent doses, time points, and organs should be in-
vestigated to generate a comprehensive gene expres-
CHALLENGES sion response to the chemical. A database that is popu-
lated with this information could then be interrogated
Although the theoretical applications seem endless, for forming associations between gene expression and
cDNA microarray technology has certain limitations. development of various efficacious or toxicity-related
One limitation is the number of samples that can be pro- phenotypes.
cessed efficiently at a time. Automation is being applied An effort to distinguish classes of toxicants, based
to microarray technology and new equipment such as solely upon cluster-type analysis of differentially
240 HAMADEH ET AL. Volume 15, Number 5, 2001

expressed genes in exposed HepG2 cells, was recently ACKNOWLEDGMENTS


reported [74] using an approximately 250-gene cDNA
microarray. Initial comparison of gene expression pro- We gratefully acknowledge the helpful comments
files for cytotoxic antiinflammatory drugs and DNA- and critical review of the manuscript by Drs. Doug
damaging agents to a database populated with gene Bell, Mike Cunningham, Jack Taylor, Rupesh Amin,
expression profiles for 100 toxicants failed to distin- Ray Tennant, and Edward Lobenhofer. We also thank
guish the two classes of toxicants. The main obstacle our collaborators at Boehringer-Ingelheim Pharmaceu-
encountered was the lack of replicates of the gene ex- ticals, Inc., Drs. Ray Stoll, Kerry Blanchard, and Supriya
pression profiles populating their database leading to Jayadev for their contribution to the studies described
low reproducibility in gene responses. This challenge in this review.
was partially overcome when a gene expression profile
was generated from 13 independent cisplatin-exposed
HepG2 samples, leading to a suite of genes that were REFERENCES
altered by toxicant exposure with relatively high repro-
ducibility [74]. This highlights the need for the gener- 1. Schena M, Shalon D, Davis RW, Brown PO. Quanti-
ation of replicate gene expression profiles for a given tative monitoring of gene expression patterns with a
sample. complementary DNA microarray. Science 1995;270:467–
470.
When designing toxicogenomic studies, the num- 2. Lipshutz RJ, Fodor SP, Gingeras TR, Lockhart DJ. High
ber of animals required for each time point or dose is density synthetic oligonucleotide arrays. Nat Genet
an important consideration. Because of economic lim- 1999;21:20–24.
itations when dealing with microarray studies, inves- 3. Fodor SP, Read JL, Pirrung MC, Stryer L, Lu AT, Solas
tigators strive to use the least number of animals that D. Light-directed, spatially addressable parallel chemical
synthesis. Science 1991;251:767–773.
produce statistically meaningful results. There are sev- 4. Pirrung MC, Huang CY. A general method for the
eral considerations that are of importance when choos- spatially defined immobilization of biomolecules on
ing the number of subjects per dose or time point for glass surfaces using “caged” biotin. Bioconjug Chem
a given study. A major consideration is the amount of 1996;7:317–321.
interanimal variation in the model system employed. 5. Bushel PR, Hamadeh HK, Bennett L, Sieber S, Martin K,
Nuwaysir EF, Johnson K, Reynolds K, Paules RS, Afshari
Figure 5 shows results of a hierarchical clustering tree CA. MAPS: A microarray project system for gene expres-
highlighting the differences between animals receiving sion experiment information and data validation. Bioin-
the same chemical treatments. Discrepancies in expres- formatics 2001;7:564–565.
sion of the suites of genes shown were not unique to 6. Ermolaeva O, Rastogi M, Pruitt KD, Schuler GD, Bittner
one of the animals, rather different animals deviated ML, Chen Y, Simon R, Meltzer P, Trent JM, Boguski MS.
Data management and analysis for gene expression ar-
from the group of animals in the expression pattern of rays. Nat Genet 1998;20:19–23.
different sets of genes (Figure 5). It is accepted that four 7. Scherf U, Ross DT, Waltham M, Smith LH, Lee JK,
to six animals per dosage group would be adequate to Tanabe L, Kohn KW, Reinhold WC, Myers TG, Andrews
derive statistically and biologically significant data that DT, Scudiero DA, Eisen MB, Sausville EA, Pommier Y,
overcomes interanimals variability. Botstein D, Brown PO, Weinstein JN. A gene expression
database for the molecular pharmacology of cancer. Nat
Genet 2000;24:236–244.
8. Chen Y, Dougherty ER, Bittner ML. Ratio-based decisions
CONCLUSIONS and the quantitative analysis of cDNA microarray im-
ages. J Biomed Opt 1997;2(4):364–374.
DNA microarrays have facilitated discovery in 9. Wittes J, Friedman HP. Searching for evidence of altered
gene expression: A comment on statistical analysis of mi-
many scientific disciplines because of the versatility of croarray data [editorial; comment]. J Natl Cancer Inst
this novel technology. We highlight the potential use 1999;91:400–401.
of this technology in discovering new aspects of mech- 10. Lee ML, Kuo FC, Whitmore GA, Sklar J. Importance of
anisms of action of toxicants, the discovery of toxico- replication in microarray gene expression studies: Sta-
logical functions of unknown genes, and in investigat- tistical methods and evidence from repetitive cDNA hy-
bridizations. Proc Natl Acad Sci USA 2000;97:9834–9839.
ing polymorphisms. The use of cDNA microarrays for 11. Casella G, Berger RL. Statistical Inference, Belmont, CA:
approaching polymorphisms in a novel fashion will Wadsworth, Inc; 1990.
accelerate the identification of polymorphisms that con- 12. Wolfinger RD, Gibson G, Wolfinger ED, Bushel P,
fer sensitivity to toxicants. Not only will these efforts Hamadeh HK, Bennett L, Paules R, Afshari CA. Assess-
help advance the field of risk assessment by better ing gene significance from cDNA microarray expression
data via mixed models. J Comput Biol 2001 (in press).
identifying susceptible populations, but will widen our 13. Hughes TR, Roberts CJ, Dai H, Jones AR, Meyer MR,
knowledge of the mechanisms of action for compounds Slade D, Burchard J, Dow S, Ward TR, Kidd MJ, Friend
that we are exposed to on a continuous basis. SH, Marton MJ. Widespread aneuploidy revealed by
Volume 15, Number 5, 2001 GENE ARRAY TECHNOLOGY IN TOXICOLOGY 241

DNA microarray expression profiling. Nat Genet 2000; 30. Ginsberg SD, Hemby SE, Lee VM, Eberwine JH,
25:333–337. Trojanowski JQ. Expression profile of transcripts in
14. Eisen MB, Spellman PT, Brown PO, Botstein D. Clus- Alzheimer’s disease tangle-bearing CA1 neurons. Ann
ter analysis and display of genome-wide expression pat- Neurol 2000;48:77–87.
terns. Proc Natl Acad Sci USA 1998;95:14863–14868. 31. Golub TR, Slonim DK, Tamayo P, Huard C, Gaasenbeek
15. Tamayo P, Slonim D, Mesirov J, Zhu Q, Kitareewan M, Mesirov JP, Coller H, Loh ML, Downing JR, Caligiuri
S, Dmitrovsky E, Lander ES, Golub TR. Interpreting MA, Bloomfield CD, Lander ES. Molecular classification
patterns of gene expression with self-organizing maps: of cancer: Class discovery and class prediction by gene
Methods and application to hematopoietic differentia- expression monitoring. Science 1999;286:531–537.
tion. Proc Natl Acad Sci USA 1999;96:2907–2912. 32. Farr S, Dunn RT. Concise review: Gene expression ap-
16. Li L, Darden TA, Weinberg CR, Pedersen LG. Gene plied to toxicology. Toxicol Sci 1999;50:1–9.
assessment and sample classification for gene expres- 33. Bartosiewicz M, Trounstine M, Barker D, Johnston R,
sion data using a genetic algorithm/k-nearest neighbor Buckpitt A. Development of a toxicological gene array
method. Combinatorial Chemistry & High Throughput and quantitative assessment of this technology. Arch
Screening 2001 (in press). Biochem Biophys 2000;376:66–73.
17. Hamadeh HK, Bushel PR, Jayadev S, Martin K, DiSorbo 34. Nuwaysir EF, Bittner M, Trent J, Barrett JC, Afshari CA.
O, Sieber S, Bennett L, Tennant R, Stoll R, Barrett JC, Microarrays and toxicology: The advent of toxicoge-
Blanchard K, Paules RS, Afshari CA. Prediction of com- nomics. Mol Carcinog 1999;24:153–159.
pound signature using high density gene expression pro- 35. Rockett JC, Dix DJ. Application of DNA arrays to toxicol-
filing. In preparation. ogy. Environ Health Perspect 1999;107:681–685.
18. Iyer VR, Eisen MB, Ross DT, Schuler G, Moore T, Lee JCF, 36. Pennie WD. Use of cDNA microarrays to probe and un-
Trent JM, Staudt LM, Hudson J Jr, Boguski MS, Lashkari derstand the toxicological consequences of altered gene
D, Shalon D, Botstein D, Brown PO. The transcriptional expression. Toxicol Lett 2000;112/113:473–477.
program in the response of human fibroblasts to serum. 37. Waring JF, Ulrich RG. The impact of genomics-based tech-
Science 1999;283:83–87. nologies on drug safety evaluation. Annu Rev Pharmacol
19. Roberts CJ, Nelson B, Marton MJ, Stoughton R, Meyer Toxicol 2000;40:335–352.
MR, Bennett HA, He YD, Dai H, Walker WL, Hughes TR, 38. Rodi CP, Bunch RT, Curtiss SW, Kier LD, Cabonce MA,
Tyers M, Boone C, Friend SH. Signaling and circuitry of Davila JC, Mitchell MD, Alden CL, Morris DL. Revolution
multiple MAPK pathways revealed by a matrix of global through genomics in investigative and discovery toxico-
gene expression profiles. Science 2000;287:873–880. logy. Toxicol Pathol 1999;27:107–110.
20. Phillips RL et al. The genetic program of hematopoietic 39. Hamadeh HK, Bushel P, Nuwaysir EF, Paules R, Barrett
stem cells. Science 2000;288:1635–1640. JC, Afshari CA. cDNA microarray technology: Merging
21. Marton MJ et al. Drug target validation and identification toxicology and genomics. In: Comments on Toxicology.
of secondary drug target effects using DNA microarrays. Amsterdam: Overseas Publishers Association; 2001.
Nat Med 1998;4:1293–1301. 40. Perret X, Fellay R, Bjourson AJ, Cooper JE, Brenner S,
22. Amundson SA et al. Identification of potential mRNA Broughton WJ. Subtraction hybridisation and shot-gun
biomarkers in peripheral blood lymphocytes for human sequencing: A new approach to identify symbiotic loci.
exposure to ionizing radiation. Radiat Res 2000;154:342– Nucleic Acids Res 1994;22:1335–1341.
346. 41. Zabarovskii ER, Dominskii DA, Kiselev LL. Physical
23. Wilson M et al. Exploring drug-induced alterations mapping of the human genome: On the way to devel-
in gene expression in Mycobacterium tuberculosis by oping an optimal strategy. Mol Biol (Mosk) 1994;28:1231–
microarray hybridization. Proc Natl Acad Sci USA 1244.
1999;96:12833–12838. 42. Zabarovsky ER, Kashuba VI, Pettersson B, Petrov N,
24. Tanaka TS et al. Genome-wide expression profiling of Zakharyev V, Gizatullin R, Lebedeva T, Bannikov V,
mid-gestation placenta and embryo using a 15,000 mouse Pokrovskaya ES, Zabarovska VI et al. Shot-gun sequenc-
developmental cDNA microarray. Proc Natl Acad Sci ing strategy for long-range genome mapping: A pilot
USA 2000;97:9127–9132. study. Genomics 1994;21:495–500.
25. Su YA et al. Identification of tumor-suppressor genes 43. Thurman RG, Marazzo DP. Mixed function oxidation and
using human melanoma cell lines UACC903, UACC903 intermediary metabolism: Metabolic interdependencies
(+6), and SRS3 by comparison of expression profiles. Mol in the liver. Adv Exp Med Biol 1975;58:355–367.
Carcinog 2000;28:119–127. 44. Lobenhofer E, Bushel P, Afshari CA, Hamadeh HK.
26. Webb GC et al. Expression profiling of pancreatic beta Progress in the application of DNA microarrays. Envi-
cells: Glucose regulation of secretory and metabolic ron Health Pespect 2001;109:877–880.
pathway genes. Proc Natl Acad Sci USA 2000;97:5773– 45. Hacia JG, Fan JB, Ryder O, Jin L, Edgemon K,
5778. Ghandour G, Mayer RA, Sun B, Hsie L, Robbins CM,
27. Stanton LW, Garrard LJ, Damm D, Garrick BL, Lam A, Brody LC, Wang D, Lander ES, Lipshutz R, Fodor
Kapoun AM, Zheng Q, Protter AA, Schreiner GF, White SP, Collins FS. Determination of ancestral alleles for
RT. Altered patterns of gene expression in response to human single-nucleotide polymorphisms using high-
myocardial infarction. Circ Res 2000;86:939–945. density oligonucleotide arrays. Nat Genet 1999;22:164–
28. Nesbit CE, Tersak JM, Grove LE, Drzal A, Choi H, 167.
Prochownik EV. Genetic dissection of c-myc apoptotic 46. Ruvkun G, Gilbert W, Horvitz HR. Detection of mutations
pathways. Oncogene 2000;19:3200–3212. and DNA polymorphisms using whole genome Southern
29. Lyons TJ, Gasch AP, Gaither LA, Botstein D, Brown PO, Cross hybridization. Nucleic Acids Res 1990;18:809–815.
Eide DJ. Genome-wide characterization of the Zap1p 47. Kajander OA, Kunnas TA, Perola M, Lehtinen SK,
zinc-responsive regulon in yeast. Proc Natl Acad Sci USA Karhunen PJ, Jacobs HT. Long-extension PCR to detect
2000;97:7957–7962. deleted mitochondrial DNA molecules is compromized
242 HAMADEH ET AL. Volume 15, Number 5, 2001

by technical artefacts. Biochem Biophys Res Commun 61. Sauer S, Lechner D, Berlin K, Lehrach H, Escary JL, Fox
1999;254:507–514. N, Gut IG. A novel procedure for efficient genotyping
48. Brookes AJ. The essence of SNPs. Gene 1999;234:177–186. of single nucleotide polymorphisms. Nucleic Acids Res
49. McCarthy JJ, Hilfiker R. The use of single-nucleotide 2000;28:E13.
polymorphism maps in pharmacogenomics. Nat Biotech- 62. Srivastava M, Eidelman O, Pollard HB. Pharmacoge-
nol 2000;18:505–508. nomics of the cystic fibrosis transmembrane conductance
50. Onalaja AO, Claudio L. Genetic susceptibility to lead regulator (CFTR) and the cystic fibrosis drug CPX using
poisoning. Environ Health Perspect 2000;108 (Suppl 1): genome microarray analysis. Mol Med 1999;5:753–767.
23–28. 63. Giaever G, Shoemaker DD, Jones TW, Liang H, Winzeler
51. Smith RL. Polymorphism in drug metabolism— EA, Astromoff A, Davis RW. Genomic profiling of drug
implications for drug toxicity. Arch Toxicol Suppl sensitivities via induced haploinsufficiency. Nat Genet
1986;9:138–146. 1999;21:278–283.
52. Boobis AR. Molecular basis for differences in sus- 64. Evans WE, Relling MV. Pharmacogenomics: Translating
ceptibility to toxicants: Introduction. Toxicol Lett functional genomics into rational therapeutics. Science
1992;64/65:109–113. 1999;286:487–491.
53. el-Zein R, Zwischenberger JB, Wood TG, Abdel-Rahman 65. Ferrari P. Pharmacogenomics: A new approach to indi-
SZ, Brekelbaum C, Au WW. Combined genetic polymor- vidual therapy of hypertension? Curr Opin Nephrol Hy-
phism and risk for development of lung cancer. Mutat pertens 1998;7:217–222.
Res 1997;381:189–200. 66. Grant DM. Pharmacogenomics and the changing face of
54. el-Zein R, Conforti-Froes N, Au WW. Interactions be- clinical pharmacology. Can J Clin Pharmacol 1999;6:131–
tween genetic predisposition and environmental toxi- 132.
cants for development of lung cancer. Environ Mol Mu- 67. Haynes PA, Yates JR III. Proteome profiling-pitfalls and
tagen 1997;30:196–204. progress. Yeast 2000;17:81–87.
55. Landi MT, Sinha R, Lang NP, Kadlubar FF. Human 68. Gevaert K, Vandekerckhove J. Protein identification
cytochrome P4501A2. IARC Sci Publ 1999;148:173– methods in proteomics. Electrophoresis 2000;21:1145–
195. 1154.
56. Vega L, Gonsebatt ME, Ostrosky-Wegman P. Aneugenic 69. Pandey A, Mann M. Proteomics to study genes and
effect of sodium arsenite on human lymphocytes in vitro: genomes. Nature 2000;405:837–846.
An individual susceptibility effect detected. Mutat Res 70. Whittemore AS. Cancer risk assessment and prevention:
1995;334:365–373. Where do we stand? Environ Health Perspect 1989;81:95–
57. Vahter M. Genetic polymorphism in the biotransforma- 101.
tion of inorganic arsenic and its role in toxicity. Toxicol 71. Kavlock RJ, Setzer RW. The road to embryologically
Lett 2000;112/113:209–217. based dose-response models. Environ Health Perspect
58. Rasmussen RE, Menzel DB. Variation in arsenic-induced 1996;104 (Suppl 1):107–121.
sister chromatid exchange in human lymphocytes and 72. Savitz DA, Harlow SD. Selection of reproductive
lymphoblastoid cell lines. Mutat Res 1997;386:299– health end points for environmental risk assess-
306. ment. Environ Health Perspect 1991;90:159–164.
59. Chiou HY, Hsueh YM, Hsieh LL, Hsu LI, Hsu YH, Hsieh 73. Burin GJ, Saunders DR. Addressing human variability in
FI, Wei ML, Chen HC, Yang HT, Leu LC, Chu TH, risk assessment—the robustness of the intraspecies un-
Chen-Wu C, Yang MH, Chen CJ. Arsenic methylation ca- certainty factor. Regul Toxicol Pharmacol 1999;30:209–
pacity, body retention, and null genotypes of glutathione 216.
S-transferase M1 and T1 among current arsenic-exposed 74. Burczynski ME, McMillian M, Ciervo J, Li L, Parker
residents in Taiwan. Mutat Res 1997;386:197–207. JB, Dunn RT II, Hicken S, Farr S, Johnson MD.
60. Chin KV, Chauhan SS, Pastan I, Gottesman MM. Regula- Toxicogenomics-based discrimination of toxic mecha-
tion of mdr RNA levels in response to cytotoxic drugs in nism in HepG2 human hepatoma cells. Toxicol Sci
rodent cells. Cell Growth Differ 1990;1:361–365. 2000;58:399–415.

También podría gustarte