microarray_CK+inflam

Embed Size (px)

Citation preview

  • 8/10/2019 microarray_CK+inflam

    1/12

    Biomarkers of acute respiratory allergen exposure: Screening for

    sensitization potential

    Cherie M. Pucheu-Haston a,, Lisa B. Copeland b, Beena Vallanat b, Elizabeth Boykin b, Marsha D.W. Ward b

    a Curriculum in Toxicology, University of North Carolina-Chapel Hill, CB# 7270, Chapel Hill, NC 27599-7270, USAb National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Research Triangle Park, NC 27711, USA

    a b s t r a c ta r t i c l e i n f o

    Article history:

    Received 9 November 2009Revised 10 December 2009

    Accepted 17 December 2009

    Available online 4 January 2010

    Keywords:

    Asthma

    Biomarkers

    Hazard screening

    Intratracheal aspiration

    Gene expression microarray

    Quantitative real-time polymerase chain

    reaction (qRT-PCR)

    Effective hazard screening will require the development of high-throughput or in vitro assays for the

    identication of potential sensitizers. The goal of this preliminary study was to identify potential biomarkers

    that differentiate the response to allergensvsnon-allergens following an acute exposure in nave individuals.

    Female BALB/c mice received a single intratracheal aspiration exposure to Metarhizium anisopliae crude

    antigen (MACA) or bovine serum albumin (BSA) in Hank's Balanced Salt Solution (HBSS) or HBSS alone. Mice

    were terminated after 1, 3, 6, 12, 18 and 24 h. Bronchoalveolar lavage uid (BALF) was evaluated to

    determine total and differential cellularity, total protein concentration and LDH activity. RNA was isolated

    from lung tissue for microarray analysis and qRT-PCR. MACA administration induced a rapid increase in BALF

    neutrophils, lymphocytes, eosinophils and total protein compared to BSA or HBSS. Microarray analysis

    demonstrated differential expression of genes involved in cytokine production, signaling, inammatory cell

    recruitment, adhesion and activation in 3 and 12 h MACA-treated samples compared to BSA or HBSS. Further

    analyses allowed identication of100 candidate biomarker genes. Eleven genes were selected for further

    assessment by qRT-PCR. Of these, 6 demonstrated persistently increased expression (Ccl17, Ccl22, Ccl7,

    Cxcl10, Cxcl2, Saa1), while C3ar1 increased from 624 h. In conclusion, a single respiratory exposure of mice

    to an allergenic mold extract induces an inammatory response which is distinct in phenotype and gene

    transcription from the response to a control protein. Further validation of these biomarkers with additional

    allergens and irritants is needed. These biomarkers may facilitate improvements in screening methods. 2009 Elsevier Inc. All rights reserved.

    Introduction

    One particularly challenging area of immunotoxicology is the

    identication of agents with a high likelihood to induce allergic

    sensitization. Traditional approaches to screening for these agents

    require time consuming multiple exposure protocols. Unfortunately,

    these methods are becoming increasingly incapable of meeting the

    screening needs of the rapidly expanding list of newly synthesized

    and bioengineered compounds, not to mention re-screening those

    agents that have been discovered to persist in the environment for

    longer than originally expected. Efcient identication of problematic

    agentswill require a shift to more high-throughput methods of hazard

    screening. It is probable that a multidisciplinary approach will be

    required, where agents will be evaluated using multiple models, with

    the nal decisions being made based upon the preponderance of

    evidence.

    We hypothesize that there is a set of differentially expressed

    gene biomarkers that distinguishes the immune response associ-

    ated with the induction of allergic sensitization from non-allergic

    immune responsiveness following a single exposure. The objectives

    of this study were two-fold: (1) to determine whether we could

    identify biomarkers of acute exposure to a known respiratory

    sensitizer as compared to a poorly allergenic compound or vehicle

    control; (2) to use this information to aid in the development of a

    more extensive validation study. The ultimate goal of this work is to

    translate these results into the development of an in vitro screening

    assay.

    To test our hypothesis, we selected the entomopathogenic fungus

    Metarhizium anisopliae extract which we have previously shown to

    induce robust responses characteristic of human allergic asthma in

    adult BALB/c mice following multiple exposures (Ward et al., 1998,

    2000). These responses can be obtained following intratracheal

    aspiration (IA) exposure to M. anisopliae crude antigen (MACA).

    Neither intraperitoneal priming nor adjuvant administration is

    required for sensitization (Ward et al., 2000). As low or non-allergic

    agent, we selected bovine serum albumin (BSA) which does not

    induceallergicor asthma-like responsesin ourmousemodel(Viana et

    al., 2002; Ward et al., 2009).

    Toxicology and Applied Pharmacology 244 (2010) 144155

    Corresponding author. c/o U.S. Environmental Protection Agency 109 T. W.

    Alexander Drive, Research Triangle Park, NC 27711, USA. Fax: +1 919 541 4284.

    E-mail addresses: [email protected],[email protected]

    (C.M. Pucheu-Haston).

    0041-008X/$ see front matter 2009 Elsevier Inc. All rights reserved.

    doi:10.1016/j.taap.2009.12.027

    Contents lists available at ScienceDirect

    Toxicology and Applied Pharmacology

    j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / y t a a p

    mailto:[email protected]:[email protected]://dx.doi.org/10.1016/j.taap.2009.12.027http://www.sciencedirect.com/science/journal/0041008Xhttp://www.sciencedirect.com/science/journal/0041008Xhttp://dx.doi.org/10.1016/j.taap.2009.12.027mailto:[email protected]:[email protected]
  • 8/10/2019 microarray_CK+inflam

    2/12

    Methods

    Animals. Eight-week-old female BALB/c mice were obtained from

    Charles River Laboratories (Raleigh, NC). All mice were housed in

    polycarbonate cages with hardwood chip bedding in an environmen-

    tally controlled, Association for Assessment and Accreditation of

    Laboratory Animal Care International (AAALAC)-accredited animal

    facility. Environmental enrichment was provided in the form of

    mouse nestlets (Ancare Corporation, Bellmore, NY). Sentinel micewere monitored serologically and were found to be free of Sendai,

    mouse pneumonia, mouse hepatitis, other murine viruses, and

    mycoplasma. Mice also were monitored for, and found to be free of,

    ectoparasites and endoparasites. The animal holding room tempera-

    ture (22 F 1.1 F) and relative humidity (50% 10%) were

    maintained at the recommended levels. Mice received standard

    rodent chow (Lab Diet 5P00, Purina Mills, St. Louis, MO) and water

    ad libitum. Mice were acclimated 1 week at this facility prior to the

    start of the experiment.

    Antigen preparation. M. anisopliae strain 1080 was obtained from

    USDA-ARS Entomopathogenic Fungus Collection in Ithaca, NY.

    Extracts of M. anisopliae mycelium and spores/conidia as well as

    inducible enzymes ltrate were produced as previously described

    (Ward et al., 1998). Briey, a fungal extract was prepared by a

    modication of the methods described by (Stankus and O'Neil,

    1988). Mycelium (hyphae growth) was grown at 27 C for 72 h in

    Sabouraud's maltose broth with aeration (150 rpm). Subsequently,

    the mycelium was collected on Whatman No. 1 lter paper, washed

    twice with saline to remove media contaminants, and air-dried

    overnight in a biosafety cabinet. The conidia, grown at 27 C on

    Sabouraud's maltose agar, were harvested by gently scraping culture

    plates of 2- to 3-week-old cultures. Each component was ground into

    a paste in saline (1:2530 w/v) and stirred overnight at 4 C. The

    supernatant was collected following centrifugation at 18,000g.

    Production of inducible proteases and chitinases by M. anisopliae is

    enhanced in the absence of readily available nitrogen and carbon.

    Therefore, a deprivation medium of unpuried chitin (Sigma

    Chemical Co., St. Louis, MO) at 3% in water was inoculated with M.anisopliaeand incubated at 27 C for 72 h with aeration (150 rpm).

    Theltrate was retained following ltration through Whatman No. 1

    lter paper. The proteins with a molecular weight greater than 3000

    were concentrated using a YM-3 lter (MWCO 3000) (Amicon,

    Beverly, MA) and assayed for total protein concentration (as

    described below). For each component, the pH was adjusted to 6.0

    followed by lter sterilization through a 0.2-um syringe lter

    (Acrodisc; Gelman Sciences, Ann Arbor, MI). Equal protein con-

    centrations of each component were combined to form the M.

    anisopliae crude antigen (MACA). Dosing aliquots were stored at

    20 C for a maximum of 10 wks until use. Extract endotoxin

    concentrations were minimal (0.0140.007 ng per 20 g dose) as

    measured by LAL assay (Lonza Group Ltd., Basel, Switzerland).

    Bovine serum albumin (BSA), fraction V, was purchased fromSigma-Aldrich (St. Louis, MO) and diluted in Hanks Balanced Salt

    Solution (HBSS Gibco/Invitrogen, Carlsbad, CA). Dosing aliquots were

    stored at 20 C until use.

    Experimental design. Female BALB/c mice were anesthetized, then

    given a single dose of MACA (20 g in 50 l HBSS), BSA (20 g in 50 l

    HBSS) or HBSS alone (50 l) by intratracheal aspiration (IA) as

    previously described (Ward et al., 2000). Briey, mice were

    anesthetized by inhalation of a mixture of isourane and oxygen,

    and then vertically suspended by their incisors to facilitate treatment

    administration. Swallowing was prevented by gently pulling the

    tongue out of the mouth in an upward direction. Antigen extract was

    deposited into the oropharynx while the nose was briey occluded,

    inducing aspiration of theextract. Sixmice from each treatmentgroup

    were terminated by sodium pentobarbital overdose (Euthasol, Virbac

    Animal Health, Fort Worth, TX) at each of six time points (1 h, 3 h, 6 h,

    12 h, 18 h and 24 h post-treatment).

    Bronchoalveolar lavage (BAL) and lung collection. Lung tissue and

    bronchoalveolar lavage uid (BALF) samples were collected from all

    mice at termination. The left lung was harvested and snap-frozen in

    liquid nitrogen immediately after euthanasia, then stored at 80 C

    until RNA isolation. The remaining lung tissue was lavaged twicewith 0.7 ml aliquots of HBSS. BALF aliquots for each animal were

    pooled and stored on ice. The BALF was centrifuged at 800 rpm

    (100g) for 15 min at 4 C. Aliquots of BALF supernatant were

    assayed for total protein and lactate dehydrogenase (LDH) activity

    (as described below), and the remainder was stored at 20 C for

    IgE quantication by ELISA. The cell pellet was resuspended in 1 ml

    HBSS and cytospin preparations of 200 l BALF were made by

    centrifugation onto glass slides (200 rpm, 10 min on a Shandon

    Cytospin; Shandon Inc., Pittsburg, PA). Following Wright-Giemsa

    staining (Fisher Scientic, Fairlawn, NJ), cells were differentially

    counted at 200 cells per slide (one slide per animal). Total BALF cell

    counts were obtained from the resuspended cells using a Coulter

    counter (Coulter Corp., Miami, FL).

    Total protein and lactate dehydrogenase (LDH) assays. BALF samples

    and fungal extract components were assayed for total protein using

    Pierce Coomassie Plus Protein Assay Reagent (Pierce/Thermo Fisher

    Scientic, Rockford, IL). Concentrations were determined from a

    standard curve using BSA standards obtained from Sigma Chemical

    Co. (St. Louis, MO). BSA works well as a general-purpose standard for

    protein quantitation. However, due to differences in amino acid

    concentrations and protein extinction coefcients, there may be some

    imprecision in the measurement of any complex protein mixture

    (such as a fungal extract or lavage uid) with any single protein

    standard.

    BALF samples were assayed for LDH activity using a commercially

    prepared kit and controls from Sigma Chemical Co. Both the total

    protein and LDH assays were modied for use on the KONELAB 30

    clinical chemistry Spectrophotometer analyzer (Thermo ClinicalLabsystems, Espoo, Finland).

    RNA isolation. Total RNA was isolated from all frozen lung tissue

    samples using a RNeasy Mini kit (Qiagen Inc., Valencia, CA) according

    to manufacturers instructions. Briey, whole left lungs were kept on

    dry ice before homogenization in 1 ml of RLT lysis buffer using an

    Omni TH homogenizer (Omni International, Marietta, GA). Lung

    homogenate (350 l) was added to an equal volume of 70% ethanol,

    vortexed and added to an RNeasy mini spin column. After

    centrifugation, the column was washed once with RW1 buffer and

    twice with RPE buffer. RNA was then eluted by the addition of RNAse-

    free water. RNasin ribonuclease inhibitor (Promega Corporation,

    Madison, WI) was added immediately after isolation to increase RNA

    stability. RNA quantication was performed using a NanoDrop ND-1000 spectrophotometer (NanoDrop Technologies, Inc., Wilmington,

    DE). RNA quality was assessed using an Agilent 2100 bioanalyzer

    with the Agilent RNA 6000 nano kit (Agilent Technologies, Santa

    Clara, CA).

    Microarray hybridization. Three to 4 unique RNA samples from all 3

    treatment groups at both the 3 and 12 h post-treatment time points

    were submitted for microarray gene expression analysis (MACA 3 h

    n =3, BSA 3 h n =3, HBSS 3 h n =3; MACA 12 h n =3, BSA 12 h

    n =4, HBSS 12 h n =4; total n =24). These sample times were

    chosen based upon a tendency for the BALF inammatory response to

    peak at 6 and 18 h, or both (see Figs. 13). The microarray chosen for

    this study was the Affymetrix Mouse Genome 430A 2.0 (Affymetrix

    Inc., Santa Clara, CA, www.affymetrix.com), which contains over

    145C.M. Pucheu-Haston et al. / Toxicology and Applied Pharmacology 244 (2010) 144155

    http://www.affymetrix.com/http://www.affymetrix.com/
  • 8/10/2019 microarray_CK+inflam

    3/12

    22,500 probe sets representing 14,500 well-characterized mouse

    genes. A total of 21 gene chips (one per mouse) were used in this

    study.

    Overall gene expression data analysis strategy. The analysis of the

    microarray data involved the comparison of a total of six data sets

    from two time points (MACA 3 h, BSA 3 h, HBSS 3 h; MACA 12 h, BSA

    12 h, HBSS 12 h). Identication of putative biomarkers was performed

    in a stepwise fashion: (1) identify sequences differentially expressed

    in either MACA-or BSA-treated animals as compared to time-matched

    HBSS controls, (2) determine sequences differentially expressed in

    MACA-treated samples only, (3) perform biomarkerlter enrichment,

    (4) perform functional expression analysis on biomarker-ltered

    data, and (5) select putative biomarkers. Additionally, differentially

    expressed sequences were subjected to pathway analysis to identify

    fundamental differences in the immune response to compounds of

    high versus low allergenicity (Fig. 1).

    Statistical analysis of microarray data. Data (.cel les) were

    analyzed and statistically ltered using Rosetta Resolver version

    7.1 software (Rosetta Inpharmatics, Kirkland, WA). For analysis

    purposes, the data from individual 25-mer oligonucleotide probe

    pairs were combined into sequence data. Each sequence represents a

    portion of a gene transcript, and is generally comprised of 11

    oligonucleotide probe pairs (reporters). A given gene may be

    represented by one or more sequences, which allows for detection of

    alternate transcripts.

    Sequence expression intensities were averaged for each treatment

    group. Sequences that were differentially expressed within a time

    point between HBSS control groups and MACA or BSA treatment

    groups were identied in Rosetta Resolver by one-way ANOVA with

    Benjamini and Hochberg False Discovery Rate (FDR) analysis(pb0.01), followed by Scheffes post hoc data analysis (pb0.05).

    Sequences identied as statistically different in intensity in either

    MACA or BSA samples as compared to HBSS were selected for ratio

    analysis. These sequences were then further ltered using an

    experimental/control ratio of2.0.

    Filtered sequence lists from MACA- and BSA-treated animals were

    then compared within a time point (i.e., MACA 3 h versus BSA 3 h;

    MACA 12 h versus BSA 12 h) using the list operations function of

    Metacore GeneGo (GeneGo, Inc., St. Joseph, MI; www.genego.com ) to

    identify sequences differentially expressed only in MACA-treated

    animals (MACA-specic sequences). A second analysis on MACA-

    specic sequences was then run to identify those sequences

    differentially expressed at both MACA post-treatment time points

    (MACA 3 h/12 h-speci

    c).

    Fig. 1.Overview of microarray data analysis strategy. RNA was harvested from snap-

    frozen lung tissue and hybridized to a gene expression microarray (Affymetrix Mouse

    Genome 430A 2.0). Sequences that were differentially expressed between HBSS control

    groups and MACA or BSA treatment groups were identied by one-way ANOVA and

    Benjamini and HochbergFDR (pb0.01)followed by Scheffe' post hocanalysis (pb0.05).

    Genes thatchanged2-fold or greater relativeto HBSSwere selected for further analysis.

    Sequences differentially expressed in MACA-treated animals were identied using

    GeneGo/Metacore software. Sequences were then ltered for biomarker potential

    using the biomarker function of Ingenuity Pathway Analysis software. Selected

    sequences weresubjected to functional analysis usingDAVID (Databasefor Annotation,

    Visualization and Integrated Discovery). Expression of putative biomarkers was then

    evaluated using quantitative real-time PCR.

    Fig. 2.BALF LDH and total protein levels. (A) Intratracheal administration (IA) of MACA

    is associated with little acute cellular toxicity (LDH). (B) IA MACA induces a rapid

    increase in BALF total protein levels, suggesting the development of pulmonary cellular

    leakage and/or edema. N=6 per treatment per time point. Pb0.01, Pb0.001

    relative to HBSS;Pb

    0.001 relative to BSA.

    Fig. 3. BALFtotal celullarity. IA MACAinduces a rapid increase in totalairway cellularity

    relative to BSA-or HBSS-treated mice.Pb0.01, Pb0.001 relative to HBSS; Pb0.05,

    Pb0.01,Pb0.001 relative to BSA.

    146 C.M. Pucheu-Haston et al. / Toxicology and Applied Pharmacology 244 (2010) 144155

    http://www.genego.com/http://www.genego.com/
  • 8/10/2019 microarray_CK+inflam

    4/12

    Biomarkerlter analysis. Sequences identied in Metacore GeneGo

    asMACA 3 h/12 h-specicwere then subjected to biomarker lter

    analysis using the Biomarker function of Ingenuity Pathway Analysis

    Software (Ingenuity Systems, Redwood City, CA; www.ingenuity.

    com). This feature facilitates prioritization of putative biomarker

    genes based upon user-dened parameters such as tissue expression,

    species of interest or disease association. For this analysis, potential

    biomarkers were selectedbasedupon thepresence in any oneor more

    of the following: immune system cells (e.g., T lymphocytes,neutrophils, dendritic cells, etc.), immune system cell lines, lung cell

    lines, epidermis, lung, spleen or thymus.

    Functional analysis. Putative biomarker sequences identied by

    Ingenuity were then queried against the gene expression databases in

    Metacore GeneGo, Ingenuity and DAVID (Database for Annotation,

    Visualization and Integrated Discovery, National Institute of Allergy

    and Infectious Diseases, NIH, USA; http://david.abcc.ncifcrf.gov/

    home.jsp) software programs. Candidate biomarkers were prioritized

    and selected based upon relevance to allergic sensitization and

    likelihood of expression in available in vitroculture systems.

    Pathway analysis. Sequences which met all the criteria for differ-

    ential expression relative to HBSS control (i.e., ANOVA/FDRp b0.01,

    Scheffe' p b0.05 and experiment/control ratio 2) were subjected to

    pathway analysis in Ingenuity software. Pathway analysis allows

    superimposition of user-specied sequence lists and expression data

    on known biological pathways and functions in the Ingenuity

    Pathways Knowledge Base. The signicance of the association

    between the data set and canonical pathways was determined in

    two ways: (1) a ratio was generated of the number of genes from the

    data set that mapped to the pathway divided by the total number of

    genes in the pathway, (2) Fischers exact test was used to calculate a

    p-value determining the probability that the association between the

    genes in the data set and the canonical pathway was explained by

    chance alone.

    Conrmation of selected microarray results using quantitative real-time

    polymerase chain reaction (qRT-PCR). Total RNA from all samples (6samples per treatment per time point; n =108) was converted to

    cDNA using a High-capacity RNA-to-cDNA kit (Applied Biosystems

    Inc., Foster City, CA) according to manufacturers instructions. Briey,

    3 g of total lung RNA was reverse transcribed using random hexamer

    primers and Multiscribe reverse transcriptase in a total reaction

    volume of 30 l. Cycling conditions were 25 C for 10 min, 37 C for

    120 min and 85 C for 5 min. All cDNA was stored at20 C until use.

    Quantitative real-time polymerase chain reaction (qRT-PCR) was

    performed on cDNA using TaqMan Universal PCR Master Mix no

    AmpErase UNG (Applied Biosystems). A 30 l total reaction volume

    was used per well, consisting of 100 g of cDNA (in 5 l RNAse-free

    water), 15 l of 2 Master Mix, 8.5 l of RNAse free water and 1.5 l of

    primers and FAM-labeled MGB probes. All primerand probe sets were

    selected from available inventoried assays and purchased fromApplied Biosystems.

    Amplication was performed on an ABI PRISM 7900 thermocyclerat

    thedefault cycling conditions (50C for2 min, 95 C for10 min, followed

    by 40 cycles of 95 C for 15 s (melt) and 60 C for 1 min (anneal)). The

    endogenous control used for these assays was 18 s. Analysis was

    performed using a comparative CT method adapted from (Livak and

    Schmittgen, 2001). The CT value for 2 replicates per sample was

    averaged foreach targetgene andthe endogenous control. Theaveraged

    CT value for 18 s was subtracted from the corresponding CT value for the

    target gene in the same sample (CT target CT endogenous control) =CT.

    Individual CT values were subtracted from the averagedCT value of the

    HBSS control group for that time point=CT. The fold change was

    calculated as 2CT. Average fold changes (relative to HBSS) and

    standard errors were then calculated for each treatmenttime point.

    Statistical analysis of BALF and qRT-PCR data. BALF LDH activity,

    total protein concentration and total and differential cell counts were

    analyzed by one-way analysis of variance (ANOVA) with Bonferroni's

    multiple comparison post-test using GraphPad Prism version 4.03 for

    Windows (GraphPad Software, San Diego, CA,www.graphpad.com).

    For these analyses, values from MACA-treated and BSA-treated

    animals were compared to HBSS-treated controls at the same time

    point. A p-value of b0.05 was considered statistically signicant.

    Reported values represent meansstandard error (SE).Gene transcription data obtained by qRT-PCR are expressed as

    mean fold changes relative to HBSS-treated controlsSE. Relative

    transcription levels for MACA-treated mice were compared to BSA-

    treated mice at each time point in Graph Pad Prism using a two-tailed

    nonparametric MannWhitney test. A value ofpb0.05 was considered

    to be signicant. Where necessary to facilitate comparison in Table 4,

    2CT valuesb1 were converted to negative fold change valuesusing

    the formula1/(2CT). However, these converted values were not

    otherwise used for display or analysis.

    Results

    BALF LDH activity and total protein concentration

    LDH activity is frequently used as an indicator of non-specic

    cellular damage. LDH activity in HBSS-treated control mice stayed

    relatively constant through the experiment (Fig. 2A). MACA-treated

    mice displayed a very small increase in LDH activity relative to HBSS

    from 6 to 24 h post-treatment, but this increase was not statistically

    signicant. LDH activity in BSA-treated mice was more variable and

    displayed no clear temporal pattern.

    BALF total protein concentration may indicate increased vascular

    permeability, increased local secretion of inammatory mediators,

    or cellular leakage secondary to damage or death. Administration of

    MACA was associated with a signicant increase (relative to both

    HBSS and BSA) in BALF total protein concentration at 12 and 18 h

    post-treatment, with a more variable increase seen at 6 h ( Fig. 2B).

    This increase in total protein in the absence of signicantly

    increased LDH activity suggests that MACA administration isassociated with the induction of pulmonary edema and inamma-

    tion but very little overt cytotoxicity during the time points

    evaluated in this study.

    Total and differential BALF cell counts

    MACA-treated mice displayed a rapid increase in total BALF cell

    numbers post-treatment (Fig. 3). These increases were signicant

    compared to both HBSS and BSA at 6, 12 and 18 h. HBSS- and BSA-

    treated mice demonstrated similar but minimal increases in BALF

    cellularity after IA, with the greatest cell counts noted at the 18 h and

    24 h time points.

    BALF cellularity was also evaluated by individual cell type.

    Macrophages demonstrated a small increase with time (up to 18 hpost-treatment) in all groups, including HBSS controls (Fig. 4A).

    Neither BSA-treated nor HBSS-treated mice demonstrated apprecia-

    ble changes in BALF neutrophils, lymphocytes or eosinophils. In

    contrast, MACA-treated mice had signicant increases in all of these

    cell types relative to both HBSS and BSA. Neutrophils were

    signicantly increased by 3 h post-treatment, while lymphocytes

    were signicantly increased by 6 h (Figs. 4B, C). Both cell types

    remained elevated for the duration of the study. Eosinophil numbers

    were signicantly increased at 12 and 24 h post-treatment ( Fig. 4D).

    When the results of the BALF analyses were evaluated as a whole,

    it was noted that most parameters rst demonstrated signicant

    perturbations at 6 h after treatment administration, with many of

    these values decreasing back towards baseline by 24 h. Thetriggers

    for these changes would be expressed before phenotypic changes

    147C.M. Pucheu-Haston et al. / Toxicology and Applied Pharmacology 244 (2010) 144155

    http://www.ingenuity.com/http://www.ingenuity.com/http://david.abcc.ncifcrf.gov/home.jsphttp://david.abcc.ncifcrf.gov/home.jsphttp://www.graphpad.com/http://www.graphpad.com/http://david.abcc.ncifcrf.gov/home.jsphttp://david.abcc.ncifcrf.gov/home.jsphttp://www.ingenuity.com/http://www.ingenuity.com/
  • 8/10/2019 microarray_CK+inflam

    5/12

    could be seen. Therefore, we selected samples from the 3 and 12 h

    post-treatment times for all three treatment groups for microarray

    gene expression analysis.

    Microarray gene expression analysis

    Our overall data analysis strategy is outlined inFig. 1. Sequences

    dened as differentially expressed had to t all of the following

    conditions: (1) sequence intensity signicantly different from time-

    matched control as determined by one-way ANOVA/Benjamini-

    Hochberg FDR pb0.01, (2) Scheffe's post hoc analysis pb0.05, and

    (3) upregulated or downregulated at least two-fold relative to time-

    matched control.

    A total of 451 sequences from MACA-treated mice were differen-

    tially expressed relative to control at the 3 h time point, with a fold

    difference range of +50.37 to 18.68 (Supplementary Table 1). This

    number increased at the 12 h time point, with 754 sequences

    differentially expressed (range +49.99 to 15.10;SupplementaryTable 2). Compared to HBSS-treated control mice, BSA-treated mice

    had a total of 124 differentially expressed sequences 3 h post-

    administration, with a fold difference range of +35.12 to 24.04

    (Supplementary Table 3). This number had decreased to 17 by 12 h

    (range +3.44 to 2.80;Supplementary Table 4).

    One objective of this study was to identify potential biomarkers of

    exposure to respiratory sensitizers. Therefore, we used the list

    operations function of GeneGo to select genes which were differen-

    tially expressed only in MACA-treated animals at either the 3 or 12 h

    time points. Genes which were also (or only) differentially expressed

    after BSA administration were excluded from further consideration as

    biomarkers, but were subjected to pathway analysis (see below). A

    total of 299 and 620 MACA-specic genes were identied at the 3 and

    12 h time points, respectively.

    Theultimategoal of this work is to translateour results into a more

    high-throughput format for hazard screening. Many high-throughput

    assays are time-sensitive and therefore will likely require the use of

    biomarkers that demonstrate persistent expression. For this reason,we used the list operations function of GeneGo to identify MACA-

    specic genes that were differentially expressed at both the 3 h and

    12 h time points (MACA 3 h/12 h genes; n =153 genes) (Table 1).

    Biomarkerlter analysis

    Our list of differentially expressed genes was generated from

    whole lung homogenate RNA. It is possible that some of these genes

    had been expressed by non-residentcellsor by cell types which might

    not be easily modeledin vitro. For this reason, we imported the list of

    MACA 3 h/12 h genes into Ingenuity Pathway Analysis Software to

    conduct biomarker lter analysis. This allowed us to select genes

    which would be expected to be expressed in a more limited selection

    of organs or cell types. We chose genes that would be expressed inlung or immune system cells (such as dendritic cells or lymphocytes),

    as well as in existing cell lines derived from these cell types. We

    included genes expressed in thymus and spleen to represent primary

    and secondary immune organs. Finally, since respiratory hypersensi-

    tivity may be induced by cutaneous sensitization (Vanoirbeek et al.,

    2004; Akei et al., 2006), we also included genes expressed in

    epidermis. Application of these limits allowed us to narrow our list

    of potential biomarkers down to 130 genes, represented by 201

    sequences (Table 2).

    Functional analysis and candidate biomarker selection

    At this point, our list of potential biomarkers was composed of

    genes that were differentially expressed in MACA-treated mice, and

    Fig. 4. BALF differential cell counts. (A)An increasein BALF macrophages wasnoted overtimein all3 treatment groups. (BD). IA MACAinducesthe rapidrecruitment of neutrophils,

    lymphocytes and eosinophils relative to BSA- or HBSS-treated mice. Pb0.05,Pb0.01,Pb0.001 relative to HBSS;Pb0.05,Pb0.01,Pb0.001 relative to BSA.

    148 C.M. Pucheu-Haston et al. / Toxicology and Applied Pharmacology 244 (2010) 144155

    http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-
  • 8/10/2019 microarray_CK+inflam

    6/12

    Table 1

    Candidate biomarker gene sequence sets differentially expressed 3 and 12 h after MACA administration.

    Sequence

    code

    Primary

    sequence

    name

    Fold

    change

    (3 h)

    Fold

    change

    (12 h)

    Sequence description Sequence

    code

    Primary

    sequence

    name

    Fold

    change

    (3 h)

    Fold

    change

    (12 h)

    Sequence description

    1450788_at Saa1 25.69 46.99 Serum amyloid A 1 1420904_at Il17ra 2.11 2.73 Interleukin 17 receptor A

    1418652_at Cxcl9 7.60 27.46 Ch emokine ( C- X- C mot if) l igan d 9 1426909_ at Uck2 2.40 2.70 Ur idine-cyt idine kinase 2

    1418930_at Cxcl10 19.11 19.91 Ch emokine ( C- X- C mot if) l igan d 10 1416111_ at Cd83 6.38 2.70 CD83 an tigen

    1419607_at Tnf 7.67 19.54 Tumor necrosis factor 1419272_at Myd88 2.17 2.68 Myeloid differentiation

    primary response gene 881428034_a_at Tnfrsf9 5.00 15.21 Tumor necrosis factor receptor

    superfamily, member 9

    1421207_ at Lif 5 .16 2.66 Leukemia inhibitor y f act or

    1420591_at Gpr84 4.04 12.09 G protein-coupled receptor 84 1418936_at Maff 2.35 2.63 v-maf musculoaponeurotic

    brosarcoma oncogene family,

    protein F (avian)

    1460227_at Timp1 4.38 12.05 Tissue inhibitor o f met allopr ot einase 1 1425374_ at Oas3 4.93 2.61 2-5oligoadenylate synthetase 3

    1419209_at Cxcl1 12.42 11.98 Chemokine (C-X-C motif) ligand 1 1449184_at Pglyrp1 2.52 2.60 Peptidoglycan recognition protein 1

    1425151_a_at Noxo1 5.83 11.43 NADPH oxidase organizer 1 1450698_at Dusp2 2.42 2.60 Dual specicity phosphatase 2

    1421228_at Ccl7 11.88 11.32 Ch emokine ( C- C motif) l igan d 7 1449233_ at Bhlhb8 2.47 2.60 Basic helix-loop-helix domain

    containing, class B, 8

    1419192_at Il4i1 13.40 11.23 Interleukin 4 induced 1 1460220_a_at Csf1 4.24 2.57 Colony stimulating factor 1

    (macrophage)

    1460282_at Trem1 9.59 11.06 Triggering receptor expressed

    on myeloid cells 1

    1425850_a_at Nek6 2.13 2.56 NIMA (never in mitosis gene a)

    -related expressed kinase 6

    1451798_at Il1rn 6.88 10.86 Interleukin 1 receptor antagonist 1418262_at Syk 1.70 2.56 Spleen tyros ine kinas e

    1419721_at Gpr109a 5.78 10.58 G protein-coupled receptor 109A 1422924_at Tnfsf9 4.77 2.55 Tumor necrosis factor (ligand)

    superfamily, member 91420380_at Ccl2 17.44 10.24 Ch emokine ( C- C motif) l igan d 2 1453924_ a_at Ptgf r 2.13 2.53 Pr ostaglandin F r eceptor

    1419413_at Ccl17 3.77 9.83 Chemokine (C-C motif) ligand 17 1450377_at Thbs1 2.70 2.51 Thrombospondin 1

    1460469_at Tnfrsf9 4.18 9.79 Tumor necrosis factor receptor

    superfamily, member 9

    1454005_at Fmo2 2.36 2.51 Flavin containing monooxygenase 2

    1423017_a_at Il1rn 6.24 9.35 Interleukin 1 receptor antagonist 1438855_x_at Tnfaip2 3.43 2.50 Tumor necrosis factor, alpha-induced

    protein 2

    1420330_at Clec4e 4.48 8.93 C-type lectin domain family 4, member e 1460231_at Irf5 2.02 2.49 Interferon regulatory factor 5

    1449227_at Ch25h 12.50 8.11 Ch olestero l 25-h ydr oxylase 1455265_ a_at Rgs16 3.79 2.47 Regulat or of G- prot ein sign alin g 16

    1425829_a_at Steap4 4.61 8.06 STEAP family member 4 1423596_at Nek6 2.50 2.47 NIMA (never in mitos is gene a)

    -related expressed kinase 6

    1420331_at Clec4e 6.36 7.58 C- type lect in domain f amily 4, member e 1427257_ at Vcan 1.55 2.46 Versican

    1417314_at Cfb 2.01 7.25 Complement factor B 1420723_at Vnn3 2.19 2.45 Vanin 3

    1419561_at Ccl3 13.72 6.96 Chemokine (C-C motif) ligand 3 1448749_at Plek 2.63 2.42 Pleckstrin

    1449399_a_at Il1b 8.75 6.88 Interleukin 1 beta 1418547_at Tfpi2 2.77 2.41 Tissue factor pathway inhibitor 2

    1426037_a_at Rgs16 10.57 6.49 Regulator of G-protein signaling 16 1421712_at Sele 16.99 2.41 Selectin, endothelial cell

    1448061_at Msr1 2.61 6.47 Macro phage scav enger recepto r 1 1438183_ x_at Sord 2.55 2.40 Sor bitol dehydrogenase 1

    1420558_at Selp 7.25 6.21 Selectin, platelet 1418261_at Syk 2.07 2.40 Spleen tyrosine kinase

    1449277_at Ccl19 5.68 6.05 Ch emokine ( C- C motif) l igan d 19 1425649_ at Slc39a14 2.30 2.39 Solut e carr ier f amily 39(zinc transporter), member 14

    1417925_at Ccl22 4.40 6.01 Ch emokine ( C- C motif) l igan d 22 1450672_ a_at Tr ex1 2.22 2.38 Thre e prime r epair e xonuclease 1

    1449984_at Cxcl2 11.22 5.99 Ch emokine ( C- X- C mot if) l igan d 2 1423006_ at Pim1 3.26 2.37 Pr oviral int egr ation sit e 1

    1421408_at Igsf6 11.88 5.91 Immunoglobulin superfamily, member 6 1451452_a_at Rgs16 8.62 2.36 Regulator of G-protein signaling 16

    1418806_at Csf3r 3.94 5.61 Colony stimulating factor 3 receptor

    (granulocyte)

    1448604_at Uck2 2.57 2.36 Uridine-cytidine kinase 2

    1416576_at Socs3 6.08 5.42 Suppressor of cytokine signaling 3 1419714_at Cd274 2.16 2.35 CD274 antigen

    1421694_a_at Vcan 2.99 5.41 Ch ondroitin sulfate prote oglycan 2 1423520_ at Lmn b1 3.15 2.34 Lamin B1

    1419532_at I l1r2 3.63 5.40 In ter leukin 1 r eceptor, t ype I I 1438097_ at Rab20 2.52 2.34 RAB20, member RAS on cog ene f amily

    1448951_at Tnfrsf1b 4.98 5.26 Tumor necrosis factor receptor

    superfamily, member 1b

    1420824_at Sema4d 1.57 2.33 Sema domain, immunoglobulin

    domain (Ig), transmembrane domain

    (TM) and short cytoplasmic domain,

    (semaphorin) 4D

    1449906_at Selp 11.66 5.26 Selectin, platelet 1451314_a_at Vcam1 2.85 2.33 Vascular cell adhesion molecule 1

    1453076_at Batf3 3.52 5.17 Basic leucine zipper transcription

    factor, ATF-like 3

    1422905_s_at Fmo2 1.49 2.33 Flavin containing monooxygenase 2

    1450318_a_at P2ry2 3.20 5.06 Purinergic receptor P2Y, G-protein coupled2

    1415899_at Junb 4.10 2.33 Jun-B oncogene

    1422062_at Msr1 2.72 5.03 Macro phage scav enger recepto r 1 1435415_ x_at Marcksl1 2.17 2.32 MARCKS- like 1

    1419697_at Cxcl11 5.01 5.01 Chemokine (C-X-C motif) ligand 11 1419394_s_at S100a8 4.58 2.32 S100 calcium binding protein

    A8 (calgranulin A)

    1450200_s_at Csf2rb 2.91 4.99 Colony stimulating factor 2 receptor, beta,

    low-afnity (granulocyte-macrophage)

    1431843_a_at Nfkbie 4.64 2.32 Nuclear factor of kappa light

    polypeptide gene enhancer in

    B-cells inhibitor, epsilon

    1425663_at I l1rn 4.24 4.98 In ter leukin 1 r eceptor antagon ist 1437226_ x_at Marcksl1 3.41 2.30 MARCKS- like 1

    1419609_at Ccr1 4.12 4.93 Ch emokine ( C- C motif) receptor 1 1438841_ s_at Ar g2 1.87 2.30 Vesicle t ranspor t t hr oug h int eraction

    with t-SNAREs 1b homolog

    1419082_at Serpinb2 2.92 4.92 Serine (or cysteine) peptidase inhibitor,

    clade B, member 2

    1417856_at Relb 3.90 2.29 Avian reticuloendotheliosis viral

    (v-rel) oncogene related B

    1416273_at Tnfaip2 8.35 4.83 Tumor necrosis factor, alpha-induced

    protein 2

    1453851_a_at Gadd45g 3.10 2.28 Growth arrest and

    DNA-damage-inducible 45 gamma

    1419482_at C3ar 1 2.09 4.78 Co mplement co mpo nent 3a r ecept or 1 1425797_ a_at Syk 1.60 2.27 Spleen t yrosine kinase

    (continued on next page)(continued on next page)

    149C.M. Pucheu-Haston et al. / Toxicology and Applied Pharmacology 244 (2010) 144155

  • 8/10/2019 microarray_CK+inflam

    7/12

    Table 1 (continued)

    Sequence

    code

    Primary

    sequence

    name

    Fold

    change

    (3 h)

    Fold

    change

    (12 h)

    Sequence description Sequence

    code

    Primary

    sequence

    name

    Fold

    change

    (3 h)

    Fold

    change

    (12 h)

    Sequence description

    1417523_at Plek 4.01 4.77 Pleckstrin 1426505_at Evi2b 2.10 2.24 Ecotropic viral integration site 2b

    1425380_at Rasgrp4 3.60 4.73 RAS guanyl releasing protein 4 1452483_a_at Cd44 2.24 2.21 CD44 antigen

    1452732_at Aspr v1 2.39 4.65 Aspart ic peptidase, r etr oviral-l ike 1 1432273_a_at D ar c 2.53 2.21 Duff y blood gr oup

    1425434_a_at Msr1 1.35 4.63 Macrophage scavenger receptor 1 1434596_at Sema4d 1.18 2.20 Sema domain, immunoglobulin

    domain (Ig), transmembrane domain

    (TM) and short cytoplasmic domain,

    (semaphorin) 4D

    1427747_a_at Lcn2 2.26 4.59 Lipocalin 2 1421173_at Irf4 2.45 2.20 Interferon regulatory factor 4

    1421578_at Ccl4 12.43 4.58 Chemokine (C-C motif) ligand 4 1427256_at Vcan 1.35 2.18 Versican

    1448914_a_at Csf1 4.77 4.55 Colony stimulating factor 1 (macrophage) 1422904_at Fmo2 1.26 2.16 Flavin containing monooxygenase 2

    1419149_at Serpine1 3.76 4.55 Serine (or cysteine) peptidase inhibitor,

    clade E, member 1

    1418099_at Tnfrsf1b 1.83 2.15 Tumor necrosis factor receptor

    superfamily, member 1b

    1431705_a_at Mcoln2 4.74 4.53 Mucolipin 2 1418830_at Cd79a 2.44 2.15 CD79A antigen (immunoglobulin-

    associated alpha)

    1435872_at Pim1 3.13 4.41 Proviral i ntegration site 1 1425154_a_at Csf1 2.77 2.15 Colony stimulating factor 1

    (macrophage)

    1419728_at Cxcl5 21.26 4.34 Chemokine (C-X-C motif) ligand 5 1448748_at Plek 2.30 2.15 Plecks trin

    1448291_at Mmp9 9.99 4.33 Matrix metallopeptidase 9 1460302_at Thbs1 2.74 2.14 Thrombospondin 1

    1422046_at Itgam 2.51 4.28 Integrin alpha M 1427683_at Egr2 2.88 2.13 Early growth response 2

    1451416_a_at Tgm1 3.53 4.15 Transglutaminase 1, K polypeptide 1416871_at Adam8 3.20 2.13 A disintegrin and metallopeptidase

    domain 8

    1425902_a_at Nfkb2 4.69 4.08 Nuclear factor of kappa light polypeptide

    gene enhancer in B-cells 2, p49/p100

    1449360_at Csf2rb2 1.70 2.13 Colony stimulating factor 2

    receptor, beta 2, low-afnity

    (granulocyte-macrophage)1420823_at Sema4d 3.95 4.08 Sema domain, immunoglobulin domain

    (Ig), transmembrane domain (TM) and

    short cytoplasmic domain, (semaphorin)

    4D

    1416304_at Lit af 2.25 2.11 LP S-induce d TN fact or

    1460197_a_at Steap4 3.09 4.04 STEAP family member 4 1420905_at Il17ra 2.14 2.10 Interleukin 17 receptor A

    1420804_s_at Clec4d 3.63 3.92 C-type lectin domain family 4, member d 1424880_at Trib1 2.76 2.10 Tribbles homolog 1 (Drosophila)

    1427429_at Csf2 10.51 3.91 Colony stimulating factor 2

    (granulocyte-macrophage)

    1451043_at Nek6 1.34 2.06 NIMA (never in mitosis gene a)-

    related expressed kinase 6

    1420349_at Ptgfr 1.50 3.73 Prostaglandin F receptor 1425197_at Ptpn2 2.33 2.05 Protei n tyrosine phosphatase, non-

    receptor type 2

    1451054_at Orm1 3.34 3.68 Orosomucoid 1 1449125_at Tnfaip8l1 2.39 2.05 Tumor necrosis factor, alpha-induced

    protein 8-like 1

    1417813_at Ikbke 3.71 3.42 Inh ibitor of kappaB kinase epsilon 1429128_x_at Nfkb2 2.28 1.91 RIKEN cDNA 1110007H17 gene

    1448181_at Klf15 2.79 3.37 Kruppel -like factor 15 1429752_x_at Clip4 1.37 1.89 CAP-GLY domain containing linker

    protein family, member 4

    1418842_at Hcls1 2.05 3.37 Hemat opoiet ic cell specic Lyn substrate 1 1427682_a_at Egr2 3.71 1.85 Early growth response 2

    1450783_at It1 2.16 3.36 Interferon-induced protein withtetratricopeptide repeats 1

    1449828_at Ptgfr 2.07 1.84 Prostaglandin F receptor

    1419483_at C3ar1 1.71 3.35 Complement component 3a receptor 1 1417140_a_at Ptpn2 1.85 1.83 Protein tyrosine phosphatase, non-

    receptor type 2

    1419610_at Ccr1 3.00 3.32 Chemokine (C-C motif) receptor 1 1418847_at Arg2 2.06 1.79 Arginase type II

    1420394_s_at Lilrb4 2.48 3.32 Leukocyte immunoglobulin-like

    receptor, subfamily B, member 4

    1450749_a_at Nr4a2 1.41 1.77 Nuclear receptor subfamily 4, group

    A, member 2

    1449450_at Ptges 3.16 3.30 Prostaglandin E synthase 1425198_at Ptpn2 2.37 1.74 Protei n tyrosine phosphatase, non-

    receptor type 2

    1418465_at Ncf4 2.08 3.25 Neutrophil cytosolic factor 4 1416303_at Litaf 2.19 1.72 LPS-induced TN factor

    1435458_at Pim1 4.11 3.25 Proviral i ntegration site 1 1453278_a_at Clip4 1.35 1.72 CAP-GLY domain containing linker

    protein family, member 4

    1450750_a_at Nr4a2 2.20 3.22 Nuclear receptor subfamily 4,

    group A, member 2

    1416298_at Mmp9 2.23 1.63 Matrix metallopeptidase 9

    1415922_s_at Marcksl1 6.98 3.21 MARCKS-like 1 1417483_at Nfkbiz 2.93 1.60 Nuclear factor of kappa light

    polypeptide gene enhancer in B-cells

    inhibitor, zeta

    1427994_at Cd300lf 2.14 3.20 CD300 antige n l ike f amily member F 1418800_at Bhlhb8 1.09 1.58 Basic he lixloophelix domain

    containing, class B, 8

    1425155_x_at Csf1 4.57 3.19 Colony stimulating factor 1 (macrophage) 1438562_a_at Ptpn2 1.73 1.55 Protein tyrosine phosphatase, non-

    receptor type 2

    1425412_at Nlrp3 2.74 3.17 NLR family, pyrin domain containing 3 1418642_at Lcp2 1.49 1.51 Lymphocyte cytosolic protein 2

    1427313_at Ptgir 2.62 3.13 Prostaglandin I receptor (IP) 1435504_at Clip4 1.01 1.50 CAP-GLY domain containing linker

    protein family, member 4

    1427911_at Tmem173 2.83 3.11 Tr ansmembran e pr ote in 173 1426584_a_at S ord 1.61 1.43 Sor bitol deh ydrogenase

    1425435_at Msr1 1.14 3.08 macrophage scavenger receptor 1 1421811_at Thbs1 1. 43 1.41 thrombos pondin 1

    1449486_at Ces1 2.79 3.03 Carboxylesterase 1 1415989_at Vcam1 3.38 1.40 Vascular cell adhesion molecule 1

    1418641_at Lcp2 2.86 2.99 Lymphocyte cytosolic protein 2 1421206_at Lif 1. 29 1.39 Leukemia inhibitory factor

    1451340_at Ar id5a 4.92 2.99 AT r ich inter act ive domain 5A (Mr f1 l ike) 1423760_at Cd44 1.56 1.39 CD44 ant igen

    1419135_at Ltb 2.72 2.90 Lymphotoxin B 1426506_at Evi2b 1.24 1.36 Ecotropic viral integration site 2b

    1456212_x_at Socs3 3.11 2.90 Cytokine inducible SH2-containing

    protein 3

    1448162_at Vcam1 2.24 1.36 Vascular cell adhesion molecule 1

    1450446_a_ at Socs1 3.35 2.88 Suppressor of cyt okine signaling 1 1448294_at Lit af 1.19 1.29 LP S-induce d TN fact or

    1455899_x_at Socs3 2.98 2.87 Cytokine inducible SH2-containing

    protein 3

    1424881_at Trib1 1.33 1.26 Tribbles homolog 1 (Drosophila)

    150 C.M. Pucheu-Haston et al. / Toxicology and Applied Pharmacology 244 (2010) 144155

  • 8/10/2019 microarray_CK+inflam

    8/12

    which had been ltered for likely expression in lung and immune

    system-related tissues (or cell lines derived from those tissues).

    However, these genes were not necessarily related to the develop-

    ment or perpetuation of allergic sensitization or asthma. To address

    this possibility, we queried the list against three gene expression

    databases to facilitate prioritization of biomarker candidates by

    known or suspected relevance to allergic sensitization, to airway

    hyperreactivity or to other pulmonary conditions associated with

    asthma (such as airway remodeling).

    The databases we used were the Ingenuity Knowledge database,

    the GeneGo database and DAVID (Database for Annotation, Visuali-

    zation and Integrated Discovery). Annotations for each of the

    potential biomarker genes were manually reviewed. Genes with low

    specicity (such as those encoding the multi-functional transcription

    factorsJunband Nfkb2) were given low priority, as were genes known

    to be associated with non-specic inammation (e.g., Tnf). Genes

    with known relevance to allergic sensitization or inammation,

    inammatory cell recruitment, airway hyperreactivity or remodeling

    were given a high priority. This process enabled the selection of a nal

    list of 11 high priority candidate biomarker genes (Table 2).

    Pathway analysis

    To gain additional insight into the nature of theimmune responses

    following exposure to agents of high versus low allergenicity,

    differentially expressed sequences were subjected to pathway

    analysis in Ingenuity. Genes differentially expressed after MACA

    exposure (either 3 h or 12 h post-administration) demonstrated a

    highly signicant association with a number of canonical pathways

    characteristic of an active and developing immuneresponse(Table 3).

    In many of these pathways, increased expression of most members of

    the NFB signaling family was seen. However, there was little to no

    change in expression of the JAK2/STAT3 or ERK, JNK or p38 map

    kinase signalingfamilies. A similar pattern of pathway association was

    observed even when only MACA-specic genes were evaluated

    (Table 3).

    Differentially expressed genes in 3 h BSA-treated mice were

    associated with pathways characteristic of a more general inamma-

    tory response and oxidative stress. No clear pathway association was

    noted in 12 h BSA samples, probably due to the low number of

    differentially expressed genes at this time point (Table 3). Neither

    time point appeared to be associated with consistent alterations in

    any of the signaling families.

    qRT-PCR analysis of candidate biomarker expression

    Expression of the 11 high-priority candidate biomarker genes was

    evaluated by qRT-PCR, both as a validation of the microarray resultsand to evaluate expression levels over the entire time course of the

    study. In general, fold difference values (relative to HBSS-treated

    mice) calculated from qRT-PCR data approximated those calculated

    from the microarray intensity data at both the 3 and 12 h time points

    in MACA-treated mice (Table 4). The largest discrepancies were seen

    for Saa1, in which microarray and PCR fold differenceswere 25.69 and

    12.59, and 46.99 and 135.74 at 3 and 12 h, respectively. Other notable

    differences between microarray and qRT-PCR values were for Cxcl2

    Table 1 (continued)

    Sequence

    code

    Primary

    sequence

    name

    Fold

    change

    (3 h)

    Fold

    change

    (12 h)

    Sequence description Sequence

    code

    Primary

    sequence

    name

    Fold

    change

    (3 h)

    Fold

    change

    (12 h)

    Sequence description

    1448728_a_at Nfkbiz 7.20 2.85 Nuclear factor of kappa light polypeptide

    gene enhancer in B-cells inhibitor, zeta

    1423521_at Lmnb1 1.52 1.21 Lamin B1

    1421326_at Csf2rb 1.97 2.85 Colony stimulating factor 2 receptor, beta,

    low-afnity (granulocyte-macrophage)

    1419698_at Cxcl11 1.23 1.19 Chemokine (C-X-C motif) ligand 11

    1427035_at Slc39a14 2.27 2.85 Solute carrier family 39 (zinc transporter),

    member 14

    1421174_at Irf4 1.75 1.12 Interferon regulatory factor 4

    1449449_at Ptges 3.11 2.82 Prostaglandin E synthase 1450160_at Lif 3.33 1.12 Leukemia inhibitory factor

    1425958_at I l1f9 5.25 2.75 I nt erleukin 1 family , member 9 1421481_at Tnfr sf 9 1.53 1.12 Tumor necrosis f acto r re cepto r

    superfamily, member 9

    1419132_at Tlr2 2.93 2.75 Toll-like receptor 2 1417137_at Uck2 2.47 1.10 Uridine-cytidine kinase 2

    1448756_at S100a9 4.03 2.73 S100 calcium binding protein A9

    (calgranulin B)

    For selection as candidate biomarkers, sequence sets had to t all of the following criteria: (1) signicantly different in expression relative to time-matched HBSS-treated mice (see

    text), (2) not differentially expressed in BSA-treated mice, (3) differentially expressed in MACA-treated mice at both the 3 and 12 h time points, and (4) selected for potential

    biomarker utility using Ingenuity Pathway Analysis Software. Values are presented as group average fold changes relative to HBSS-treated mice for each sequence set.

    Table 2

    Overview of high-priority candidate biomarkers of acute respiratory exposure to sensitizing agents.

    Primary

    sequence name

    Sequence description Chemotactic Associated with

    allergic disease

    Associated with

    pulmonary disease

    Persistently expressed

    (qRT-PCR)

    CCL17 Chemokine (C-C motif) ligand 17; thymus and activation regulated chemokine

    (TARC)

    X X X

    CCL22 Chemokine (C-C motif) ligand 22; macrophage derived chemokine (MDC) X X X

    CCL19 Chemokine (C-Cmotif) ligand19;macrophage inhibitoryprotein3- (MIP-3) X

    CCL7 Chemokine (C-C motif) ligand 7; monocyte chemoattractant protein 3 (MCP-3) X X

    CXCL2 Chemokine (C-X-C motif) ligand 2; macrophage inammatory protein 2

    (MIP-2)

    X X

    CXCL10 Chemokine (C-X-C motif) ligand 10; interferon inducible protein 10 (IP-10) X X X

    SAA1 Serum amyloid A1 X X

    C3aR1 Complement component 3a receptor 1 X X

    VCAM1 Vascular cell adhesion molecule 1 X

    SOCS3 Suppressor of cytokine signaling 3 X

    Arg2 Arginase 2 X X

    Biomarker andfunctional analysis of genes differentiallyexpressed at both3 and12 h timepoints allowed identication of 11 candidatebiomarker genes. Selectedgenes hadto meet

    at least one of the following criteria: (1) involved in chemotaxis, (2) associated with allergic disease, and (3) known or strongly suspected associated with allergic or non-allergic

    pulmonary disease. Candidate genes were also evaluated for persistence of gene expression (as determined by qRT-PCR) over the course of the study.

    151C.M. Pucheu-Haston et al. / Toxicology and Applied Pharmacology 244 (2010) 144155

  • 8/10/2019 microarray_CK+inflam

    9/12

    (11.22 and 24.01, and 5.99 and 17.44 at 3 and 12 h), Ccl7 (11.32 and

    23.10 at 12 h) and Ccl19 (6.05 and 12.06 at 12 h). In contrast, direct

    comparison between microarray and qRT-PCR values was usually not

    possible for BSA-treated mice, as almost none of these sequence sets

    met the statistical cutoff parameters during initial microarray analysis

    (data not shown).

    Table 3

    Summary of pathway analyses.

    Name p-value No. of genes in data set

    mapping to pathway/Total

    no. of genes in pathway

    Ratio Net effect

    MACA 3 h

    IL-10 signaling 9.43E-14 16/71 0.225 Increased IL-1 signaling via NFB; decreased IL-10 signaling

    IL-6 signaling 5.10E-12 17/96 0.177 Increased IL-1 and TNF signaling via NFB; increased

    production of inammatory mediators

    LXR/RXR activation 8.84E-12 15/85 0.176 Increased IL-1 and TNF signaling via NFB; increased productionof inammatory mediators

    Dendritic cell maturation 3.61E-11 20/165 0.121 Increased production of inammatory mediators, adhesion and

    costimulatory molecules

    Acute phase response signaling 5.27E-11 21/178 0.118 Increased IL-1 and TNF signaling via NFB; increased production

    of inammatory mediators

    MACA 12 h

    Acute phase response signaling 6.30E-14 30/178 0.169 Increased IL-1 and TNF signaling via NFB; increased production

    of inammatory mediators

    TREM1 signaling 3.65E-12 17/69 0.246 Increased IL-1 and TNF signaling via AKT and NFB; increased

    production of inammatory mediators

    Dendritic cell maturation 2.87E-11 25/165 0.152 Increased production of inammatory mediators, adhesion and

    costimulatory molecules

    Role of pattern recognition receptors

    in recognition of bacteria and viruses

    1.04E- 09 17/88 0.193 I ncreased signaling by TLR-2 and complemen t r eceptor s; incr ease d

    production of inammatory mediators

    Hepaticbro sis/hepatic stellate cell activat ion 1.67E- 09 21/135 0.156 I ncre ased product ion of inammatory mediators

    MACA-specic 3 h/12 h

    Acute phase response signaling 6.48E-13 16/178 0.09 Increased IL-1 and TNF signaling via NFB; increased production ofinammatory mediators

    Dendritic cell maturation 1.82E-11 14/165 0.085 Increased production of inammatory mediators, adhesion and

    costimulatory molecules

    TREM1 signaling 4.05E-11 10/69 0.145 Increased IL-1 and TNF signaling via AKT and NFB; increased

    production of inammatory mediators

    LXR/RXR activation 4.34E-10 10/85 0.118 Increased IL-1 and TNF signaling via NFB; increased production

    of inammatory mediators

    Role of pattern recognition receptors in

    recognition of bacteria and viruses

    9.66E- 10 10/88 0.114 I ncreased signaling by TLR-2 and complemen t r eceptor s; incr ease d

    production of inammatory mediators

    BSA 3 h

    Xenobi otic metabolism signaling 3.22E-04 7/254 0.028 Enhanced redox regulation; decreased oxidation

    Aryl hydrocarbon receptor signaling 8.60E-04 5/155 0.032 Mixed effects on redox status

    NRF-2 mediated oxidative stress response 2.34E-03 5/185 0.027 Increased antioxidant response

    LPS/I L- 1 mediat ed inhibition of RX R function 3.43E- 03 5/198 0.025 I ncre ased l ipid me tabo lism

    IL-10 signaling 4.28E-03 3/71 0.042 Small increase in IL-6 production

    BSA 12 h

    Coagulation system 3.14E-02 1/37 0.027 Increased production of coagulation factor X

    CD40 signaling 5.59E-02 1/70 0.014 Not signicantMacropinocytosis 5.67E-02 1/72 0.014 Not signicant

    CCR5 signaling in macrophages 5.76E-02 1/87 0.011 N ot signicant

    IL-4 signaling 5.76E-02 1/72 0.014 Not signicant

    Genes that were signicantly expressed in MACA- or BSA-treated mice were evaluated to determine canonical pathway association using Ingenuity Pathway Analysis software.

    Table 4

    Microarray and PCR comparison.

    Sequence description Primary

    sequence name

    MACA fold

    change 3 h

    microarray

    MACA fold

    change 3 h

    qRT-PCR

    MACA fold

    change 12 h

    microarray

    MACA fold

    change 12 h

    qRT-PCR

    BSA fold

    change 3 h

    microarray

    BSA fold

    change 3 h

    qRT-PCR

    BSA fold

    change 12 h

    microarray

    BSA fold

    change 12 h

    qRT-PCR

    Serum amyloid A 1 Saa1 25.69 12.59 46.99 135.74 N/aa 1.50 N/a 6.55

    Chemokine (C-X-C motif) ligand 10 Cxcl10 19.11 19.23 19.91 20.76 N/a 1.51 N/a 1.94

    Chemokine (C-C motif) ligand 7 Ccl7 11.88 11.44 11.32 23.10 N/a 2.04 N/a 1.83

    Chemokine (C-X-C motif) ligand 2 Cxcl2 11.22 24.01 5.99 17.44 N/a 1.05 N/a 7.19

    Chemokine (C-C motif) ligand 19 Ccl19 5.68 5.20 6.05 12.06 N/a 1.07 N/a 2.09

    Chemokine (C-C motif) ligand 22 Ccl22 4.40 3.08 6.01 7.75 N/a 1.67 N/a 2.20

    Suppressor of cytokine signaling 3 Socs3 4.06b 3.62 3.73 2.89 1.49 1.09 N/a 1.92

    Chemokine (C-C motif) ligand 17 Ccl17 3.77 5.46 9.83 10.13 N/a 2.52 N/a 2.41

    Vascular cell adhesion molecule 1 Vcam1 2.82 4.55 1.70 1.03 N/a 1.01 N/a 1.09

    Arginase type II Arg2 1.97 2.37 2.04 2.02 1.47 1.01 N/a 1.21

    Complement component 3a receptor 1 C3ar1 1.90 2.10 4.07 2.36 N/a 1.21 N/a 1.03

    For validation purposes, average microarray gene fold-expression values for 11 candidate biomarker genes were compared to the corresponding values obtained by qRT-PCR

    analysis. qRT-PCR values were calculated relative to HBSS-treated control samples for each time point using the 2Ct method. For comparison purposes, 2Ct values b 1 were

    converted to negative fold change values using the formula: 1/2Ct.a N/aindicates that the expression intensity for this parameter failed to meet the initial statistical cutoff.b

    Where necessary, the expression values of genes represented by multiple sequence sets have been averaged.

    152 C.M. Pucheu-Haston et al. / Toxicology and Applied Pharmacology 244 (2010) 144155

  • 8/10/2019 microarray_CK+inflam

    10/12

    Fig. 5.Transcription of candidate biomarker genes as determined by qRT-PCR. Transcript levels were calculated using the CTmethod. Values are presented as average fold chang

    (dened as fold change of 1 and indicated by a dotted line).

  • 8/10/2019 microarray_CK+inflam

    11/12

  • 8/10/2019 microarray_CK+inflam

    12/12

    Disclaimer

    This research paper has been reviewed by the National Health and

    Environmental Effects Research Laboratory, US Environmental Pro-

    tection Agency and approved for publication. Approval does not

    signify that the contents necessarily reect the views and policies of

    the agency, nor does mention of trade names or commercial products

    constitute endorsement or recommendation for use.

    This work was supported by UNC/EPA training agreementCR83323701.

    Declaration of interest

    The Authors report no conicts of interest. The Authors alone are

    responsible for the content and writing of the paper.

    Acknowledgments

    The Authors would like to thank Debora Andrews, Judy Richards,

    Richard Jaskot and Dr. Yong Joo Chung of US EPA for technical and

    intellectual assistance. Additionally, we would like to thank Drs.

    Robert Luebke, MaryJane Selgrade, and Susan Hester for their critical

    review of the manuscript.

    Appendix A. Supplementary data

    Supplementary data associated with this article can be found, in

    the online version, atdoi:10.1016/j.taap.2009.12.027.

    References

    Akei, H.S., Brandt, E.B., Mishra, A., Strait, R.T., Finkelman, F.D., Warrier, M.R., Hershey,G.K., Blanchard, C., Rothenberg, M.E., 2006. Epicutaneous aeroallergen exposureinduces systemic TH2 immunity that predisposes to allergic nasal responses.

    J. Allergy Clin. Immunol. 118, 6269.Ali, H., Panettieri Jr., R.A., 2005. Anaphylatoxin C3a receptors in asthma. Respir. Res. 6, 19.Bochner, B.S., Hudson, S.A., Xiao, H.Q., Liu, M.C., 2003. Release of both CCR4-active and

    CXCR3-active chemokines during human allergic pulmonary late-phase reactions.J. Allergy Clin. Immunol. 112, 930934.

    Buyukozturk, S., Gelinic, A.A., Genc, S., Kocak, H., Oneriyidogan, Y., Erdan, S., Dal, M.,Colakoglu, B.,2004. Acute phase reactants in allergic airway disease. Tohoku. J. Exp.Med. 204 (3), 209213.

    Carey, M.A., Card, J.W., Voltz, J.W., Germolec, D.R., Korach, K.S., Zeldin, D.C., 2007. Theimpact of sex and sex hormones on lung physiology and disease: lessons fromanimal studies. Am. J. Physiol. Lung Cell Mol. Physiol. 293, L272 L278.

    Drouin, S.M., Corry, D.B., Hollman, T.J., Kildsgaard, J., Wetsel, R.A., 2002. Absence of thecomplement anaphylatoxin C3a receptor suppresses Th2 effector functions in amurine model of pulmonary allergy. J. Immunol. 169, 59265933.

    Fulkerson, P.C., Zimmermann, N., Hassman, L.M., Finkelman, F.D., Rothenberg, M.E.,2004.Pulmonary chemokine expression is coordinately regulatedby STAT1, STAT6,and IFN-gamma. J. Immunol. 173, 75657574.

    Hijnen, D., de Bruin-Weller, M., Oosting, B., Lebre, C., de Jong, E., Bruijnzeel-Koomen, C.,Knol, E., 2004. Serum thymus and activation-regulated chemokine (TARC) and

    cutaneous T cell-attracting chemokine (CTACK) levels in allergic diseases: TARCand CTACK are disease-specic markers for atopic dermatitis. J. Allergy Clin.Immunol. 113, 334340.

    Humbles, A.A., Lu,B., Nilsson, C.A., Lilly,C., Israel, E.,Fujiwara, Y.,Gerard,N.P.,Gerard,C.,2000. A role for the C3a anaphylatoxin receptor in the effector phase of asthma.Nature 406, 9981001.

    Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25, 402408.

    McKinley, L., Kim, J., Bolgos, G.L., Siddiqui, J., Remick, D.G., 2005. CXC chemokinesmodulate IgE secretion and pulmonary inammation in a model of allergic asthma.Cytokine 32, 178185.

    Michalec, L., Choudhury, B.K.,Postlethwait, E., Wild,J.S., Alam,R., Lett-Brown, M., Sur,S.,2002. CCL7 and CXCL10 orchestrate oxidative stress-induced neutrophilic lunginammation. J. Immunol. 168, 846852.

    Ozseker, F., Buyukozturk, S., Depboylu, B., Yilmazbayhan, D., Karayigit, E., Gelincik, A.,Genc, S., Colakoglu, B., Dal, M., Issever, H., 2006. Serum amyloid A (SAA) in inducedsputum of asthmatics: A new look to an old marker. Int. Immunopharmaco. 6,15691576.

    Postma, D.S., 2007. Gender differences in asthma development and progression. Gen.Med. 4, S133S146.

    Prussin, C., Metcalfe, D.D., 2006. 5. IgE, mast cells, basophils, and eosinophils. J. AllergyClin. Immunol. 117, S450S456.

    Pucheu-Haston, C.M., Shuster, D., Olivry, T., Brianceau, P., Lockwood, P., McClanahan, T.,de Waal Malefyt, R., Mattson, J.D., Hammerberg, B., 2006. A canine model ofcutaneous late-phase reactions: prednisolone inhibition of cellular and cytokineresponses. Immunology 117, 177187.

    Seymour, B.W.,Friebertshauser,K.E., Peake, J.L.,Pinkerton, K.E.,Coffman, R.L.,Gershwin,L.J., 2002. Gender differences in the allergic response of mice neonatally exposed toenvironmental tobacco smoke. Dev. Immunol. 9, 4754.

    Shang, X.-Z., Chiu, B.-C., Stolberg, V., Lukacs, N.W., Kunkel, S.L., Murphy, H.S., Chensue,

    S.W., 2002. Eosinophil recruitment in type-2 hypersensitivity pulmonary granu-lomas: source and contribution of monocyte chemotactic protein-3 (CCL7). Am. J.Pathol. 161, 257266.

    Stankus, R.P., O'Neil, C.E., 1988. Antigenic/allergenic characterization of American andGerman cockroach extracts. J. Allergy Clin. Immunol. 81, 563570.

    Tillie-Leblond, I., Hammad, H., Desurmont, S., Wallaert,B., Tonnel, A.-B., Gosset, P., 2000.CC chemokines and interleukin-5 in bronchial lavage uid from patients withstatus asthmaticus. Potential implication in eosinophil recruitment. Am. J. Respir.Crit. Care Med. 162, 586592.

    Vanoirbeek, J.A.J., Tarkowski, M., Ceuppens, J.L., Verbeken, E.K., Nemery, B., Hoet, P.H.M.,2004. Respiratory response to toluene diisocyanate depends on prior frequency andconcentration of dermal sensitization in mice. Toxicol. Sci. 80, 310321.

    Viana, M.E., Coates, N.H., Gavett, S.H., Selgrade, M.K., Vesper, S.J., Ward, M.D., 2002. Anextract of Stachybotrys chartarum causes allergic asthma-like responses in a BALB/c mouse model. Toxicol. Sci. 70, 98109.

    Ward, M.D.W., Chung, Y.J., Haykal-Coates, N., Copeland, L., 2009. Differential allergyresponses to Metarhizium anisopliae fungal component extracts in BALB/c mice.

    J. Immunotoxicol. 6, 6273.Ward, M.D.W., Madison, S.L., Andrews, D.L., Sailstad, D.M., Gavett, S.H., Selgrade, M.J.,

    2000. Comparison of respiratory responses to Metarhizium anisopliaeextract usingtwo different sensitization protocols. Toxicology 147, 133145.

    Ward, M.D.W., Sailstad, D.M., Selgrade, M.K., 1998. Allergic responses to thebiopesticide metarhizium anisopliae in Balb/c mice. Toxicol. Sci. 45, 195203.

    Whitehead, G.S., Walker, J.K.L., Berman, K.G., Foster, W.M., Schwartz, D.A., 2003.Allergen-induced airway disease is mouse strain dependent. Am. J. Physiol. Lung.Cell. Mol. Physiol. 285, L32L42.

    Wood, S.H., Clements, D.N., Ollier, W.E., Nuttall, T., McEwan, N.A., Carter, S.D., 2009.Gene expression in canine atopic dermatitis and correlation with clinical severityscores. J. Dermatol. Sci. 55, 2733.

    Zwirner, J., Gtze, O., Begemann, G., Kapp, A., Kirchhoff, K., Werfel, T., 1999. Evaluationof C3a receptor expression on human leucocytes by the use of novel monoclonalantibodies. Immunology 97, 166172.

    155C.M. Pucheu-Haston et al. / Toxicology and Applied Pharmacology 244 (2010) 144155