predominance of interferon-related responses in the brain during … · molecular immunopathology...

13
INFECTION AND IMMUNITY, May 2008, p. 1812–1824 Vol. 76, No. 5 0019-9567/08/$08.000 doi:10.1128/IAI.01650-07 Copyright © 2008, American Society for Microbiology. All Rights Reserved. Predominance of Interferon-Related Responses in the Brain during Murine Malaria, as Identified by Microarray Analysis Jenny Miu,‡ Nicholas H. Hunt,* and Helen J. Ball Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December 2007/Returned for modification 2 February 2008/Accepted 18 February 2008 Cerebral malaria (CM) can be a fatal manifestation of Plasmodium falciparum infection. We examined global gene expression patterns during fatal murine CM (FMCM) and noncerebral malaria (NCM) by microarray analysis. There was differential expression of a number of genes, including some not yet characterized in the pathogenesis of FMCM. Some gene induction was observed during Plasmodium berghei infection regardless of the development of CM, and there was a predominance of genes linked to interferon responses, even in NCM. However, upon real-time PCR validation and quantitation, these genes were much more highly expressed in FMCM than in NCM. The observed changes included genes belonging to pathways such as interferon signaling, major histocompatibility complex processing and presentation, apoptosis, and immunomodulatory and antimicrobial processes. We further characterized differentially expressed genes by examining the cellular source of their expression as well as their temporal expression patterns during the course of malaria infection. These data identify a number of novel genes that represent interesting candidates for further investigation in FMCM. Malaria is a devastating disease affecting developing coun- tries in the tropical and subtropical regions of the world. Ce- rebral malaria (CM), a severe manifestation of Plasmodium falciparum infection, can lead to neurological complications and death (64). Murine models of malaria are important tools for studying the pathogenesis of malaria, with numerous clin- ical and histopathological similarities between human and mu- rine malaria having been described (19). Comparisons between fatal murine CM (FMCM) and noncerebral malaria (NCM) provide valuable insights into the pathogenesis of CM. A number of significant changes in the murine brain that characterize CM have been described, including breakdown of the blood-brain barrier (62), redistribution and activation of glia (31, 32, 34), apoptosis of endothelial cells and astrocytes (46, 47), metabolic changes (44, 48), and distinct patterns of chemokine gene expression (38). A recent approach to identifying novel changes within the brain during CM is to profile gene expression changes using microarray technology (9, 30). Microarray studies can generate a plethora of data that pinpoint involvement of unknown or poorly characterized genes, stimulating investigation of their biological functions. Some studies have used microarrays to examine global gene expression patterns during murine ma- laria, defining gene expression profiles that are associated with susceptibility or resistance to murine CM (26, 27), identifying genes in the brain that discriminate between different out- comes of Plasmodium infection (5), or describing trends in gene expression within the brain (55). Studies on the spleen have been performed to find genes that differentiate between cerebral and noncerebral (55) and between lethal and nonle- thal (52) forms of murine malaria. Microarrays also have been used to examine Plasmodium infection in humans (14, 41) and monkeys (68). In these studies, the majority of genes identified have well defined roles in such functions as immunity, eryth- ropoiesis, and metabolism. In the current study we used microarrays to examine global gene expression patterns in the brains of Plasmodium berghei- infected mice. In contrast to previous studies, which examined the response of genetically divergent murine hosts to the par- asite that causes FMCM, P. berghei ANKA (PbA), we investi- gated the response of genetically identical murine hosts to different, but related, parasite strains, PbA and P. berghei K173 (PbK), the latter being one that results in NCM. Furthermore, we used two different microarray technologies, oligonucleotide and Affymetrix arrays, and compared the outcomes. Using either technique, we found similar patterns, though different degrees, of gene expression in groups irrespective of progres- sion to CM. These included genes belonging to interferon (IFN) signaling pathways and major histocompatibility com- plex (MHC) processing and presentation, as well as those associated with apoptotic, immunomodulatory, and antimicro- bial processes. However, we found that very strong induction of these genes was associated with fatal outcome due to FMCM, as confirmed by real-time PCR. These results contrib- ute to our understanding of the processes occurring in the brain during CM, and the roles of the genes identified deserve further attention in FMCM and other neurological diseases. MATERIALS AND METHODS Murine models of malaria. Female CBA/T6 mice (6 to 8 weeks old) were housed in the Blackburn Animal House, University of Sydney, and given food and water ad libitum. Intraperitoneal inoculation of CBA/T6 mice with 1 10 6 * Corresponding author. Mailing address: Molecular Immunopa- thology Unit, Bosch Institute, Medical Foundation Building (K25), University of Sydney, Sydney, New South Wales 2006, Australia. Phone: 61-2-9036-3242. Fax: 61-2-9036-3286. E-mail: nicholas.hunt @bosch.org.au. † Supplemental material for this article may be found at http://iai .asm.org/. ‡ Present address: Department of Medicine, McGill University, Montreal, Quebec, Canada. Published ahead of print on 25 February 2008. 1812 on November 19, 2020 by guest http://iai.asm.org/ Downloaded from

Upload: others

Post on 15-Aug-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Predominance of Interferon-Related Responses in the Brain during … · Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December

INFECTION AND IMMUNITY, May 2008, p. 1812–1824 Vol. 76, No. 50019-9567/08/$08.00�0 doi:10.1128/IAI.01650-07Copyright © 2008, American Society for Microbiology. All Rights Reserved.

Predominance of Interferon-Related Responses in the Brain duringMurine Malaria, as Identified by Microarray Analysis�†

Jenny Miu,‡ Nicholas H. Hunt,* and Helen J. BallMolecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia

Received 11 December 2007/Returned for modification 2 February 2008/Accepted 18 February 2008

Cerebral malaria (CM) can be a fatal manifestation of Plasmodium falciparum infection. We examined globalgene expression patterns during fatal murine CM (FMCM) and noncerebral malaria (NCM) by microarrayanalysis. There was differential expression of a number of genes, including some not yet characterized in thepathogenesis of FMCM. Some gene induction was observed during Plasmodium berghei infection regardless ofthe development of CM, and there was a predominance of genes linked to interferon responses, even in NCM.However, upon real-time PCR validation and quantitation, these genes were much more highly expressed inFMCM than in NCM. The observed changes included genes belonging to pathways such as interferonsignaling, major histocompatibility complex processing and presentation, apoptosis, and immunomodulatoryand antimicrobial processes. We further characterized differentially expressed genes by examining the cellularsource of their expression as well as their temporal expression patterns during the course of malaria infection.These data identify a number of novel genes that represent interesting candidates for further investigation inFMCM.

Malaria is a devastating disease affecting developing coun-tries in the tropical and subtropical regions of the world. Ce-rebral malaria (CM), a severe manifestation of Plasmodiumfalciparum infection, can lead to neurological complicationsand death (64). Murine models of malaria are important toolsfor studying the pathogenesis of malaria, with numerous clin-ical and histopathological similarities between human and mu-rine malaria having been described (19). Comparisons betweenfatal murine CM (FMCM) and noncerebral malaria (NCM)provide valuable insights into the pathogenesis of CM.

A number of significant changes in the murine brain thatcharacterize CM have been described, including breakdown ofthe blood-brain barrier (62), redistribution and activation ofglia (31, 32, 34), apoptosis of endothelial cells and astrocytes(46, 47), metabolic changes (44, 48), and distinct patterns ofchemokine gene expression (38).

A recent approach to identifying novel changes within thebrain during CM is to profile gene expression changes usingmicroarray technology (9, 30). Microarray studies can generatea plethora of data that pinpoint involvement of unknown orpoorly characterized genes, stimulating investigation of theirbiological functions. Some studies have used microarrays toexamine global gene expression patterns during murine ma-laria, defining gene expression profiles that are associated withsusceptibility or resistance to murine CM (26, 27), identifyinggenes in the brain that discriminate between different out-

comes of Plasmodium infection (5), or describing trends ingene expression within the brain (55). Studies on the spleenhave been performed to find genes that differentiate betweencerebral and noncerebral (55) and between lethal and nonle-thal (52) forms of murine malaria. Microarrays also have beenused to examine Plasmodium infection in humans (14, 41) andmonkeys (68). In these studies, the majority of genes identifiedhave well defined roles in such functions as immunity, eryth-ropoiesis, and metabolism.

In the current study we used microarrays to examine globalgene expression patterns in the brains of Plasmodium berghei-infected mice. In contrast to previous studies, which examinedthe response of genetically divergent murine hosts to the par-asite that causes FMCM, P. berghei ANKA (PbA), we investi-gated the response of genetically identical murine hosts todifferent, but related, parasite strains, PbA and P. berghei K173(PbK), the latter being one that results in NCM. Furthermore,we used two different microarray technologies, oligonucleotideand Affymetrix arrays, and compared the outcomes. Usingeither technique, we found similar patterns, though differentdegrees, of gene expression in groups irrespective of progres-sion to CM. These included genes belonging to interferon(IFN) signaling pathways and major histocompatibility com-plex (MHC) processing and presentation, as well as thoseassociated with apoptotic, immunomodulatory, and antimicro-bial processes. However, we found that very strong inductionof these genes was associated with fatal outcome due toFMCM, as confirmed by real-time PCR. These results contrib-ute to our understanding of the processes occurring in thebrain during CM, and the roles of the genes identified deservefurther attention in FMCM and other neurological diseases.

MATERIALS AND METHODS

Murine models of malaria. Female CBA/T6 mice (6 to 8 weeks old) werehoused in the Blackburn Animal House, University of Sydney, and given foodand water ad libitum. Intraperitoneal inoculation of CBA/T6 mice with 1 � 106

* Corresponding author. Mailing address: Molecular Immunopa-thology Unit, Bosch Institute, Medical Foundation Building (K25),University of Sydney, Sydney, New South Wales 2006, Australia.Phone: 61-2-9036-3242. Fax: 61-2-9036-3286. E-mail: [email protected].

† Supplemental material for this article may be found at http://iai.asm.org/.

‡ Present address: Department of Medicine, McGill University,Montreal, Quebec, Canada.

� Published ahead of print on 25 February 2008.

1812

on Novem

ber 19, 2020 by guesthttp://iai.asm

.org/D

ownloaded from

Page 2: Predominance of Interferon-Related Responses in the Brain during … · Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December

PbA parasitized red blood cells (PRBCs) (courtesy of G. Grau, University ofSydney, Australia) is a well-described model of FMCM, in which mice displayneurological symptoms by day 6 to 7 postinoculation (p.i.). On the other hand,mice inoculated with PbK PRBCs (courtesy of I. Clark, Australian NationalUniversity, Australia) do not show these neurological signs but instead succumbto acute anemia and hyperparasitemia at 2 to 3 weeks p.i. (40). Control micewere uninfected. All procedures involving the use of animals were approved by,and carried out according to the regulations of, the Animal Ethics Committee,University of Sydney.

Tissue extraction and histopathology. Mice were anesthetized by isofluraneinhalation and euthanized at each stated time point. Mice were perfused with 10ml cold sterile phosphate-buffered saline to clear the vessels of blood, and organswere removed. Formalin-fixed, paraffin-embedded sections (7 �m) were used toexamine histopathological features, as described previously (47).

RNA extraction. Brain tissue from animals was collected in 1 ml TRIzolreagent (Invitrogen, Victoria, Australia) and immediately homogenized usingZirconia beads (Biospec Products, OK) and a Fast Prep homogenizer (Q-Bio-gene, CA). Total RNA was isolated according to the manufacturer’s instructions(Invitrogen). The integrity of the RNA samples was tested using BioanalyzerNano chips (Agilent Technologies) according to the manufacturer’s instructions.Samples were DNase treated using a DNA-free kit (Ambion, TX). The sameRNA samples were hybridized to the oligonucleotide and Affymetrix arrays aswere used in the real-time PCR analyses.

Experimental design of microarray studies. All experiments were designedto be compliant with Minimum Information About a Microarray Experiment(MIAME) standards. For oligonucleotide array experiments, control sampleswere pooled and used as a common reference for the test samples. Experimentalgroups of five mice were either inoculated with PbA and sacrificed for samplepreparation at day 6 p.i. [PbA(6)] or inoculated with PbK and sacrificed forsample preparation at day 6 p.i. [PbK(6)] or day 14 p.i. [PbK(14)]. Five individualtest samples were used per group and directly compared to the pooled (n � 5)control reference sample. For Affymetrix array experiments, biological replicates(n � 5) from all groups [control, PbA(6), PbK(6), and PbK(14)] were pooled andhybridized to an individual slide. These were compared with the pooled controlsample in downstream computational analyses.

Oligonucleotide arrays: cDNA labeling, hybridization, scanning, and normal-ization. For oligonucleotide arrays, indirect labeling was used to incorporatecyanine (Cy3 and Cy5) dyes into cDNA samples. Labeled probes were thenapplied to 22.5K Compugen mouse oligonucleotide arrays (�22,500 genes)(Ramaciotti Centre, University of New South Wales). Slides were washed andthen scanned, and spot intensities were determined using an Axon scanner(GenePix 3.0; Molecular Devices, CA). Basic normalization procedures for oli-gonucleotide arrays were carried out in GenePix 3.0, including backgroundsubtraction to remove data generated from noise and removal of poor-qualityspots due to hybridization artifacts or morphology. Data were then imported intoLimma (R, Bioconductor project; http://www.bioconductor.org/) (12) for loessnormalization. For full experimental details, including raw data sets, refer to theGene Expression Omnibus website (http://www.ncbi.nlm.nih.gov/geo/index.cgi),accession number GSE9808.

Prior to normalization, the majority of the arrays revealed systemic biases (i.e.,more spots with negative M values), as well as low-intensity spots (i.e., spots withlow A values), which can affect ratios. Some arrays also showed fanning effects(i.e., spots with low intensity and high background), which can lead to very largebut erroneous ratios. MA plots were used to visualize these spatial and intensityeffects (see Fig. S1a in the supplemental material), while box plots were used toview the spread of M values (see Fig. S1c in the supplemental material). Manyarrays showed variations across the M values, particularly across each print-tipgroup. These systemic biases suggested that local and not global methods ofnormalization are more appropriate in this situation. From the raw data, localnormalization methods were employed to correct for technical effects that cancontribute to differential gene expression. This was performed using loess nor-malization in the Limma program (57). Normalized data showed a better spreadacross A values, without extreme variation across M values, as viewed in MA plots(see Fig. S1b in the supplemental material). Similarly, in box plots, normalized Mvalues were found to cluster more tightly around M � 0, with less spread than forraw data (see Fig. S1d in the supplemental material).

Affymetrix arrays: cDNA labeling, hybridization, scanning, and normaliza-tion. For Affymetrix arrays, cRNA was labeled using the one-cycle target labelingprotocol as described in the Affymetrix GeneChip Expression Analysis Manual(Affymetrix). Labeled cRNA samples were then hybridized to AffymetrixGeneChip Mouse Genome 430 2.0 arrays (�43,000 genes) (Ramaciotti Centre,University of New South Wales) before being scanned using the GeneArrayScanner (Affymetrix). Affymetrix arrays were initially assessed and single-array

analyses carried out in the GeneChip Operating Software program according tothe Affymetrix GeneChip Expression Analysis Data Analysis Fundamentals Manual(Affymetrix) on data generated from the image analyses. For full experimentaldetails, including raw data sets, refer to the Gene Expression Omnibus website(http://www.ncbi.nlm.nih.gov/geo/index.cgi), accession number GSE9808.

Microarray data analysis. Following basic normalization procedures carriedout using Limma and GeneChip Operating Software, data from both oligonu-cleotide arrays and Affymetrix arrays were imported into Genespring (AgilentTechnologies) for further analyses. Within the Genespring program, genes areassigned individual P values, based on a one-sample Student t test and assuminga baseline of 0 on a log scale. For oligonucleotide arrays, genes were selected asdifferentially expressed if they had a normalized value greater than 2 (upregu-lated) or less than 0.5 (downregulated) on a log scale, with at least three slidesin each group satisfying these criteria. Genes were then ranked according to theirP values to generate lists of upregulated and downregulated genes. However,one-way analyses of variance were found to be too stringent when applied tothese lists, as no genes were found to be differentially expressed, and thus werenot used. For Affymetrix arrays, following normalization using robust multiarrayanalysis, arrays from P. berghei-infected mice were compared to the uninfectedcontrol array. Genes were selected as differentially expressed if they had anormalized value greater than 2 (upregulated) or less than 0.5 (downregulated)on a log scale. Genes were then ranked according to their fold change values togenerate lists of upregulated and downregulated genes.

Functional annotation of genes. Functional annotation of genes was per-formed using the Database for Annotation, Visualization, and Integrated Dis-covery (DAVID) (http://david.abcc.ncifcrf.gov/home.jsp) (6). Genes were anno-tated based on the three structured vocabularies (ontologies), describing geneproducts in terms of their “biological processes,” “molecular functions,” and“cellular compartments,” which are considered independent of each other.DAVID has the advantage of clustering genes with similar gene ontology (GO)terms together, thus removing the high level of redundancy that exists in theontologies and making interpretation easier.

Real-time PCR. Real-time PCR was performed as described previously (2),except that PCRs were performed with the Corbett Rotor-Gene (Corbett Re-search, New South Wales, Australia) and using Platinum Sybr green qPCRSuperMix UDG (Invitrogen). mRNA levels were normalized to a housekeepinggene, that for hypoxanthine guanine phosphoribosyltransferase (HPRT), andexpressed relative to the mean for uninfected control samples using the deltathreshold cycle method. The specificity of primers was checked using meltingcurve analyses, and the primer sets used (Table 1) had similar amplificationefficiencies.

Laser capture microdissection. Laser capture microdissection was performedas previously described (38). Briefly, cerebral microvessels were stained with afluorescent lectin [fluorescein Griffonia (Bandeiraea) Simplicifolia lectin; Vector,CA]. Laser capture was performed on a PixCell II microscope (Arcturus, TX)

TABLE 1. Primer sequences used

Primer Sequences (forward, reverse)

�2-m............................5�-TGATCATATGCCAAACCCTCTG-3�,5�-CAGACTTCAATTAGGCCTCTTTGC-3�

CTLA-2� ...................5�-AGCTGTGGCTTGACTGGTAACA-3�,5�-ATGGCAGAGTGCTTGCTTCAC-3�

CTSW.........................5�-GTTGTCCAATAATGAGCAGGCA-3�,5�-TTGATGGTCACGGTGATAGGTC-3�

Fas ..............................5�-GCGATGAAGAGCATGGTTTAGA-3�,5�-TCGCAGTAGAAGTCTGGTTTGC-3�

GZMA .......................5�-CAGAAATGTGGCTATCCTTCACC-3�,5�-CCTGGTTTCACATCATCTCCCT-3�

GZMB........................5�-ATGTGTGCTATGTGGCTGGTTG-3�,5�-GCGTGTTTGAGTATTTGCCCAT-3�

HPRT.........................5�-GCTTTCCCTGGTTAAGCAGTACA-3�,5�-CAAACTTGTCTGGAATTTCAAATC-3�

IGTP ..........................5�-AGGCAACAGCTTGTAATGATGC-3�,5�-ACCTTGGCTGATTATCCTGGC-3�

Ii..................................5�-GACACGGCAAATGAAGTCAGAA-3�,5�-TTAGATGCGGTGCCAGCAA-3�

ISG15 .........................5�-TTCTGACGCAGACTGTAGACACG-3�,5�-ACTTGTTCCGCTGGACACCTT-3�

OAS-1b ......................5�-AAGACAGGAAGGTGCCCAAA-3�,5�-GGACAGAAGCCTGGAGTCTGA-3�

VOL. 76, 2008 MICROARRAY ANALYSIS OF PLASMODIUM INFECTION 1813

on Novem

ber 19, 2020 by guesthttp://iai.asm

.org/D

ownloaded from

Page 3: Predominance of Interferon-Related Responses in the Brain during … · Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December

using high-sensitivity caps (CapSure HSLCM; Arcturus). Vessels and parenchy-mal cells were captured onto separate caps, and the RNA was isolated usingcolumns as described in the RNeasy MicroKit (Qiagen, Victoria, Australia).RNA was amplified according to a previously described protocol for degradedRNA (66), and real-time PCR was carried out as described above.

Statistical analyses. Statistical analyses for gene expression data were per-formed using the Kruskal-Wallis test. Where statistical differences were ob-served, probabilities were then calculated using the Mann-Whitney test on se-lected comparisons. These analyses were carried out using GraphPad Prismversion 4.00 for Windows (GraphPad Software, CA).

Microarray accession number. The Gene Expression Omnibus accession num-ber for the microarray data reported in this paper, which also includes details ofthe MIAME-compliant experiments, is GSE9808.

RESULTS

Many genes are differentially expressed in P. berghei infec-tion. Many genes were differentially expressed in the brains ofmice inoculated with P. berghei (Fig. 1), irrespective of whetherthe animal developed CM. Genes were either upregulated

(normalized value greater than 2, red) or downregulated (nor-malized value less than 0.5, green) according to criteria out-lined in Materials and Methods. Similar sets of genes werefound to be upregulated on the oligonucleotide (see Table S1in the supplemental material) and Affymetrix (see Table S3 inthe supplemental material) arrays. There were differences inthe lists of downregulated genes on oligonucleotide (see TableS2 in the supplemental material) and Affymetrix (see Table S4in the supplemental material) arrays, partially due to the greendye bias observed in the oligonucleotide arrays. This com-monly occurs due to the higher instability of the green dye,resulting in less efficient incorporation of this dye into cDNA.

IFN-stimulated genes (ISGs) associated with signaling andimmune functions were predominant on lists of upregulatedgenes, including IFN regulatory factor 1 (IRF1), Ia-associatedinvariant chain (Ii), �2-microglobulin (�2-m), granzymes A andB, and IFN--induced GTPase (IGTP), as well as poorly an-

FIG. 1. Line graphs of array gene expression data as visualized in Genespring, for oligonucleotide arrays, showing upregulated genes (foldchange of 2.0) in PbA(6) (A), PbK(6) (B), and PbK(14) (C) samples and downregulated genes (fold change of �0.5) in PbA(6) (D), PbK(6) (E),and PbK(14) (F) samples, and also for Affymetrix arrays, showing color-coded gene expression in PbA(6) (G), PbK(6) (H), and PbK(14)(I) samples (red, expression of 1.0 [upregulated]; green, expression of �1.0 [downregulated]).

1814 MIU ET AL. INFECT. IMMUN.

on Novem

ber 19, 2020 by guesthttp://iai.asm

.org/D

ownloaded from

Page 4: Predominance of Interferon-Related Responses in the Brain during … · Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December

notated ISGs such as IFN-induced protein with tetratricopep-tide repeats 2 (IFIT2) and IFN-induced transmembrane pro-tein 3 (IFITM3), IFN-induced gene 30 (Ifi30), Ifi35, and Ifi44(see Tables S1 and S3 in the supplemental material). Genesassociated with cellular compartments as well as unannotatedgenes were found on lists of downregulated genes (see TablesS2 and S4 in the supplemental material).

Genes are differentially expressed between the PbA(6),PbK(6), and PbK(14) groups. On closer inspection, manygenes were common to each group [i.e., PbA(6), PbK(6), orPbK(14)], indicating that gene expression patterns may over-lap. In order to identify genes specific to each group, differentlists were directly compared using Venn diagrams in the Gene-spring program (Fig. 2; see Tables S5 and S6 in the supple-mental material).

Altogether, there were 205 upregulated genes on the oligo-nucleotide arrays due to P. berghei infection. Of these, 88 geneswere specifically upregulated 6 days after PbA infection (groupA), including genes associated with signaling (e.g., GATAbinding protein 2, signal transducers and activators of tran-scription 2, and transcription factor Id1B), immune functions(e.g., cathepsin S, glial fibrillary acidic protein, ISG15, low-molecular-weight polypeptide 7, and 2�-5� oligoadenylate syn-thetase-like 2 [OAS-2]), and metabolism (e.g., fructosebisphosphatase and lactate dehydrogenase). Seventeen geneswere specifically upregulated 6 days after PbK infection (groupB), and 50 genes were specifically upregulated 14 days afterPbK infection (group C), including genes associated with im-mune functions (e.g., CCL5 and vascular cell adhesion mole-cule 1) and hemopoiesis (e.g., �-globin and hemoglobin �).Four genes were upregulated in both PbA(6) and PbK(6)(group D), 16 genes were upregulated in both PbA(6) andPbK(14) (group E), 6 genes were upregulated in both PbK(6)and PbK(14) (group F), and 24 genes were commonly upregu-lated in all three groups (group G). In these intersectinggroups (i.e., D, E, F, and G), genes were commonly associatedwith immune functions (e.g., �2-m, CCL12, CXCL9, CXCL11,

cytotoxic-T-lymphocyte-associated protein 2� [CTLA-2�],granzymes A and B, guanylate nucleotide binding 1, histocom-patibility 2, Ii, IFIT1, IFIT3, IGTP, IRF1, and metallothionein1) (Fig. 2; see Table S5 in the supplemental material).

Similarly, many of these genes were found on lists of up-regulated genes on the Affymetrix arrays following P. bergheiinfection (see Table S6 in the supplemental material).

Functional clustering reveals distinct groups of differen-tially expressed genes. Upregulated genes were clustered usingthe DAVID program, based on level 5 GO terms. For oligo-nucleotide arrays, some clusters were nonspecifically associ-ated with P. berghei infection, including clusters of genes forbiological processes related to immune functions (e.g., antigenprocessing and response to pathogen) as well as molecularfunctions associated with signaling (e.g., nucleotide binding).There were a few clusters of genes that were specific toPbA(6), including those for biological processes (e.g., catabo-lism, negative regulation of biological and cellular processes,and signal transduction) and cellular compartments (e.g., nu-clear lumen and non-membrane-bound organelle). While noclusters were specific to PbK(6), a cluster for a cellular com-partment (e.g., hemoglobin complex) was found to be specificto PbK(14) (see Table S7 in the supplemental material).

Clustering of Affymetrix arrays revealed similar groups ofgenes, particularly genes for biological processes related toimmune functions (e.g., antigen processing and response topathogen) and molecular functions associated with signaling(e.g., nucleotide binding and receptor binding). Again therewere a few clusters of genes that were specific to PbA(6),including those for biological processes (e.g., cytokine biosyn-thesis, regulation of enzyme activity, and endocytosis) and mo-lecular functions (e.g., ligase activity) (see Table S8 in thesupplemental material).

Clustering groups were diverse in downregulated genes, andunannotated genes were prevalent (see Tables S7 and S8 in thesupplemental material). There was no discernible pattern andlittle overlap in the annotations of downregulated genes during

FIG. 2. Venn diagrams of oligonucleotide arrays, showing the intersection (�) between different groups, where group A includes genesupregulated only in PbA(6) (n � 88), group B includes genes upregulated only in PbK(6) (n � 17), group C includes genes upregulated only inPbK(14) (n � 50), group D includes genes upregulated in PbA(6) � PbK(6) (n � 4), group E includes genes upregulated in PbA(6) � PbK(14)(n � 16), group F includes genes upregulated in PbK(6) � PbK(14) (n � 6), and group G includes genes upregulated in all groups (n � 24). GroupH includes genes downregulated only in PbA(6) (n � 65), group I includes genes downregulated only in PbK(6) (n � 168), group J includes genesdownregulated only in PbK(14) (n � 81), group K includes genes downregulated in PbA(6) � PbK(6) (n � 22), group L includes genesdownregulated in PbA(6) � PbK(14) (n � 11), group M includes genes downregulated in PbK(6) � PbK(14) (n � 10), and group N includes genesdownregulated in all groups (n � 9).

VOL. 76, 2008 MICROARRAY ANALYSIS OF PLASMODIUM INFECTION 1815

on Novem

ber 19, 2020 by guesthttp://iai.asm

.org/D

ownloaded from

Page 5: Predominance of Interferon-Related Responses in the Brain during … · Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December

TABLE 2. Selected lists of functionally annotated genes that are upregulated during PbA infection

GO term and gene GO term and gene

Transcription Proteosome (prosome, macropain) subunit, beta type 8 (large multifunctional peptidase 7)Activating transcription factor 3 Proteosome (prosome, macropain) subunit, beta type 9 (large multifunctional peptidase 2)B-cell translocation gene 2, antiproliferative TAP-binding proteinCaudal type homeobox 1 Transporter 1, ATP-binding cassette, subfamily B (MDR/TAP)CCAAT/enhancer binding protein, beta Transporter 2, ATP-binding cassette, subfamily B (MDR/TAP)CCAAT/enhancer binding protein, deltaChromobox homolog 1 (Drosophila HP1 beta) Innate immunityCLPB caseinolytic peptidase b homolog (Escherichia coli) Complement component 1, Q subcomponent, alpha polypeptideEarly growth response 4 Complement component 1, Q subcomponent, beta polypeptideEstrogen-related receptor gamma Complement component 1, Q subcomponent, C chainETS variant gene 4 (E1a enhancer binding protein, E1af) Complement component 1, R subcomponentFBJ osteosarcoma oncogene Complement component 2 (within H-2S)FBJ osteosarcoma oncogene B Complement component 3GATA binding protein 2 Complement component 4B (Childo blood group)General transcription factor II I repeat domain-containing 1 Complement factor BHairy and enhancer of split 1 (Drosophila) DEAD (Asp-Glu-Ala-Asp) box polypeptide 58Inhibitor of DNA binding 1 Fc receptor, IgE, high affinity I, gamma polypeptideIFN-activated gene 204 Fc receptor, IgG, high affinity IIFN-dependent positive-acting transcription factor 3 gamma Fc receptor, IgG, low affinity IIIIRF1 IFN induced with helicase C domain 1IRF7 Serine (or cysteine) peptidase inhibitor, clade G, member 1IRF8 Toll-like receptor 3Lipopolysaccharide-induced TN factorMethyl-CPG binding domain protein 2 ChemotaxisMinichromosome maintenance deficient 5, cell division cycle 46 Chemokine (C-C motif) ligand 2Neuronal D4 domain family member Chemokine (C-C motif) ligand 3Nuclear X box binding factor 1 Chemokine (C-C motif) ligand 5Procollagen (type III) N-endopeptidase Chemokine (C-C motif) ligand 6Retinoblastoma-like 1 (P107) Chemokine (C-C motif) ligand 7Signal transducer and activator of transcription 1 Chemokine (C-C motif) ligand 8Signal transducer and activator of transcription 2 Chemokine (C-C motif) ligand 12Signal transducer and activator of transcription 3 Chemokine (C-X-C motif) ligand 9SIN3-associated polypeptide Chemokine (C-X-C motif) ligand 10Timeless homolog (Drosophila) Chemokine (C-X-C motif) ligand 11Transcription factor 20Tripartite motif protein 30 Immune functionV-MAF musculoaponeurotic fibrosarcoma oncogene family, protein F OAS-1Zinc finger protein, subfamily 1a, 1 (Ikaros) OAS-2

Adenosine deaminaseApoptosis B-cell linker

Axin1 up-regulated 1 CD14 antigenBaculoviral Iap repeat-containing 3 CD3 antigen, delta polypeptideB-cell leukemia/lymphoma 2 related-protein A1A CD40 antigenB-cell translocation gene 2, antiproliferative Colony-stimulating factor 1 (macrophage)Caspase 4, apoptosis-related cysteine peptidase C-type lectin domain family 4, member ECaspase 7 Cyclin-dependent kinase inhibitor 1A (P21)Caspase 8 Eukaryotic translation initiation factor 2-alpha kinase 2Caspase 12 FYN-binding proteinCD74 antigen (invariant polypeptide of MHC, class II antigen associated) Growth arrest and DNA damage-inducible 45 gammaGranzyme A Guanylate nucleotide-binding protein 1Granzyme B Guanylate nucleotide-binding protein 2Growth arrest and DNA damage-inducible 45 beta Guanylate nucleotide-binding protein 4Growth arrest and DNA damage-inducible 45 gamma Integrin alpha LIFN induced with helicase C domain 1 Integrin beta 2Lipopolysaccharide-induced TN factor IFN-activated gene 202Peptidoglycan recognition protein 1 IFN-activated gene 203Phosphatidylserine receptor IFN-activated gene 204PYD and CARD domain containing IFN-dependent positive-acting transcription factor 3 gammaSerine (or cysteine) peptidase inhibitor, clade A, member 3G IFN-Serum/glucocorticoid-regulated kinase IFN--inducible protein 30Tumor necrosis factor receptor superfamily, member 1A IFN induced with helicase C domain 1

IRF1Antigen presentation IRF7

�2-m IRF8Cathepsin S IFN-�-inducible proteinCD74 antigen (invariant polypeptide of MHC, class II antigen associated) IFN-induced protein 35Heat shock protein 1B IFIT1Histocompatibility 2, class II, antigen A, alpha IFIT2Histocompatibility 2, class II, antigen A, beta 1 IFIT3Histocompatibility 2, class II, antigen E beta Interleukin 1 receptor antagonistHistocompatibility 2, class II, locus DMA Leukocyte immunoglobulin-like receptor, subfamily B (with TM and ITIM domains), member 3Histocompatibility 2, class II, locus MB1 Leukocyte immunoglobulin-like receptor, subfamily B, member 4Histocompatibility 2, class II, locus MB2 Lymphocyte antigen 86Histocompatibility 2, D region locus 1 Lymphocyte cytosolic protein 2Histocompatibility 2, D region locus 4 LysozymeHistocompatibility 2, K1, K region Macrophage activation 2Histocompatibility 2, Q region locus 2 Macrophage activation 2 likeHistocompatibility 2, Q region locus 5 Myeloid differentiation primary response gene 88Histocompatibility 2, Q region locus 7 Neutrophil cytosolic factor 1Histocompatibility 2, Q region locus 10 Nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, zetaHistocompatibility 2, T region locus 9 P lysozyme structuralHistocompatibility 2, T region locus 10 Peptidoglycan recognition protein 1Histocompatibility 2, T region locus 22 Ring finger protein 125Histocompatibility 2, T region locus 23 SAM domain and HD domain, 1Histocompatibility 2, T region locus 24 Serine (or cysteine) peptidase inhibitor, clade A, member 3GIFN--inducible protein 30 Serine (or cysteine) peptidase inhibitor, clade A, member 3NMHC A.CA/J(H-2K-F) class I antigen Serine (or cysteine) peptidase inhibitor, clade G, member 1Proteasome (prosome, macropain) 28 subunit, alpha Serum amyloid A 3Proteasome (prosome, macropain) 28 subunit, beta T-cell specific GTPase

1816 MIU ET AL. INFECT. IMMUN.

on Novem

ber 19, 2020 by guesthttp://iai.asm

.org/D

ownloaded from

Page 6: Predominance of Interferon-Related Responses in the Brain during … · Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December

P. berghei infection, due in part to the green bias on oligonu-cleotide arrays. Moreover, since many of the genes found inthese lists were not annotated, these were not explored furtherhere.

Annotations and classes of differentially expressed genes.To gain a clearer overview of genes differentially expressedduring P. berghei infection, gene lists from oligonucleotide andAffymetrix arrays were combined. Importantly, both oligonu-cleotide and Affymetrix array determinations were performedusing the same RNA samples obtained from the same biolog-ical experiment. Genes were annotated using level 3 GO termsin DAVID and the percentage of functional classes deter-mined, together with their statistical significance (see Table S9in the supplemental material).

There was a large overlap in the annotations for genes thatwere upregulated during P. berghei infection, many of whichwere overrepresented. These were associated with GO termsfrom biological processes such as “immune response” [PbA(6),110 genes, P � 1.98 � 10�49; PbK(6), 56 genes, P � 1.19 �10�38; and PbK(14), 74 genes, P � 1.56 � 10�39], molecularfunctions such as “cytokine activity” [PbA(6), 16 genes, P �2.63 � 10�4; PbK(6), 5 genes, P � 6.84 � 10�2; and PbK(14),10 genes, P � 4.13 � 10�3], and cellular compartments such as“immunological synapse” [PbA(6), 13 genes, P � 3.45 � 10�8;PbK(6), 13 genes, P � 1.80 � 10�14; and PbK(14), 14 genes, P �3.68 � 10�12] (see Table S9 in the supplemental material).Overall, there were more genes upregulated by PbA infectionthan by PbK infection. Genes belonging to selected annotationcategories are shown in Table 2.

When the expression patterns in the oligonucleotide andAffymetrix arrays were compared, a high degree of concor-dance between the two platforms was observed, although ahigher number of upregulated genes were observed with theAffymetrix arrays (Table 3).

PCR validation of differentially expressed genes on the ar-rays. Real-time PCR was carried out to validate the resultsobtained from the oligonucleotide and Affymetrix arrays. Ini-tially, validation by real-time PCR was used to confirm theoligonucleotide array data and to determine at which pointgenes were no longer differentially expressed, by randomlyselecting genes at set intervals within the ranked gene lists.This improved the selection criteria for differentially expressedgenes and increased confidence in the selected lists of differ-entially expressed genes (data not shown).

Genes of interest that were upregulated during PbA infec-tion from both oligonucleotide and Affymetrix arrays werethen selected for real-time PCR validation. These includedgenes with known immune functions such as Ii, �2-m, andgranzymes A and B, as well as some genes with interestingannotations, such as OAS-1b, ISG15, IGTP, cathepsin W(CTSW), and CTLA-2�. Many genes were upregulated duringPbA infection but not PbK infection. Figure 3 shows represen-tative graphs of genes upregulated at the mRNA level.

OAS-1b mRNA (Fig. 3A) was upregulated in PbA at day6 p.i. (26.1- 3.1-fold compared with uninfected [mean standard error of the mean {SEM}]) compared with PbK atday 6 p.i. (6.8- 0.6-fold) and PbK at day 14 p.i. (6.7- 1.8-fold). ISG15 mRNA (Fig. 3B) was strongly upregulated inPbA(6) (121.3- 17.4-fold) compared with PbK(6) (20.7- 2.5-fold) and PbK(14) (18.2- 6.0-fold), and similarly, IGTP

mRNA (Fig. 3C) was greatly upregulated in PbA(6) (133.5- 7.8-fold) compared with PbK(6) (18.4- 2.2-fold) andPbK(14) (23.6- 2.6-fold).

There also were some genes that were predominantly up-regulated at the end stage of the disease [PbA(6) andPbK(14)]. These included Ii mRNA (Fig. 3D), which wasstrongly induced in PbA(6) (54.1- 3.9-fold) and PbK(14)(58.4- 16.2-fold) compared with PbK(6) (19.8- 3.2-fold).CTSW mRNA (Fig. 3E) was upregulated in the PbA(6) (31.5- 9.1-fold) and PbK(14) (29.2- 11.4-fold) compared with PbK(6)(11.5- 0.7-fold). CTLA-2� mRNA (Fig. 3F) had a similar ex-pression pattern, being upregulated in PbA(6) (28.0- 4.8-fold)and PbK(14) (44.2- 17.8-fold) compared with PbK(6) (4.0- 0.2-fold).

Overall, the real-time PCR data confirmed the upregulationof genes found on both the oligonucleotide and Affymetrixarrays and showed concordance between these two differentplatforms. However, the real-time PCR results also revealedthat differential expression was not always easily identified byarrays, particularly by oligonucleotide arrays, which are lesssensitive than Affymetrix arrays. By comparing the results fromthe arrays with the real-time PCR results, it is evident that,during hybridization of the samples, a threshold is reached onthe oligonucleotide arrays that does not allow highly upregu-lated or downregulated genes to be distinguished from genesthat are also differentially expressed at lower levels. Thus, anumber of genes found to be upregulated by P. berghei infec-tion on the arrays also were found to be differentially expressedbetween PbA(6), PbK(6), and PbK(14) upon validation withreal-time PCR (Fig. 3).

There also appeared to be a significant problem associatedwith the detection of genes that are of low abundance. Forexample, tumor necrosis factor is known to be highly upregu-lated in the central nervous system during PbA infection (35)but was not identified as an upregulated gene on the arrays,despite being represented. With real-time PCR, tumor necro-sis factor mRNA (data not shown) was found to be stronglyupregulated in PbA(6) compared with PbK(6) and PbK(14).These findings suggest that a number of differentially ex-pressed genes are not detected above background levels eventhough they may play critical roles during PbA infection.

Cellular expression of differentially expressed genes. Thecellular sources of a number of these genes were then deter-mined using laser capture microdissection. Vessels were sepa-rated from parenchymal cells of the brain (neurons and glia) inPbA-infected mice and real-time PCR performed to determinerelative gene expression.

The majority of genes, including Ii, �2-m, OAS-1b, ISG15,and IGTP, were found to be expressed predominantly by ves-sel-associated cells compared with parenchymal cells, whichexpressed mRNAs for these genes at much lower levels. How-ever, CTLA-2� and Fas were very highly expressed by vessel-associated cells but were not detectable in parenchymal cells(Table 4).

Temporal expression of differentially expressed genes. Theexpression patterns of a number of genes validated by real-time PCR also were examined during the progression of PbAinfection. OAS-1b (Fig. 4A) and ISG15 (Fig. 4B) mRNAs wereboth found to increase dramatically at day 4 p.i. (22.1- 5.5-fold compared with uninfected [mean SEM] for OAS-1b

VOL. 76, 2008 MICROARRAY ANALYSIS OF PLASMODIUM INFECTION 1817

on Novem

ber 19, 2020 by guesthttp://iai.asm

.org/D

ownloaded from

Page 7: Predominance of Interferon-Related Responses in the Brain during … · Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December

TABLE 3. Comparisons between selected gene expression data from oligonucleotide and Affymetrix arrays

Category and GenBankaccession no. Ifi no. Name Synonym(s)

Expression on:

Oligonucleotide arraya: Affymetrix arrayb:

PbA(6) PbK(6) PbK(14) PbA(6) PbK(6) PbK(14)

IFN signalingNM_008337 IFN- �NM_009283 STAT1 �� � �NM_019963 STAT2 �� �NM_213659 STAT3 �NM_008390 IRF1 �� �� �NM_016850 IRF7 �� �� �� � �NM_008320 IRF8 ICSBP �� � �NM_008394 IRF9 p48, ISGF3 �� �� �� � �NM_007707 SOCS3 �

MHC class IINM_010378 H2-Aa �� �� ��NM_010381 H2-Ea (Aa/Ea) �� �� ��NM_010387 H2-DMb1/DMb2 � �X00496 Ii �� �� ��� �� �� ��NM_009982 CTSC �� � �NM_021281 CTSS �� � � �

MHC class INM_001001892 H2-K1/K2 � �� � � �NM_010380 H2-D1/Q1 �� �� �� � � ��NM_008200 H2-D3/D4 � � ��NM_010393 H2-Q5 � �� � �X16214 H2-T8/T14 � �NM_010398 H2-T23/T24 �� � ��NM_009735 �2-m ��� �� ��� �� � �NM_010724 PSMB8 LMP-7 �� �� � ��NM_013585 PSMB9 LMP-2 ��� � ��NM_013640 PSMB10 MECL-1 �� �NM_011189 PSME1 PA28� �NM_011190 PSME2 PA28� �NM_134013 PSME4 TEMO �NM_013683 TAP1 �� � �NM_011530 TAP2 �� � �

ApoptosisNM_009807 CASP1 �NM_007611 CASP7 �NM_009812 CASP8 �Y13089 CASP11 � � �NM_009808 CASP12 �NM_010370 GZMA � �� �� � �NM_013542 GZMB ��� �� ��� ��� � ��NM_007601 CAPN3 ��NM_016816 OAS-1 �NM_145211 OAS-1a � �NM_011854 OAS-L2 �� ��� �� ��

ImmunomodulationNM_010851 MyD88 ��NM_126166 TLR3 �NM_008324 IDO �NM_007796 CTLA-2� �� ��� �� � ��

Apoptosis/antiproliferationNM_011940 Ifi202 Ifi202b, Ifi202a ��� �� �NM_008328 Ifi203 � �NM_008329 Ifi204 Ifi16 �� �NM_172648 Ifi205 D3 �

ImmunomodulationNM_015783 ISG15 G1p2 � �� � �

Continued on following page

1818 MIU ET AL. INFECT. IMMUN.

on Novem

ber 19, 2020 by guesthttp://iai.asm

.org/D

ownloaded from

Page 8: Predominance of Interferon-Related Responses in the Brain during … · Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December

and 43.9- 6.7-fold compared with uninfected [mean SEM]for ISG15) before reaching maximal expression at day 6 p.i.(42.6- 8.7-fold for OAS-1b and 76.1- 8.6-fold, ISG15).

On the other hand, IGTP (Fig. 4C) mRNA expressionshowed a more gradual increase from day 4 p.i. (34.0- 2.1-fold), increasing at day 5 p.i. (87.8- 28.2-fold) before reach-ing a maximal expression level at day 6 p.i. (261.3- 85.3-fold).

DISCUSSION

ISGs are induced during P. berghei infection. These resultsshow that ISGs (7, 8) are predominantly induced during P.berghei infection, particularly genes associated with IFN signal-ing pathways and immune responses. mRNAs for many ofthese genes were highly upregulated in the brain during PbAinfection (FMCM), but some also were induced during PbKinfection (NCM), largely at the end stage of the disease. Therelative induction of many genes was generally found to begreater in PbA (6 days p.i.) than in PbK (6 days p.i.) or PbK (14days p.i.) upon real-time PCR validation, highlighting theimportance of IFN- during the pathogenesis of FMCM (1,13, 51).

Some processes that were evident from gene expression pat-terns on the arrays included antigen processing and presenta-tion, apoptosis, and antimicrobial pathways. While some ofthese genes previously have been established to play a role inthe pathogenesis of FMCM, for example, intercellular adhe-sion molecule-1 (28) and granzyme B (46), there also were a

number of upregulated genes, previously unreported, that rep-resent interesting candidates for further investigation.

(i) IFN signaling pathways. Microarray analysis demon-strated that genes for essential components of the IFN signal-ing pathway, including members of the signal transducers andactivators of transcription family and the IRF family, wereinduced during P. berghei infection. IRFs are secondary tran-scription factors that are induced early following IFN- stim-ulation and drive the transcription of IFN--responsive genes,including other ISGs (54). IRF1, a transcriptional activator,was upregulated on the arrays, whereas IRF2, a transcriptionalrepressor, was not. IRF1 is required for the development of aTh1-type immune response, and its absence leads to the induc-tion of a Th2-type response (25, 59). Negative regulators of theIFN signaling pathway, for example, the family of suppressorof cytokine signaling proteins, also were upregulated (see Ta-bles S1 and S3 in the supplemental material).

(ii) MHC processing and presenting pathways. Componentsof the MHC I and MHC II pathways were found by microarrayanalysis to be expressed during P. berghei infection (see TablesS1 and S3 in the supplemental material). This was not surpris-ing given the upregulation of genes associated with IFN sig-naling pathways, particularly IRF1, and the importance ofCD4� and CD8� T cells in malaria immunity and immunopa-thology (16, 46, 50, 58, 67). The expression of genes involved inantigen-processing and -presenting pathways known to be in-volved in the pathogenesis of murine malaria, for example,MHC peptides and costimulatory molecules (24), lends confi-

TABLE 3—Continued

Category and GenBankaccession no. Ifi no. Name Synonym(s)

Expression on:

Oligonucleotide arraya: Affymetrix arrayb:

PbA(6) PbK(6) PbK(14) PbA(6) PbK(6) PbK(14)

AntimicrobialNM_018738 IGTP ��� �� ���� �� ��NM_021792 IIGP1 �� �� ���� ��� ���NM_019440 IIGP2 GTPI �� �� ��� �� ��NM_008326 Ifi1 LRG47 IIGP3, IRGM �� ��� ��� �� ��NM_008330 Ifi47 IRG47 IIGP4, OLFR56 ��� �� ��NM_011579 TGTP GTP2 ��� ��� ��� ���� ��� ��NM_029000 VLIG1 IISG1, GVIN �� �� ��

Unknown functionNM_010259 GBP1 MPA1 ��� �� �� �NM_010260 GBP2 �� ���� ��� ���NM_008620 GBP4 MPA2 ��� ��� ��� �� �� ��NM_027835 IFIH1 MDA5 �� � �NM_029803 IFI27 ISG12 �� � �NM_020583 ISG20 �� � �NM_013562 IFRD1 �NM_008332 IFI56 IFIT1 GARG-16 ��� ��� �� ��� �� ��NM_010501 IFI54 IFIT2 GARG-49 �� �� � �NM_008331 IFI49 IFIT3 GARG-39 �� �� �� ��� �� ��NM_026820 IFITM1 MIL2 �NM_030694 IFITM2 MIL3 � �NM_025378 IFITM3 MIL1, IP15, CD225 ��� ��� ��� �� ��NM_023065 IFI30 �NM_027320 IFI35 �� � �NM_133871 IFI44 ��� �� ��

a �, 1- to 2-fold change; ��, 2- to 4-fold change; ���, 4-fold change.b �, 2- to 5-fold change; ��, 6- to 20-fold change; ���, 21-to 60-fold change; ����, 61- to 100-fold change.

VOL. 76, 2008 MICROARRAY ANALYSIS OF PLASMODIUM INFECTION 1819

on Novem

ber 19, 2020 by guesthttp://iai.asm

.org/D

ownloaded from

Page 9: Predominance of Interferon-Related Responses in the Brain during … · Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December

dence to the validity of the upregulation of novel genes foundby microarray analysis without further confirmation.

(iii) Apoptosis and cell proliferation. A number of genesassociated with apoptotic pathways were expressed on the ar-rays following P. berghei infection. These included caspases,granzymes, cathepsins, calpains, protease inhibitors, OASs,

and members of the p200 family (see Tables S1 and S3 in thesupplemental material). Significantly, these were much morestrongly induced in PbA infection than in PbK infection. Apop-tosis occurs in the brain during human and murine CM inendothelial cells (45, 46, 65), astrocytes (47), and neurons (23,53, 65). Both the death receptor-mediated pathway and the

FIG. 3. mRNA expression of OAS-1b (A), ISG15 (B), IGTP (C), Ii (D), CTSW (E), and CTLA-2� (F) in whole-brain homogenates ofuninfected control mice and PbA(6)-, PbK(6)-, and PbK(14)-infected mice. Expression, measured by real-time PCR as described in Materials andMethods, is relative to the mean expression value in uninfected control samples. Values are means SEMs (n � 6). Samples were initiallydetermined to be significantly different using the Kruskal-Wallis test (P � 0.001), and then the Mann-Whitney test was used: ��, P � 0.01 (valuescompared with uninfected controls); ##, P � 0.01 [values compared with PbA(6)]; **, P � 0.01 [values compared with PbK(6)]; *, P � 0.05 [valuescompared with PbK(6)].

TABLE 4. Relative mRNA expression in microdissected cells during FMCMa

mRNAExpression (mean SEM; n � 4)b

�2-m CTLA-2� Fas IGTP Ii ISG15 OAS-1b

Parenchymal 1.0 NDc ND 1.0 1.0 1.0 1.0Vessel 5.3 2.3 35.21 � 104 22.6 � 104 7.0 � 104 1.9 � 104 20.0 10.5 17.5 6.5 5.1 1.4 5.1 2.3

a Vessels were separated from parenchymal cells (neurons and glia) in PbA-infected mouse brains as described previously (38).b Expression, measured by real-time PCR, is relative to the mean expression value in PbA-infected parenchymal cell samples.c ND, not detected. When PCR products were not detected after 50 cycles of PCR, an arbitrary threshold value of 50 was assigned for the purposes of calculating

a relative induction.

1820 MIU ET AL. INFECT. IMMUN.

on Novem

ber 19, 2020 by guesthttp://iai.asm

.org/D

ownloaded from

Page 10: Predominance of Interferon-Related Responses in the Brain during … · Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December

stress-induced pathway of apoptosis have been implicated inFMCM (21, 42, 45, 47). Both pathways converge on caspase 3,which also is involved in the pathogenesis of FMCM (23, 36,45–47).

Other proteases, including granzymes, cathepsins, and cal-pains, also play a role in apoptosis, primarily by activatingcaspases. Some of these genes also have been implicated inneuronal death in neurological diseases (11, 17, 60), includingCM (33, 56). A number of protease inhibitors were expressedon the arrays, including CTLA-2� (22), and may represent ahost mechanism to limit the damage mediated by proteases,particularly since these proteolytic processes can lead to de-velopment of intracerebral hemorrhaging (63), which is aninvariable finding in human and murine CM.

(iv) Immunomodulatory and antimicrobial genes. Genes in-volved in immune responses, such as Fc receptors and compo-nents of the complement system, were upregulated during P.berghei infection (see Tables S1 and S3 in the supplementalmaterial), reflecting the important role of the immune systemin this disease (19). Genes involved in adaptive immunity andother immunodulatory functions, including cytokines, chemo-kines, leukocyte surface antigens, and adhesion molecules, alsowere induced. There were a number of upregulated ISGs withinteresting but poorly defined functions, including the family ofIFIT genes and the family of IFITM genes. It remains to beseen whether these genes play other roles in host defense,particularly against parasites.

A number of interesting immunomodulatory genes ex-pressed on the arrays have not yet been assigned a role in CM.

For example, ISG15 is released by lymphocytes and monocytes(4, 20) and is inducible by IFN (20) but can itself induce thesecretion of IFN- from T cells and stimulate the activity ofindoleamine 2,3-dioxygenase 1 (49), which has complex rolesin malaria infection (18), as well as lymphocyte and NK pro-liferation, and can induce cytolytic activity (3). Other ISGsidentified by arrays belong to the p47 and p65 GTPase (i.e.,IGTP) families (29, 61). These GTPases have antimicrobialfunctions and are preferentially induced by IFN-, a cytokinethat is essential for the pathogenesis of FMCM through ill-understood mechanisms. While the roles of these genes are notwell described, specific gene knockout mice have differences intheir susceptibilities to infectious diseases, including those ofthe central nervous system (29, 61).

Real-time PCR Validation. Selected genes were validatedusing real-time PCR (e.g., OAS-1b [Fig. 3A], ISG15 [Fig. 3B],IGTP [Fig. 3C], Ii [Fig. 3D], CTSW [Fig. 3E], and CTLA-2�[Fig. 3F]), confirming the pattern of gene expression seen inthe arrays (i.e., upregulated in P. berghei infection). However,real-time PCR also revealed that the expression of some ofthese mRNAs was greater in PbA(6) and PbK(14) than inPbK(6).

Overall these results suggest that there are similar processesoccurring in the brain at the end stages of P. berghei infection,independent of the occurrence of cerebral symptoms. Theseinclude those associated with MHC antigen-processing and-presenting pathways, although there was a striking differencein the mRNA upregulation of costimulatory molecules on thearrays. While PbA infection was associated with increased

FIG. 4. mRNA expression of OAS-1b (A), ISG15 (B), and IGTP (C) in whole-brain homogenates of uninfected control and PbA-infected miceover the course of the disease. Expression, measured by real-time PCR as described in Materials and Methods, is relative to the mean expressionvalue in uninfected control samples. Values are means SEMs (n � 4 to 8). Samples were initially determined to be significantly different usingthe Kruskal-Wallis test (P � 0.001), and then the Mann-Whitney test was used: ���, P � 0.001 (values compared to uninfected controls, day 0);��, P � 0.01 (values compared to uninfected controls, day 0); �, P � 0.05 (values compared to uninfected controls, day 0); **, P � 0.01 (valuescompared to day 4 p.i.); *, P � 0.05 (values compared to day 4 p.i.).

VOL. 76, 2008 MICROARRAY ANALYSIS OF PLASMODIUM INFECTION 1821

on Novem

ber 19, 2020 by guesthttp://iai.asm

.org/D

ownloaded from

Page 11: Predominance of Interferon-Related Responses in the Brain during … · Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December

mRNAs for a number of costimulatory molecules (e.g., CD40and CD86; see Table S3 in the supplemental material), this wasnot seen during PbK infection. In the absence of costimulation,T cells often undergo anergy and apoptosis instead of prolif-eration and differentiation. Since costimulation is an essentialcomponent of the antigen presentation pathway, this suggeststhat there is an absence of T-cell activation during PbK infec-tion.

The location of gene expression in the brain clearly is im-portant. The expression of Ii and �2-m mRNAs was greater invessel-associated cells than in parenchymal cells (Table 4).This suggests that endothelial cells are most likely to expressmalaria antigens associated with either MHC I or MHC II.Indeed, brain endothelial cells are capable of expressing MHCI and MHC II, and their expression levels are associated withthe development of FMCM (39). Microglia also can expressMHC I, but not costimulatory molecules, during FMCM (43).Similarly, mRNAs of genes associated with apoptotic pro-cesses, such as Fas, OAS-1b, and CTLA-2�, were localizedpredominantly to vessel-associated cells compared with paren-chymal cells (Table 4), suggesting that important interactionsoccur at the blood-brain barrier during FMCM. Furthermore,ISG15 and IGTP mRNAs also are predominantly localized tocells associated with cerebral vessels, and given the cytokine-like activities of ISG15 and its potential nonimmune roles inthe brain, as well as the antimicrobial actions of IGTP, theseare interesting candidates to examine in malaria infection.

These findings emphasize the key role played by cells of theblood-brain barrier, particularly endothelial cells, duringFMCM. These cells are located at the key biological interfacebetween the blood compartment, including the circulating in-traerythrocytic malaria parasite, and the brain parenchyma.Changes to the blood-brain barrier during malaria duringFMCM are well described (18, 19) and are thought to be botha cause and an effect of inflammatory processes. The promi-nent localization of gene induction to cells of the blood-brainbarrier supports the idea that key interactions between theparasite and the host occur at the endothelium during FMCM,and as a consequence, pathological events are concentratedprimarily within cerebral microvessels, with lesser and laterinvolvement of cells from the brain parenchyma.

Concluding remarks. Overall, these data are supportive of aninflammatory reaction occurring in the brain during FMCM,which is typified by high circulating levels of inflammatory medi-ators, an activated endothelium, and adherent leukocytes (19).There was a high degree of concordance between results obtainedfrom the oligonucleotide and Affymetrix arrays, which revealedsimilar patterns of gene expression.

Some gene expression changes typical of inflammatory re-sponses also occurred at the end stage of PbK infection, in theabsence of neurological signs. These changes likely result fromhigh circulating levels of PRBCs and cytokines (37) but, over-all, were small compared with changes due to PbA infectionand presumably are insufficient for immunopathological pro-cesses. A number of genes, such as intercellular adhesion mol-ecule-1 and CXCL9, were upregulated at the end stage of PbAand PbK infection and are largely indicative of an activatedendothelium. Moreover, indoleamine 2,3-dioxygenase-1 is verystrongly induced in endothelial cells during the late stages ofPbK (15), perhaps as a tissue-protective response (18), sug-

gesting that important responses also can occur in the brainduring NCM.

These results support the view that leukocytes play a crucialrole in the pathogenesis of FMCM. Leukocytes do not adhere tothe microvasculature during noncerebral models of malaria, in-cluding late stages of PbK infection, and during PbA infection ingenetically resistant mice, for example, those deficient in IFN- orits receptor (1, 51), LT-� (10), and CXCR3 (38), despite contin-ued expression of adhesion molecules by the endothelium. Thissuggests that leukocytes are unable to adhere to the endotheliumduring PbK infection, perhaps because of a lack of integrin acti-vation or an absence of costimulation by antigen-presenting cells,thus leading to leukocyte anergy.

Overall, these data highlight the significant gene expressionchanges in the brain during PbA infection (FMCM). Further-more, certain critical mediators that were upregulated in PbAwere absent during PbK infection. In FMCM, several inducedpathways were evident from the gene expression profiles on thearrays involving signaling and transcription, apoptosis, immu-nomodulation, and antimicrobial activities. Some of these havebeen identified previously in FMCM using arrays (5, 55), butwithout comparisons having been made with late stages ofNCM. The results from this study contribute to our under-standing of the complex pathogenic processes that occur dur-ing malaria.

ACKNOWLEDGMENTS

We thank Bronwyn Robertson and Helen Speirs for their technicalhelp.

This work was supported by grants from the National Health andMedical Research Council of Australia and the Sir Zelman CowenUniversities Fund to N.H.H. J.M. was supported by an AustralianPostgraduate Award. H.J.B. was a Rolf Edgar Lake Research Fellowof the Faculty of Medicine, University of Sydney.

REFERENCES

1. Amani, V., A. M. Vigario, E. Belnoue, M. Marussig, L. Fonseca, D. Mazier,and L. Renia. 2000. Involvement of IFN-gamma receptor-medicated signal-ing in pathology and anti-malarial immunity induced by Plasmodium bergheiinfection. Eur. J. Immunol. 30:1646–1655.

2. Ball, H. J., H. G. MacDougall, I. S. McGregor, and N. H. Hunt. 2004.Cyclooxygenase-2 in the pathogenesis of murine cerebral malaria. J. Infect.Dis. 189:751–758.

3. D’Cunha, J., E. Knight, Jr., A. L. Haas, R. L. Truitt, and E. C. Borden. 1996.Immunoregulatory properties of ISG15, an interferon-induced cytokine.Proc. Natl. Acad. Sci. USA 93:211–215.

4. D’Cunha, J., S. Ramanujam, R. Wagner, P. Witt, E. Knight, Jr., and E.Borden. 1996. In vitro and in vivo secretion of human ISG15, an IFN-induced immunomodulatory cytokine. J. Immunol. 157:4100–4108.

5. Delahaye, N. F., N. Coltel, D. Puthier, L. Flori, R. Houlgatte, F. A. Iraqi, C.Nguyen, G. E. Grau, and P. Rihet. 2006. Gene-expression profiling discrim-inates between cerebral malaria (CM)-susceptible mice and CM-resistantmice. J. Infect. Dis. 193:312–321.

6. Dennis, G., Jr., B. T. Sherman, D. A. Hosack, J. Yang, W. Gao, H. C. Lane,and R. A. Lempicki. 2003. DAVID: Database for Annotation, Visualization,and Integrated Discovery. Genome Biol. 4:P3.

7. Der, S. D., A. Zhou, B. R. Williams, and R. H. Silverman. 1998. Identificationof genes differentially regulated by interferon-alpha, -beta, or -gamma usingoligonucleotide arrays. Proc. Natl. Acad. Sci. USA 95:15623–15628.

8. de Veer, M. J., M. Holko, M. Frevel, E. Walker, S. Der, J. M. Paranjape,R. H. Silverman, and B. R. G. Williams. 2001. Functional classification ofinterferon-stimulated genes identified using microarrays. J. Leukoc Biol.69:912–920.

9. Duggan, D. J., M. Bittner, Y. Chen, P. Meltzer, and J. M. Trent. 1999.Expression profiling using cDNA microarrays. Nat. Genet. 21:10–14.

10. Engwerda, C. R., T. L. Mynott, S. Sawhney, J. B. De Souza, Q. D. Bickle, andP. M. Kaye. 2002. Locally up-regulated lymphotoxin-alpha, not systemictumor necrosis factor-alpha, is the principle mediator of murine cerebralmalaria. J. Exp. Med. 195:1371–1377.

11. Felbor, U., B. Kessler, W. Mothes, H. H. Goebel, H. L. Ploegh, R. T. Bronson,

1822 MIU ET AL. INFECT. IMMUN.

on Novem

ber 19, 2020 by guesthttp://iai.asm

.org/D

ownloaded from

Page 12: Predominance of Interferon-Related Responses in the Brain during … · Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December

and B. R. Olsen. 2002. Neuronal loss and brain atrophy in mice lackingcathepsins B and L. Proc. Natl. Acad. Sci. USA 99:7883–7888.

12. Gentleman, R. C., V. J. Carey, D. M. Bates, B. Bolstad, M. Dettling, S.Dudoit, B. Ellis, L. Gautier, Y. Ge, J. Gentry, K. Hornik, T. Hothorn, W.Huber, S. Iacus, R. Irizarry, F. Leisch, C. Li, M. Maechler, A. J. Rossini, G.Sawitzki, C. Smith, G. Smyth, L. Tierney, J. Y. Yang, and J. Zhang. 2004.Bioconductor: open software development for computational biology andbioinformatics. Genome Biol. 5:R80.

13. Grau, G. E., H. Heremans, P. F. Piguet, P. Pointaire, P. H. Lambert, A.Billiau, and P. Vassalli. 1989. Monoclonal antibody against interferon-gamma can prevent experimental cerebral malaria and its associated over-production of tumor necrosis factor. Proc. Natl. Acad. Sci. USA 86:5572–5574.

14. Griffiths, M. J., M. J. Shafi, S. J. Popper, C. A. Hemingway, M. M. Kortok,A. Wathen, K. A. Rockett, R. Mott, M. Levin, C. R. Newton, K. Marsh, D. A.Relman, and D. P. Kwiatkowski. 2005. Genomewide analysis of the hostresponse to malaria in Kenyan children. J. Infect. Dis. 191:1599–1611.

15. Hansen, A. M., H. J. Ball, A. J. Mitchell, J. Miu, O. Takikawa, and N. H.Hunt. 2004. Increased expression of indoleamine 2,3-dioxygenase in murinemalaria infection is predominantly localised to the vascular endothelium. Int.J. Parasitol. 34:1309–1319.

16. Hermsen, C., T. van de Wiel, E. Mommers, R. Sauerwein, and W. Eling.1997. Depletion of CD4� or CD8� T-cells prevents Plasmodium bergheiinduced cerebral malaria in end-stage disease. Parasitology 114:7–12.

17. Huang, Y., and K. K. W. Wang. 2001. The calpain family and human disease.Trends Mol. Med. 7:355–362.

18. Hunt, N. H., J. Golenser, T. Chan-Ling, S. Parekh, C. Rae, S. Potter, I. M.Medana, J. Miu, and H. J. Ball. 2006. Immunopathogenesis of cerebralmalaria. Int. J. Parasitol. 36:569–582.

19. Hunt, N. H., and G. E. Grau. 2003. Cytokines: accelerators and brakes in thepathogenesis of cerebral malaria. Trends Immunol. 24:491–499.

20. Knight, E., Jr., and B. Cordova. 1991. IFN-induced 15-kDa protein is re-leased from human lymphocytes and monocytes. J. Immunol. 146:2280–2284.

21. Kumar, K. A., and P. P. Babu. 2002. Mitochondrial anomalies are associatedwith the induction of intrinsic cell death proteins-Bcl(2), Bax, cytochrome-cand p53 in mice brain during experimental fatal murine cerebral malaria.Neurosci. Lett. 329:319–323.

22. Kurata, M., M. Hirata, S. Watabe, M. Miyake, S. Y. Takahashi, and Y.Yamamoto. 2003. Expression, purification, and inhibitory activities of mousecytotoxic T-lymphocyte antigen-2alpha. Protein Expr. Purif. 32:119–125.

23. Lackner, P., C. Burger, K. Pfaller, V. Heussler, R. Helbok, M. Morandell, G.Broessner, E. Tannich, E. Schmutzhard, and R. Beer. 2007. Apoptosis inexperimental cerebral malaria: spatial profile of cleaved caspase-3 and ul-trastructural alterations in different disease stages. Neuropathol. Appl. Neu-robiol. 33:560–571.

24. Langhorne, J., F. R. Albano, M. Hensmann, L. Sanni, E. Cadman, C.Voisine, and A. M. Sponaas. 2004. Dendritic cells, pro-inflammatory re-sponses, and antigen presentation in a rodent malaria infection. Immunol.Rev. 201:35–47.

25. Lohoff, M., D. Ferrick, H.-W. Mittrucker, G. S. Duncan, S. Bischof, M.Rollinghoff, and T. W. Mak. 1997. Interferon regulatory factor-1 is requiredfor a T helper 1 immune response in vivo. Immunity 6:681–689.

26. Lovegrove, F. E., S. A. Gharib, S. N. Patel, C. A. Hawkes, K. C. Kain, andW. C. Liles. 2007. Expression microarray analysis implicates apoptosis andinterferon-responsive mechanisms in susceptibility to experimental cerebralmalaria. Am. J. Pathol. 171:1894–1903.

27. Lovegrove, F. E., L. Pena-Castillo, N. Mohammad, W. C. Liles, T. R. Hughes,and K. C. Kain. 2006. Simultaneous host and parasite expression profilingidentifies tissue-specific transcriptional programs associated with susceptibil-ity or resistance to experimental cerebral malaria. BMC Genomics 7:295.

28. Ma, N., N. H. Hunt, M. C. Madigan, and T. Chan-Ling. 1996. Correlationbetween enhanced vascular permeability, up-regulation of cellular adhesionmolecules and monocyte adhesion to the endothelium in the retina duringthe development of fatal murine cerebral malaria. Am. J. Pathol. 149:1745–1762.

29. MacMicking, J. D. 2004. IFN-inducible GTPases and immunity to intracel-lular pathogens. Trends Immunol. 25:601–609.

30. Mandruzzato, S. 2007. Technological platforms for microarray gene expres-sion profiling. Adv. Exp. Med. Biol. 593:12–18.

31. Medana, I. M., T. Chan-Ling, and N. H. Hunt. 2000. Reactive changes ofretinal microglia during fatal murine cerebral malaria: effects of dexameth-asone and experimental permeabilization of the blood-brain barrier. Am. J.Pathol. 156:1055–1065.

32. Medana, I. M., T. Chan-Ling, and N. H. Hunt. 1996. Redistribution anddegeneration of retinal astrocytes in experimental murine cerebral malaria:relationship to disruption of the blood-retinal barrier. Glia 16:51–64.

33. Medana, I. M., N. P. Day, T. T. Hien, N. T. Mai, D. Bethell, N. H. Phu, G. D.Turner, J. Farrar, N. J. White, and M. M. Esiri. 2007. Cerebral calpain infatal falciparum malaria. Neuropathol. Appl. Neurobiol. 33:179–192.

34. Medana, I. M., N. H. Hunt, and T. Chan-Ling. 1997. Early activation ofmicroglia in the pathogenesis of fatal murine cerebral malaria. Glia 19:91–103.

35. Medana, I. M., N. H. Hunt, and G. Chaudhri. 1997. Tumor necrosis factor-alpha expression in the brain during fatal murine cerebral malaria: evidencefor production by microglia and astrocytes. Am. J. Pathol. 150:1473–1486.

36. Medana, I. M., N. T. Mai, N. P. Day, T. T. Hien, D. Bethell, N. H. Phu, J.Farrar, N. J. White, and G. D. Turner. 2001. Cellular stress and injuryresponses in the brains of adult Vietnamese patients with fatal Plasmodiumfalciparum malaria. Neuropathol. Appl. Neurobiol. 27:421–433.

37. Mitchell, A. J., A. M. Hansen, L. Hee, H. J. Ball, S. M. Potter, J. C. Walker,and N. H. Hunt. 2005. Early cytokine production is associated with protec-tion from murine cerebral malaria. Infect. Immun. 73:5645–5653.

38. Miu, J., A. J. Mitchell, M. Muller, S. L. Carter, P. M. Manders, J. A.McQuillan, B. M. Saunders, H. J. Ball, B. Lu, I. L. Campbell, and N. H.Hunt. 2008. Chemokine gene expression during fatal murine cerebral ma-laria and protection due to CXCR3 deficiency. J. Immunol. 180:1217–1230.

39. Monso-Hinard, C., J. N. Lou, C. Behr, P. Juillard, and G. E. Grau. 1997.Expression of major histocompatibility complex antigens on mouse brainmicrovascular endothelial cells in relation to susceptibility to cerebral ma-laria. Immunology 92:53–59.

40. Neill, A. L., and N. H. Hunt. 1992. Pathology of fatal and resolving Plasmo-dium berghei cerebral malaria in mice. Parasitology 105:165–175.

41. Ockenhouse, C. F., W. C. Hu, K. E. Kester, J. F. Cummings, A. Stewart, D. G.Heppner, A. E. Jedlicka, A. L. Scott, N. D. Wolfe, M. Vahey, and D. S. Burke.2006. Common and divergent immune response signaling pathways discov-ered in peripheral blood mononuclear cell gene expression patterns inpresymptomatic and clinically apparent malaria. Infect. Immun. 74:5561–5573.

42. Ohno, T., F. Kobayashi, and M. Nishimura. 2005. Fas has a role in cerebralmalaria, but not in proliferation or exclusion of the murine parasite in mice.Immunogenetics 57:293–296.

43. Pais, T. F., and S. Chatterjee. 2005. Brain macrophage activation in murinecerebral malaria precedes accumulation of leukocytes and CD8� T cellproliferation. J. Neuroimmunol. 163:73–83.

44. Parekh, S. B., W. A. Bubb, N. H. Hunt, and C. Rae. 2006. Brain metabolicmarkers reflect susceptibility status in cytokine gene knockout mice withmurine cerebral malaria. Int. J. Parasitol. 36:1409–1418.

45. Pino, P., I. Vouldoukis, J. P. Kolb, N. Mahmoudi, I. Desportes-Livage, F.Bricaire, M. Danis, B. Dugas, and D. Mazier. 2003. Plasmodium falciparum-infected erythrocyte adhesion induces caspase activation and apoptosis inhuman endothelial cells. J. Infect. Dis. 187:1283–1290.

46. Potter, S., T. Chan-Ling, H. J. Ball, H. Mansour, A. Mitchell, L. Maluish,and N. H. Hunt. 2006. Perforin mediated apoptosis of cerebral microvascularendothelial cells during experimental cerebral malaria. Int. J. Parasitol. 36:485–496.

47. Potter, S. M., T. Chan-Ling, E. Rosinova, H. J. Ball, A. J. Mitchell, and N. H.Hunt. 2006. A role for Fas-Fas ligand interactions during the late-stageneuropathological processes of experimental cerebral malaria. J. Neuroim-munol. 173:96–107.

48. Rae, C., J. A. McQuillan, S. B. Parekh, W. A. Bubb, S. Weiser, V. J. Balcar,A. M. Hansen, H. J. Ball, and N. H. Hunt. 2004. Brain gene expression,metabolism, and bioenergetics: interrelationships in murine models of cere-bral and noncerebral malaria. FASEB J. 18:499–510.

49. Recht, M., E. Borden, and E. Knight, Jr. 1991. A human 15-kDa IFN-induced protein induces the secretion of IFN-gamma. J. Immunol. 147:2617–2623.

50. Renia, L., S. M. Potter, M. Mauduit, D. S. Rosa, M. Kayibanda, J.-C.Deschemin, G. Snounou, and A. C. Gruner. 2006. Pathogenic T cells incerebral malaria. Int. J. Parasitol. 36:547–554.

51. Rudin, W., N. Favre, G. Bordmann, and B. Ryffel. 1997. Interferon-gammais essential for the development of cerebral malaria. Eur. J. Immunol. 27:810–815.

52. Schaecher, K., S. Kumar, A. Yadava, M. Vahey, and C. F. Ockenhouse. 2005.Genome-wide expression profiling in malaria infection reveals transcrip-tional changes associated with lethal and nonlethal outcomes. Infect. Im-mun. 73:6091–6100.

53. Schluesener, H. J., P. G. Kremsner, and R. Meyermann. 1998. Widespreadexpression of MRP8 and MRP14 in human cerebral malaria by microglialcells. Acta Neuropathol. (Berlin) 96:575–580.

54. Schroder, K., P. J. Hertzog, T. Ravasi, and D. A. Hume. 2004. Interferon-gamma: an overview of signals, mechanisms and functions. J. Leukoc. Biol.75:163–189.

55. Sexton, A. C., R. T. Good, D. S. Hansen, M. C. D’Ombrain, L. Buckingham,K. Simpson, and L. Schofield. 2004. Transcriptional profiling reveals sup-pressed erythropoiesis, up-regulated glycolysis, and interferon-associated re-sponses in murine malaria. J. Infect. Dis. 189:1245–1256.

56. Shukla, M., Y. Rajgopal, and P. P. Babu. 2006. Activation of calpains,calpastatin and spectrin cleavage in the brain during the pathology of fatalmurine cerebral malaria. Neurochem. Int. 48:108–113.

57. Smyth, G. K., and T. Speed. 2003. Normalization of cDNA microarray data.Methods 31:265–273.

58. Stephens, R., and J. Langhorne. 2006. Priming of CD4� T cells and devel-opment of CD4� T cell memory; lessons for malaria. Parasite Immunol.28:25–30.

VOL. 76, 2008 MICROARRAY ANALYSIS OF PLASMODIUM INFECTION 1823

on Novem

ber 19, 2020 by guesthttp://iai.asm

.org/D

ownloaded from

Page 13: Predominance of Interferon-Related Responses in the Brain during … · Molecular Immunopathology Unit, Bosch Institute, University of Sydney, Sydney, Australia Received 11 December

59. Taki, S., T. Sato, K. Ogasawara, T. Fukuda, M. Sato, S. Hida, G. Suzuki, M.Mitsuyama, E.-H. Shin, and S. Kojima. 1997. Multistage regulation of Th1-type immune responses by the transcription factor IRF-1. Immunity 6:673–679.

60. Tang, C.-H., J.-W. Lee, M. G. Galvez, L. Robillard, S. E. Mole, and H. A.Chapman. 2006. Murine cathepsin F deficiency causes neuronal lipofusci-nosis and late-onset neurological disease. Mol. Cell. Biol. 26:2309–2316.

61. Taylor, G. A., C. G. Feng, and A. Sher. 2004. p47 GTPases: regulators ofimmunity to intracellular pathogens. Nat. Rev. Immunol. 4:100–109.

62. Thumwood, C. M., N. H. Hunt, I. A. Clark, and W. B. Cowden. 1988.Breakdown of the blood-brain barrier in murine cerebral malaria. Parasitol-ogy 96:579–589.

63. Wang, J., and S. E. Tsirka. 2005. Contribution of extracellular proteolysisand microglia to intracerebral hemorrhage. Neurocrit. Care 3:77–85.

64. WHO. 2000. Severe falciparum malaria. World Health Organization, Com-

municable Diseases Cluster. Trans. R. Soc. Trop. Med. Hyg. 94(Suppl. 1):S1–S90.

65. Wiese, L., J. A. Kurtzhals, and M. Penkowa. 2006. Neuronal apoptosis,metallothionein expression and proinflammatory responses during cerebralmalaria in mice. Exp. Neurol. 200:216–226.

66. Xiang, C. C., M. Chen, L. Ma, Q. N. Phan, J. M. Inman, O. A. Kozhich, andM. J. Brownstein. 2003. A new strategy to amplify degraded RNA from smalltissue samples for microarray studies. Nucleic Acids Res. 31:e53.

67. Yanez, D. M., D. D. Manning, A. J. Cooley, W. P. Weidanz, and H. C. van derHeyde. 1996. Participation of lymphocyte subpopulations in the pathogenesisof experimental murine cerebral malaria. J. Immunol. 157:1620–1624.

68. Ylostalo, J., A. C. Randall, T. A. Myers, M. Metzger, D. J. Krogstad, andF. B. Cogswell. 2005. Transcriptome profiles of host gene expression in amonkey model of human malaria. J. Infect. Dis. 191:400–409.

Editor: J. F. Urban, Jr.

1824 MIU ET AL. INFECT. IMMUN.

on Novem

ber 19, 2020 by guesthttp://iai.asm

.org/D

ownloaded from