Gene expression in blood changes rapidly in

0 downloads 0 Views 547KB Size Report
Jan 4, 2006 - Gene expression changes in human blood after stroke. Y Tang et .... 7 software, Silicon ...... systemically after permanent middle cerebral artery.
Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102 & 2006 ISCBFM All rights reserved 0271-678X/06 $30.00 www.jcbfm.com

Gene expression in blood changes rapidly in neutrophils and monocytes after ischemic stroke in humans: a microarray study Yang Tang1, Huichun Xu1, XinLi Du1, Lisa Lit1, Wynn Walker1, Aigang Lu1, Ruiqiong Ran1, Jeffrey P Gregg2, Melinda Reilly3, Art Pancioli4, Jane C Khoury3, Laura R Sauerbeck3, Janice A Carrozzella3, Judith Spilker4, Joseph Clark3, Kenneth R Wagner3, Edward C Jauch4, Dongwoo J Chang5, Piero Verro1, Joseph P Broderick3 and Frank R Sharp1 1

MIND Institute and Departments of Neurology, University of California at Davis, Sacramento, California, USA; 2MIND Institute and Departments of Pathology, University of California at Davis, Sacramento, California, USA; 3Departments of Neurology, University of Cincinnati, Cincinnati, Ohio, USA; 4Departments of Emergency Medicine, University of Cincinnati, Cincinnati, Ohio, USA; 5MIND Institute and Departments of Neurological Surgery, University of California at Davis, Sacramento, California, USA

Ischemic brain and peripheral white blood cells release cytokines, chemokines and other molecules that activate the peripheral white blood cells after stroke. To assess gene expression in these peripheral white blood cells, whole blood was examined using oligonucleotide microarrays in 15 patients at 2.460.5, 5 and 24 h after onset of ischemic stroke and compared with control blood samples. The 2.4-h blood samples were drawn before patients were treated either with tissue-type plasminogen activator (tPA) alone or with tPA plus Eptifibatide (the Combination approach to Lysis utilizing Eptifibatide And Recombinant tPA trial). Most genes induced in whole blood at 2 to 3 h were also induced at 5 and 24 h. Separate studies showed that the genes induced at 2 to 24 h after stroke were expressed mainly by polymorphonuclear leukocytes and to a lesser degree by monocytes. These genes included: matrix metalloproteinase 9; S100 calcium-binding proteins P, A12 and A9; coagulation factor V; arginase I; carbonic anhydrase IV; lymphocyte antigen 96 (cluster of differentiation (CD)96); monocarboxylic acid transporter (6); ets-2 (erythroblastosis virus E26 oncogene homolog 2); homeobox gene Hox 1.11; cytoskeleton-associated protein 4; N-formylpeptide receptor; ribonuclease-2; N-acetylneuraminate pyruvate lyase; BCL6; glycogen phosphorylase. The fold change of these genes varied from 1.6 to 6.8 and these 18 genes correctly classified 10/15 patients at 2.4 h, 13/15 patients at 5 h and 15/15 patients at 24 h after stroke. These data provide insights into the inflammatory responses after stroke in humans, and should be helpful in diagnosis, understanding etiology and pathogenesis, and guiding acute treatment and development of new treatments for stroke. Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102doi:10.1038/sj.jcbfm.9600264; published online 4 January 2006 Keywords: genomics; inflammation; ischemia; leukocytes; microarrays; neutrophils

Introduction Correspondence: Dr FR Sharp, MIND Institute and Department of Neurology, University of California at Davis, 2805 50th Street – Room 2416, Sacramento, CA 95817, USA. E-mail: [email protected] This study was supported by National Institute of Health grants: NS043252, NS028167, NS042774, NS044283 (FR Sharp) and by P50 NS44283 (JP Broderick). We thank the UCD Genomic Center for running the arrays for this study and the Cincinnati stroke team for obtaining the blood samples. Received 10 September 2005; revised 11 October 2005; accepted 7 November 2005; published online 4 January 2006

There is substantial evidence for an inflammatoryimmune response to ischemic brain mediated by a variety of molecules expressed by leukocytes as well as by the brain (Allan and Rothwell, 2003; Danton and Dietrich, 2003; del Zoppo et al, 2000, del Zoppo et al, 2001; Dirnagl, 2004; Fagan et al, 2004; Iadecola and Alexander, 2001; Lo et al, 2002; Mattila et al, 1998; Zheng and Yenari, 2004). After experimental strokes, there is adhesion of leukocytes to venules within 1 h in a rat model (Ritter et al, 2000) and within the first 2 h in a pig model (Gidday et al,

Gene expression changes in human blood after stroke Y Tang et al 1090

1997). An antibody to the CD18 adhesion molecule blocked the leukocyte adherence to the vessel walls (Gidday et al, 1997). This implies that changes in gene regulation occur in the leukocytes and endothelial cells within this short period of time. Moreover, polymorphonuclear leukocytes (PMN) are detectable within areas of infarction in human strokes within 3 to 6 h (Akopov et al, 1996), with the accumulation being quite marked by 6 to 15 h after ischemic stroke (Akopov et al, 1996; Lindsberg et al, 1996; Lindsberg and Grau, 2003). Direct assessment of white blood cell gene expression has also been studied after stroke. IL1, IL-8, IL10, IL-17, interferon gamma and macrophage inflammatory protein (MIP) mRNAs are elevated in peripheral blood mononuclear cells in patients and animals with ischemic stroke from 1 day up to 10 days after the strokes (Kostulas et al, 1998, 1999; Li et al, 2001; Pelidou et al, 1999). Similarly, leukocyte proteinases, interleukins, matrix metalloproteinases (MMPs) and integrins are elevated in patients after both acute stroke and transient ischemic attacks (Beschorner et al, 2002; Caimi et al, 2001; Elneihoum et al, 1996; Falke et al, 2000; Schwab et al, 2001). At 12 h after stroke, the CD18 adhesion molecule is highly upregulated on leukocytes of peripheral blood of patients with ischemic stroke (Fiszer et al, 1998; Prestigiacomo et al, 1999). In experimental stroke studies, IL1, TNF, TNF receptor, adhesion molecules and various interleukins are elevated in peripheral blood leukocytes within a few hours of ischemic stroke (Barone and Feuerstein, 1999; DeGraba, 1998; del Zoppo et al, 2000; Li et al, 2001). Though it is still unclear whether the immune response in stroke is helpful or harmful (del Zoppo et al, 2001; Dirnagl, 2004; Iadecola and Alexander, 2001; Kazmierski et al, 2004), there is no question that there are marked changes of gene expression in leukocytes in response to stroke with marked infiltration of ischemic areas by inflammatory cells. It is notable that smoking, an important risk factor for cerebral ischemia and hemorrhage, activates neutrophils (Pitzer et al, 1996). Thus, ample evidence supports the notion that changes of gene expression will occur in white blood cells after ischemic stroke. Although changes of body temperature, C-reactive protein and white blood cell count are documented (Audebert et al, 2004; Emsley and Tyrrell, 2002; Smith et al, 2004), the role of inflammation in human ischemic brain is still unclear. This is due in part to the inability to directly study human brain. The difficulty in translating results of animal studies to clinical trials was apparent in the unexpected failure of an antiinflammatory clinical trial (Unnamed, 2001). Therefore, further investigation into inflammatory processes after stroke in humans seemed warranted. Microarray technology makes it possible to examine the expression of thousands of genes. Using this technology, we have identified characteristic blood ribonucleic acid (RNA) expression profiles for Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102

neurological conditions, including epilepsy (Tang et al, 2004a), migraine (Hershey et al, 2004), Tourette’s syndrome (Tang et al, 2005) and several neurogenetic disorders, including Down’s syndrome (Tang et al, 2004b, c). It is likely that this approach could provide surrogate markers for diagnostic and mechanistic purposes, including diseases where there is acute brain injury and an associated inflammatory response. Accordingly, we examined gene expression profiles in the blood of rats subjected to ischemic stroke, hemorrhagic stroke, sham surgeries, kainateinduced seizures, hypoxia and insulin-induced hypoglycemia (Tang et al, 2001). We found injuryspecific gene expression changes in blood using microarrays 24 h after each injury. In addition, there was a common blood gene expression profile that correlated with the occurrence of neuronal cell death regardless of the cause (Tang et al, 2003). This led to a recent human study by Moore et al (2005), who found a genomic signature from peripheral blood that was diagnostic for ischemic stroke in humans. This landmark study examined patients at roughly 1 to 4 days after ischemic stroke, and studied peripheral blood mononuclear cell (PBMC) gene expression. The aims in our study were to extend the animal and human work in stroke and examine the genomic changes of whole blood before 3 h, at 5 h and at 24 h after the onset of ischemic stroke in adult patients. We also compared the genes expressed after stroke to those of several leukocyte subsets, including neutrophils, monocytes, B cells, CD4 + helper T cells, CD8 + cytotoxic T cells and natural killer cells, to determine the cellular origin of the genes regulated by ischemic stroke. The data show that changes of gene expression occur in whole blood before 3 h in many patients and in most patients by 5 h after ischemic stroke, and that the changes of gene expression in blood are accounted for mainly by neutrophils (PMNs) and to a lesser extent by monocytes.

Materials and methods Study Participants A total of 15 patients with acute ischemic stroke enrolled in a clinical trial (Combination approach to Lysis utilizing Eptifibatide And Recombinant tissue-type plasminogen activator (r-tPA) (CLEAR) in acute ischemic stroke participated in this study. The CLEAR trial is a study evaluating the safety and efficacy of a combination of eptifibatide (Integrilin) and low-dose r-tPA (Activase) compared with standard-dose r-tPA (Activase) for acute ischemic stroke in patients treated within 3 h of onset. For the combination treatment arm, eptifibatide is infused intravenously over 2 h and the low dose of r-tPA of 0.6 mg/ kg is infused over 30 mins. Standard-dose r-tPA of 0.9 mg/ kg is infused over an hour with 10% of the dose given as a bolus. Patients are randomized in a double-blind fashion

Gene expression changes in human blood after stroke Y Tang et al 1091

to combined therapy versus the standard dose of intravenons r-tPA in a 3:1 ratio. Ischemic stroke was diagnosed clinically, based on the presence of focal neurological signs and symptoms. Computed tomography brain scan was performed before treatment and patients with hemorrhage excluded from the study. After informed consent, 15 mL blood was obtained from each patient at three time points: before 3 h – designated ‘3 h’ after-stroke (on admission before treatment), ‘5 h’ after-stroke (after the infusion of thrombolytic drugs) and ‘24 h’ after-stroke. Stroke severity was assessed using the National Institute of Health Stroke Scale (NIHSS) at 3, 5, 24 h and 5 days after stroke. Eight healthy subjects were recruited as controls. Each healthy subject was asked to give 15 mL blood at two different times. The Institutional Review Boards at the participating institutions approved the study protocols and consent forms. A total of 60 separate blood samples (45 stroke, 15 control) were analyzed in this study.

Sample Processing and Array Hybridization

Ribonucleic Acid Isolation from Whole Blood: A total of 15 mL blood was collected into 6 PAXgene vacutainers (PreAnalytiX, Hilden, Germany) via antecubital fossa venipuncture at each time. PAXgene tubes contain a proprietary reagent that immediately stabilizes RNA, thus reducing RNA degradation and inhibiting gene induction after phlebotomy (Rainen et al, 2002). Total RNA was isolated using the PAXgene blood RNA kit (PreAnalytiX, Germany) according to the manufacturer’s protocol. The RNA isolated with this protocol comes from all blood cells, including PMN (neutrophil, basophil and eosinophil), mononuclear cells (PBMC) (lymphocytes, macrophage/monocytes), platelets and red blood cells.

red blood corpuscles (RBCs). The desired cells, not labeled with antibody, were collected from the plasma:ficoll interface, washed once with phosphate-buffered saline and the RNA isolated using Trizol reagent. The purity of cells separated with this protocol is 90%75% for CD4 + T cells, 76%78% for CD8 + T cells, 81%78% for CD19 + B cells, 74%710% for CD56 + natural killer (NK) cells and 69%712% for CD14 + monocytes (mean7s.d.) based on flow cytometry data from our laboratory and data provided by the manufacturer. The relative impurity of the cell types obtained is compensated to some degree by the gene selection criteria that were designed to detect genes differentially expressed in the different cell types using cluster analyses. Sample labeling, hybridization to chips and image scanning were performed using standard Affymetrix protocols (Affymetrix Expression Analysis Technical Manual). For each whole blood sample from stroke and control subjects, 10 mg total RNA was labeled using the One-Cycle Target Labeling protocol and hybridized to one array. For each white blood cell subset, 25 ng total RNA was labeled using a Two-Cycle Target Labeling protocol and hybridized to arrays. Affymetrix Human U133 Plus2 arrays that contain more that 54,000 probe sets that sample over 39,000 genes were used for each blood sample in this study. Reverse transcriptase-polymerase chain reaction (RTPCR) confirmation was not performed in this study, because of the limited amount of RNA obtained in a number of the patients. The RNA that is currently available from the patients in this report will be saved, and once the entire CLEAR study is completed, RT-PCR will then be performed on all of the stroke and control subjects as a group.

Probe-Level Data Analysis

Ribonucleic Acid Isolation from Leukocyte Subsets: Blood was drawn from three healthy donors for all leukocyte subsets. Neutrophils were separated from 6 mL sodium citrate, anticoagulated venous blood by centrifugation in leukocyte separation media (Histopaque-1119 and Histopaque-1077; Sigma-Aldrich, St Louis, MO, USA). Ribonucleic acid was isolated using Trizol reagent (Invitrogen, CA, USA) according to the manufacturer’s directions. The purity of the PMNs was B98% based on microscopic examination after Wright-Giemsa staining. Subsets of mononuclear cells, including CD4 + T cells, CD8 + T cells, CD19 + B cells, CD56 + natural killer cells and CD14 + monocytes, were enriched using RosetteSept negative selection methods (StemCell Technologies, Vancouver, BC, Canada). For each subset, 8 mL venous blood was drawn into Vacutainers CPTt Cell Preparation Tubes with sodium citrate (Becton & Dickinson, Bridgeport, NY, USA). (400 mL) RosetteSep TM antibody cocktail was added to each tube of whole blood and incubated for 20 mins at room temperature. The antibody cocktail crosslinks unwanted cells in whole blood to red blood cells, forming immunorosettes with increased density. After incubation, the blood collected in CPTTM tubes is centrifuged for 20 mins at 1800g to precipitate the unwanted cells and free

After scanning the array, the raw expression values (probe level data) for each gene were saved in Affymetrix.cel. files. The probe level data were then collated using Robust Multi-array Average (RMA) software (http://www.bioconductor.org/). This involved nonlinear background reduction, quantile normalization and summarization by median polishing (Bolstad et al, 2003; Irizarry et al, 2003). One array from one healthy control was found to be an outlier and eliminated from further analysis as its average intensity was significantly different from other arrays.

Gene-Level Data Analysis

Genes Regulated by Stroke Compared with Healthy Controls: Three supervised statistical analyses were used with a priori knowledge of sample categories (i.e. 3, 5, 24 h and controls) to identify genes regulated by stroke at each time point compared with healthy controls. The purpose was to identify biologically meaningful genomic changes by adjusting the level of statistical stringency. We first used the one-way analysis of variance (ANOVA) followed by the Student–Newman–Keuls post hoc test with time Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102

Gene expression changes in human blood after stroke Y Tang et al 1092

after stroke onset as the variable (healthy control, 3-, 5and 24-h stroke) (GENESPRINGs 7 software, Silicon Genetics, Redwood City, CA, USA). The type I error probability value (P-value) was corrected with Benjamini– Hochberg false discovery technique (false detection rate (FDR)) for multiple test comparisons. False detection rate < 0.05 was considered significant. We also directly compared each time point after stroke with healthy controls (3 h versus control, 5 h versus control, 24 h vs. control) using three unpaired-sample t-tests instead of ANOVA. The P-value was corrected by Benjamini–Hochberg procedure with FDR < 0.05 as the significance threshold. Prediction analysis of microarray (PAM) software used the ‘nearest shrunken centroids’ method to identify a minimum set of signature genes that distinguish stroke from healthy controls (Tibshirani et al, 2002). The accuracy of the classifier was tested using a tenfold crossvalidation.

Genes Regulated Between Different Times After Stroke: To identify genes that were significantly regulated between different time points after stroke (5 versus 3 h, 24 versus 5 h), a class comparison tool (BRB-Array Tools 2.0, developed by Dr Richard Simon and Amy Peng at the National Cancer Institute and Emmes Corporation) was used. This tool performed a paired-sample t-test for each gene and P < 0.01 was considered significant. Though the P-values were not corrected for multiple test comparisons, the global significance of the genomic changes between different times after stroke was then assessed using a permutation analysis, which performed 1000 random permutations of the class labels (i.e. which arrays correspond to which classes, i.e. 3, 5 and 24 h) and computed the proportion of the random permutations that gave as many genes significant at P < 0.01 level. This estimated probability of randomness provided a global test of whether the expression profiles at two time points were significantly different. In this global testing situation, obtaining a P-value of less than 0.05 is sufficient to establish that expression profiles between two classes were different (BRB-Array Tools 2.0, National Cancer Institute).

Genes that Correlate with Stroke Severity: To identify genes that correlated with clinical stroke severity, we used quantitative traits analysis in BRB-Array Tools. This analysis found genes that were significantly correlated with the NIHSS score at 3 h (quantitative variable), which was used as an index of initial stroke severity. Spearman correlation coefficients were used as a measure of correlation and used to compute parametric P-values for each gene. A parametric P-value of < 0.05 was considered significant. A permutation analysis similar to that used in class comparison was used to evaluate the overall significance of the correlation of gene expression with NIHSS. Genes that Correlate with Outcome of Recanalization Therapy: We next determined whether the blood genomic changes after stroke correlated with the outcome of recanalization therapy. The outcome was defined as the Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102

decrease of NIHSS from 3 to 24 h (DNIHSS) (Broderick et al, 2000). Patients with DNIHSS > 6 were considered as ‘good outcome’ (n = 6), DNIHSS < 3 was considered as ‘relatively bad outcome’ (n = 6), DNIHSS between 3 and 6 was considered as ‘indeterminant’ (n = 3). For each patient, the gene expression value at 24 h was used for the comparison between ‘good outcome’ and ‘bad outcome.’ The class comparison tool of the BRB-Array Tools was used and the significance was assessed by permutation analysis.

Results Blood Genomic Changes at 3, 5 and 24 h After Stroke

We used three different methods to examine the genomic changes after stroke. With ANOVA and the Student–Newman–Keuls Post hoc test (Benjamini– Hochberg FDR < 0.05), a total of 18, 22 and 24 genes are significantly regulated at 3, 5 and 24 h, respectively, compared with healthy controls. Since this approach might be too stringent, we chose to directly compare stroke samples at each time point with controls (3 h versus control, 5 h versus control, 24 h versus control). A total of 104, 1106 and 906 genes were significant at 3, 5 and 24 h, respectively (unpaired t-test, Benjamini–Hochberg FDR < 0.05, fold change > 1.5). The gene lists are included in Supplementary data. We found that many genes were significantly regulated at more than one time point and that nearly all of the genes regulated at 3 h after stroke continued to be regulated at 5 and 24 h (Figure 1A). However, many more genes are significantly regulated at 5 and 24 h than at 3 h. Lastly, examination of the patterns of gene regulation indicated that the majority of the genes that were significantly regulated at 5 h and 24 h had started to change, but had not achieved statistical significance at 3 h. Temporal Genomic Changes After Stroke

To examine the changes of gene expression over time after stroke, the permutation-based, pairedsample t-test analysis was employed (BRB-Array Tools). We found that, among 1335 genes regulated for at least one poststroke time point, 134 genes were regulated between 3 and 5 h (parametric P < 0.01, permutation P = 0.035), and 93 genes were regulated between 5 and 24 h (parametric P < 0.01, permutation P = 0.039). This suggests that the blood genomic pattern was evolving with time after stroke. Among these genes, 23 were significantly changed at 5 h compared with both 3 and 24 h (Figure 1B). This pattern might be related to stroke or may reflect the effects of medication (t-PA7platelet antagonist) given just after the 3 h (2.4270.48 h; see Table 1) blood sample, but before the 5 h blood sample. It is also noteworthy that two genes - IL1b and SOCS3, which were not significantly regulated at 3 h

Gene expression changes in human blood after stroke Y Tang et al 1093

A 3h vs Control

5h vs Control 7

4

438

Prediction Analysis of Blood Genomic Markers for Stroke

89 4

572

241 24h vs Control

B 5h vs 3h

24h vs 5h 21

95

48

2 16

22

50 24h vs 3h

Figure 1 (A) Venn diagram of a total of 1355 genes regulated at 3, 5 and 24 h after stroke compared with healthy controls. These gene lists were generated using the following criteria: (1) Benjamini–Hochberg FDR < 0.05, unpaired t-test. (2) The average fold increase or decrease is at least 1.5-fold compared with healthy controls. (B) Venn diagram of genes significantly regulated between 3 and 5 h, between 5 and 24 h and between 3 h and 24 h after stroke. These gene lists were generated using the following criteria: P < 0.01, paired t-test. 3 h—blood drawn between 2 and 3 h after stroke, but before treatment; Control— healthy control people without neurological or other diseases.

Table 1 Demographic summary of study participants Stroke

Control

Age (year)

64714

49711

Gender Male Female

73.30% 26.70%

87.50% 12.50%

Race White Black Other

80% 20% 0%

75% 0% 25%

Aspirin before stroke 46.60% N/A Time from onset to first blood draw (hour) 2.4270.48 N/A National Institute of Health Stroke Scale 1st blood draw 2nd blood draw 3rd blood draw

compared with healthy controls, were significantly increased at 5 h and increased further at 24 h.

1577 1278 977

N/A N/A N/A

We used PAM, which used the ‘nearest shrunken centroids’ method, to identify the minimum number of signature genes that distinguished ischemic stroke from healthy controls. A threshold is adjusted to produce the fewest number of genes with the least misclassification error (Tibshirani et al, 2002). At a threshold of 4.5, with a total of 29 probe sets which represented 18 genes (Table 2), PAM achieved the lowest error after cross-validation (Figure 2A). All of the control samples are identified correctly, while 5/15 of 3-h samples, 2/15 of 5-h samples and 1/15 of 24-h samples were misclassified (Figures 2B and 3). It is noted that among the five misclassified samples at 3 h, three can be correctly classified at 5 h and all can be correctly classified at 24 h (Figures 3A and 3B). This attests to increased robustness of the blood genomic pattern after stroke over time. However, one sample correctly classified at 3 and 5 h was misclassified at 24 h, which points to interindividual heterogeneity. It is important to emphasize that of the 29 probe sets and 18 genes identified by PAM, none of these genes were regulated as a function of age, race, gender, aspirin or NIH stroke scale as noted below. Cellular Origin of Genes Regulated by Stroke

We next compared the expression of genes regulated by stroke among different blood cell types. Among the 29 probe sets identified by PAM, most were highly expressed by neutrophils compared with the other cell types, while some were also highly expressed by monocytes (Figure 4A). This held true when a more inclusive gene list (1204 genes upregulated by stroke at any of the three time points, Benjamini–Hochberg FDR < 0.05 and fold change > 1.5) was used (Figure 4B). This suggests that neutrophils and monocytes are the major blood cell types involved in the human blood genomic responses to acute ischemic stroke within the 2.4 to 24-h time window examined in this study. Confirmation of these results will require direct measurements of the gene expression in these cell types in stroke patients in future studies. Blood Genomic Patterns Related to Stroke Severity, Outcome and Aspirin

Among 1335 genes regulated by stroke, using an unpaired t-test with 3 samples from each individual treated as replicates, we found that 1186 genes have significantly different expression between individual patients (P < 0.05, Benjamini–Hochberg Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102

Gene expression changes in human blood after stroke Y Tang et al 1094

Table 2 PAM gene list Gene name

3h

5h

24 h

Fold

P-value

Fold

P-value

Fold

Hox 1.11

2.3

6.74E 05

2.9

8.83E 08

2.4

CKAP4 S100A9

2.0 1.6

4.38E 05 2.01E 05

2.4 1.9

3.57E 08 9.50E 10

2.0 1.9

MMP9

3.2

3.54E 05

3.7

4.69E 07

2.7

S100P F5

2.6 1.8

1.11E 04 5.32E 05

3.9 1.9

2.31E 07 1.75E 06

3.0 2.0

FPR1

2.1

2.37E 04

2.2

3.79E 05

2.1

S100A12

2.2

2.59E 04

3.1

1.75E 07

2.9

RNASE2

1.9

1.46E 04

2.0

2.37E 05

2.8

ARG1 CA4 LY96 SLC16A6

3.8 2.2 2.1 1.6

5.03E 3.54E 3.67E 2.37E

04 05 03 04

6.8 2.8 3.4 1.9

6.31E 3.41E 6.43E 2.66E

06 07 06 06

3.3 2.3 3.6 1.9

HIST2H2AA ets-2

1.7 1.8

5.32E 05 1.12E 04

1.9 1.9

5.42E 07 4.69E 07

1.9 2.1

BCL6 PYGL NPL

1.8 2.5 1.9

8.02E 05 8.54E 04 3.54E 05

2.1 4.1 2.3

2.00E 06 4.39E 08 5.68E 09

2.5 3.9 2.2

Description P-value 1.17E 06 Clone IMAGE:5019307, mRNA, homologous to mouse homeobox protein (Hox-1.11) gene 7.88E 08 Cytoskeleton-associated protein 4 5.59E 09 S100 calcium-binding protein A9 (calgranulin B) 1.32E 05 Matrix metalloproteinase 9 (gelatinase B, 92 kDa gelatinase, 92 kDa type IV collagenase) 2.14E 06 S100 calcium-binding protein P 5.71E 07 coagulation factor V (proaccelerin, labile factor) 5.93E 05 Human N-formylpeptide receptor (fMLP-R98) mRNA, complete cds 1.51E 07 S100 calcium-binding protein A12 (calgranulin C) 1.65E 09 Ribonuclease, RNase A family, 2 (liver, eosinophil-derived neurotoxin) 1.17E 06 Arginase I 1.76E 07 Carbonic anhydrase IV 1.51E 07 Lymphocyte antigen 96 3.13E 07 Solute carrier family 16 (monocarboxylic acid transporters), member 6 1.51E 07 Histone 2, H2aa 6.30E 07 v-ets erythroblastosis virus E26 oncogene homolog 2 (avian) 1.51E 07 B-cell CLL/lymphoma 6 3.13E 08 Glycogen phosphorylase 2.06E 07 N-acetylneuraminate pyruvate lyase (dihydrodipicolinate synthase)

A total of 29 probe sets, which represent 18 known genes, were identified by PAM analysis at a threshold of 4.5.

correction). This indicates that the stroke genomic profile is associated with considerable variability between individuals. To begin to address some of the causes of this heterogeneity, we examined the effects of several factors on the expression of the 1335 stroke-regulated genes, including initial stroke severity, response to revascularization therapy and previous use of aspirin. Other factors, including brain hemorrhage after thrombolytic therapy, were not assessed in this first report because of a limited sample size (60 blood samples total in this study). There were 82 genes that significantly correlated with NIHSS at 3 h (P < 0.05, Spearman correlation). However, the permutation P-value was 0.21 (BRBArray Tools), suggesting that with this multiple comparison correction this was not a significant effect with this sample size. This list of genes is included in the appendix to this study. Similarly, a total of 16 genes correlated with the outcome of recanalization therapy (P < 0.01, unpaired t-test ‘good outcome’ versus ‘bad outcome’). Again, because the permutation P-value was 0.19 (BRBArray Tools), this was not a significant effect with this sample size. Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102

For stroke patients on aspirin compared with those not on aspirin, using an FDR of 0.05 and a fold change of 1.5, then 424 genes are regulated between the stroke patients on aspirin versus those not on aspirin. However, of these genes, only 143 genes were significantly different between patients on aspirin before the onset of their strokes (24 h earlier) compared with those who were not on aspirin before their strokes (unpaired t-test, P < 0.01). The permutation P-value was 0.016 (BRB-Array Tools), suggesting that this was a significant effect. Notably, of the 424 aspirin-regulated genes identified using FDR, only 30 genes are among the 906 stroke-regulated genes at 24 h. Most importantly, none of the 424 aspirin-regulated genes were among the strokeregulated genes at 3 h, and none of the aspirinregulated genes are among the 18 genes predictive of stroke. Thus, though aspirin does influence gene expression, aspirin does not have a significant effect on gene expression profiles after acute stroke. This can be appreciated at least visually, in that the blood genomic profiles for the stroke patients on aspirin (A) versus those not on aspirin (not labeled) appear to be similar to each other at each time, as shown in Figures 3A and 3B.

Gene expression changes in human blood after stroke Y Tang et al 1095

Figure 2 Class prediction analysis of all subjects that uses PAM software. Prediction analysis of microarray used the ‘nearest shrunken centroid’ method to identify genes that best distinguish stroke from controls. The number of the signature genes was adjusted by tuning the shrinkage parameter (threshold). The misclassification error for each threshold was computed using a 10-fold crossvalidation. (A) Misclassification error is displayed as a function of the threshold parameter. Error bar represents standard deviation. (B) Crossvalidated probabilities for all samples. Patient ID and time of blood draw were listed on the x-axis (bottom), with stroke samples displayed in the left panel and control samples displayed in the right panel. For each sample, the probability of being predicted as ‘stroke’ (denoted as a pink square) or as ‘control’ (denoted as a blue diamond) was displayed on the y-axis.

Patient Demographics—Effects of Age, Gender and Race

The characteristics of the ischemic stroke patients and healthy control subjects are listed in Table 1. There is a significant difference in age and race between the stroke and control groups. However, we have previously shown that there is no significant difference in gene expression in the blood of 36 younger adults (ages 15 to 50) compared with 22 older (ages 50 to 80) adults (Tang et al, 2004b). In addition, though there are profound effects of gender on blood genomic profiles in our previous studies (Tang et al, 2004b); genes markedly regulated as a function of gender are entirely different from those regulated after acute ischemic stroke in the present study. The list of the most highly regulated genes as a function of gender is included in the appendix to this study. A larger cohort will be needed to test for age and gender differences during acute stroke. Though it was not possible to control for race in this study, we did compare the gene expression of

the 3 black patients with stroke to the 12 Caucasian patients with stroke at each time point of 3, 5 and 24 h. There were 54 genes that were significantly different between the black patients with stroke compared with the Caucasian patients with stroke at 2 to 24 h after stroke—few compared with the 906 genes regulated at 24 h after stroke (see above). Of these 54 genes, only 6 were among the 906 genes regulated at 24 h, none were among the 104 genes regulated at 3 h (see above) and none were among the 18 genes predictive of stroke noted above. The list of the race-regulated genes is attached in the appendix. Though larger sample sizes will need to be studied to carefully address this question, race does not appear to be a major regulator of gene expression after acute stroke in this preliminary study. This is shown pictorially in Figures 3A and 3B, where the gene profiles for the black patients (B) appear to be similar to the Caucasian patients (not labeled) after stroke, at each time point after stroke. The gene lists obtained in this study are included either as an appendix to this publication and/or in the website for this journal. The gene lists will also be provided to anyone who requests them. The lists include genes regulated at each time, genes regulated by gender and race, and genes regulated by aspirin. The lists based on both Bonferroni (very stringent) and FDR multiple comparison corrections are included.

Discussion The major finding of this study is that gene expression changes rapidly in blood after ischemic stroke in humans, occurring before 3 h in 10 of 15 patients by 5 h in 13 of 15 patients, and by 24 h in all 15 patients in this study. Moreover, comparison of gene expression in patients with ischemic stroke with gene expression in different cell types shows that the changes of gene expression after ischemic stroke occur predominantly in PMNs and to a lesser degree in peripheral blood monocytes. This temporal and cell-specific whole genome data from blood is among the first of its kind to be obtained in the first few hours after ischemic stroke either from humans or animals (Moore et al, 2005; Tang et al, 2001).

Comparison to Previous Studies

The first study of whole genome expression in human blood after ischemic stroke, however, was just reported by Moore et al (2005), who found that ischemic stroke could be diagnosed using PBMC with a sensitivity of 78% and specificity of 80%. The mean time for that study was 32.4717.8 h after stroke onset, with one-third of the blood draws performed within 24 h. The current study and that of Moore et al (2005) provide the first proof of principle studies in humans that changes of gene Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102

Gene expression changes in human blood after stroke Y Tang et al 1096

expression occur at very early times in the peripheral blood of patients after ischemic stroke. The current results show changes before 3 h after stroke,

Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102

suggesting that these rapid changes of gene expression might be useful for making the early diagnosis of ischemic stroke in humans. It is notable that the

Gene expression changes in human blood after stroke Y Tang et al 1097

genes regulated in the blood of rats after stroke do not predict the genes that are regulated in the blood of humans after stroke (Tang et al, 2001). There are also significant differences between the genes identified in this study and in that by Moore et al (2005) in spite of using similar Affymetrix microarray technology. Using the Benjamini–Yekutieli multiple test correction, Moore et al identified 771 stroke-regulated genes from PBMC. To make a direct comparison, we employed similar criteria (Benjamini–Hochberg test and at least 1.5-fold change) and identified 1355 genes from whole blood, among which 11% (156 genes) overlap with Moore et al’s (2005) list. Similarly, our study identified 18 known genes using PAM analysis, only two of which were also identified by Moore et al using PAM. The differences between our results and those of Moore et al are likely due in large part to the fact that different cell populations were isolated and studied. Moore et al studied PBMC. Our study utilized whole blood that includes PMNs and PBMCs (which includes lymphocytes and monocytes). Among all blood cell types, PMNs are the first to infiltrate the brain after stroke in humans and animal stroke models (Lindsberg et al, 1996; Akopov et al, 1996; Gidday et al, 1997; Lindsberg and Grau, 2003; Ritter et al, 2000). In addition, our cell-specific data show the following: the large majority of the genes that are induced between 2 and 24 h after stroke in humans are induced in PMNs; only a minority of the induced genes are induced in monocytes; almost all of the 18 genes that are the best predictors of stroke at 3, 5 and 24 h are expressed in PMNs. Hence, since the study of Moore et al (2005) only sampled PBMCs, that study would not have detected the changes of gene expression in PMNs detected in the present study. The other major difference between the present study and that of Moore et al (2005) was that all of the patients in this study were treated either with tissue-type plasminogen activator (tPA) alone or with tPA plus eptifibatide as part of the CLEAR trial. Since patients were treated by 3 h in the present study, the marked increases in the numbers of regulated genes at 5 and 24 h in the present study could be due in part to these medications. However,

the changes of gene expression at the first time point before 3 h in the current study are not due to medication, since this blood sample was drawn before treatment, on average at 2.4 h after the stroke but before treatment that was given by 3 h after the stroke. It is for this reason that we have concentrated on the genes that were most regulated before 3 h after the stroke, that would not be affected by medications, and that continued to be induced at 5 and 24 h after the stroke. Moreover, genes regulated at later times may represent the downstream targets of ‘initiator’ molecules regulated early by stroke, and it is the genes regulated before 3 h that are likely to be most useful for diagnosis, prognosis and guiding treatment. Genes Regulated at 3 h After Stroke: Consistent with the finding that neutrophils (PMNs) infiltrate early after stroke, many genes regulated at 3 h are expressed predominantly by activated PMNs and may contribute to the tissue damage after stroke. Below we discuss the genes regulated at 3 h since these are unaffected by drug treatment in the trial, and the 3-h genes would be most relevant for early responses predictive of stroke. Two molecules— formyl peptide receptor 1 (FPR1) and peptidoglycan recognition protein 1 (PGLYRP1)—represent initial steps leading to PMN activation. FPR1 is a receptor that specifically binds formyl peptides originating from the debris of necrotic cells. Its activation leads to the accumulation of granulocytes at the site of inflammation (Le et al, 2002). PGLYRP1 recognizes the peptidoglycan of bacteria and activates the Toll pathway (Steiner, 2004). Several other molecules may serve as crucial mediators of inflammatory damage. Matrix metalloproteinase 9, largely expressed by neutrophils (Opdenakker et al, 2001), participates in the degradation of extracellular matrix and blood–brain barrier breakdown. Matrix metalloproteinase 9 plasma levels have been related to the infarct volume (Montaner et al, 2003b) and predict hemorrhagic complications after thrombolysis (Castellanos et al, 2003; Montaner et al, 2003a). S100 proteins including S100P, A8, A9 and A12, are also expressed by activated granulocytes (Roth et al, 2003; Vogl et al, 1999). These proteins, bind

Figure 3 Cluster analysis of genes regulated by stroke in patients on aspirin (A) and in patients not on aspirin before stroke (not labeled) in 12 Caucasian (not labeled) and 3 black (B) patients. (A) A panel of 29 genes identified by PAM and (B) a total of 1355 genes regulated at 3, 5 or 24 h after stroke were subjected to hierarchical cluster analyses (Eisen et al, 1998) and displayed from top to bottom. The relative expression in each sample was indicated by the fold change relative to the median of controls, as represented by the color of the squares. The samples were displayed from left to right, with samples misclassified by PAM analysis denoted by pink squares. id—patient identifying number in the study. A = patients on aspirin before the stroke. B = black patients. F5— coagulation Factor 5—EST—expressed sequence tag; PYGL—glycogen phosphorylase; BCL6—B-cell CLL/lymphoma 6; ETS-2—vets erythroblastosis virus E26 oncogene homolog 2 (avian); NPL- N-acetylneuraminate pyruvate lyase; SLC16A6—monocarboxylic acid transporter, member 6; CKAP4—cytoskeleton-associated protein 4; Hox 1.11—homologous to mouse homeobox protein (Hox 1.11) gene; S100P—S100 calcium-binding protein P; S100A9—calgranulin B, S100 calcium-binding protein A9; S100A12— calgranulin C; CA4—carbonic anyhydrase IV; MMP9—matrix metalloproteinase 9 (gelatinase B); ARG1—arginase I; FPR1—human N-formylpeptide receptor; HIST2H2AA—histone 2; RNASE2—ribonuclease, Rnase A family, 2; LY96—lymphocyte antigen 96. Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102

Gene expression changes in human blood after stroke Y Tang et al 1098

Figure 4 Cellular origin of genes regulated by stroke. Blood cell subtypes, including B cells (B), monocytes (Mono), PMN or neutrophil, CD4 + T cells (CD4 + ), CD8 + T cells (CD8 + ) and NK cells were separated from a single healthy donor. Ribonucleic acid isolated from each subset was hybridized to three genechips as technical replicates. Expression levels of genes regulated by stroke were compared between different cell subtypes. (A) 29 probe sets identified by PAM; (B) 1204 genes upregulated by any of three poststroke time points (Benjamini–Hochberg FDR < 0.05, fold change > 1.5). B—CD19 + antibody-producing cells; Mono—CD14 + monocytes; PMN—polymorphonuclear cells, neutrophils; CD4 + —T cells; CD8 + —T cells; NK—CD56 + natural killer T cells. Gene abbreviations are the same as in Figure 3. Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102

Gene expression changes in human blood after stroke Y Tang et al 1099

specifically to vascular endothelial cells and induce a thrombogenic and inflammatory response (Viemann et al, 2004). Neutrophil cytosolic factor-4 (NCF-4) is also induced by 3 h after stroke, a gene that codes for p40(phox) protein that is a component of the nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase complex that produces superoxide-generated free radicals in neutrophils, and is related to NCF-1 and NCF-2, which are mutated in neutrophils of patients with chronic granulomatous disease (Kuribayashi et al, 2002). Coagulation factor V and VIII are also upregulated after ischemic stroke in this study. Plasma levels of these two factors are increased in acute coronary syndromes and correlate with the magnitude of inflammation (Apetrei et al, 2004). These procoagulants might be expressed by monocytes/macrophages and participate in the inflammatory process (Edwards and Rickles, 1984). Several genes regulated at 3 h, including pellino 1, IRAK and NALP12, play important roles in IL-1 signaling (Jiang et al, 2003; Tschopp et al, 2003). However, the blood expression of IL-1b itself was not increased until 5 h. Therefore, cytokines released from endogenous microglia may play a role in priming the IL-1 pathway of peripheral leukocytes. Besides the proinflammatory molecules, several molecules that negatively regulate the innate immune responses were also expressed. For example, the prototypic decoy receptor IL-1RII, which is the major IL-1 scavenger on PMNs (Bourke et al, 2003), is increased at 3 h after stroke. SOCS3, a suppressor of cytokine signaling, correlates highly with the expression of IL-1b in this study and may represent an important mechanism for limiting the immune response (Starr et al, 1997). Stroke Blood Test—Previous Studies: Since changes of gene expression are detected before 3 h after stroke, and 10/15 patients can be predicted as having stroke at 3 h compared with healthy controls, it is possible that these changes of gene expression could be useful in developing a diagnostic test for stroke. Many attempts have been made at identifying serum or plasma protein markers of stroke, including MMP9, GFAP, S100 proteins, neuronspecific enolase, N-acetyl-aspartic acid and many others (Ali et al, 2001, 2000; Bertsch et al, 2001; Buttner et al, 1999, 1997; Castellanos et al, 2003; Elting et al, 2000; Herrmann et al, 2000; Lindsberg and Grau, 2003; Lynch et al, 2004; Martens et al, 1998; Montaner et al, 2003a, b; Persson et al, 1987; Ribo et al, 2004; Rosen et al, 1998; Schoerkhuber et al, 1999; Smith et al, 2004; Stevens et al, 1999; Strachan et al, 1999; Sulter et al, 1998; Ueda et al, 1995; Vila et al, 2000). Indeed, MMP9, alone or with a panel of four proteins, can predict stroke with high accuracy (Lynch et al, 2004; Montaner et al, 2003b). However, most of the proposed markers have not been examined in the very acute patient between 2 and 12 h when acute diagnosis and treatment are

most important. Whether gene expression or protein levels will eventually be used for a diagnostic test, the present study suggests that there are very early changes that could be useful in diagnosis and potentially predict the hemorrhagic complications of tPA and other drugs (Montaner et al, 2003a). Ribonucleic acid expression might be useful because it is induced before proteins, though it is easier to measure proteins rapidly using current technology. Effects of Comorbidities and Other Risk Factors on Gene Expression in Blood

It is important to emphasize that the 18 genes identified in this study might or might not be those most useful in eventually diagnosing stroke in a 3-h time window. The genes derived in this study compared ischemic strokes with healthy controls poorly matched for age, race and vascular risk factors. We have previously characterized the genomic changes in blood related to age and gender (Tang et al, 2004b), and hence these were not included in the lists of genes identified in the current study. Our preliminary results indicate that race may modulate gene expression after acute stroke, but this is not a major factor. The effects of common vascular risk factors, including smoking, hypertension, diabetes and hyperlipidemia, have been controlled to some degree by Moore et al (2005), but need further study. Even if risk factors were controlled for, the current data does not answer whether this panel of genes would distinguish ischemic stroke from intracerebral hemorrhage or other medical conditions like myocardial infarction. Thus, the changes of gene expression must be interpreted with caution. However, it is clear that the evolving genomic pattern identified in the current study is mostly caused by stroke, because the effects of coexisting confounding factors would be relatively stable for a particular patient and would not progress over a 24-h period of time. The progression of gene expression over time suggests that the gene changes identified here were related, at least in large part, to evolving cerebral infarction. How Genes in Blood Relate to the Pathophysiology of Ischemic Brain Injury: The mechanisms for the extremely rapid changes in gene expression after ischemic stroke remain to be elucidated, but may involve direct interactions between the white blood cells and ischemic brain endothelium. In addition, it is possible that the blood genomic response is preprogrammed to some extent once cerebral ischemia occurs. The current data also shows that genomic changes of peripheral granulocytes precede their detectable accumulation in brain by nuclear imaging techniques (Akopov et al, 1996; Price et al, 2004). This is expected since signals between cerebral endothelial cells and passing leukocytes Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102

Gene expression changes in human blood after stroke Y Tang et al 1100

must first result in chemoattraction, which is then followed by adhesion and migration into the brain. The genomic changes that occur in the leukocytes during this time should help us understand this process in humans. If the response of neutrophils and monocytes after stroke is harmful, then strategies for knocking down the genes identified here in peripheral blood leukocytes might decrease this inflammatory response and thereby improve tissue survival after ischemia and perhaps decrease reperfusion induced cerebral hemorrhage.

References Akopov SE, Simonian NA, Grigorian GS (1996) Dynamics of polymorphonuclear leukocyte accumulation in acute cerebral infarction and their correlation with brain tissue damage. Stroke 27:1739–43 Ali C, Docagne F, Nicole O, Lesne S, Toutain J, Young A, Chazalviel L, Divoux D, Caly M, Cabal P, Derlon JM, MacKenzie ET, Buisson A, Vivien D (2001) Increased expression of transforming growth factor-beta after cerebral ischemia in the baboon: an endogenous marker of neuronal stress? J Cereb Blood Flow Metab 21:820–7 Ali MS, Harmer M, Vaughan R (2000) Serum S100 protein as a marker of cerebral damage during cardiac surgery. Br J Anaesth 85:287–98 Allan SM, Rothwell NJ (2003) Inflammation in central nervous system injury. Philos Trans R Soc Lond B Biol Sci 358:1669–77 Apetrei E, Ciobanu-Jurcut R, Rugina M, Gavrila A, Uscatescu V (2004) C-reactive protein, prothrombotic imbalance and endothelial dysfunction in acute coronary syndromes without ST elevation. Rom J Intern Med 42:95–102 Audebert HJ, Rott MM, Eck T, Haberl RL (2004) Systemic inflammatory response depends on initial stroke severity but is attenuated by successful thrombolysis. Stroke 35:2128–33 Barone FC, Feuerstein GZ (1999) Inflammatory mediators and stroke: new opportunities for novel therapeutics. J Cereb Blood Flow Metab 19:819–34 Bertsch T, Casarin W, Kretschmar M, Zimmer W, Walter S, Sommer C, Muehlhauser F, Ragoschke A, Kuehl S, Schmidt R, Eden BP, Nassabi C, Nichterlein T, Fassbender K (2001) Protein S-100B: a serum marker for ischemic and infectious injury of cerebral tissue. Clin Chem Lab Med 39:319–23 Beschorner R, Schluesener HJ, Gozalan F, Meyermann R, Schwab JM (2002) Infiltrating CD14+ monocytes and expression of CD14 by activated parenchymal microglia/macrophages contribute to the pool of CD14+ cells in ischemic brain lesions. J Neuroimmunol 126:107–15 Bolstad BM, Irizarry RA, Astrand M, Speed TP (2003) A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics 19:185–93 Bourke E, Cassetti A, Villa A, Fadlon E, Colotta F, Mantovani A (2003) IL-1 beta scavenging by the type II IL-1 decoy receptor in human neutrophils. J Immunol 170:5999–6005 Broderick JP, Lu M, Kothari R, Levine SR, Lyden PD, Haley EC, Brott TG, Grotta J, Tilley BC, Marler JR, Frankel M (2000) Finding the most powerful measures of the Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102

effectiveness of tissue plasminogen activator in the NINDS tPA stroke trial. Stroke 31:2335–41 Buttner T, Lack B, Jager M, Wunsche W, Kuhn W, Muller T, Przuntek H, Postert T (1999) Serum levels of neuronspecific enolase and s-100 protein after single tonic– clonic seizures. J Neurol 246:459–61 Buttner T, Weyers S, Postert T, Sprengelmeyer R, Kuhn W (1997) S-100 protein: serum marker of focal brain damage after ischemic territorial MCA infarction. Stroke 28:1961–5 Caimi G, Canino B, Ferrara F, Montana M, Musso M, Porretto F, Carollo C, Catania A, Lo Presti R (2001) Granulocyte integrins before and after activation in acute ischaemic stroke. J Neurol Sci 186:23–6 Castellanos M, Leira R, Serena J, Pumar JM, Lizasoain I, Castillo J, Davalos A (2003) Plasma metalloproteinase-9 concentration predicts hemorrhagic transformation in acute ischemic stroke. Stroke 34:40–6 Danton GH, Dietrich WD (2003) Inflammatory mechanisms after ischemia and stroke. J Neuropathol Exp Neurol 62:127–36 DeGraba TJ (1998) The role of inflammation after acute stroke: utility of pursuing anti-adhesion molecule therapy. Neurology 51:S62–8 del Zoppo G, Ginis I, Hallenbeck JM, Iadecola C, Wang X, Feuerstein GZ (2000) Inflammation and stroke: putative role for cytokines, adhesion molecules and iNOS in brain response to ischemia. Brain Pathol 10:95–112 del Zoppo GJ, Becker KJ, Hallenbeck JM (2001) Inflammation after stroke: is it harmful? Arch Neurol 58:669–72 Dirnagl U (2004) Inflammation in stroke: the good, the bad, and the unknown. Ernst Schering Res Found Workshop 47:87–99 Edwards RL, Rickles FR (1984) Macrophage procoagulants. Prog Hemost Thromb 7:183–209 Eisen MB, Spellman PT, Brown PO, Botstein D (1998) Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci USA 95:14863–8 Elneihoum AM, Falke P, Axelsson L, Lundberg E, Lindgarde F, Ohlsson K (1996) Leukocyte activation detected by increased plasma levels of inflammatory mediators in patients with ischemic cerebrovascular diseases. Stroke 27:1734–8 Elting JW, de Jager AE, Teelken AW, Schaaf MJ, Maurits NM, van der Naalt J, Sibinga CT, Sulter GA, De Keyser J (2000) Comparison of serum S-100 protein levels following stroke and traumatic brain injury. J Neurol Sci 181:104–10 Emsley HC, Tyrrell PJ (2002) Inflammation and infection in clinical stroke. J Cereb Blood Flow Metab 22: 1399–419 Fagan SC, Hess DC, Hohnadel EJ, Pollock DM, Ergul A (2004) Targets for vascular protection after acute ischemic stroke. Stroke 35:2220–5 Falke P, Elneihoum AM, Ohlsson K (2000) Leukocyte activation: relation to cardiovascular mortality after cerebrovascular ischemia. Cerebrovasc Dis 10: 97–101 Fiszer U, Korczak-Kowalska G, Palasik W, Korlak J, Gorski A, Czlonkowska A (1998) Increased expression of adhesion molecule CD18 (LFA-1beta) on the leukocytes of peripheral blood in patients with acute ischemic stroke. Acta Neurol Scand 97:221–4 Gidday JM, Park TS, Gonzales ER, Beetsch JW (1997) CD18-dependent leukocyte adherence and vascular injury in pig cerebral circulation after ischemia. Am J Physiol 272:H2622–9

Gene expression changes in human blood after stroke Y Tang et al

Herrmann M, Vos P, Wunderlich MT, de Bruijn CH, Lamers KJ (2000) Release of glial tissue-specific proteins after acute stroke: a comparative analysis of serum concentrations of protein S-100B and glial fibrillary acidic protein. Stroke 31:2670–7 Hershey AD, Tang Y, Powers SW, Kabbouche MA, Gilbert DL, Glauser TA, Sharp FR (2004) Genomic abnormalities in patients with migraine and chronic migraine: preliminary blood gene expression suggests platelet abnormalities. Headache 44:994–1004 Iadecola C, Alexander M (2001) Cerebral ischemia and inflammation. Curr Opin Neurol 14:89–94 Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, Speed TP, Astrand M (2003) Summaries of Affymetrix GeneChip probe level data: a comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Nucleic Acids Res 31:e15 Jiang Z, Johnson HJ, Nie H, Qin J, Bird TA, Li X (2003) Pellino 1 is required for interleukin-1 (IL-1)-mediated signaling through its interaction with the IL-1 receptorassociated kinase 4 (IRAK4)-IRAK-tumor necrosis factor receptor-associated factor 6 (TRAF6) complex. J Biol Chem 278:10952–6 Kazmierski R, Guzik P, Ambrosius W, Ciesielska A, Moskal J, Kozubski W (2004) Predictive value of white blood cell count on admission for in-hospital mortality in acute stroke patients. Clin Neurol Neurosurg 107:38–43 Kostulas N, Kivisakk P, Huang Y, Matusevicius D, Kostulas V, Link H (1998) Ischemic stroke is associated with a systemic increase of blood mononuclear cells expressing interleukin-8 mRNA. Stroke 29:462–6 Kostulas N, Pelidou SH, Kivisakk P, Kostulas V, Link H (1999) Increased IL-1beta, IL-8, and IL-17 mRNA expression in blood mononuclear cells observed in a prospective ischemic stroke study. Stroke 30:2174–9 Kuribayashi F, Nunoi H, Wakamatsu K, Tsunawaki S, Sato K, Ito T, Sumimoto H (2002) The adaptor protein p40(phox) as a positive regulator of the superoxideproducing phagocyte oxidase. EMBO J 21:6312–20 Le Y, Murphy PM, Wang JM (2002) Formyl-peptide receptors revisited. Trends Immunol 23:541–8 Li HL, Kostulas N, Huang YM, Xiao BG, van der Meide P, Kostulas V, Giedraitas V, Link H (2001) IL-17 and IFNgamma mRNA expression is increased in the brain and systemically after permanent middle cerebral artery occlusion in the rat. J Neuroimmunol 116:5–14 Lindsberg PJ, Carpen O, Paetau A, Karjalainen-Lindsberg ML, Kaste M (1996) Endothelial ICAM-1 expression associated with inflammatory cell response in human ischemic stroke. Circulation 94:939–45 Lindsberg PJ, Grau AJ (2003) Inflammation and infections as risk factors for ischemic stroke. Stroke 34:2518–32, (Epub 2003 Sep 2518) Lo EH, Wang X, Cuzner ML (2002) Extracellular proteolysis in brain injury and inflammation: role for plasminogen activators and matrix metalloproteinases. J Neurosci Res 69:1–9 Lynch JR, Blessing R, White WD, Grocott HP, Newman MF, Laskowitz DT (2004) Novel diagnostic test for acute stroke. Stroke 35:57–63 Martens P, Raabe A, Johnsson P (1998) Serum S-100 and neuron-specific enolase for prediction of regaining consciousness after global cerebral ischemia. Stroke 29:2363–6 Mattila KJ, Valtonen VV, Nieminen MS, Asikainen S (1998) Role of infection as a risk factor for athero-

sclerosis, myocardial infarction, and stroke. Clin Infect Dis 26:719–34 Montaner J, Molina CA, Monasterio J, Abilleira S, Arenillas JF, Ribo M, Quintana M, Alvarez-Sabin J, Castellanos M, Leira R, Serena J, Pumar JM, Lizasoain I, Castillo J, Davalos A (2003a) Matrix metalloproteinase9 pretreatment level predicts intracranial hemorrhagic complications after thrombolysis in human stroke. Circulation 107:598–603 Montaner J, Rovira A, Molina CA, Arenillas JF, Ribo M, Chacon P, Monasterio J, Alvarez-Sabin J (2003b) Plasmatic level of neuroinflammatory markers predict the extent of diffusion-weighted image lesions in hyperacute stroke. J Cereb Blood Flow Metab 23:1403–7 Moore DF, Li H, Jeffries N, Wright V, Cooper Jr RA, Elkahloun A, Gelderman MP, Zudaire E, Blevins G, Yu H, Goldin E, Baird AE (2005) Using peripheral blood mononuclear cells to determine a gene expression profile of acute ischemic stroke: a pilot investigation. Circulation 111:212–21 Opdenakker G, Van den Steen PE, Dubois B, Nelissen I, Van Coillie E, Masure S, Proost P, Van Damme J (2001) Gelatinase B functions as regulator and effector in leukocyte biology. J Leukoc Biol 69:851–9 Pelidou SH, Kostulas N, Matusevicius D, Kivisakk P, Kostulas V, Link H (1999) High levels of IL-10 secreting cells are present in blood in cerebrovascular diseases. Eur J Neurol 6:437–42 Persson L, Hardemark HG, Gustafsson J, Rundstrom G, Mendel-Hartvig I, Esscher T, Pahlman S (1987) S-100 protein and neuron-specific enolase in cerebrospinal fluid and serum: markers of cell damage in human central nervous system. Stroke 18:911–8 Pitzer JE, Del Zoppo GJ, Schmid-Schonbein GW (1996) Neutrophil activation in smokers. Biorheology 33:45–58 Prestigiacomo CJ, Kim SC, Connolly Jr ES, Liao H, Yan SF, Pinsky DJ (1999) CD18-mediated neutrophil recruitment contributes to the pathogenesis of reperfused but not nonreperfused stroke. Stroke 30:1110–7 Price CJ, Menon DK, Peters AM, Ballinger JR, Barber RW, Balan KK, Lynch A, Xuereb JH, Fryer T, Guadagno JV, Warburton EA (2004) Cerebral neutrophil recruitment, histology, and outcome in acute ischemic stroke: an imaging-based study. Stroke 35:1659–64 Rainen L, Oelmueller U, Jurgensen S, Wyrich R, Ballas C, Schram J, Herdman C, Bankaitis-Davis D, Nicholls N, Trollinger D, Tryon V (2002) Stabilization of mRNA expression in whole blood samples. Clin Chem 48:1883–90 Ribo M, Montaner J, Molina CA, Arenillas JF, Santamarina E, Quintana M, Alvarez-Sabin J (2004) Admission fibrinolytic profile is associated with symptomatic hemorrhagic transformation in stroke patients treated with tissue plasminogen activator. Stroke 35:2123–7. (Epub 2004 Jul 2128) Ritter LS, Orozco JA, Coull BM, McDonagh PF, Rosenblum WI (2000) Leukocyte accumulation and hemodynamic changes in the cerebral microcirculation during early reperfusion after stroke. Stroke 31:1153–61 Rosen H, Rosengren L, Herlitz J, Blomstrand C (1998) Increased serum levels of the S-100 protein are associated with hypoxic brain damage after cardiac arrest. Stroke 29:473–7 Roth J, Vogl T, Sorg C, Sunderkotter C (2003) Phagocytespecific S100 proteins: a novel group of proinflammatory molecules. Trends Immunol 24:155–8

1101

Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102

Gene expression changes in human blood after stroke Y Tang et al 1102

Schoerkhuber W, Kittler H, Sterz F, Behringer W, Holzer M, Frossard M, Spitzauer S, Laggner AN (1999) Time course of serum neuron-specific enolase. A predictor of neurological outcome in patients resuscitated from cardiac arrest. Stroke 30:1598–603 Schwab JM, Nguyen TD, Meyermann R, Schluesener HJ (2001) Human focal cerebral infarctions induce differential lesional interleukin-16 (IL-16) expression confined to infiltrating granulocytes, CD8+ T-lymphocytes and activated microglia/macrophages. J Neuroimmunol 114:232–41 Smith CJ, Emsley HC, Gavin CM, Georgiou RF, Vail A, Barberan EM, del Zoppo GJ, Hallenbeck JM, Rothwell NJ, Hopkins SJ, Tyrrell PJ (2004) Peak plasma interleukin-6 and other peripheral markers of inflammation in the first week of ischaemic stroke correlate with brain infarct volume, stroke severity and long-term outcome. BMC Neurol 4:2 Starr R, Willson TA, Viney EM, Murray LJ, Rayner JR, Jenkins BJ, Gonda TJ, Alexander WS, Metcalf D, Nicola NA, Hilton DJ (1997) A family of cytokine-inducible inhibitors of signalling. Nature 387:917–21 Steiner H (2004) Peptidoglycan recognition proteins: on and off switches for innate immunity. Immunol Rev 198:83–96 Stevens H, Jakobs C, de Jager AE, Cunningham RT, Korf J (1999) Neurone-specific enolase and N-acetyl-aspartate as potential peripheral markers of ischaemic stroke. Eur J Clin Invest 29:6–11 Strachan MW, Abraha HD, Sherwood RA, Lammie GA, Deary IJ, Ewing FM, Perros P, Frier BM (1999) Evaluation of serum markers of neuronal damage following severe hypoglycaemia in adults with insulin-treated diabetes mellitus. Diabetes Metab Res Rev 15:5–12 Sulter G, Elting JW, De Keyser J (1998) Increased serum neuron specific enolase concentrations in patients with hyperglycemic cortical ischemic stroke. Neurosci Lett 253:71–3 Tang Y, Gilbert DL, Glauser TA, Hershey AD, Sharp FR (2005) Blood gene expression profiling of neurologic diseases: a pilot microarray study. Arch Neurol 62:210–5 Tang Y, Glauser TA, Gilbert DL, Hershey AD, Privitera MD, Ficker DM, Szaflarski JP, Sharp FR (2004a) Valproic acid blood genomic expression patterns in children with epilepsy—a pilot study. Acta Neurol Scand 109:159–68 Tang Y, Lu A, Aronow BJ, Sharp FR (2001) Blood genomic responses differ after stroke, seizures, hypoglycemia,

and hypoxia: blood genomic fingerprints of disease. Ann Neurol 50:699–707 Tang Y, Lu A, Ran R, Aronow BJ, Schorry EK, Hopkin RJ, Gilbert DL, Glauser TA, Hershey AD, Richtand NW, Privitera M, Dalvi A, Sahay A, Szaflarski JP, Ficker DM, Ratner N, Sharp FR (2004b) Human blood genomics: distinct profiles for gender, age and neurofibromatosis type 1. Brain Res Mol Brain Res 132:155–67 Tang Y, Nee AC, Lu A, Ran R, Sharp FR (2003) Blood genomic expression profile for neuronal injury. J Cereb Blood Flow Metab 23:310–9 Tang Y, Schapiro MB, Franz DN, Patterson BJ, Hickey FJ, Schorry EK, Hopkin RJ, Wylie M, Narayan T, Glauser TA, Gilbert DL, Hershey AD, Sharp FR (2004c) Blood expression profiles for tuberous sclerosis complex 2, neurofibromatosis type 1, and Down’s syndrome. Ann Neurol 56:808–14 Tibshirani R, Hastie T, Narasimhan B, Chu G (2002) Diagnosis of multiple cancer types by shrunken centroids of gene expression. Proc Natl Acad Sci USA 99:6567–72 Tschopp J, Martinon F, Burns K (2003) NALPs: a novel protein family involved in inflammation. Nat Rev Mol Cell Biol 4:95–104 Ueda T, Hatakeyama T, Sakaki S, Ohta S, Kumon Y, Uraoka T (1995) Changes in coagulation and fibrinolytic system after local intra-arterial thrombolysis for acute ischemic stroke. Neurol Med Chir (Tokyo) 35:136–43 Unnamed (2001) Use of anti-ICAM-1 therapy in ischemic stroke: results of the Enlimomab Acute Stroke Trial. Neurology 57:1428–34 Viemann D, Strey A, Janning A, Jurk K, Klimmek K, Vogl T, Hirono K, Ichida F, Foell D, Kehrel B, Gerke V, Sorg C, Roth J (2004) Myeloid-related protein 8 and 14 induce a specific inflammatory response in human microvascular endothelial cells. Blood 105: 2955–62 Vila N, Castillo J, Davalos A, Chamorro A (2000) Proinflammatory cytokines and early neurological worsening in ischemic stroke. Stroke 31:2325–9 Vogl T, Propper C, Hartmann M, Strey A, Strupat K, van den Bos C, Sorg C, Roth J (1999) S100A12 is expressed exclusively by granulocytes and acts independently from MRP8 and MRP14. J Biol Chem 274: 25291–6 Zheng Z, Yenari MA (2004) Post-ischemic inflammation: molecular mechanisms and therapeutic implications. Neurol Res 26:884–92

Supplementary Information accompanies the paper on Journal of Cerebral Blood Flow website (http://www.nature.com/jcbfm)

Journal of Cerebral Blood Flow & Metabolism (2006) 26, 1089–1102