Altered gene expression profiles in stable versus acute childhood asthma

Information

  • Patent Application
  • 20070141585
  • Publication Number
    20070141585
  • Date Filed
    December 21, 2005
    18 years ago
  • Date Published
    June 21, 2007
    17 years ago
Abstract
Differences in gene expression in control and asthma patients to profile, differentiate, evaluate, etc. patients with exacerbated asthma and stable asthma.
Description

In accordance with 37 CFR §§ 1.77, 1.821-1.824, and 1.52(e), applicants state that the Sequence Listing for this application is submitted only in electronic form on a CD-ROM and herein incorporate by reference the entire Sequence Listing contained on the CD-ROM. The only material on the CD-ROM is the Sequence Listing for the instant patent application, which was created on Dec. 20, 2005 and has 616 KB. Applicants further state that the Sequence Listing information recorded and submitted herein in computer readable form is identical to the written (CD-ROM) sequence listing.


FIELD OF THE INVENTION

The invention is directed to asthma gene expression profiles.


BACKGROUND

Asthma is the most common chronic disease of childhood and has a strong genetic component. Microarray technology has been used previously to identify gene profiles associated with asthma but were limited to adult patients and to RNA derived from peripheral blood mononuclear cells. Because asthma most often begins in childhood, genes identified in adults may not represent genes important for asthma development.


SUMMARY OF THE INVENTION

Microarray technology was applied to childhood asthma. Gene profiles, also referred to herein as signatures, associated with childhood stable and exacerbated asthma, also referred to herein as acute asthma, were determined. It was also determined whether the same genes induced during stable asthma were expressed during asthma exacerbations, or whether a distinct set of genes were activated during an asthma exacerbation. Microarray technology in a group-averaged approach, and confirmatory reverse transcription polymerase chain reaction (RT-PCR) at an individual patient level, provided global gene expression profiles in respiratory epithelial cells derived from nasal respiratory epithelial cells in normal and asthmatic children.


Children with stable asthma (asthma-S), children experiencing an asthma exacerbation (asthma-E), and non-asthmatic children were evaluated. RNA was prepared from nasal respiratory epithelial cells isolated from each child, initially analyzed as pooled samples from the three groups. Further validation was performed on individual patient samples using microarrays and RT-PCR.


Distinct gene clusters were identifiable in individual and pooled asthma-S and asthma-E samples. Asthma-E samples demonstrated the strongest and most reproducible signatures, with 314 genes of 34,886 measured as Present on the chip, demonstrating induction or repression of greater than two-fold with p<0.05 in each of four individual samples. Asthma-S-regulated genes encompassed genes that overlapped with those of Asthma-E, but were fewer (166) and less consistent with respect to their behavior across the Asthma-E patient samples. Independent gene expression signatures were reflective of cells and genes poised or committed to activation by an asthma attack.


The information is useful for asthma diagnosis, evaluation of status and treatment response, and design of prophylaxis and therapy. These and other advantages will be apparent in light of the following figures, tables, and detailed description.




BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows hierarchical clustering and relative expression of genes highly expressed in childhood asthma.



FIG. 2 shows quantitative reverse transcription polymerase chain reaction analysis of selected genes.



FIG. 3 shows hierarchical clustering of genes highly expressed in individual children with stable and acute asthma compared with controls.



FIG. 4 shows selected upregulated and downregulated genes compared with normal controls.




DETAILED DESCRIPTION

This application contains at least one drawing executed in color. A Petition under 37 C.F.R. § 1.84 requesting acceptance of the color drawings is filed separately on even date herewith. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.


Applicants incorporate by reference in its entirety Guajardo et al., Altered gene expression profiles in nasal respiratory epithelium reflect stable versus acute childhood asthma, J. Allergy Clin Immunol 2005; 115:243-53.


Healthy and asthmatic children attending the clinics and emergency department of Cincinnati Children's Hospital Medical Center (CCHMC) were evaluated. Asthmatic children were either stable, indicated by the substantial absence of wheezing, or exacerbated, indicated by wheezing. Asthma was diagnosed according to American Thoracic Society criteria. Participants were included in one of three groups: 1. Stable allergic asthma (asthma-S group, N=10) inclusion criteria: a. younger than 18 years of age, b. physician-diagnosed asthma currently stable (not wheezing), and c. positive skin prick testing to any of the allergens from an environmental panel that included dust mite, molds, cat, dog, feathers, weeds and ragweed, tree pollens and grass allergen extracts (Hollister-Stier Laboratories, Spokane Wash.); 2. Exacerbation of asthma (asthma-E group, N=10) included children acutely wheezing with similar inclusion criteria to the asthma-S group with the exception of skin test positivity (skin testing was not performed in this group because it could further deteriorate the acute status of asthma); and 3. Healthy children (control group, N=10) inclusion criteria: a. younger than 18 years of age, b. healthy with no acute infections or major chronic illnesses, and c. negative results to the above-described environmental skin test prick panel. Exclusion criteria to participate in the study included a. 18 years of age or older, b. the use of nasal or systemic steroids within the last 30 days, c. nasal malformations/tumors, and d. acute infectious disease present in the asthma-S or control groups. Children with a concurrent diagnosis of allergic rhinitis were excluded if they had used nasal steroids within 30 days. The use of inhaled steroids was not interrupted for this study. Skin prick testing was performed in the asthma-S and control groups using DermaPiks (Greer Laboratories, Lenoir N.C.). Histamine (1 mg/ml) and normal saline (0.9% NaCl) were used as positive and negative controls. Reactions were considered positive if there was an erythematous base with a wheal ≧3 mm in diameter.


Nasal mucosa sampling was performed using a CytoSoft Brush (Medical Packaging Corp., Camarillo Calif.) and the sample was immediately taken to the laboratory for processing. Samples from children in the group experiencing an asthma exacerbation (asthma-E) were taken within one hour of arrival to the emergency room and before any steroids were given. The cells were suspended in phosphare buffered saline (PBS) and an aliquot was stained with Diff-Quick (Dade Behring Inc, Newark Del.). Cell counting was performed in five high power fields (hpf) and the relative percentages of cell types were calculated. A total eosinophil cell count was also performed.


RNA was isolated from the nasal mucosa sample using TRIZOL according to manufacturer instructions (TRIZOL Reagent, Invitrogen Corporation, Carlsbad Calif.). Average RNA yield was 42.6 μg. In one embodiment, two micrograms of RNA from each subject were pooled to form a group sample containing 20 μg. The three samples (Control, Asthma-S, and Asthma-E) were then submitted to the Affymetrix Genechip Core Facility at Cincinnati Children's Hospital Medical Center for processing and microarray hybridization using the HG-U133A GeneChip (Affymetrix, Santa Clara Calif.) according to Affymetrix guidelines. The HG-U133A chip microarray had a total of 22,215 probe sets (excluding controls) that identified 14,285 genes of which 12,735 are known. In addition, four individual samples from each group (Control, Asthma-S and Asthma-E) were submitted to the Affymetrix Genechip Core Facility at CCHMC for microarray hybridization using the HG-U133_plus 2 GeneChip (Affymetrix) according to Affymetrix guidelines. The HG-U133_plus 2 chip microarray had a total of 54,675 probe sets.


Scanned output files were analyzed using Microarray Suite 5.0 software (Affymetrix). From cell image data files, gene transcript levels were estimated as the Signal strength using the MAS5.0 (Affymetrix). Global scaling was performed to compare genes from chip to chip. Arrays were scaled to the same target intensity value (Tgt=1500) per gene and analyzed independently.


Second-stage data analyses were performed using GeneSpring software (Silicon Genetics, Redwood City Calif.). Initially, data from pooled asthma-S and asthma-E groups were analyzed by looking for genes that were most different in expression relative to the control group sample. A total of 299 cDNAs corresponding to genes that were three-fold up- or down-regulated in the asthma-S or asthma-E groups when compared to the control group were identified. These 299 highly expressed cDNAs were clustered according to their expression profiles along the GI A-P axis by using hierarchical clustering algorithms as implemented in the GeneSpring program. Clustering of the dataset was by several different normalization methods. The use of raw ratios of hybridization versus reference or the log2 of these ratios provided an assessment of genes based on their levels of expression. These 299 highly expressed cDNAs were clustered according to their relative expression profiles using normalization of raw ratios or the log2 of these ratios to the expression of the control group. The number of immune-related genes was obtained for each cluster.


GeneChip data from individual RNA samples were examined by a statistical approach designed to test the hypothesis that genes whose expression was altered in the individual samples would parallel that observed in the pooled samples. Next-generation human HG-U133-plus2 GeneChips (human HG-U133_plus2) were analyzed using MicroArraySuite 5 and the resulting GeneChip intensities were exported to GeneSpring 7.0 and normalized to the median expression level among the four control samples. Genes whose expression varied according to diagnostic group were obtained by first selecting genes that the Affymetrix algorithm reported to be “Present” in at least two gene chips. This returned 32,435 genes from the 54,675 on the chip. Next, an approximately normal distribution of gene expression values was generated by representing each gene's expression as the log of its expression Signal as measured by Microarray Suite. ANOVA was applied to the conditions with a probability of less than 0.01 (acute vs. stable vs. control) to obtain genes differentially expressed between conditions in at least three out of five chips. Each of these lists were further filtered for median expression being at least two-fold different between the two conditions. The resulting gene list of 1378 genes was then subjected to cluster analysis using Standard Correlation as implemented in GeneSpring.


Gene specific primers (designed using Beacon Designer software) were chosen to span at least one intron in the genomic sequence to enable the mRNA-derived product to be distinguished from any possible contaminating genomic product. The sequences of primers for the target genes were as follows: lymphotactin (NM003175) AATCAAGACCTACACCATCAC SEQ ID NO: 1 (sense) and TTCCTGTCCATGCTCCTG SEQ ID NO: 2 (anti-sense); histamine 4 (H4) receptor (NM21624) GGTGTGATCTCCATTCCTTTG SEQ ID NO: 3 (sense) and GCCACCATCAGAGTAACAATC SEQ ID NO: 4 (anti-sense); retinoic acid receptor (RARα) (hCT2294851) AGGAGACTGAGATTAGC SEQ ID NO: 5 (sense) and AAGAAGAAGGCGTAGG SEQ ID NO: 6 (anti-sense); CXCL11 (NM005409) GCTACAGTTGTTCAAGGCTTCC SEQ ID NO: 7 (sense) and TTGGGATTTAGGCATCGTTGTC SEQ ID NO: 8 (anti-sense); and ubiquitin C (UBC) (M26880) ATTTGGGTCGCGGTTCTTGSEQ ID NO: 9 (sense) and TGCCTTGACATTCTCGATGGT SEQ ID NO: 10 (anti-sense). Prior to cDNA synthesis (using SuperScript U, RNase H, Invitrogen), 1-2 ug of each RNA sample was pretreated with DNase I (Invitrogen) to eliminate any potential contaminating genomic DNA. RT-PCR analysis was conducted with the iCycler (Bio-Rad) using the “iQ SYBR Green Supermix” Taq polymerase mix (Bio-Rad). The amount of double-stranded DNA product, indicated by SYBR Green fluorescence, was measured at the end of each extension cycle. The relative message levels of each target gene were normalized to the UBC housekeeping gene.


Statistical differences between the relative expression levels of RARα, H4R, CXCL11, and lymphotactin genes among the different groups were determined by ANOVA (one-way) of the means and standard error values. This was followed by the Bonferroni procedure to allow for multiple comparisons. A p-value of <0.05 was considered significant.


The mean age in years for the Control (n=10), Asthma-S (n=10), and Asthma-E (n=10) group subjects was 11.7 (SD±2.3), 11.4 (SD±3.4), and 10.1 (SD±6.17) respectively. The gender (male:female) and race (AfricanAmerican:Caucasian) ratios were 7:3 and 8:2 for the control group; 7:3 and 5:5 for the Asthma-S group, and 6:4 and 9:1 for the Asthma-E group. There was no statistical difference between the groups. The children in the asthma-S and asthma-E groups were predominantly African American males, which agreed with published data regarding the racial and gender distributions of childhood asthma in urban environments.


The cellular composition of the nasal respiratory epithelial sample was determined for each subject. For the control group, the average number of cells/hpf (400×) was 265 (SD±104) with 97.7% epithelial cells, 1.84% polymorphonuclear leukocyte cells (PMN), 0.36% squamous cells, and 0.07% eosinophils. For the Asthma-S group the average number of cells/hpf was 219 (SD±87) with 96.3% epithelial cells, 3.32% PMN, 0.30% squamous cells, and 0.09% eosinophils; and for the Asthma-E group the average number of cells/hpf was 154 (SD±69) with 92.3% epithelial cells, 7.24% PMN, 0.18% squamous cells, and 0.25% eosinophils. Epithelial cells represented greater than 92% of the total cells isolated in all groups. There were higher percentages of PMN and eosinophils in samples from children experiencing an asthma exacerbation, however, they remained minor populations (7.2% and 0.25%, respectively) compared with the percentage of respiratory cells (92.3%) and the differences were not statistically significant. Overall, a lower average of total cells was recovered from the nasal samples of children experiencing an asthma exacerbation. While not being bound by a specific theory, this may be due to excessive mucus. RNA was isolated from each sample with an average yield of 42.6 μg per sample from one nostril. Two μg of RNA were pooled from each subject in each group and the three pools were subjected to microarray analysis (HG-U133A Affymetrix GeneChip).


In the asthma-S and asthma-E pooled groups, the expression of 253 (2.0%) of the known genes changed by at least three-fold in either group. The mean raw expression of these genes was 2076 (SD 4322) with a range of 17.4 to 67068 and a median and mode of 1192 and 1903, respectively.


The microarray data are summarized in FIG. 1. Hierarchical clustering and relative expression of genes highly expressed in childhood asthma are shown in panel A. Colors are graded to indicate increased (red) or decreased (blue) expression relative to reference. Relative expression in a given cluster is shown in panel B. The y-axis represents expression normalized to control expression. Cluster analysis examining gene profiles revealed eight distinct clusters of genes regulated in stable and acute childhood asthma. The genes in cluster or group A (N=33) were similarly upregulated in both asthma-S and asthma-E. In cluster B (N=55), genes were upregulated in asthma-S, and further induced in asthma-E. Cluster C genes (N=25) were unchanged in asthma-S, but upregulated in asthma-E. Cluster D genes (N=77) were downregulated in asthma-S, but unchanged in asthma-E. Cluster E genes (N=31) were upregulated in asthma-S, but unchanged in asthma-E. Cluster F genes (N=4) were upregulated in asthma-S, but downregulated in asthma-E. Cluster G genes (N=35) were unchanged in asthma-S, but downregulated in asthma-E. Cluster H genes (N=39) were downregulated in both asthma-S and asthma-E.


Cluster A (N=33), representing genes similarly upregulated in both asthma-S and asthma-E, contained 32 known genes (0.25% of the total known genes). Of these genes, 27.3% were immune-related and 21.2% were involved in signal transduction. The genes in Cluster B that were upregulated during asthma exacerbations (asthma-E) to a higher extent than in asthma-S were comprised of 43.6% immune-related genes. The genes in Cluster C that were upregulated during asthma exacerbations (asthma-E), but unchanged in stable asthma, were comprised of 44% immune-related genes. Clusters D-H were each comprised of less than 6.5% immune-related genes. Genes in Cluster E that were upregulated in children with stable asthma, but unchanged during asthma exacerbations, included 6.5% immune related genes. The genes in this profile included mainly signal transduction genes and cell function enzymes. Clusters D, F, G and H, which contain genes that were downregulated, consist largely of genes involved in basic cell functions and unknown genes.


Distinct clusters of genes that were differentially regulated in childhood asthma were identified, as shown in Tables 1-14. Distinct sets of genes were activated during stable vs. exacerbated asthma, establishing that exacerbated asthma status is distinguished based on the occurrence of strong gene expression signatures in nasal epithelial samples. Stable asthma status also exhibited differential signatures but with more variability. While not bound by any theory, this may suggest clinical and or mechanistic heterogeneity among the patients.


In the exacerbated asthma pooled sample, 12 genes were upregulated at least two-fold (Table 15) and 50 genes were upregulated at least three-fold (Table 16) as compared to the control pooled sample. Fourteen genes in the exacerbated asthma pooled sample were downregulated by at least two-fold (Table 17) and 7 genes were downregulated by at least three-fold (Table 18), compared to the control pooled sample. In the stable asthma pooled sample, 7 genes were upregulated by at least two-fold (Table 19) and 11 genes were upregulated by at least three-fold (Table 20) compared to the control pooled sample. Also, 6 genes in the stable asthma pooled sample were downregulated by at least two-fold (Table 21) and 4 genes were downregulated by at least three-fold (Table 22) compared to the control pooled sample.


Reverse transcription polymerase chain reaction (RT-PCR) analysis confirmed expression of genes identified by microarray. For the microarray analysis, equivalent amounts of RNA were pooled from individuals in each group. All individual RNA samples isolated from nasal mucosal cells from participants from the control (N=10), asthma-S(N=10), and asthma-E (N=10) groups were analyzed by RT-PCR for expression of each gene. Four genes (CXCL11, RARα, H4R, and lymphotactin) were examined: one induced in the asthma-E group exclusively (Cluster C), one induced in the asthma-S group exclusively (Cluster E), and two simultaneously induced in both groups (Cluster A).


Quantitative RT-PCR analysis is shown in FIG. 2, with the y-axis in each graph representing relative message levels, normalized to the average of duplicate UBC message levels. RT-PCR confirmed increased expression of the selected genes in asthma-S and/or asthma-E. CXCL11 expression was increased in asthma-E to a greater extent than asthma-S. RARα was induced in asthma-S, but not asthma-E, and lymphotactin was induced equally in asthma-S and asthma-E. The RT-PCR data validated the genes identified by chip array and confirmed differential expression in the asthma-E and asthma-S groups.



FIG. 3 shows hierarchical clustering of genes highly expressed in individual children with stable and exacerbated asthma compared with controls, with colors graded to indicate increased (red) or decreased (blue) expression relative to reference. The clustering results from the individual samples are presented beside the data from the RNA samples pooled from each group (N=10). As shown, the data from the individual samples were substantially consistent both between individuals and when compared to the pooled sample data. A stepwise filtering method was used to derive a total of 161 genes whose expression was significantly different between the groups (ANOVA, p<0.01), and in addition was of sufficient magnitude to increase or decrease by at least two-fold in 3 of the 4 individual samples.


In the exacerbated asthma individual group (versus the pooled group), 88 genes were upregulated and 53 genes were downregulated compared to the control group. In the stable asthma group, 21 genes were upregulated and 12 genes were downregulated. More specifically, in the exacerbated asthma group, 9 genes exhibited at least a two-fold increase (Table 1) and 79 genes showed at least a three-fold increase (Table 2) compared to controls. Also, 36 genes were decreased by at least two-fold (Table 3) and 15 genes were decreased by at least three-fold (Table 4) compared to controls. In the stable asthma group, 11 genes were upregulated by at least two-fold (Table 5) and 10 genes were upregulated by at least three-fold (Table 6) compared to controls. Also, 8 genes were downregulated by at least two-fold (Table 7) and 4 genes were downregulated by at least three-fold (Table 8) compared to controls. Many of the changes in gene expression were specific to either stable or exacerbated asthma in that gene expression did not change in the other group or had the opposite change in expression. For example, 70 genes that exhibited at least a two-fold increase in the exacerbated asthma group showed no change of expression in the stable asthma group (Table 9). Also, 50 genes were downregulated by at least two-fold in the exacerbated asthma group but were unchanged in the stable asthma group (Table 10). For stable asthma specific genes, 4 genes were upregulated by at least two-fold while unchanged in the exacerbated asthma group (Table 11) and 9 genes were downregulated by at least two-fold in the stable asthma group and remained unchanged in the exacerbated asthma group (Table 12). In some cases, the direction of change in gene expression was the same for both the stable and exacerbated asthma groups. For example, 16 genes showed at least a two-fold increase in expression in both groups compared to control groups (Table 13). Four genes exhibited an inverse in expression levels, with a gene upregulated by at least two-fold in the exacerbated asthma group and downregulated by at least two-fold in the stable asthma group, or vice versa (Table 14).


Among the 161 most upregulated and downregulated genes (at least three-fold change, p<0.01) that were consistent in at least 3 of the 4 samples, classes of genes were evaluated to determine if a particular class of genes was overrepresented. Two classes of genes were noted as shown in FIG. 4. Selected upregulated immune-related genes (A) and downregulated cilia-related genes (B) compared with controls in at least 3 out of 4 samples. Among the upregulated genes, 37 immune-related genes were consistently upregulated at least three-fold (FIG. 4A). Among the downregulated genes, 9 cilia-related genes were consistently downregulated at least three-fold (FIG. 4B).


Respiratory epithelial cells serve as an accessible alternative proxy for lower respiratory epithelium, even if they may not fully represent the genes that are expressed in the lungs of children with asthma. Many of the genes that were found to be induced in childhood asthma have been implicated in the pathogenesis of asthma in other studies, including arginase, SOCS-3, complement 3a receptor, and lymphotactin. For the chip array analysis, both pooled samples derived from equivalent amounts of RNA from each individual in each group, as well as individual samples, were utilized. The gene expression signatures obtained from the individual samples agreed with the pooled samples. RT-PCR of the individual RNA samples further validated findings and RT-PCR confirmed the chip array data.


The percentage of immune related genes was examined in each cluster. In the pooled samples, clusters that included genes induced specifically in either asthma exacerbations (Cluster C) or genes that were induced at a higher level during asthma exacerbations (Cluster D) contained the highest percentages and absolute numbers of immune-related genes. This was confirmed by further chip array analyses of the individual samples. The immune-related genes were overrepresented among the most upregulated genes. Genes that are not classified as immune genes may have direct or indirect effects on the immune system. Asthma-E samples demonstrated the strongest and most reproducible signatures and these signatures were distinct from Asthma-S. Among the most downregulated genes, cilia-related genes were overrepresented. Because the respiratory epithelium is often damaged in asthma and there is an overproduction of mucus, one might predict that genes important in ciliary function would be induced. While not being bound by any theory, downregulation in cilia-related genes may contribute to asthma pathogenesis by impairing mucus clearance. Alternatively, downregulation of cilia genes may be a response to damage and may be used for repair or remodeling.


Each cluster identifies novel potential target candidate genes for childhood asthma. In Cluster A, the H4 receptor gene was induced nearly ten-fold. This gene was recently cloned and found to be expressed in leukocytes, including eosinophils, as well as the lung and it is involved in childhood asthma. In contrast, the H1, H2, and H3 receptors were not induced. Also in Cluster A, SOCS-3 was induced nearly nine-fold. SOCS-3 expression correlates strongly with the pathology of asthma and atopic dermatitis, as well as serum IgE levels in allergic human patients. The complement 3α receptor 1 gene SEQ ID NO: 65 in Table 2 was induced. In a previous study examining the role of C3α in asthma, C3α levels were increased following segmental airway challenge in sensitized adults with asthma, and the receptor is also regulated during the effector phase of asthma. Several IFN-induced proteins were also induced in this cluster of genes induced in asthma-E to a greater level than asthma-S. Because asthma exacerbations in children can be associated with upper respiratory viral infections, some of these may represent an IFN-mediated anti-viral response. Genes that were induced exclusively during asthma exacerbations (Table 9) included integrin α4 as well as several chemokines and chemokine receptors. Integrin α4 (CD49d), which is important for eosinophil survival and recruitment, was induced 8.6 fold in children experiencing asthma exacerbation, but not in children with stable asthma. In contrast, genes that were induced in asthma-S, but not asthma-E (Table 11) did not include chemokine receptors nor chemokines. The most strongly induced gene in this cluster was the gene encoding RARα, which was induced approximately 28-fold compared to non-asthmatic children. Retinoids exert multiple effects upon lung differentiation and growth, and this receptor may contribute to lung repair or remodeling in children with ongoing stable asthma. Another gene in this cluster is arginase, supporting its roles as a mediator of childhood asthma. In a recent study, adults with stable asthma had increased arginase expression in their lungs and BALF compared with normal controls. In a previous study utilizing microarray analysis to identify genes important in asthma RNA isolated from peripheral blood mononuclear cells from adults with atopic asthma, allergic rhinitis but not asthma, and healthy controls was analyzed. Decreased levels of interferon α/β receptor (ratio 0.42) were found, similar to results in Cluster H.


Although environmental factors likely contribute to the different prevalence rates, genes with large allele frequency differences between a Caucasian population and a Han Chinese population may be partly responsible for the current variation in asthma susceptibility. One-hundred and sixty-one known genes identified by microarray data were examined for large allele frequency differences between Caucasian and Han Chinese populations. Allele frequencies of the single nucleotide polymorphisms (SNPs) within each gene in Caucasians and in Han Chinese were retrieved from the public HapMap database (http://www.hapmap.org). FST was calculated as FST2/pq, where σ2 is the variance in allele frequency, and p and q are the average allele frequency of each allele among subpopulations, respectively. When comparing two subpopulations with two alleles in each, the variance in allele frequency equals to a 2=(p1−p2)2/4, where p1 is the allele frequency in the first subpopulation, and p2 is the frequency of the same allele in the second subpopulation. Therefore, the FST was calculated by FST=(p1−p2)2/(4p(1−p)). The sample size obtained from HapMap project was large, i.e., 60 and 45 unrelated Caucasians and Han Chinese were genotyped, respectively, and allowed for calculation of FST without additional corrections. FST has a theoretical minimum of 0, indicating no genetic divergence, and a theoretical maximum of 1, indicating fixation for alternative alleles in different subpopulations. The observed FST is usually much less than 1 in human subpopulations. The following qualitative guidelines were used for the interpretation of FST: 0<FST<0.05: little genetic differentiation; 0.05<FST<0.15: moderate genetic differentiation; 0.15<FST<0.25: great genetic differentiation; and FST>0.25: very great genetic differentiation.


The FST for each identified SNPs (from HapMap database) was calculated in each of the 161 most regulated genes related to childhood asthma based on the gene expression profile comparisons. Among the asthma signature genes from the chip array results, there were 43 genes (39%) with large allele frequency differences (FST>0.15) between Caucasian and Han Chinese populations. Of these 43 genes, 17 had a FST>0.25 (Table 23) and 26 had a 0.15>FST>0.25 (Table 24). This proportion is higher than the average genetic differentiation between these two subpopulations.


Among these 43 genes, six genes (PDE4B SEQ. ID NO. 27; SPRR2B SEQ. ID NO. 109; ADCY2 SEQ. ID NO. 46; KIF3A SEQ. ID NO. 80; DNAH5 SEQ ID NO. 115; and PLAUSEQ. ID NO. 98) are located in chromosomal regions that have been linked to asthma phenotypes and atopy phenotypes and have been shown to either be regulated during allergic inflammation and/or to regulate release of Th2 cytokines including IL-13. These six genes have been shown to either be regulated during allergic inflammation and/or to regulate release of Th2 cytokines including IL-13. The following summarizes of each gene and its relationship to allergic inflammation and IL-13.


Phosphodiesterase 4B (PDE4B): The cyclic nucleotides, cAMP and cGMP, are important second messengers known to control many cellular processes. In inflammatory cells, activation of cAMP signaling has negative modulatory effects on numerous steps required for immune inflammatory responses, including T cell activation and proliferation, cytokine recruitment and recruitment of leukocyte. The cyclic nucleotide signaling system is complex and interlinked with many other pathways. Their signals are tightly controlled by regulating the synthesis and breakdown of these molecules. Phosphodiesterases are the enzymes that degrade and inactivate cyclic nucleotides. The phosphodiesterase 4 (PDE4) family consists of four genes (PDE4A-D) and each gene encodes multiple variants generated from alternate splicing and different transcriptional promoters. The phenotypes of the different PDE4 null mice support unique functions for each PDE4 gene. PDE4B null mice are generally healthy, but peripheral blood leukocytes derived from PDE4B null mice produce very little TNFα in response to LPS. Extensive studies using specific PDE4B inhibitors both in vitro and in vivo have demonstrated a potent anti-inflammatory effect as well as regulation of airway smooth muscle by PDE4B. Given the broad inhibitory effects, pharmacologic manipulation of PDE4 is a promising approach for treating chronic inflammatory conditions including asthma. In animal models of asthma, PDE4 inhibitors have been shown to inhibit airway inflammation and remodeling. A key feature of chronic inflammatory airway diseases such as asthma is mucus hypersecretion. MUC5AC is the predominant mucin gene expressed in healthy airways and is increased in asthmatic patients. Selective PDE4 inhibition was shown to be effective in decreasing EGF-induced MUC5AC expression in human airway epithelial cells. In a placebo-controlled, randomized clinical trial, PDE4 inhibition was found to be efficacious in exercise-induced asthma; the mean percentage fall of FEV1 after exercise was reduced by 41% as compared to placebo.


One mechanism by which PDE4 inhibitors exert an anti-inflammatory effect is by inhibiting IL-13 production in allergic diseases by T cells and basophils. In one study, phytohaemagglutinin (PHA)-induced IL-13 release from peripheral blood mononuclear cells from atopic asthma patients was inhibited by rolipram, a PDE4 inhibitor. In another study, rolipram inhibited IL-13 production from PHA- or anti-CD3 plus anti-CD28-stimulated human T cells. Similarly, PDE4 inhibition blocked Dermatophagoides pteronyssinus-induced interleukin-13 secretion in atopic dermatitis T cells.


Small proline-rich protein 2B (SPRR2B): SPRR genes encode a class of small proline rich proteins that are strongly induced during differentiation of human epidermal keratinocytes in vitro and in vivo. They are encoded by closely related members of a gene family closely linked within a 300-kb DNA segment on human chromosome 1q21-q22 in a region that has been linked to atopy phenotypes. These genes are expressed predominantly in squamous epithelium, where they contribute to the formation of the insoluble cornified crosslinked envelope that provides structural integrity and limits permeability through transglutaminase-induced N-glutamyl)lysine isopeptide crosslinks and interchain disulfide bonds. Studies using primary human and murine cells grown at the air-liquid interface demonstrated that IL-13 directly induces SPRR2B. In fact SPRR2B was one of only four genes that was induced more strongly by IL-13 than IL-4. The fact that SPRR2B was strongly induced by IL-13 supported that it may be important in the pathogenesis of allergic disease. This role for SPRR2b substantiated in a recent study by Drs. Rothenberg and Wills-Karp where SPRR2B was shown to be induced in lungs of mice in a mouse model of asthma in a Stat6-dependent fashion. Thus, SPRR2 is an allergen- and IL-13-induced gene in experimental allergic responses that may be involved in disease pathophysiology.


Adenylate Cyclase 2 (ADCY2): This gene encodes a member of the family of adenylate cyclases, which are membrane-associated enzymes that catalyze the formation of the secondary messenger cyclic adenosine monophosphate (cAMP). This enzyme is insensitive to Ca(2+)/calmodulin, and is stimulated by the G protein beta and gamma subunit complex. It is located on chromosome 5p15 in a region that has been linked to atopy phenotypes. Cyclic AMP has a broad range of anti-inflammatory effects on a variety of effector cells involved in asthma. Pharmacologic inhibition of adenylate cyclases inhibited IL-13 production from PHA- or anti-CD3 plus anti-CD28-stimulated human T cells.


Kinesin family member 3A (KIF3A): KIF3 is a heterotrimeric member of the kinesin superfamily of microtubule associated motors. This functionally diverse family of proteins mediates transport between the endoplasmic reticulum and the Golgi, transports protein complexes within cilia and flagella, and is involved in anterograde transport of membrane bound organelles in neurons and melanosomes. Embryos lacking KIF3A die at 10 days postcoitum, exhibit randomized establishment of L-R asymmetry, and display numerous structural abnormalities. Interestingly, the KIF3A gene is located on 5q31 in a region that has demonstrated linkage to asthma and atopy in multiple studies. It is located immediately upstream of IL4 and IL13 and conserved non-coding sequences in this area have been implicated in the coordinate transcriptional regulation of IL-4 and IL-13.


Dynein, axonemal, heavy polypeptide 5 (DNAH5): DNAH5 is a component of the outer dynein arm of cilia. Mutations in DNAH5 resulting in non-functional proteins have been found to be responsible for primary ciliary dyskinesia. Similar to ADCY2, the DNAH5 gene is located on chromosome 5p15 in a region that has been linked to atopy phenotypes. IL-13 has direct effects on ciliary function, specifically it alters mucociliary differentiation and decreases ciliary beat frequency of ciliated epithelial cells.


Plasminogen activator, urokinase (PLAU): PLAU is located on chromosome 10q24 and its gene product, urokinase, is serine protease involved in degradation of the extracellular matrix. Urokinase converts plasminogen to plasmin by specific cleavage of an Arg-Val bond in plasminogen. Recent studies suggest that the plasmin system plays an active role in tissue remodeling by influencing the production of inflammatory mediators and growth factors. Plasmin also degrades the extracellular matrix (ECM), either directly removing glycoproteins from ECM or by activating matrix metalloproteinases (MMPs). Urokinase is synthesized by airway cells, and inflammatory mediators affect its expression. In mouse models of asthma urokinase is induced in the lungs of mice, and in monocytes urokinase is induced in response to IL-4 and IL-13. The role of urokinase in inflammation was further defined in a recent study whereby urokinase gene-targeted mice fail to generate a Th2 immune response following schistosomal antigen challenge. The plasmin system is also involved in eotaxin-mediated chemotaxis of eosinophils.


Each of the gene products is expressed in respiratory epithelium. None of these epithelial genes have been studied as potential candidate genes for asthma.


In summary, the distinct gene expression profiles in nasal respiratory epithelial cells of children with stable asthma (asthma-S) and children experiencing an asthma exacerbation (asthma-E) provided an overview of the genetic portrait of childhood asthma and the differences in genes important in promoting the development of asthma versus promoting the ongoing phenotype of asthma. Strong gene expression signatures that reflect clinical asthma attack status in readily sampled patient tissues provide a new opportunity for molecular sub-classification and clinical management of asthma patients. Both stabilized and acutely affected asthmatic children exhibited characteristic expression profiles that affect understanding of disease status, treatment response, and new therapies.

TABLE 1Individual samplesExacerbated Asthma two-fold increaseSeq. IDCommonDescriptionGenbankNumberEMR2egf-like module containing, mucin-like, hormoneNM_01344722receptor-like 2THBDthrombomodulinNM_00036123ZNF407zinc finger protein 407NM_01775731ALDH1A3aldehyde dehydrogenase 1 family, member A3NM_00069339OLR1oxidized low density lipoprotein (lectin-like) receptor 1NM_00254369SCELsciellinNM_14477777PLAUplasminogen activator, urokinaseNM_00265898DCP2decapping enzyme hDcp2NM_152624126SLC30A7solute carrier family 30 (zinc transporter), member 7NM_133496152









TABLE 2










Individual samples


Exacerbated Asthma three-fold increase













Seq. ID


Common
Description
Genbank
Number













KRT24
keratin 24
NM_019016
14


SOSTDC1
sclerostin domain containing 1
NM_015464
15


BM039
uncharacterized bone marrow protein BM039
NM_018455
16


KRT6A
keratin 6A
NM_005554
17


CALB1
calbindin 1, 28 kDa
NM_004929
18


GPR65
G protein-coupled receptor 65
NM_003608
20


FPRL1
formyl peptide receptor-like 1
NM_001462
24


PROK2
prokineticin 2
NM_021935
26


PDE4B
phosphodiesterase 4B, cAMP-specific (phosphodiesterase
NM_002600
27



E4 dunce homolog, Drosophila)


FOS
v-fos FBJ murine osteosarcoma viral oncogene homolog
NM_005252
28


TREM1
triggering receptor expressed on myeloid cells 1
NM_018643
29


HAL
histidine ammonia-lyase
NM_002108
30


SLC2A14
solute carrier family 2 (facilitated glucose transporter),
NM_153449
32



member 14


AQP9
aquaporin 9
NM_020980
33


BCL2A1
BCL2-related protein A1
NM_004049
34


CYP1A1
cytochrome P450, family 1, subfamily A, polypeptide 1
NM_000499
35


SERPINB4
serine (or cysteine) proteinase inhibitor, clade B
NM_002974
36



(ovalbumin), member 4


CLECSF6
C-type (calcium dependent, carbohydrate-recognition
NM_016184
40



domain) lectin, superfamily member 6


IL1B
interleukin 1, beta
NM_000576
41


LILRB1
leukocyte immunoglobulin-like receptor, subfamily B
NM_D06669
42



(with TM and ITIM domains), member 1


SLC2A3
solute carrier family 2 (facilitated glucose transporter),
NM_006931
43



member 3


S100A9
S100 calcium binding protein A9 (calgranulin B)
NM_002965
44


PLEK
pleckstrin
NM_002664
45


DTNA
dystrobrevin, alpha
NM_001390
47


ARNTL2
aryl hydrocarbon receptor nuclear translocator-like 2
NM_020183
48


RAI3
retinoic acid induced 3
NM_003979
49


GOS2
putative lymphocyte G0/G1 switch gene
NM_015714
50


RPEL1
RPEL repeat containing 1
AB051520
51


CSF2RB
colony stimulating factor 2 receptor, beta, low-affinity
NM_000395
52



(granulocyte-macrophage)


PLAUR
plasminogen activator, urokinase receptor
NM_002659
53


HIST1H2BH
histone 1, H2bh
NM_003524
54


EMP1
epithelial membrane protein 1
NM_001423
55


TF
transferrin
NM_001063
56


PRG1
proteoglycan 1, secretory granule
NM_002727
57



unnamed protein product; diaminopimelate



decarboxylase (AA 1-327); Bacillus subtilis lys gene for
X17013
58



diaminopimelate decarboxylase (EC 4.1.1.20).


PLEK
pleckstrin
NM_002664
59


GPR43
G protein-coupled receptor 43
NM_005306
60


HIST1H2BC
histone 1, H2bc
NM_003526
61


SPRR1A
small proline-rich protein 1A
NM_005987
62



integrin, alpha M (complement component receptor 3,
NM_000632
65


ITGAM
alpha; also known as CD11b (p170), macrophage antigen



alpha polypeptide)


SOD2
superoxide dismutase 2, mitochondrial
NM_000636
67


GPR97
G protein-coupled receptor 97
NM_170776
70


SPEC1
small protein effector 1 of Cdc42
NM_020239
72


MMP25
matrix metalloproteinase 25
NM_022468
73



synonyms: HIS, HSI, ARL1, ARL-1, ALDRLn, AKR1B11,



AKR1B12, MGC14103; aldose reductase-like 1; aldo-keto


AKR1B10
reductase family 1, member B11 (aldose reductase-like);
NM_004812
74



aldose reductase-like peptide; aldose reductase-related



protein; small intestine reductase; go



synonyms: HIS, HSI, ARL1, ARL-1, ALDRLn, AKR1B11,



AKR1B12, MGC14103; aldose reductase-like 1; aldo-keto


AKR1B10
reductase family 1, member B11 (aldose reductase-like);
NM_020299
75



aldose reductase-like peptide; aldose reductase-related



protein; small intestine reductase; go


EMP1
epithelial membrane protein 1
NM_001423
76


HM74
putative chemokine receptor
NM_006018
78


NR4A1
nuclear receptor subfamily 4, group A, member 1
NM_002135
79


HIST1H1C
histone 1, H1c
NM_005319
83




Homo sapiens transcribed sequences

BQ010718
84


SERPINB13
serine (or cysteine) proteinase inhibitor, clade B (ovalbumin), member 13
NM_012397
86


HIST1H2BK
histone 1, H2bk
NM_080593
88


IL1R2
interleukin 1 receptor, type II
NM_004633
90


DUSP5
dual specificity phosphatase 5
NM_004419
92


TLR4
toll-like receptor 4
NM_003266
93


CCR1
chemokine (C—C motif) receptor 1
NM_001295
94


RAI3
retinoic acid induced 3
NM_003979
96


KRT13
keratin 13
NM_153490
97




Homo sapiens transcribed sequences

BQ277484
101


SLC11A1
solute carrier family 11 (proton-coupled divalent metal
NM_000578
105



ion transporters), member 1


C1QR1
complement component 1, q subcomponent, receptor 1
NM_012072
106


IL1A
interleukin 1, alpha
NM_000575
107


SPRR2B
small proline-rich protein 2B
NM_001017418
109


SPRR3
small proline-rich protein 3
NM_005416
110


MAD
MAX dimerization protein 1
NM_002357
112


CLECSF12
C-type (calcium dependent, carbohydrate-recognition
NM_197953
114



domain) lectin, super-family member 12


SPRR1B
small proline-rich protein 1B (cornifin)
NM_003125
117


EAT2
SH2 domain-containing molecule EAT2
NM_053282
120


PRV1
polycythemia rubra vera 1
NM_020406
129


RAB35
RAB35, member RAS oncogene family
NM_006861
132


S100A2
S100 calcium binding protein A2
NM_005978
134


CD44
CD44 antigen (homing function and Indian blood group
NM_000610
135



system)


JUNB
jun B proto-oncogene
NM_002229
139


LILRB2
leukocyte immunoglobulin-like receptor, subfamily B
NM_005874
145



(with TM and ITIM domains), member 2


C4.4A
GPI-anchored metastasis-associated protein homolog
NM_014400
146


CPA4
carboxypeptidase A4
NM_016352
153


LILRB1
leukocyte immunoglobulin-like receptor, subfamily B
NM_006669
160



(with TM and ITIM domains), member 1


FCGR2A
Fc fragment of IgG, low affinity IIa, receptor for (CD32)
NM_021642
163







* Denotes a “corrected” accession number found in Genbank














TABLE 3










Individual samples


Exacerbated Asthma two-fold decrease













Seq. ID


Common
Description
Genbank
Number













SF1
splicing factor 1
NM_004630
11


KCTD7
potassium channel tetramerisation domain containing 7
NM_153033
12


DNAH7
dynein, axonemal, heavy polypeptide 7
NM_018897
63


FLJ23505

Homo sapiens cDNA: FLJ23505 fis, clone LNG03017

NM_024716
68


KIF3A
kinesin family member 3A
NM_007054
80


IGFBP7
insulin-like growth factor binding protein 7
NM_001553
81


TSAP6
tumor suppressor pHyde; Homo sapiens dudulin 2
NM_182915
82



(TSAP6), mRNA.


IPW

Homo sapiens mRNA; cDNA DKFZp686M12165 (from

BX648788
89



clone DKFZp686M12165)


PLTP
phospholipid transfer protein
NM_006227
91



gb: BC029442.1/DB_XREF = gi: 20809535/TID = Hs2Affx.1.373/



CNT = 1/FEA = FLmRNA/TIER = FL/STK = 1/



NOTE = sequence(s) not in UniGene/DEF = Homo sapiens,
BC029442
99



Similar to immunity associated protein 1, clone MGC: 32707



IMAGE: 4618467, mRNA, complete cds./PROD = Similar to immu


SIAH1
seven in absentia homolog 1 (Drosophila)
NM_003031
100




Homo sapiens transcribed sequences

BX107340
103


FLJ40427
hypothetical protein FLJ40427
NM_178504
108


DNAH5
dynein, axonemal, heavy polypeptide 5
NM_001369
115


DNAH9
dynein, axonemal, heavy polypeptide 9
NM_001372
116




Homo sapiens cDNA FLJ12411 fis, clone MAMMA1002964.

AK022473
118


LOC123872
similar to RIKEN cDNA 4930457P18
NM_178452
121


CHST5
carbohydrate (N-acetylglucosamine 6-O) sulfotransferase 5
NM_024533
122



UI-H-BW0-aim-d-12-0-UI.s1 NCI_CGAP_Sub6 Homo
AW294722
123




sapiens cDNA clone IMAGE: 2729902 3′, mRNA sequence.



KIAA1688
KIAA1688 protein
AB051475
125


DCDC2
doublecortin domain containing 2
NM_016356
127




Homo sapiens transcribed sequences

CD698727
128


SPAG8
sperm associated antigen 8
NM_012436
130


ATM
ataxia telangiectasia mutated (includes complementation
NM_000051
138



groups A, C and D)


FLJ13621
hypothetical protein FLJ13621
NM_025009
140


HPX
hemopexin
NM_000613
141


DNAI2
dynein, axonemal, intermediate polypeptide 2
NM_023036
143




Homo sapiens mRNA; cDNA DKFZp761D2417 (from clone

AL831856
148



DKFZp761D2417)

148


MGC16202
hypothetical protein MGC16202
NM_032373
156




Homo sapiens LOH11CR1P gene, loss of heterozygosity,

CA428747
157



11, chromosomal region 1 gene P product


LOC146177

Homo sapiens hypothetical protein LOC146177

NM_175059
158



(LOC146177), mRNA.


MGC40053
hypothetical protein MGC40053
NM_152583
161


LOC339005

Homo sapiens cDNA FLJ33935 fis, clone CTONG2017910.

AK091254
162



alternatively spliced; beta-1 form; possible membrane-


C18orf1
spanning protein; clone 22; Homo sapiens chromosome
NM_181481
164



18 open reading frame 1 (C18orf1), mRNA.


MYEF2
myelin expression factor 2
NM_016132
165


DNAH3
dynein, axonemal, heavy polypeptide 3
NM_017539
167
















TABLE 4










Individual samples


Exacerbated Asthma three-fold decrease













Seq. ID


Common
Description
Genbank
Number













CLGN
calmegin
NM_004362
13


ADCY2
adenylate cyclase 2 (brain)
NM_020546
46




Homo sapiens transcribed sequence with weak similarity




to protein sp: P39194 (H. sapiens) ALU7_HUMAN Alu
BX361062
66



subfamily SQ sequence contamination warning entry


MGC26733
hypothetical protein MGC26733
NM_144992
104


MYLK
myosin, light polypeptide kinase
NM_053025
111


SLC26A7
solute carrier family 26, member 7
NM_052832
119




Homo sapiens cDNA: FLJ22631 fis, clone HSI06451.

AK026284
124


LOC138428
hypothetical protein LOC286207
AL833241
133




Homo sapiens cDNA: FLJ22781 fis, clone KAIA1958.

AK026434
136




Homo sapiens cDNA FLJ12093 fis, clone HEMBB1002603.

AK022155
137




Homo sapiens cDNA: FLJ23502 fis, clone LNG02862

AK027155
142



UI-H-EZ1-bbk-j-02-0-UI.s1 NCI_CGAP_Ch2 Homo


LOC165186

sapiens cDNA clone UI-H-EZ1-bbk-j-02-0-UI 3′, mRNA

NM_199280
144



sequence.; ESTs, Weakly similar to T00057 hypothetical



protein KIAA0423 - human (fragment) [H. sapiens]



UI-E-EJ0-ahi-c-20-0-UI.r1 UI-E-EJ0 Homo sapiens cDNA
BM712946
149



clone UI-E-EJ0-ahi-c-20-0-UI 5′, mRNA sequence.


FLJ14665
Start codon is not identified.; Homo sapiens mRNA for
AB058767
155



KIAA1864 protein, partial cds.


SCARF2
scavenger receptor class F, member 2
NM_153334
159
















TABLE 5










Individual samples


Stable Asthma two-fold increase













Seq. ID


Common
Description
Genbank
Number













SOSTDC1
sclerostin domain containing 1
NM_015464
15


BM039
uncharacterized bone marrow protein BM039
NM_018455
16


CALB1
calbindin 1, 28 kDa
NM_004929
18


CYP1A1
cytochrome P450, family 1, subfamily A, polypeptide 1
NM_000499
35




Homo sapiens transcribed sequences

AI476722
37


EMP1
epithelial membrane protein 1
NM_001423
76


SCEL
sciellin
NM_144777
77



UI-H-BW0-aim-d-12-0-UI.s1 NCI_CGAP_Sub6 Homo
AW294722
123




sapiens cDNA clone IMAGE: 2729902 3′, mRNA sequence.



DCP2
decapping enzyme hDcp2
NM_152624
126


RAB35
RAB35, member RAS oncogene family
NM_006861
132


SLC30A7
solute carrier family 30 (zinc transporter), member 7
NM_133496
152
















TABLE 6










Individual samples


Stable Asthma three-fold increase













Seq. ID


Common
Description
Genbank
Number













FOS
v-fos FBJ murine osteosarcoma viral oncogene homolog
NM_005252
28


ZNF407
zinc finger protein 407
NM_017757
31


ZCCHC2
zinc finger, CCHC domain containing 2
NM_017742
38


ARNTL2
aryl hydrocarbon receptor nuclear translocator-like 2
NM_020183
48



unnamed protein product; diaminopimelate decarboxylase



(AA 1-327); Bacillus subtilis lys gene for diaminopimelate
X17013
58



decarboxylase (EC 4.1.1.20).


SPEC1
small protein effector 1 of Cdc42
NM_020239
72




Homo sapiens transcribed sequences

BQ277484
101


CBX5
chromobox homolog 5 (HP1 alpha homolog, Drosophila)
NM_012117
131


C4.4A
GPI-anchored metastasis-associated protein homolog
NM_014400
146


FLJ10525
hypothetical protein FLJ10525
NM_018126
154
















TABLE 7










Individual samples


Stable Asthma two-fold decrease













Seq. ID


Common
Description
Genbank
Number













BCL6
B-cell CLL/lymphoma 6 (zinc finger protein 51)
NM_138931
21


TREM1
triggering receptor expressed on myeloid cells 1
NM_018643
29


RPEL1
RPEL repeat containing 1
AB051520
51


DNCI1
dynein, cytoplasmic, intermediate polypeptide 1
NM_004411
85


EPOR
erythropoietin receptor
NM_000121
87


LILRB2
leukocyte immunoglobulin-like receptor, subfamily B
NM_005874
145



(with TM and ITIM domains), member 2


HLA-DPB1
major histocompatibility complex, class II, DP beta 1
NM_002121
147


KCNC3
potassium voltage-gated channel, Shaw-related subfamily,
NM_004977
166



member 3
















TABLE 8










Individual samples


Stable Asthma three-fold decrease













Seq. ID


Common
Description
Genbank
Number















Homo sapiens mRNA; cDNA DKFZp566D053 (from

AW611486
71



clone DKFZp566D053)


LOC253559
nectin-like protein 3
NM_153184
95


ZNF483
zinc finger protein 483
NM_007169
150


ZNF483
zinc finger protein 483
NM_133464
151
















TABLE 9










Individual samples


Exacerbated Asthma increases, Stable Asthma no change










Common
Description
Genbank
Seq. ID Number













KRT24
keratin 24
NM_019016
14


KRT6A
keratin 6A
NM_005554
17


GPR65
G protein-coupled receptor 65
NM_003608
20


EMR2
egf-like module containing, mucin-like, hormone receptor-
NM_013447
22



like 2


THBD
thrombomodulin
NM_000361
23


FPRL1
formyl peptide receptor-like 1
NM_001462
24


PROK2
prokineticin 2
NM_021935
26


PDE4B
phosphodiesterase 4B, cAMP-specific (phosphodiesterase
NM_002600
27



E4 dunce homolog, Drosophila)


HAL
histidine ammonia-lyase
NM_002108
30


SLC2A14
solute carrier family 2 (facilitated glucose transporter),
NM_153449
32



member 14


AQP9
aquaporin 9
NM_020980
33


BCL2A1
BCL2-related protein A1
NM_004049
34


SERPINB4
serine (or cysteine) proteinase inhibitor, clade B
NM_002974
36



(ovalbumin), member 4


ALDH1A3
aldehyde dehydrogenase 1 family, member A3
NM_000693
39


CLECSF6
C-type (calcium dependent, carbohydrate-recognition
NM_016184
40



domain) lectin, superfamily member 6


IL1B
interleukin 1, beta
NM_000576
41


LILRB1
leukocyte immunoglobulin-like receptor, subfamily B (with
NM_006669
42



TM and ITIM domains), member 1


SLC2A3
solute carrier family 2 (facilitated glucose transporter),
NM_006931
43



member 3


S100A9
S100 calcium binding protein A9 (calgranulin B)
NM_002965
44


PLEK
pleckstrin
NM_002664
45


DTNA
dystrobrevin, alpha
NM_001390
47


RAI3
retinoic acid induced 3
NM_003979
49


G0S2
putative lymphocyte G0/G1 switch gene
NM_015714
50


CSF2RB
colony stimulating factor 2 receptor, beta, low-affinity
NM_000395
52



(granulocyte-macrophage)


PLAUR
plasminogen activator, urokinase receptor
NM_002659
53


HIST1H2BH
histone 1, H2bh
NM_003524
54


EMP1
epithelial membrane protein 1
NM_001423
55


TF
transferrin
NM_001063
56


PRG1
proteoglycan 1, secretory granule
NM_002727
57


PLEK
pleckstrin
NM_002664
59


GPR43
G protein-coupled receptor 43
NM_005306
60


HIST1H2BC
histone 1, H2bc
NM_003526
61


SPRR1A
small proline-rich protein 1A
NM_005987
62



integrin, alpha M (complement component receptor 3,


ITGAM
alpha; also known as CD11b (p170), macrophage antigen
NM_000632
65



alpha polypeptide)


SOD2
superoxide dismutase 2, mitochondrial
NM_000636
67


OLR1
oxidised low density lipoprotein (lectin-like) receptor 1
NM_002543
69


GPR97
G protein-coupled receptor 97
NM_170776
70


MMP25
matrix metalloproteinase 25
NM_022468
73



synonyms: HIS, HSI, ARL1, ARL-1, ALDRLn, AKR1B11,
NM_004812
74



AKR1B12, MGC14103; aldose reductase-like 1; aldo-keto


AKR1B10
reductase family 1, member B11 (aldose reductase-like);
NM_020299*



aldose reductase-like peptide; aldose reductase-related



protein; small intestine reductase; go



synonyms: HIS, HSI, ARL1, ARL-1, ALDRLn, AKR1B11,



AKR1B12, MGC14103; aldose reductase-like 1; aldo-keto


AKR1B10
reductase family 1, member B11 (aldose reductase-like);
NM_020299
75



aldose reductase-like peptide; aldose reductase-related



protein; small intestine reductase; go


HM74
putative chemokine receptor
NM_006018
78


NR4A1
nuclear receptor subfamily 4, group A, member 1
NM_002135
79


HIST1H1C
histone 1, H1c
NM_005319
83




Homo sapiens transcribed sequences

BQ010718
84


SERPINB13
serine (or cysteine) proteinase inhibitor, clade B (ovalbumin), member 13
NM_012397
86


HIST1H2BK
histone 1, H2bk
NM_080593
88


IL1R2
interleukin 1 receptor, type II
NM_004633
90


DUSP5
dual specificity phosphatase 5
NM_004419
92


TLR4
toll-like receptor 4
NM_003266
93


CCR1
chemokine (C—C motif) receptor 1
NM_001295
94


RAI3
retinoic acid induced 3
NM_003979
96


KRT13
keratin 13
NM_153490
97


PLAU
plasminogen activator, urokinase
NM_002658
98


SLC11A1
solute carrier family 11 (proton-coupled divalent metal ion transporters), member 1
NM_000578
105


C1QR1
complement component 1, q subcomponent, receptor 1
NM_012072
106


IL1A
interleukin 1, alpha
NM_000575
107


SPRR2B
small proline-rich protein 2B
NM_001017418
109


SPRR3
small proline-rich protein 3
NM_005416
110


MAD
MAX dimerization protein 1
NM_002357
112


CLECSF12
C-type (calcium dependent, carbohydrate-recognition
NM_197953
114



domain) lectin, super-family member 12


SPRR1B
small proline-rich protein 1B (cornifin)
NM_003125
117


EAT2
SH2 domain-containing molecule EAT2
NM_053282
120


PRV1
polycythemia rubra vera 1
NM_020406
129


S100A2
S100 calcium binding protein A2
NM_005978
134


CD44
CD44 antigen (homing function and Indian blood group
NM_000610
135



system)


JUNB
jun B proto-oncogene
NM_002229
139


LILRB2
leukocyte immunoglobulin-like receptor, subfamily B (with
NM_005874
145



TM and ITIM domains), member 2


CPA4
carboxypeptidase A4
NM_016352
153


LILRB1
leukocyte immunoglobulin-like receptor, subfamily B (with
NM_006669
160



TM and ITIM domains), member 1


FCGR2A
Fc fragment of IgG, low affinity IIa, receptor for (CD32)
NM_021642
163







*corrected accession number in Genbank














TABLE 10










Individual samples


Exacerbated Asthma decreases, Stable Asthma no change













Seq. ID


Common
Description
Genbank
Number













SF1
splicing factor 1
NM_004630
11


KCTD7
potassium channel tetramerisation domain containing 7
NM_153033
12


CLGN
calmegin
NM_004362
13


ADCY2
adenylate cyclase 2 (brain)
NM_020546
46


DNAH7
dynein, axonemal, heavy polypeptide 7
NM_018897
63




Homo sapiens transcribed sequence with weak similarity to


66



protein sp: P39194 (H. sapiens) ALU7_HUMAN Alu subfamily
BX361062



SQ sequence contamination warning entry


FLJ23505

Homo sapiens cDNA: FLJ23505 fis, clone LNG03017

NM_024716
68


KIF3A
kinesin family member 3A
NM_007054
80


IGFBP7
insulin-like growth factor binding protein 7
NM_001553
81


TSAP6
tumor suppressor pHyde; Homo sapiens dudulin 2 (TSAP6),
NM_182915
82



mRNA.


IPW

Homo sapiens mRNA; cDNA DKFZp686M12165 (from clone

BX648788
89



DKFZp686M12165)


PLTP
phospholipid transfer protein
NM_006227
91



gb: BC029442.1/DB_XREF = gi: 20809535/TID = Hs2Affx.1.373/



CNT = 1/FEA = FLmRNA/TIER = FL/STK = 1/



NOTE = sequence(s) not in UniGene/DEF = Homo sapiens,
BC029442
99



Similar to immunity associated protein 1, clone MGC: 32707



IMAGE: 4618467, mRNA, complete cds./PROD = Similar to immu


SIAH1
seven in absentia homolog 1 (Drosophila)
NM_003031
100




Homo sapiens transcribed sequences

BX107340
103


MGC26733
hypothetical protein MGC26733
NM_144992
104


FLJ40427
hypothetical protein FLJ40427
NM_178504
108


MYLK
myosin, light polypeptide kinase
NM_053025
111


DNAH5
dynein, axonemal, heavy polypeptide 5
NM_001369
115


DNAH9
dynein, axonemal, heavy polypeptide 9
NM_001372
116




Homo sapiens cDNA FLJ12411 fis, clone MAMMA1002964.

AK022473
118


SLC26A7
solute carrier family 26, member 7
NM_052832
119


LOC123872
similar to RIKEN cDNA 4930457P18
NM_178452
121


CHST5
carbohydrate (N-acetylglucosamine 6-O) sulfotransferase 5
NM_024533
122




Homo sapiens cDNA: FLJ22631 fis, clone HSI06451.

AK026284
124


KIAA1688
KIAA1688 protein
AB051475
125


DCDC2
doublecortin domain containing 2
NM_016356
127




Homo sapiens transcribed sequences

CD698727
128


SPAG8
sperm associated antigen 8
NM_012436
130


LOC138428
hypothetical protein LOC286207
AL833241
133




Homo sapiens cDNA: FLJ22781 fis, clone KAIA1958.

AK026434
136




Homo sapiens cDNA FLJ12093 fis, clone HEMBB1002603.

AK022155
137


ATM
ataxia telangiectasia mutated (includes complementation
NM_000051
138



groups A, C and D)


FLJ13621
hypothetical protein FLJ13621
NM_025009
140


HPX
hemopexin
NM_000613
141




Homo sapiens cDNA: FLJ23502 fis, clone LNG02862

AK027155
142


DNAI2
dynein, axonemal, intermediate polypeptide 2
NM_023036
143



UI-H-EZ1-bbk-j-02-0-UI.s1 NCI_CGAP_Ch2 Homo sapiens


LOC165186
cDNA clone UI-H-EZ1-bbk-j-02-0-UI
NM_199280
144



3′, mRNA sequence.;



ESTs, Weakly similar to T00057 hypothetical protein



KIAA0423 - human (fragment) [H. sapiens]




Homo sapiens mRNA; cDNA DKFZp761D2417 (from clone




DKFZp761D2417)
AL831856
148



UI-E-EJO-ahi-c-20-0-UI.r1 UI-E-EJO Homo sapiens cDNA
BM712946
149



clone UI-E-EJO-ahi-c-20-0-UI 5′, mRNA sequence.


FLJ14665
Start codon is not identified.; Homo sapiens mRNA for
AB058767
155



KIAA1864 protein, partial cds.


MGC16202
hypothetical protein MGC16202
NM_032373
156




Homo sapiens LOH11CR1P gene, loss of heterozygosity, 11,

CA478747
157



chromosomal region 1 gene P product


LOC146177

Homo sapiens hypothetical protein LOC146177

NM_175059
158



(LOC146177), mRNA.


SCARF2
scavenger receptor class F, member 2
NM_153334
159


MGC40053
hypothetical protein MGC40053
NM_152583
161


LOC339005

Homo sapiens cDNA FLJ33935 fis, clone CTONG2017910.

AK091254
162



alternatively spliced; beta-1 form; possible membrane-


C18orf1
spanning protein; clone 22; Homo sapiens chromosome 18
NM_181481
164



open reading frame 1 (C18orf1), mRNA.


MYEF2
myelin expression factor 2
NM_016132
165


DNAH3
dynein, axonemal, heavy polypeptide 3
NM_017539
167
















TABLE 11










Individual samples


Stable Asthma increases, Exacerbated Asthma no change













Seq. ID


Common
Description
Genbank
Number















Homo sapiens transcribed sequences

AI476722
37


ZCCHC2
zinc finger, CCHC domain containing 2
NM_017742
38


CBX5
chromobox homolog 5 (HP1 alpha homolog, Drosophila)
NM_012117
131


FLJ10525
hypothetical protein FLJ10525
NM_018126
154
















TABLE 12










Individual samples


Stable Asthma decreases, Exacerbated Asthma no change













Seq. ID


Common
Description
Genbank
Number













BCL6
B-cell CLL/lymphoma 6 (zinc finger protein 51)
NM_138931
21




Homo sapiens mRNA; cDNA DKFZp566D053 (from clone

AW611486
71



DKFZp566D053)


DNCI1
dynein, cytoplasmic, intermediate polypeptide 1
NM_004411
85


EPOR
erythropoietin receptor
NM_000121
87


LOC253559
nectin-like protein 3
NM_153184
95


HLA-DPB1
major histocompatibility complex, class II, DP beta 1
NM_002121
147


ZNF483
zinc finger protein 483
NM_007169
150


ZNF483
zinc finger protein 483
NM_133464
151


KCNC3
potassium voltage-gated channel, Shaw-related subfamily,
NM_004977
166



member 3
















TABLE 13










Individual samples


Stable Asthma increases, Exacerbated Asthma increases













Seq. ID


Common
Description
Genbank
Number













SOSTDC1
sclerostin domain containing 1
NM_015464
15


BM039
uncharacterized bone marrow protein BM039
NM_018455
16


CALB1
calbindin 1, 28 kDa
NM_004929
18


FOS
v-fos FBJ murine osteosarcoma viral oncogene homolog
NM_005252
28


ZNF407
zinc finger protein 407
NM_017757
31


CYP1A1
cytochrome P450, family 1, subfamily A, polypeptide 1
NM_000499
35


ARNTL2
aryl hydrocarbon receptor nuclear translocator-like 2
NM_020183
48



unnamed protein product; diaminopimelate decarboxylase



(AA 1-327); Bacillus subtilis lys gene for diaminopimelate
X17013
58



decarboxylase (EC 4.1.1.20).


SPEC1
small protein effector 1 of Cdc42
NM_020239
72


EMP1
epithelial membrane protein 1
NM_001423
76


SCEL
sciellin
NM_144777
77




Homo sapiens transcribed sequences

BQ277484
101


DCP2
decapping enzyme hDcp2
NM_152624
126


RAB35
RAB35, member RAS oncogene family
NM_006861
132


C4.4A
GPI-anchored metastasis-associated protein homolog
NM_014400
146


SLC30A7
solute carrier family 30 (zinc transporter), member 7
NM_133496
152
















TABLE 14










Individual samples


Inverse Changes


Stable Asthma increases, Exacerbated Asthma Decrease or Stable Asthma decreases,


Exacerbated Asthma increases













Seq. ID


Common
Description
Genbank
Number













TREM1
triggering receptor expressed on myeloid cells 1
NM_018643
29


RPEL1
RPEL repeat containing 1
AB051520
51



UI-H-BW0-aim-d-12-0-UI.s1 NCI_CGAP_Sub6 Homo sapiens
AW294722
123



cDNA clone IMAGE: 2729902 3′, mRNA sequence.


LILRB2
leukocyte immunoglobulin-like receptor, subfamily B (with TM
NM_005874
145



and ITIM domains), member 2
















TABLE 15










Pooled samples


Exacerbated Asthma two-fold increase













Seq. ID


Common
Description
Genbank
Number





GPR65
G protein-coupled receptor 65
NM_003608
20


HAL
histidine ammonia-lyase
NM_002108
30


SLC2A14
solute carrier family 2 (facilitated glucose transporter),
NM_153449
32



member 14


PLAUR
plasminogen activator, urokinase receptor
NM_002659
53


TF
transferrin
NM_001063
56


PLEK
pleckstrin
NM_002664
59


HIST1H2BC
histone 1, H2bc
NM_003526
61


GPR97
G protein-coupled receptor 97
NM_170776
70


HIST1H2BK
histone 1, H2bk
NM_080593
88


IL1R2
interleukin 1 receptor, type II
NM_004633
90


DUSP5
dual specificity phosphatase 5
NM_004419
92


TLR4
toll-like receptor 4
NM_003266
93
















TABLE 16










Pooled samples


Exacerbated Asthma three-fold increase













Seq. ID


Common
Description
Genbank
Number













KRT24
keratin 24
NM_019016
14


SOSTDC1
sclerostin domain containing 1
NM_015464
15


KRT6A
keratin 6A
NM_005554
17


CALB1
calbindin 1, 28 kDa
NM_004929
18


FCGR2B
Fc fragment of IgG, low affinity IIb, receptor for (CD32)
NM_004001
19


EMR2
egf-like module containing, mucin-like, hormone receptor-
NM_013447
22



like 2


FPRL1
formyl peptide receptor-like 1
NM_001462
24


PDE4B
phosphodiesterase 4B, cAMP-specific (phosphodiesterase E4
NM_002600
27



dunce homolog, Drosophila)


FOS
v-fos FBJ murine osteosarcoma viral oncogene homolog
NM_005252
28


TREM1
triggering receptor expressed on myeloid cells 1
NM_018643
29


ZNF407
zinc finger protein 407
NM_017757
31


AQP9
aquaporin 9
NM_020980
33


BCL2A1
BCL2-related protein A1
NM_004049
34


CYP1A1
cytochrome P450, family 1, subfamily A, polypeptide 1
NM_000499
35


ALDH1A3
aldehyde dehydrogenase 1 family, member A3
NM_000693
39


CLECSF6
C-type (calcium dependent, carbohydrate-recognition
NM_016184
40



domain) lectin, superfamily member 6


IL1B
interleukin 1, beta
NM_000576
41


LILRB1
leukocyte immunoglobulin-like receptor, subfamily B (with
NM_006669
42



TM and ITIM domains), member 1


SLC2A3
solute carrier family 2 (facilitated glucose transporter),
NM_006931
43



member 3


S100A9
S100 calcium binding protein A9 (calgranulin B)
NM_002965
44


PLEK
pleckstrin
NM_002664
45


DTNA
dystrobrevin, alpha
NM_001390
47


RAI3
retinoic acid induced 3
NM_003979
49


G0S2
putative lymphocyte G0/G1 switch gene
NM_015714
50


RPEL1
RPEL repeat containing 1
AB051520
51


CSF2RB
colony stimulating factor 2 receptor, beta, low-affinity
NM_000395
52



(granulocyte-macrophage)


EMP1
epithelial membrane protein 1
NM_001423
55


PRG1
proteoglycan 1, secretory granule
NM_002727
57



unnamed protein product; diaminopimelate decarboxylase



(AA 1-327); Bacillus subtilis lys gene for diaminopimelate
X17013
58



decarboxylase (EC 4.1.1.20).


GPR43
G protein-coupled receptor 43
NM_005306
60


SPRR1A
small proline-rich protein 1A
NM_005987
62


SOD2
superoxide dismutase 2, mitochondrial
NM_000636
67


OLR1
oxidised low density lipoprotein (lectin-like) receptor 1
NM_002543
66



synonyms: HIS, HSI, ARL1, ARL-1, ALDRLn, AKR1B11,



AKR1B12, MGC14103; aldose reductase-like 1; aldo-keto


AKR1B10
reductase family 1, member B11 (aldose reductase-like);
NM_004812
74



aldose reductase-like peptide; aldose reductase-related



protein; small intestine reductase; go



synonyms: HIS, HSI, ARL1, ARL-1, ALDRLn, AKR1B11,



AKR1B12, MGC14103; aldose reductase-like 1; aldo-keto


AKR1B10
reductase family 1, member B11 (aldose reductase-like);
NM_020299
75



aldose reductase-like peptide; aldose reductase-related



protein; small intestine reductase; go


EMP1
epithelial membrane protein 1
NM_001423
76


HM74
putative chemokine receptor
NM_006018
78


HIST1H1C
histone 1, H1c
NM_005319
83


CCR1
chemokine (C—C motif) receptor 1
NM_001295
94


KRT13
keratin 13
NM_153490
97


PLAU
plasminogen activator, urokinase
NM_002658
98


IL1A
interleukin 1, alpha
NM_000575
107


SPRR2B
small proline-rich protein 2B
NM_001017418
109


SPRR1B
small proline-rich protein 1B (cornifin)
NM_003125
117


S100A2
S100 calcium binding protein A2
NM_005978
134


LILRB2
leukocyte immunoglobulin-like receptor, subfamily B (with
NM_005874
145



TM and ITIM domains), member 2


C4.4A
GPI-anchored metastasis-associated protein homolog
NM_014400
146


CPA4
carboxypeptidase A4
NM_016352
153


LILRB1
leukocyte immunoglobulin-like receptor, subfamily B (with
NM_006669
160



TM and ITIM domains), member 1


FCGR2A
Fc fragment of IqG, low affinity IIa, receptor for (CD32)
NM_021642
163







* corrected accession number in Genbank














TABLE 17










Pooled samples


Exacerbated Asthma two-fold decrease













Seq. ID


Common
Description
Genbank
Number













CLGN
calmegin
NM_004362
13


FLJ13615
hypothetical protein FLJ13615
NM_025114
25


FLJ23505

Homo sapiens cDNA: FLJ23505 fis, clone LNG03017

NM_024716
68


KIF3A
kinesin family member 3A
NM_007054
80


IGFBP7
insulin-like growth factor binding protein 7
NM_001553
81


TSAP6
tumor suppressor pHyde; Homo sapiens dudulin 2 (TSAP6),
NM_182915
82



mRNA.


PLTP
phospholipid transfer protein
NM_006227
91


SIAH1
seven in absentia homolog 1 (Drosophila)
NM_003031
100


TCF2
transcription factor 2, hepatic; LF-B3; variant hepatic
NM_006481
113



nuclear factor


LOC123872
similar to RIKEN cDNA 4930457P18
NM_178452
121


DCDC2
doublecortin domain containing 2
NM_016356
127


HPX
hemopexin
NM_000613
141


DNAI2
dynein, axonemal, intermediate polypeptide 2
NM_023036
143



alternatively spliced; beta-1 form; possible membrane-


C18orf1
spanning protein; clone 22; Homo sapiens chromosome 18
NM_181481
164



open reading frame 1 (C18orf1), mRNA.
















TABLE 18










Pooled samples


Exacerbated Asthma three-fold decrease













Seq. ID


Common
Description
Genbank
Number













SF1
splicing factor 1
NM_004630
11


KCTD7
potassium channel tetramerisation domain containing 7
NM_153033
12


MYLK
myosin, light polypeptide kinase
NM_053025
111




Homo sapiens cDNA FLJ12411 fis, clone MAMMA1002964.

AK022473
118
















TABLE 19










Pooled samples


Stable Asthma two-fold increase













Seq. ID


Common
Description
Genbank
Number













FOS
v-fos FBJ murine osteosarcoma viral oncogene homolog
NM_005252
28


AQP9
aquaporin 9
NM_020980
33


IL1B
interleukin 1, beta
NM_000576
41


S100A9
S100 calcium binding protein A9 (calgranulin B)
NM_002965
44


PLEK
pleckstrin
NM_002664
45


OLR1
oxidised low density lipoprotein (lectin-like) receptor 1
NM_002543
69


JUNB
jun B proto-oncogene
NM_002229
139
















TABLE 20










Pooled samples


Stable Asthma three-fold increase













Seq. ID


Common
Description
Genbank
Number













KRT6A
keratin 6A
NM_005554
17


CALB1
calbindin 1, 28 kDa
NM_004929
18


PDE4B
phosphodiesterase 4B, cAMP-specific (phosphodiesterase E4
NM_002600
27



dunce homolog, Drosophila)


HAL
histidine ammonia-lyase
NM_002108
30


ZNF407
zinc finger protein 407
NM_017757
31


CYP1A1
cytochrome P450, family 1, subfamily A, polypeptide 1
NM_000499
35


LILRB1
leukocyte immunoglobulin-like receptor, subfamily B (with TM
NM_006669
42



and ITIM domains), member 1


DTNA
dystrobrevin, alpha
NM_001390
47



unnamed protein product; diaminopimelate decarboxylase (AA



1-327); Bacillus subtilis lys gene for diaminopimelate
X17013
58



decarboxylase (EC 4.1.1.20).


IL1A
interleukin 1, alpha
NM_000575
107


LILRB2
leukocyte immunoglobulin-like receptor, subfamily B (with TM
NM_005874
145



and ITIM domains), member 2
















TABLE 21










Pooled samples


Stable Asthma two-fold decrease













Seq. ID


Common
Description
Genbank
Number













SF1
splicing factor 1
NM_004630
11


FCGR2B
Fc fragment of IgG, low affinity IIb, receptor for (CD32)
NM_004001
19


BCL6
B-cell CLL/lymphoma 6 (zinc finger protein 51)
NM_138931
21


IPW

Homo sapiens mRNA; cDNA DKFZp686M12165 (from clone

BX648788
89



DKFZp686M12165)


TCF2
transcription factor 2, hepatic; LF-B3; variant hepatic nuclear
NM_006481
113



factor


SPAG8
sperm associated antigen 8
NM_012436
130
















TABLE 22










Pooled samples


Stable Asthma three-fold decrease













Seq. ID


Common
Description
Genbank
Number













SOSTDC1
sclerostin domain containing 1
NM_015464
15


RPEL1
RPEL repeat containing 1
AB051520
51


PLAU
plasminogen activator, urokinase
NM_002658
98


C12orf6
chromosome 12 open reading frame 6
NM_020367
102
















TABLE 23










Genes FST > 0.25 between Caucasians and Han Chinese from 161 gene dataset













Seq. ID


Common
Description
Genbank
Number













PDE4B
phosphodiesterase 4B, cAMP-specific (phosphodiesterase E4
NM_002600
27



dunce homolog, Drosophila)


ADCY2
adenylate cyclase 2 (brain)
NM_020546
46


KIF3A
kinesin family member 3A
NM_007054
80


HIST1H1C
histone 1, H1c
NM_005319
83


PLAU
plasminogen activator, urokinase
NM_002658
98


FLJ40427
hypothetical protein FLJ40427
NM_178504
108


SPRR2B
small proline-rich protein 2B
NM_001017418
109


MYLK
myosin, light polypeptide kinase
NM_053025
111


DNAH5
dynein, axonemal, heavy polypeptide 5
NM_001369
115


SLC26A7
solute carrier family 26, member 7
NM_052832
119


CHST5
carbohydrate (N-acetylglucosamine 6-O) sulfotransferase 5
NM_024533
122


ZNF483
zinc finger protein 483
NM_007169
150


ZNF483
zinc finger protein 483
NM_133464
151


SCARF2
scavenger receptor class F, member 2
NM_153334
159


C18orf1
alternatively spliced; beta-1 form; possible membrane-
NM_181481
164



spanning protein; clone 22; Homo sapiens chromosome 18



open reading frame 1 (C18Sorf1), mRNA.


MYEF2
myelin expression factor 2
NM_016132
165


KCNC3
potassium voltage-gated channel, Shaw-related subfamily,
NM_004977
166



member 3







* corrected accession number in Genbank














TABLE 24










Genes 0.15 < FST < 0.25 between Caucasians and Han Chinese from 161 gene dataset













Seq. ID


Common
Description
Genbank
Number













CALB1
calbindin 1, 28 kDa
NM_004929
18


HAL
histidine ammonia-lyase
NM_002108
30


ZNF407
zinc finger protein 407
NM_017757
31


SLC2A14
solute carrier family 2 (facilitated glucose transporter),
NM_153449
32



member 14


BCL2A1
BCL2-related protein A1
NM_004049
34


ZCCHC2
zinc finger, CCHC domain containing 2
NM_017742
38


ALDH1A3
aldehyde dehydrogenase 1 family, member A3
NM_000693
39


PLEK
pleckstrin
NM_002664
45


DTNA
dystrobrevin, alpha
NM_001390
47


ARNTL2
aryl hydrocarbon receptor nuclear translocator-like 2
NM_020183
48


EMP1
epithelial membrane protein 1
NM_001423
55


PRG1
proteoglycan 1, secretory granule
NM_002727
57


DNAH7
dynein, axonemal, heavy polypeptide 7
NM_018897
63


SCEL
sciellin
NM_144777
77


DNCI1
dynein, cytoplasmic, intermediate polypeptide 1
NM_004411
85


MGC26733
hypothetical protein MGC26733
NM_144992
104


TCF2
transcription factor 2, hepatic; LF-B3; variant hepatic
NM_006481
113



nuclear factor


CLECSF12
C-type (calcium dependent, carbohydrate-recognition
NM_197953
114



domain) lectin, superfamily member 12


DNAH9
dynein, axonemal, heavy polypeptide 9
NM_001372
116


EAT2
SH2 domain-containing molecule EAT2
NM_053282
120


PRV1
polycythemia rubra vera 1
NM_020406
129


DNAI2
dynein, axonemal, intermediate polypeptide 2
NM_023036
143



UI-H-EZ1-bbk-j-02-0-UI.s1 NCI_CGAP_Ch2 Homo sapiens


LOC165186
cDNA clone UI-H-EZ1-bbk-j-02-0-UI 3′, mRNA sequence.;
NM_199280
144



ESTs, Weakly similar to T00057 hypothetical protein



KIAA0423 - human (fragment) [H. sapiens]


SLC30A7
solute carrier family 30 (zinc transporter), member 7
NM_133496
152


LILRB1
leukocyte immunoglobulin-like receptor, subfamily B (with
NM_006669
160



TM and ITIM domains), member 1


DNAH3
dynein, axonemal, heavy polypeptide 3
NM_017539
167









Other variations or embodiments of the invention will also be apparent to one of ordinary skill in the art from the above figures, tables, and description. Thus, the forgoing embodiments are not to be construed as limiting the scope of this invention.

Claims
  • 1. A method to evaluate a patient comprising: obtaining from a tissue an expression profile of at least one gene activated in lungs of a human patient with asthma, comparing the patient's gene expression profile with a gene expression profile for stable asthma and a gene expression profile for exacerbated asthma, and determining whether the patient has a propensity for at least one of stable asthma or exacerbated asthma based on at least a two-fold difference between at least one gene in the expression profile.
  • 2. The method of claim 1 based on at least a three-fold difference.
  • 3. The method of claim 1 wherein the tissue is nasal respiratory epithelium.
  • 4. The method of claim 1 comparing a cluster of genes.
  • 5. The method of claim 1 or claim 2 wherein the difference between at least one of SEQ ID NOS: 11-18, 20, 22-24, 26-36, 39-63, 65-70, 72-84, 86, 88-94, 96-101, 103-112, 114-130, 132-146, 148-149, 152-153, 155-165, or 167 gene indicates a propensity for exacerbated asthma.
  • 6. The method of claim 1 or claim 2 wherein the difference between at least one of SEQ ID NOS: 15-16, 18, 21, 28, 29, 31, 35, 37-38, 48, 51, 58, 71-72, 76-77, 85, 87, 95, 101, 123, 126, 131-132, 145-147, 150-152, 154, or 166 gene indicates a propensity for stable asthma.
  • 7. The method of claim 1 wherein the patient is a child.
  • 8. A diagnostic method comprising obtaining a gene expression profile from nasal respiratory epithelium of a patient, and diagnosing exacerbated asthma in the patient if at least a two-fold increase between at least one of SEQ ID NOS: 14-18, 20, 22-24, 26-36, 39-45, 47-62, 65, 67, 69-70, 72-79, 83-84, 86, 88, 90, 92-94, 96-98, 101, 105-107, 109-110, 112, 114, 117, 120, 126, 129, 132, 134-135, 139, 145-146, 152-153, 160, or 163 in the patient tissue expression profile over a control tissue expression profile is present.
  • 9. A diagnostic method comprising obtaining a gene expression profile from nasal respiratory epithelium of a patient, and diagnosing exacerbated asthma in the patient if at least a two-fold decrease in a at least one of SEQ ID NO: 11-13, 46, 63, 66, 68, 80-82, 89, 91, 99-100, 103-104, 108, 111, 115-116, 118-119, 121-125, 127-128, 130, 133, 136-138, 140-144, 148-149, 155-159, 161-162, 164-165, or 167 in the patient tissue expression profile over a control tissue expression profile is present.
  • 10. A diagnostic method comprising obtaining a gene expression profile from nasal respiratory epithelium of a patient, and diagnosing stable asthma in the patient if at least a two-fold increase between at least one of SEQ ID NOS: 15-16, 18, 28, 31, 35, 37-38, 48, 58, 72, 76-77, 101, 123, 126, 131-132, 146, 152, or 154 in the patient tissue expression profile over a control tissue expression profile is present.
  • 11. A diagnostic method comprising obtaining a gene expression profile from nasal respiratory epithelium of a patient, and diagnosing stable asthma in the patient if at least a two-fold decrease in at least one of SEQ ID NOS: 21, 29, 51, 71, 85, 87, 95, 145, 147, 150-151, or 166 in the patient tissue expression profile over a control tissue expression profile is present.
  • 12. The method of any one of claims 8, 9, 10, or 11 further comprising identifying at least one gene for prophylactic or therapeutic intervention.
  • 13. A cluster of genes differentially regulated in at least one of stable asthma or acute asthma comprising SEQ ID NOS: 11-167.
  • 14. The cluster of genes of claim 10 having an allele frequency difference >0.15 between a Caucasian population and a Han Chinese population, the genes comprising SEQ ID NOS: 18, 27, 30-32, 34, 38-39, 45-48, 55, 57, 63, 77, 80, 83, 85, 98, 104, 108-109, 111, 113-116, 119-120, 122, 129, 143-144, 150-152, 159-160, and 164-167.
  • 15. The cluster of genes of claim 10 located in a chromosomal region linked to at least one of an asthma phenotype or an atopy phenotype.
  • 16. The cluster of genes of claim 13 comprising PDE4B SEQ. ID NO: 27, SPRR2B SEQ. ID NO: 109, ADCY2 SEQ. ID NO: 46, KIF3A SEQ. ID NO: 80, DNAH5 SEQ ID NO: 115, and PLAU SEQ. ID NO: 98.
  • 17. A cluster of genes induced at least two-fold in a patient with exacerbated asthma over a patient with stable asthma, the cluster comprising SEQ ID NOS: 14, 17, 20, 22-24, 26-27, 30, 32-34, 36, 39-45, 47, 49-50, 52-57, 59-62, 65, 67, 69-70, 73-75, 78-79, 83-84, 86, 88, 90, 92-94, 96-98, 105-107, 109-110, 112, 114, 117, 120, 129, 134-135, 139, 145, 153, 160, and 163.
  • 18. A cluster of genes repressed at least two-fold in a patient with exacerbated asthma over a patient with stable asthma, the cluster comprising SEQ ID NOS: 11-13, 46, 63, 66, 68, 80-82, 89, 91, 99-100, 103-104, 108, 111, 115-116, 118-119, 121-122, 124-125, 127-128, 130, 133, 136-138, 140-144, 148-149, 155-159, 161-162, 164-165, and 167.
  • 19. A target candidate gene for preferential intervention in childhood exacerbated asthma over childhood stable asthma comprising integrin α4 for preferential intervention in childhood exacerbated asthma over childhood stable asthma.
  • 20. A cluster of genes induced at least two-fold in a patient with stable asthma over a patient with exacerbated asthma, the cluster comprising SEQ ID NOS: 37, 38, 131, and 154.
  • 21. A cluster of genes repressed at least two-fold in a patient with stable asthma over a patient with exacerbated asthma, the cluster comprising SEQ ID NOS: 21, 71, 85, 87, 95, 147, 150, 151, and 166.
  • 22. A cluster of genes induced at least two-fold in a patient with stable asthma over a patient with exacerbated asthma wherein the cluster lacks genes encoding chemokine receptors or chemokines.
  • 23. A gene for preferential intervention in childhood stable asthma over childhood exacerbated asthma comprising a gene encoding retinoic acid receptor α for preferential intervention in childhood stable asthma over childhood exacerbated asthma.
  • 24. A cluster of genes induced at least two-fold in both exacerbated asthma and stable asthma comprising SEQ ID NOS: 15-16, 18, 28, 31, 35, 48, 58, 72, 76-77, 101, 126, 132, 146, and 152.
  • 25. A cluster of genes inversely expressed between patients with exacerbated asthma and patients with stable asthma, the expression being at least a two-fold difference, the cluster comprising SEQ ID NOS: 29, 51, 123, and 145.
  • 26. A gene for preferential intervention in at least one of childhood exacerbated asthma or childhood stable asthma, the gene comprising at least one of histamine 4 receptor gene or SOCS-3 gene.
  • 27. A cluster of genes induced at least three-fold in a patient during asthma exacerbation and induced less than three-fold in the patient during stable asthma, the genes comprising SEQ ID NOS: 15, 16, 18, 35, 76, and 132.
  • 28. An evaluation method comprising identifying at least one gene expression profile in a nasal epithelial cell of a child patient with asthma altered over a gene expression profile in a nasal epithelial cell of a child control, and using the identification to evaluate the child patient as having a propensity for at least one of stable asthma or exacerbated asthma based on at least a two-fold difference in at least one gene in the expression profile.
  • 29. The method of claim 28 based on at least a three-fold difference.
  • 30. The method of claim 28 further comprising treating the patient for stable asthma or exacerbated asthma based on the evaluation.
  • 31. The method of claim 28 further comprising prophylactically treating the patient for stable asthma or exacerbated asthma based on the evaluation.
  • 32. A method to assess lower respiratory epithelial cell DNA comprising obtaining DNA from nasal mucosa cells and assessing the DNA, the nasal epithelial cell DNA as an accessible proxy for DNA from lower respiratory epithelial cells.
  • 33. The method of claim 32 wherein nasal mucosa cells are obtained by nasal mucosa sampling.
  • 34. A method to classify an asthmatic patient comprising obtaining an expression profile from nasal respiratory epithelium of a human patient with asthma, comparing the patient's gene expression profile with a gene expression profile for stable asthma and a gene expression profile for exacerbated asthma, and determining the patient's asthma classification based on at least a two-fold difference between the compared expression profiles.
  • 35. The method of claim 34 further comprising providing prophylaxis or treatment to the patient based on the classification.
Government Interests

The U.S. Government has a paid-up license in this invention and the right in limited circumstances to require the patent owner to license others on reasonable terms as provided for by the terms of Grant Nos. R01A146652-01A1 and R01HL72987 awarded by the National Institutes of Health.