Skip to main content
Molecular Medicine logoLink to Molecular Medicine
. 2007 Jan-Feb;13(1-2):40–58. doi: 10.2119/2006-000056.Edwards

Molecular Profile of Peripheral Blood Mononuclear Cells from Patients with Rheumatoid Arthritis

Christopher J Edwards 1,2,*,, Jeffrey L Feldman 3,*, Jonathan Beech 1, Kathleen M Shields 3, Jennifer A Stover 5, William L Trepicchio 4, Glenn Larsen 6, Brian MJ Foxwell 1, Fionula M Brennan 1, Marc Feldmann 1, Debra D Pittman 3
PMCID: PMC1869619  PMID: 17515956

Abstract

Rheumatoid arthritis (RA) is a chronic inflammatory arthritis. Currently, diagnosis of RA may take several weeks, and factors used to predict a poor prognosis are not always reliable. Gene expression in RA may consist of a unique signature. Gene expression analysis has been applied to synovial tissue to define molecularly distinct forms of RA; however, expression analysis of tissue taken from a synovial joint is invasive and clinically impractical. Recent studies have demonstrated that unique gene expression changes can be identified in peripheral blood mononuclear cells (PBMCs) from patients with cancer, multiple sclerosis, and lupus. To identify RA disease-related genes, we performed a global gene expression analysis. RNA from PBMCs of 9 RA patients and 13 normal volunteers was analyzed on an oligonucleotide array. Compared with normal PBMCs, 330 transcripts were differentially expressed in RA. The differentially regulated genes belong to diverse functional classes and include genes involved in calcium binding, chaperones, cytokines, transcription, translation, signal transduction, extracellular matrix, integral to plasma membrane, integral to intracellular membrane, mitochondrial, ribosomal, structural, enzymes, and proteases. A k-nearest neighbor analysis identified 29 transcripts that were preferentially expressed in RA. Ten genes with increased expression in RA PBMCs compared with controls mapped to a RA susceptibility locus, 6p21.3. These results suggest that analysis of RA PBMCs at the molecular level may provide a set of candidate genes that could yield an easily accessible gene signature to aid in early diagnosis and treatment.

INTRODUCTION

Rheumatoid arthritis (RA) is a chronic inflammatory disease causing synovial joint damage, disability, and a shortened life expectancy (1,2). An awareness of the destructive potential of RA has led to more aggressive use of disease-modifying anti-rheumatic drugs (DMARDs) (3) and the development of immune therapies targeted to molecules and cells important in the pathogenesis of RA. These include the TNF inhibitors infliximab, etanercept, and adalimumab (4). Synovial joint damage occurs early in the disease course, and many patients demonstrate erosions within a few months after becoming symptomatic (5). Recent evidence suggests that early aggressive therapy (infliximab and methotrexate) yields greater benefit than similar therapy after failure of other drugs (68). To initiate early aggressive therapy requires reliable and rapid determination of diagnosis and prognosis. In addition, factors used to predict a poor prognosis, including sex, age of onset, multiple joint involvement, rheumatoid factor, and the presence of the shared epitope of HLA-DR4, are not always reliable (912).

Gene expression profiling may allow early diagnosis, aid in identifying factors that predict poor prognosis, and help focus early, aggressive, and expensive therapy to those that would benefit the most. Expression analysis of tissues taken at the site of disease within a synovial joint is invasive and impractical on a routine basis. However, recent studies have demonstrated unique gene expression changes in peripheral blood mononuclear cells (PBMCs) from patients with cancer, multiple sclerosis, and lupus (1317). In this study, a genome-wide scan of PBMCs from normal volunteers and RA PBMCs was performed using oligonucleotide arrays representing 6800 human genes to explore gene expression in the PBMCs of individuals with RA.

MATERIALS AND METHODS

Patient Selection

Patients with RA, defined by American College of Rheumatology (ACR) criteria (18), were identified in a rheumatology clinic with approval from the local research ethics committee. Demographic data including age, sex, and time since diagnosis were collected. A tender joint count (TJC 0-28), swollen joint count (JC 0-28), patient’s best global assessment (visual analog scale), and erythrocyte sedimentation rate (ESR) were performed to calculate a 28-joint disease activity score (DAS28). The presence of rheumatoid factor (RF) and the use of DMARDs were recorded. Blood was also collected from healthy volunteers with no previous diagnosis of RA or other chronic inflammatory diseases.

Isolation of RNA and Preparation of Labeled Hybridization Solutions

An 8-mL sample of venous blood was collected into CPT Vacutainer cell purification tubes (Becton Dickinson, Franklin Lakes, NJ, USA) and refrigerated immediately. Samples were immediately transferred to the laboratory, and PBMCs from the 9 RA and 13 normal volunteers were separated according to the manufacturer’s recommendations. Briefly, the tube was centrifuged at 1500g (2700 rpm) at room temperature, and PBMCs were isolated before being washed twice in PBS. Total RNA was extracted using the RNeasy minikit (Qiagen, Valencia, CA, USA). For each sample, 2 μg total RNA was used to generate cDNA as described (19). RNA quality was determined by observing distinct 28S and 18S ribosomal bands on an agarose gel. First-strand cDNA synthesis was performed under the following buffer conditions: 1× 1st-strand buffer (Invitrogen Life Technologies, Carlsbad, CA, USA), 10 mM DTT (Gibco/Invitrogen), 500 μM of each dNTP (Invitrogen Life Technologies), 400 units Superscript RT II (Invitrogen Life Technologies), and 40 units RNase inhibitor (Ambion, Austin, TX, USA). The reaction proceeded at 47°C for 1 h. Second-strand cDNA was synthesized with the addition of the following reagents at the final concentrations listed: 1× 2nd-strand buffer (Invitrogen Life Technologies), an additional 200 μM of each dNTP (Invitrogen Life Technologies), 40 units E. coli DNA polymerase I (Invitrogen Life Technologies), 2 units E. coli RNaseH (Invitrogen Life Technologies), and 10 units E. coli DNA ligase. The reaction proceeded at 15°C for 2 h; during the last 5 min of this reaction, 6 units T4 DNA polymerase (New England Biolabs, Beverly, MA, USA) was added. The resulting double-stranded cDNA was purified with the use of BioMag carboxyl-terminated particles as follows: 0.2 mg BioMag particles (Polysciences, Warrington, PA, USA) were equilibrated by washing three times with 0.5 M EDTA and resuspended at a concentration of 22.2 mg/mL in 0.5 M EDTA. The double-stranded cDNA reaction was diluted to a final concentration of 10% PEG/1.25 M NaCl, and the bead suspension was added to a final bead concentration of 0.614 mg/mL. The reaction was incubated at room temperature for 10 min. The cDNA/bead complexes were washed with 300 μL of 70% ethanol, the ethanol was removed, and the tubes were allowed to air dry. The cDNA was eluted with the addition of 20 μL of 10 mM Tris-acetate, pH 7.8, and incubated for 2 to 5 min, and the cDNA-containing supernatant was removed.

Purified double stranded cDNA (10 μL) was added to an in vitro transcription (IVT) solution which contained 1× IVT buffer (Ambion), 5000 units T7 RNA polymerase (Epicentre Technologies, Madison, WI, USA), 3mM GTP, 1.5 mM ATP, 1.2 mM CTP, and 1.2 mM UTP (Amersham/Pharmacia), 0.4 mM each bio-16 UTP and bio-11 CTP (Enzo Diagnostics, Farmingdale, NY, USA), and 80 units RNase inhibitor (Ambion). The reaction proceeded at 37°C for 16 h. Labeled RNA was purified with the use of an RNeasy kit (Qiagen). The RNA yield was quantified by measuring absorbance at 260 nm.

Hybridization to Affymetrix Microarrays and Detection of Fluorescence

Eleven in vitro synthesized transcripts from segments of bacterial genes were included in each hybridization reaction to generate a global standard curve to normalize the oligonucleotide microarrays to each other and estimate the sensitivity of the arrays (20). Purified biotinylated cRNA (10 μg) was hybridized to oligonucleotide arrays comprised of 6937 human gene qualifiers (human FL6800 array P/N900183, Affymetrix, Santa Clara, CA).

Raw fluorescent intensity values were collected and reduced with GeneChip v3.2 software (Affymetrix) as described (Affymetrix GeneChip Analysis Suite User Guide). This determined the probability of each gene qualifier represented on the array being absent, present, or marginal, as well as calculating a specific hybridization intensity value, or average difference, for each transcript. The relative abundances of the 11 bacterial control cRNA transcripts ranged from 1:300,000 (3 ppm) to 1:1000 (1000 ppm) stated in terms of the number of control transcripts per total transcripts. As determined by the signal response from these control transcripts, the sensitivity of detection of the arrays ranged between ~1:300,000 and 1:100,000 copies/million. The average difference for each transcript was normalized to frequency values as described (20).

Transcripts designated absent in all samples were excluded from the analysis; 3295 (49%) of the transcripts remained. Further analysis of the processed data was performed with GeneSpring version 7.1 (Agilent Technologies, Redwood City, CA, USA). To identify transcripts that were increased in the RA samples compared with controls, >50% (at least 5 of 9) of the samples had to be called present, with a frequency of 10 ppm or greater, and have a change in expression, relative to the average expression of the controls, of at least 2-fold. The resulting data set had 324 gene qualifiers. To find gene qualifiers whose expression was decreased, a list was generated of gene qualifiers from normal samples that were called present with a frequency of ≥ 10 ppm. The resulting list was filtered for an average decrease in expression, relative to the controls, of at least 2-fold in the disease samples. Six gene qualifiers met these criteria; 330 transcripts were used for the analyses. Annotation for each gene was determined based on GO, Entrez Gene, PubMed, and literature searches.

Statistical and Clustering Analyses

An unsupervised hierarchical clustering was performed on the 330 genes to group the samples on the basis of similarity of their expression profiles (21). Statistically significant differences in expression were determined using Welch ANOVA (22) coupled with two different multiple testing corrections. The Benjamini and Hochberg false discovery rate (FDR) (23) was applied with a P value <0.05, with 326 genes passing this criterion. The Bonferroni family-wise error rate (FWER) (24,25) was applied with a P value <0.05, with 189 gene qualifiers passing this criterion. Finally, a class prediction using the k-nearest neighbor method (26) was applied to the filtered data to determine which genes had the highest discrimination between normal and RA samples.

RESULTS

Characteristics of the RA patients used in the study, including demographics, disease activity scores, and DMARD use, are illustrated in Table 1. In total, 324 transcripts increased by at least two-fold between the RA and control subjects, and six transcripts decreased by at least twofold between the RA and control subjects (Table 2).

Table 1.

Characteristics of RA patients including demographics, disease activity scores, and DMARD use.

ID Age, years Sex RF titer Erosions DMARD use DAS
RA1 61 F + Prednisolone 5 mg/d, methotrexate 15 mg/wk 3.9
RA2 66 F 1/5120 + Prednisolone 7.5 mg/d, methotrexate 20 mg/wk, sulfasalazine 500 mg BD 4.5
RA3 46 F 1/640 + Methotrexate 15 mg/wk, hydroxychloroquine 200 mg BD 3.5
RA4 52 F 1/640 Methotrexate 12.5 mg/wk 3.6
RA5 55 F 1/320 + Methotrexate 12.5 mg/wk, hydroxychloroquine 200 mg BD 3.3
RA6 35 F 1/2560 + 3.0
RA7 74 M 1/320 + Methotrexate 7.5 mg/wk 3.5
RA8 77 M 1/20400 + Prednisolone 7.5 mg/d, methotrexate 17.5 mg/wk 3.2
RA 9 49 F 1/1250 + Methotrexate 10 mg/wk 2.9

Table 2.

Differentially regulated transcripts.

Symbol Name GenBank acc. no. Map Freq RA Freq control % disease samples, ≥ 2x or ″–2x Fold changea t test (freq) ANOVA – FDR ANOVA – FWER Class predictor gene: strength
Calcium Binding
ANXA11 Annexin A11 L19605 10q23 97.6 ± 9.1 39.1 ± 1.8 89% 2.5 ± 0.2 2.8E–02 1.5E–05 6.0E–04 1.954
S100A4 S100 calcium binding protein A4 M80563 1q21 213.2 ± 19.9 87.6 ± 7.8 89% 2.4 ± 0.2 3.2E–02 5.3E–05 5.3E–03
S100A12 S100 calcium binding protein A12 D83657 1q21 65.4 ± 14.0 22.8 ± 4.8 67% 2.6 ± 0.8 5.2E–01 3.3E–03
CALM3 Calmodulin 3 J04046 19q13.2-q13.3 29.6 ± 1.8 13.8 ± 1.0 67% 2.1 ± 0.1 6.9E–02 4.7E–06 1.1E–04
CACYBP Calcyclin binding protein BC001431 1q24-q25 179.2 ± 23.4 72.2 ± 8.3 67% 2.3 ± 0.5 1.1E–01 4.0E–04
Chaperone
HYOU1 Hypoxia up-regulated 1 U65785 11q23.1-q23.3 33.7 ± 3.6 11.4 ± 0.7 78% 3.0 ± 0.3 4.2E–02 2.0E–05 1.1E–03
NAP1L4 Nucleosome assembly protein 1-like 4 U77456 11p15.5 26.6 ± 3.6 9.8 ± 0.8 78% 2.7 ± 0.4 5.7E–02 1.9E–04 3.3E–02
TTC1 Tetratricopeptide repeat domain 1 U46570 5q32-q33.2 41.7 ± 4.7 16.1 ± 1.3 67% 2.6 ± 0.3 2.9E–02 5.3E–05 5.4E–03
DNAJC7 DnaJ (Hsp40) homolog, subfamily C, member 7 U46571 17q11.2 17.8 ± 2.3 6.5 ± 0.5 67% 2.7 ± 0.3 1.6E–01 1.0E–04 1.2E–02
Cytokine/chemokine
IGF2 Insulin growth factor 2 gene, intron 7 S73149 34.1 ± 4.9 8.9 ± 0.7 89% 3.8 ± 0.5 8.0E–02 1.9E–05 1.0E–03 1.874
CSF1 Human macrophage-specific colony-stimulating factor M11296 1p21-p13 28.0 ± 3.6 9.6 ± 0.7 78% 2.9 ± 0.4 9.6E–02 4.3E–05 3.7E–03
CCL5 Chemokine (C-C motif) ligand 5 M21121 17q11.2-q12 156.8 ± 14.9 53.7 ± 3.7 78% 2.9 ± 0.3 2.7E–02 2.5E–05 1.5E–03
CCL22 Chemokine (C-C motif) ligand 22 U83239 16q13 31.2 ± 5.0 11.2 ± 0.9 67% 2.8 ± 0.4 1.3E–01 4.4E–04
CCL22 (duplicate) Chemokine (C-C motif) ligand 22 U83171 16q13 15.2 ± 2.1 6.2 ± 0.2 67% 2.5 ± 0.3 1.8E–02 6.3E–04
TGFB1 Transforming growth factor, β 1 M38449 19q13.2 26.6 ± 4.0 10.1 ± 1.1 67% 2.4 ± 0.6 7.2E–03 1.3E–03
MLN Motilin X15393 6p21.3 13.4 ± 1.8 6.0 ± 0.4 67% 2.2 ± 0.3 9.5E–02 8.9E–04
PF4 Platelet factor 4 (chemokine (C-X-C motif) ligand 4) M25897 4q12-q21 278.3 ± 14.2 136.5 ± 13.7 56% 2.0 ± 0.1 1.2E–01 1.2E–05 4.7E–04
IL7R Interleukin 7 receptor M29696 5p13 24.0 ± 10.8 30.2 ± 9.6 67% −2.3 ± 1.0 1.3E–01
DNA binding
HMGB1 High-mobility group box 1 D63874 13q12 83.2 ± 9.3 27.3 ± 4.3 78% 3.0 ± 0.3 2.5E–01 3.3E–05 2.6E–03
MKRN4 Ring zinc-finger protein U41315 Xp21.1 15.0 ± 1.9 5.8 ± 0.3 67% 2.6 ± 0.3 1.3E–01 3.1E–04
HIST2H2AA Histone 2, H2aa L19779 1q21.3 71.9 ± 8.2 32.9 ± 3.0 56% 2.2 ± 0.2 5.2E–02 6.6E–05 7.2E–03
DDB1 Damage-specific DNA binding protein 1 U32986 11q12-q13 24.7 ± 3.3 11.0 ± 0.6 67% 2.0 ± 0.5 6.2E–02 1.5E–03
Enzyme
LYZ Lysozyme M21119 12q14.3 53.0 ± 27.2 69.2 ± 20.0 67% −6.6 ± 2.2 8.0E–02
AGPAT1 1-acylglycerol-3-phosphate O-acyltransferase 1 U56417 6p21.3 26.3 ± 2.7 7.2 ± 0.4 89% 3.7 ± 0.4 6.6E–02 3.1E–05 2.3E–03 2.006
DIA1 NADH-cytochrome b5 reductase M28713 33.3 ± 3.4 8.9 ± 0.5 100% 3.7 ± 0.4 5.1E–02 2.5E–06 3.7E–05 2.462
KIAA0220 PI-3-kinase-related kinase SMG-1-like D86974 16p12.2 239.2 ± 33.3 73.4 ± 8.9 78% 3.3 ± 0.5 1.5E–02 1.2E–04 1.6E–02
GSTZ1 Glutathione transferase zeta 1 U86529 14q24.3 25.6 ± 3.3 7.8 ± 0.6 78% 3.3 ± 0.4 1.0E–01 2.5E–05 1.5E–03
PYGB Phosphorylase, glycogen U47025 20p11.2-p11.1 25.2 ± 2.7 7.8 ± 0.4 89% 3.2 ± 0.3 5.1E–02 1.3E–04 1.9E–02
SAT Spermidine/spermine N1-acetyltransferase U40369 Xp22.1 37.7 ± 4.0 12.3 ± 2.0 89% 3.1 ± 0.3 4.1E–01 1.9E–05 9.6E–04
UROD Uroporphyrinogen decarboxylase X89267 1p34 42.7 ± 6.2 14.2 ± 1.1 78% 3.0 ± 0.4 1.1E–01 2.1E–04 3.7E–02
GPI Glucose phosphate isomerase K03515 19q13.1 35.8 ± 4.5 12.1 ± 1.0 67% 3.0 ± 0.4 2.3E–02 3.3E–05 2.6E–03
GSTO1 Glutathione S-transferase omega 1 U90313 10q25.1 43.2 ± 4.2 14.3 ± 1.5 89% 3.0 ± 0.3 8.9E–02 5.4E–06 1.4E–04
DDX11 DEAD/H (Asp-Glu-Ala-Asp/His) box polypeptide 11 U75968 12p11 20.6 ± 2.8 7.1 ± 0.6 100% 2.9 ± 0.4 6.3E–02 1.5E–05 6.7E–04
IMPDH1 IMP (inosine monophosphate) dehydrogenase 1 J05272 7q31.3-q32 31.4 ± 4.7 10.9 ± 0.9 78% 2.9 ± 0.4 3.7E–02 2.3E–04 4.3E–02
PIB5PA Phosphatidylinositol (4,5) bisphosphate 5-phosphatase, A U45975 22q11.2-q13.2 20.0 ± 2.6 7.2 ± 0.4 67% 2.8 ± 0.4 6.6E–02 2.7E–04
CNP 2′,3′-cyclic nucleotide 3′ phosphodiesterase D13146 17q21 49.9 ± 6.4 17.8 ± 1.4 78% 2.8 ± 0.4 1.3E–01 4.9E–05 4.8E–03
UPP1 Uridine phosphorylase 1 X90858 7p12.3 21.4 ± 1.3 7.6 ± 0.6 89% 2.8 ± 0.2 5.1E–02 2.4E–07 6.1E–07 2.246
AMPD2 Adenosine monophosphate deaminase 2 (isoform L) M91029 1p13.3 30.3 ± 4.0 11.2 ± 0.8 78% 2.7 ± 0.4 6.6E–02 8.6E–05 9.8E–03
BCAT2 Branched chain aminotransferase 2, mitochondrial U62739 19q13 18.8 ± 2.0 7.1 ± 0.3 78% 2.7 ± 0.3 3.1E–02 1.0E–04 1.2E–02
HSD17B3 Hydroxysteroid (17-beta) dehydrogenase 3 U05659 9q22 21.4 ± 2.6 8.1 ± 0.5 78% 2.7 ± 0.3 8.0E–02 1.6E–04 2.5E–02
GARS Glycyl-tRNA synthetase U09587 7p15 29.7 ± 2.5 11.2 ± 0.7 78% 2.7 ± 0.2 7.6E–02 3.7E–06 7.1E–05
PTGS1 Prostaglandin-endoperoxide synthase 1 M59979 9q32-q33.3 14.2 ± 2.3 5.5 ± 0.4 67% 2.6 ± 0.4 7.9E–02 1.6E–03
TGM1 Transglutaminase 1 L34840 14q11.2, 3p22-p21.33 18.4 ± 2.3 7.2 ± 0.7 67% 2.6 ± 0.3 1.9E–01 9.2E–05 1.1E–02
AOAH Acyloxyacyl hydrolase M62840 7p14-p12 25.2 ± 5.4 9.2 ± 0.8 67% 2.5 ± 0.7 2.3E–01 4.8E–03
GNPDA1 Glucosamine-6-phosphate deaminase 1 D31766 5q21 13.8 ± 2.0 5.5 ± 0.3 56% 2.5 ± 0.4 6.7E–02 6.4E–04
AARS Alanyl-tRNA synthetase D32050 16q22 18.1 ± 2.5 7.4 ± 0.3 67% 2.5 ± 0.3 1.6E–01 9.7E–04
SETDB1 SET domain, bifurcated 1 D31891 1q21 18.6 ± 2.5 7.4 ± 0.7 67% 2.5 ± 0.3 8.9E–02 1.5E–04 2.3E–02
DCTD Deoxycytidylate deaminase gene L39874 4q35.1 25.0 ± 3.0 9.8 ± 0.8 67% 2.5 ± 0.3 8.0E–02 5.4E–05 5.5E–03
HUMNOSB Inducible nitric oxide synthase D29675 14.1 ± 2.6 5.3 ± 0.2 67% 2.4 ± 0.6* 2.6E–01 4.1E–03
MGLL Monoglyceride lipase U67963 3q21.3 18.9 ± 2.0 7.2 ± 0.5 89% 2.4 ± 0.5 9.8E–02 2.3E–04 4.4E–02
DAO D-amino-acid oxidase D11370 12q24 14.9 ± 2.2 6.2 ± 0.2 56% 2.4 ± 0.4 9.7E–02 1.1E–03
DDT D-dopachrome tautomerase U49785 22q11.23 24.9 ± 2.9 10.5 ± 0.6 56% 2.4 ± 0.3 4.9E–02 1.1E–04 1.5E–02
TPI1 Triosephosphate isomerase 1 M10036 12p13 73.3 ± 8.5 30.0 ± 2.6 78% 2.4 ± 0.3 2.7E–02 1.0E–04 1.3E–02
SULT1A3 Sulfotransferase family, cytosolic, 1A, phenol-preferring, member 3 U20499 16p11.2 20.3 ± 2.3 8.3 ± 0.6 67% 2.4 ± 0.3 1.7E–01 7.6E–05 8.5E–03
GLA Galactosidase, alpha X14448 Xq22 20.6 ± 2.2 8.6 ± 0.9 78% 2.4 ± 0.3 2.8E–01 1.0E–04 1.3E–02
PPGB Protective protein for β-galactosidase M22960 20q13.1 83.4 ± 8.6 35.2 ± 2.4 78% 2.4 ± 0.2 2.6E–02 4.9E–05 4.8E–03
FAH Fumarylacetoacetate hydrolase M55150 15q23-q25 18.7 ± 1.1 7.9 ± 0.5 89% 2.4 ± 0.1 4.4E–02 6.8E–07 3.4E–06 2.008
CDA Cytidine deaminase L27943 1p36.2-p35 26.9 ± 5.8 10.5 ± 1.0 56% 2.3 ± 0.7 6.4E–02 4.6E–03
HYAL2 Hyaluronoglucosaminidase 2 AJ000099 3p21.3 24.9 ± 3.7 10.9 ± 0.8 67% 2.3 ± 0.3 6.4E–02 1.7E–03
INPP5D Inositol polyphosphate-5-phosphatase U57650 2q36-q37 42.8 ± 5.6 18.8 ± 1.3 67% 2.3 ± 0.3 8.9E–02 4.6E–04
CES1 Carboxylesterase 1 L07765 16q13-q22.1 14.0 ± 2.0 6.2 ± 0.2 56% 2.3 ± 0.3 1.0E–01 1.2E–03
ACADVL Acyl-Coenzyme A dehydrogenase D43682 17p13-p11 41.4 ± 3.8 18.2 ± 1.6 67% 2.3 ± 0.2 2.0E–02 3.2E–05 2.4E–03
SULT1A1 Sulfotransferase family, cytosolic, 1A, phenol-preferring, member 1 L19999 16p12.1 21.0 ± 2.3 9.2 ± 1.1 56% 2.3 ± 0.2 2.9E–01 9.3E–05 1.1E–02
PRDX6 Peroxiredoxin 6 D14662 1q24.1 36.4 ± 2.7 16.1 ± 2.1 67% 2.3 ± 0.2 1.0E–01 4.4E–05 4.0E–03
UQCRC1 Ubiquinol-cytochrome c reductase core protein I L16842 3p21.3 24.1 ± 3.3 10.1 ± 0.8 78% 2.2 ± 0.5 7.5E–02 5.4E–04
GUSB Glucuronidase, beta M15182 7q21.11 18.9 ± 2.2 8.5 ± 0.5 56% 2.2 ± 0.3 4.9E–02 1.2E–04 1.6E–02
HK3 Hexokinase 3 U51333 5q35.2 47.1 ± 7.3 20.0 ± 2.0 56% 2.1 ± 0.5 3.4E–01 1.6E–03
CHKL Choline kinase-like U62317 22q13.33 60.8 ± 7.0 26.6 ± 1.5 67% 2.1 ± 0.4 1.1E–01 5.0E–04
PGM1 Phosphoglucomutase 1 M83088 1p31 16.7 ± 1.5 7.8 ± 0.5 56% 2.1 ± 0.2 2.4E–01 4.6E–05 4.2E–03
PCSK6 Proprotein convertase subtilisin/kexin type 6 M80482 15q26 10.7 ± 1.1 5.1 ± 0.1 44% 2.1 ± 0.2 3.7E–02 1.7E–04 2.6E–02
PMM1 Phosphomannomutase 1 U86070 22q13.2 13.8 ± 1.3 6.6 ± 0.2 56% 2.1 ± 0.2 3.6E–02 1.7E–04 2.7E–02
TALDO1 Transaldolase 1 L19437 11p15.5-p15.4 85.3 ± 9.9 37.7 ± 3.0 78% 2.0 ± 0.5 1.2E–01 6.4E–04
Extracellular matrix
EPB49 Erythrocyte membrane protein band 4.9 (dematin) L19713 8p21.1 38.7 ± 4.3 9.9 ± 0.8 100% 3.9 ± 0.4 2.8E–02 3.1E–06 5.2E–05 2.064
SPARC Secreted protein, acidic, cysteine-rich (osteonectin) J03040 5q31.3-q32 57.8 ± 8.2 24.4 ± 3.5 56% 2.4 ± 0.3 1.1E–01 4.0E–04
Integral intracellular membrane
CPT1B Carnitine palmitoyltransferase 1B (muscle) Y08682 22q13.33 13.6 ± 1.9 4.9 ± 0.2 67% 2.8 ± 0.4 8.0E–02 3.3E–04
STX5A Syntaxin 5A U26648 11q12.3 19.9 ± 2.2 7.9 ± 0.6 89% 2.5 ± 0.3 1.8E–02 1.9E–05 9.0E–04
HAX1 HS1 binding protein U68566 1q22 31.0 ± 2.9 13.9 ± 1.2 67% 2.2 ± 0.2 7.7E–02 4.9E–05 4.6E–03
BAP1 BRCA1 associated protein-1 D87462 3p21.31-p21.2 12.9 ± 1.0 6.2 ± 0.3 56% 2.1 ± 0.2 1.4E–02 5.4E–05 5.6E–03
BZRP Benzodiazapine receptor (peripheral) L21954 22q13.31 127.9 ± 26.1 50.5 ± 4.3 56% 2.3 ± 0.6 1.9E–01 4.8E–03
BCL2 B-cell CLL/lymphoma 2 M14745 18q21.33 16.2 ± 2.1 7.5 ± 0.6 44% 1.9 ± 0.5 1.4E–01 8.0E–04
HERPUD1 Homocysteine-inducible, endoplasmic reticulum stress-inducible, ubiquitin-like domain member 1 D14695 16q12.2-q13 16.6 ± 3.1 7.0 ± 0.6 56% 1.9 ± 0.7 4.2E–01 7.8E–03
Integral plasma membrane
STAB1 Stabilin 1 D87433 3p21.31 30.2 ± 6.7 4.7 ± 0.2 100% 6.4 ± 1.4* 1.8E–01 6.3E–05 6.8E–03 2.164
IGHG3 Immunoglobulin heavy constant γ 3 M87789 14q32.33 105.6 ± 38.7 15.8 ± 2.5 67% 6.3 ± 2.6 1.2E–01 1.4E–02
ZYX Zyxin X95735 7q32 38.0 ± 6.5 10.5 ± 1.0 78% 3.6 ± 0.6 4.1E–02 4.7E–04
SELPLG Selectin P ligand U25956 12q24 75.9 ± 10.0 21.1 ± 1.8 78% 3.6 ± 0.5 4.4E–02 2.2E–05 1.3E–03
PRF1 Perforin M31951 10q22 16.9 ± 6.6 25.0 ± 3.8 67% −3.4 ± 1.2 1.0E + 00
CD151 CD151 antigen D29963 11p15.5 29.2 ± 4.9 8.7 ± 0.8 78% 3.4 ± 0.6 2.9E–01 1.3E–04 1.8E–02
CD63 CD63 antigen X62654 12q12-q13 41.8 ± 6.7 12.4 ± 1.3 89% 3.4 ± 0.5 1.4E–01 1.0E–04 1.2E–02
IFNGR2 Interferon γ receptor 2 U05875 21q22.11 37.9 ± 6.9 11.5 ± 1.1 56% 3.3 ± 0.6 1.3E–01 3.4E–04
TRIP12 Thyroid hormone receptor interactor 12 D28476 2q37.1 27.4 ± 3.7 8.9 ± 1.0 78% 3.1 ± 0.4 2.4E–01 1.4E–04 2.1E–02
LENG4 Leukocyte receptor cluster member 4 S82470 19q13.4 26.8 ± 3.9 8.7 ± 0.6 78% 3.1 ± 0.4 8.6E–02 1.6E–04 2.5E–02
CSF3R Colony stimulating factor 3 receptor M59820 1p35-p34.3 33.3 ± 6.9 10.2 ± 0.8 67% 3.0 ± 0.8 5.9E–02 4.2E–03
GP1BB Glycoprotein Ib (platelet), β polypeptide U59632 22q11.21 97.2 ± 16.2 30.5 ± 3.3 67% 3.0 ± 0.7 6.9E–02 7.1E–04
FLOT2 Flotillin 2 M60922 17q11-q12 50.4 ± 7.9 16.1 ± 1.2 78% 2.8 ± 0.7 2.8E–02 3.9E–03
VAT1 Vesicle amine transport protein 1 homolog U18009 17q21 12.3 ± 1.2 4.5 ± 0.1 78% 2.8 ± 0.3* 1.1E–01 2.5E–05 1.6E–03 2.327
MCL1 Myeloid cell leukemia sequence 1 L08246 1q21 88.7 ± 9.7 32.2 ± 3.9 78% 2.8 ± 0.3 6.6E–02 5.7E–05 6.0E–03
HLA-DQA1 Major histocompatibility complex, class II, DQ α 1 M34996 6p21.3 80.6 ± 18.7 25.1 ± 3.7 56% 2.7 ± 1.0 2.2E–01 6.8E–03
MICB MHC class I chain-related gene B U65416 6p21.3 17.3 ± 2.1 6.5 ± 0.2 67% 2.7 ± 0.3 1.2E–01 2.0E–04 3.5E–02
CD7 CD7 antigen D00749 17q25.2-q25.3 69.9 ± 8.8 25.8 ± 2.1 67% 2.7 ± 0.3 4.3E–03 1.7E–04 2.9E–02
LILRA2 Leukocyte immunoglobulin-like receptor, subfamily A (with TM domain), member 2 U82275 19q13.4 25.4 ± 4.7 8.8 ± 0.8 67% 2.6 ± 0.7 1.4E–01 2.1E–03
HEM1 Hematopoietic protein 1 M58285 12q13.1 38.4 ± 5.1 14.6 ± 2.0 67% 2.6 ± 0.3 1.2E–01 1.7E–04 2.7E–02
FCGRT Fc fragment of IgG, receptor, transporter, alpha U12255 19q13.3 78.6 ± 13.7 29.2 ± 2.5 67% 2.5 ± 0.6 1.1E–01 2.8E–03
SPG7 Spastic paraplegia 7 homolog X65784 16q24.3 21.6 ± 2.2 8.5 ± 0.5 78% 2.5 ± 0.3 6.4E–02 3.2E–05 2.4E–03
IL10RB Interleukin 10 receptor, beta Z17227 21q22.1-q22.2 15.8 ± 1.6 6.2 ± 0.4 78% 2.5 ± 0.3 3.7E–02 3.6E–05 2.9E–03
SELP Selectin P M25322 1q22-q25 15.0 ± 1.2 5.9 ± 0.4 78% 2.5 ± 0.2 1.0E–01 1.5E–06 1.4E–05 2.018
CEACAM4 Carcinoembryonic antigen-related cell adhesion molecule 4 D90276 19q13.2 13.3 ± 2.1 5.5 ± 0.2 56% 2.4 ± 0.4* 5.8E–02 1.7E–03
CD3E CD3E antigen, epsilon polypeptide M23323 11q23 45.8 ± 5.8 18.9 ± 1.0 67% 2.4 ± 0.3 4.0E–02 4.0E–04
AAMP Angio-associated, migratory cell protein M95627 2q35 22.1 ± 3.2 9.4 ± 0.7 56% 2.4 ± 0.3 5.1E–02 1.1E–03
LAMP1 Lysosomal-associated membrane protein 1 J04182 13q34 47.8 ± 4.5 20.2 ± 2.0 67% 2.4 ± 0.2 7.4E–02 3.3E–05 2.6E–03
BST2 Bone marrow stromal cell antigen 2 D28137 19p13.2 46.3 ± 9.5 16.4 ± 1.6 78% 2.3 ± 0.8 2.5E–01 9.3E–03
CD33 CD33 antigen M23197 19q13.3 19.4 ± 3.1 7.6 ± 0.7 56% 2.3 ± 0.5 2.3E–01 1.3E–03
ACRV1 Intra-acrosomal protein S65583 11p12-q13 12.8 ± 2.3 5.5 ± 0.2 56% 2.3 ± 0.4* 7.3E–02 2.9E–03
PTTG1IP Pituitary tumor-transforming 1 interacting protein Z50022 21q22.3 30.0 ± 3.3 13.0 ± 0.9 56% 2.3 ± 0.3 6.0E–02 1.7E–04 2.7E–02
HDLBP High density lipoprotein binding protein (vigilin) M64098 2q37 18.8 ± 2.3 8.3 ± 0.6 67% 2.3 ± 0.3 5.3E–02 2.9E–04
ICAM3 Intercellular adhesion molecule 3 X69819 19p13.3-p13.2 52.2 ± 4.4 22.6 ± 1.7 67% 2.3 ± 0.2 8.2E–03 2.0E–05 1.1E–03
OS-9 Amplified in osteosarcoma U41635 12q13 58.6 ± 7.7 24.2 ± 1.8 67% 2.2 ± 0.5 8.3E–02 9.9E–04
EMP3 Epithelial membrane protein 3 U52101 19q13.3 159.3 ± 21.9 65.2 ± 4.9 67% 2.2 ± 0.5 1.5E–03 1.1E–03
HA-1 Minor histocompatibility antigen HA-1 D86976 19p13.3 103.1 ± 11.3 43.5 ± 2.0 78% 2.2 ± 0.4 3.5E–02 1.7E–04 2.8E–02
EBI3 Epstein-Barr virus induced gene 3 L08187 19p13.3 12.8 ± 1.7 5.8 ± 0.2 56% 2.2 ± 0.3 3.1E–02 5.4E–04
SPN Sialophorin M61827 16p11.2 33.2 ± 4.5 15.0 ± 1.3 67% 2.2 ± 0.3 8.3E–02 6.9E–04
CD19 CD19 antigen M84371 16p11.2 14.6 ± 1.3 6.7 ± 0.3 67% 2.2 ± 0.2 1.0E–02 1.1E–04 1.5E–02
ITGAX Integrin, α X M81695 16p11.2 32.2 ± 4.8 13.8 ± 1.0 44% 2.1 ± 0.5 2.1E–01 1.1E–03
P2RX5 Purinergic receptor P2X, ligand-gated ion channel, 5 U49395 17p13.3 17.4 ± 1.8 8.2 ± 0.7 56% 2.1 ± 0.2 3.1E–02 1.7E–04 2.8E–02
HLA-DOA Major histocompatibility complex, class II, DO alpha M31525 6p21.3 13.6 ± 1.6 6.4 ± 0.4 56% 2.1 ± 0.2 7.8E–02 3.5E–04
KLRK1 Killer cell lectin-like receptor subfamily K, member 1 X54870 12p13.2-p12.3 31.3 ± 3.8 13.4 ± 1.5 67% 2.0 ± 0.5 1.2E–03 6.8E–04
ITGB2 Integrin, β 2 (antigen CD18 (p95), lymphocyte function-associated antigen 1 M15395 21q22.3 86.1 ± 7.9 37.8 ± 3.9 78% 2.0 ± 0.4 1.2E–01 1.8E–04 3.1E–02
LAPTM5 Lysosomal-associated multispanning membrane protein-5 U51240 1p34 146.9 ± 21.8 61.8 ± 5.4 67% 1.9 ± 0.6 1.2E–01 2.8E–03
Mitochondrial
UCP2 Uncoupling protein 2 U94592 11 50.2 ± 6.6 22.5 ± 2.0 56% 2.0 ± 0.5 4.8E–03 7.9E–04
Other
IER2 Immediate early response 2 M62831 19p13.13 105.7 ± 17.3 28.7 ± 1.7 78% 3.5 ± 0.7 6.4E–02 3.7E–04
PTMA Prothymosin, alpha M14483 2q35-q36 65.9 ± 17.1 109.3 ± 8.0 44% −3.4 ± 1.5 9.7E–01 3.9E–02
CRIP2 Cysteine-rich protein 2 D42123 14q32.3 25.6 ± 3.2 8.1 ± 0.6 78% 3.2 ± 0.4 1.5E–01 1.1E–04 1.4E–02
PHC2 Polyhomeotic-like 2 U89278 1p34.3 23.4 ± 3.0 7.8 ± 0.3 89% 3.0 ± 0.4 6.3E–02 1.7E–04 2.7E–02
FTH1 Ferritin, heavy polypeptide 1 L20941 11q13 279.0 ± 16.1 122.8 ± 5.7 67% 2.3 ± 0.1 2.9E–02 2.4E–07 7.2E–07 2.452
PFC Properdin P factor, complement M83652 Xp11.3-p11.23 46.6 ± 8.4 19.1 ± 1.8 56% 2.2 ± 0.6 1.8E–01 1.2E–02
IFI44 Interferon-induced protein 44 D28915 1p31.1 13.6 ± 2.5 5.5 ± 0.3 56% 2.0 ± 0.7 7.2E–01 5.5E–03
Proteases or inhibitors
PCOLN3 Procollagen (type III) N-endopeptidase U58048 16q24.3 17.3 ± 2.3 5.4 ± 0.2 78% 3.2 ± 0.4 8.0E–02 1.4E–04 2.0E–02
MME Membrane metallo-endopeptidase J03779 3q25.1-q25.2 24.6 ± 3.4 7.8 ± 0.7 67% 3.1 ± 0.4 2.2E–01 1.9E–04 3.1E–02
ADAM8 A disintegrin and metalloproteinase domain 8 D26579 10q26.3 31.6 ± 4.1 10.6 ± 0.9 67% 3.0 ± 0.4 6.7E–02 4.9E–05 4.5E–03
SERPINB6 Serine (or cysteine) proteinase inhibitor, clade B (ovalbumin), member 6 S69272 6p25 24.7 ± 2.8 8.7 ± 0.5 89% 2.8 ± 0.3 3.0E–02 1.9E–05 9.5E–04
TIMP2 Tissue inhibitor of metalloproteinase 2 M32304 17q25 25.7 ± 3.3 8.8 ± 0.8 78% 2.7 ± 0.5 6.2E–02 2.0E–04 3.3E–02
CTSD Cathepsin D M63138 11p15.5 82.9 ± 16.7 30.3 ± 2.0 56% 2.7 ± 0.6 6.4E–02 1.0E–03
SPINT2 Serine protease inhibitor, Kunitz type, 2 U78095 19q13.1 23.9 ± 1.9 10.5 ± 0.9 89% 2.3 ± 0.2 1.0E–01 7.2E–06 2.1E–04
NOMO1 PM5 protein, centromeric copy X57398 16p13.11 27.3 ± 2.9 11.5 ± 0.9 78% 2.4 ± 0.2 5.6E–02 1.2E–04 1.5E–02
PSME1 Proteasome (prosome, macropain) activator subunit 1 L07633 14q11.2 84.3 ± 11.3 34.9 ± 3.2 56% 2.2 ± 0.5 1.1E–01 5.5E–04
PSMD2 Proteasome 26S subunit, non-ATPase, 2 D78151 3q27.3 36.2 ± 4.5 16.0 ± 1.2 56% 2.3 ± 0.3 1.3E–01 2.1E–04 3.7E–02
CTSB Cathepsin B M14221 8p22 71.3 ± 11.1 29.8 ± 2.8 56% 2.1 ± 0.6 2.2E–01 6.6E–03
PSMA4 Proteasome subunit, α type, 4 D00763 15q24.1 42.8 ± 5.9 17.8 ± 2.6 67% 2.2 ± 0.5 2.9E–01 5.7E–04
Ribosomal
MRPL28 Mitochondrial ribosomal protein L28 U19796 16p13.3 22.0 ± 2.7 6.2 ± 0.3 100% 3.5 ± 0.4 1.2E–01 6.2E–06 1.7E–04 2.299
RPL39 Ribosomal protein L39 D79205 Xq22-q24 465.1 ± 21.5 226.3 ± 24.3 22% 2.1 ± 0.1 1.3E–01 1.9E–05 8.7E–04
RPS4Y1 Ribosomal protein S4, Y-linked 1 M58459 Yp11.3 15.1 ± 7.7 32.2 ± 6.4 78% −5.5 ± 1.4 9.7E–01
Signal transduction
CSRP1 Cysteine and glycine-rich protein 1 M76378 1q32 21.0 ± 2.7 5.7 ± 0.4 89% 3.7 ± 0.5 1.3E–01 2.9E–05 2.0E–03 1.937
GNAZ Guanine nucleotide binding protein (G protein), α z polypeptide J03260 22q11.22 24.9 ± 3.9 7.6 ± 0.5 67% 3.3 ± 0.5 1.0E–01 2.1E–04 3.6E–02
CDC25B Cell division cycle 25B S78187 20p13 63.9 ± 8.5 19.1 ± 1.5 89% 3.3 ± 0.4 8.5E–02 5.6E–05 5.9E–03
ILK Integrin-linked kinase U40282 11p15.5-p15.4 29.2 ± 4.1 8.3 ± 0.7 78% 3.3 ± 0.7 5.6E–02 2.8E–04
PTPRN Protein tyrosine phosphatase, receptor type, N L18983 2q35-q36.1 20.8 ± 2.9 6.4 ± 0.4 78% 3.3 ± 0.5 9.5E–02 1.1E–04 1.4E–02
TSC2 Tuberous sclerosis 2 L48546 16p13.3 30.4 ± 3.3 9.2 ± 0.6 78% 3.3 ± 0.4 3.8E–02 1.2E–05 4.3E–04 1.984
BRD2 Bromodomain containing 2 X62083 6p21.3 70.9 ± 8.0 21.2 ± 2.8 78% 3.4 ± 0.4 1.2E–01 1.9E–05 9.8E–04
CLU Clusterin M63379 8p21-p12 222.8 ± 26.1 72.8 ± 6.8 78% 3.1 ± 0.4 3.6E–02 1.6E–05 7.2E–04
INPP5E Inositol polyphosphate-5-phosphatase U45974 9q34.3 15.1 ± 1.3 4.7 ± 0.2 89% 3.2 ± 0.3* 1.1E–01 3.9E–06 8.9E–05 2.575
LTK Leukocyte tyrosine kinase D16105 15q15.1-q21.1 23.8 ± 3.2 7.2 ± 0.8 89% 3.3 ± 0.4 4.1E–01 2.1E–05 1.2E–03
NRGN Neurogranin X99076 11q24 230.1 ± 27.2 76.5 ± 7.2 78% 3.0 ± 0.4 3.4E–02 2.5E–05 1.6E–03
PLCB2 Phospholipase C, β 2 M95678 15q15 84.0 ± 7.8 25.6 ± 1.9 100% 3.3 ± 0.3 1.9E–02 6.4E–07 2.6E–06 2.064
PKM2 Pyruvate kinase, muscle X56494 15q22 60.0 ± 6.5 18.8 ± 1.5 78% 3.2 ± 0.3 1.3E–02 7.2E–06 2.2E–04
PSD Pleckstrin and Sec7 domain containing X99688 10q24 23.4 ± 3.7 7.8 ± 0.6 67% 3.0 ± 0.5 2.0E–01 2.8E–04
MAP2K3 Mitogen-activated protein kinase kinase 3 D87116 17q11.2 32.9 ± 4.1 11.2 ± 1.1 78% 2.9 ± 0.4 8.5E–02 4.9E–05 4.6E–03
IKBKE Inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase epsilon D63485 1q32.1 18.9 ± 2.1 6.7 ± 0.4 78% 2.8 ± 0.3 1.2E–01 1.4E–04 2.0E–02
FASTK FAST kinase X86779 7q35 20.3 ± 2.4 7.0 ± 0.5 78% 2.9 ± 0.3 2.8E–02 8.5E–05 9.7E–03
LSP1 Lymphocyte-specific protein 1 M33552 11p15.5 48.1 ± 7.2 15.9 ± 1.8 78% 3.0 ± 0.5 4.5E–04 6.9E–05 7.5E–03
RABGGTA Rab geranylgeranyltransferase, α subunit Y08200 14q11.2 23.6 ± 3.1 8.6 ± 0.5 67% 2.7 ± 0.4 5.4E–02 1.4E–04 2.1E–02
MADD MAP-kinase activating death domain AB002356 11p11.2 31.7 ± 4.1 11.2 ± 0.7 67% 2.8 ± 0.4 1.5E–01 1.0E–04 1.3E–02
CSNK2A2 Casein kinase 2, α prime polypeptide M55268 16p13.3-p13.2 17.3 ± 1.8 6.1 ± 0.2 78% 2.9 ± 0.3 1.7E–01 4.3E–05 3.8E–03 2.056
CCND3 Cyclin D3 M92287 6p21 68.3 ± 6.2 23.6 ± 2.3 78% 2.9 ± 0.3 5.0E–02 3.9E–06 8.5E–05
CENTB1 Centaurin, β 1 D30758 17p13.2 61.9 ± 6.3 22.6 ± 1.7 67% 2.7 ± 0.3 1.2E–02 2.5E–05 1.6E–03
PRKACG Protein kinase, cAMP-dependent, catalytic, γ M34182 9q13 41.6 ± 3.1 15.1 ± 1.5 89% 2.8 ± 0.2 1.2E–02 1.7E–06 2.2E–05
YWHAH Tyrosine 3-monooxygenase/ tryptophan 5-monooxygenase activation protein, eta polypeptide D78577 22q12.3 85.7 ± 5.7 31.1 ± 4.3 89% 2.8 ± 0.2 8.9E–02 2.8E–05 1.9E–03
NCF1 Neutrophil cytosolic factor 1 M55067 7q11.23 72.3 ± 13.7 22.1 ± 1.7 78% 3.1 ± 0.8 1.3E–02 1.2E–03
ARHGEF2 Rho/rac guanine nucleotide exchange factor 2 U72206 1q21-q22 25.6 ± 5.7 8.7 ± 0.7 56% 2.7 ± 0.8 2.9E–02 8.8E–03
TRAF1 TNF receptor-associated factor 1 U19261 9q33-q34 20.4 ± 2.6 7.3 ± 0.3 78% 2.6 ± 0.5 1.0E–01 3.4E–04
TRAF4 TNF receptor-associated factor 4 X80200 17q11-q12 15.1 ± 2.2 6.0 ± 0.3 56% 2.5 ± 0.4 6.0E–02 4.6E–04
ARAF1 V-raf murine sarcoma 3611 viral oncogene homolog U01337 Xp11.4-p11.2 32.7 ± 2.8 11.8 ± 0.9 89% 2.8 ± 0.2 1.6E–02 1.6E–06 1.7E–05
CSK C-src tyrosine kinase X59932 15q23-q25 62.4 ± 8.0 22.5 ± 1.6 78% 2.8 ± 0.4 9.4E–03 1.2E–04 1.6E–02
FKBP4 FK506 binding protein 4, 59kDa M88279 12p13.33 23.1 ± 3.2 8.8 ± 0.4 67% 2.6 ± 0.4 4.8E–02 4.8E–04
GNG10 Guanine nucleotide binding protein (G protein), γ 10 U31383 9q32 18.3 ± 3.0 6.7 ± 0.7 56% 2.7 ± 0.4 2.8E–01 3.7E–04
TNFRSF14 Tumor necrosis factor receptor superfamily, member 14 U70321 1p36.3-p36.2 28.1 ± 2.6 10.5 ± 0.7 78% 2.7 ± 0.2 7.5E–03 5.4E–06 1.4E–04
ARHGEF16 Rho guanine exchange factor 16 D89016 1p36.3 18.8 ± 3.0 6.6 ± 0.4 67% 2.6 ± 0.6 3.4E–01 2.9E–03
TNFRSF1B Tumor necrosis factor receptor superfamily, member 1B M32315 1p36.3-p36.2 67.4 ± 11.4 24.6 ± 2.6 67% 2.7 ± 0.5 4.9E–02 2.6E–04 4.9E–02
PIM1 Pim-1 oncogene M16750 6p21.2 34.9 ± 4.5 13.9 ± 1.1 67% 2.3 ± 0.5 1.0E–01 6.0E–04
STK19 Serine/threonine kinase 19 BC016916 6p21.3 12.8 ± 1.5 5.4 ± 0.3 78% 2.4 ± 0.3* 4.3E–02 1.7E–04 2.8E–02
NDRG1 N-myc downstream regulated gene 1 D87953 8q24.3 39.6 ± 5.4 15.5 ± 1.1 67% 2.6 ± 0.3 1.3E–01 4.7E–04
PXN Paxillin U14588 12q24 39.1 ± 3.1 15.3 ± 1.8 89% 2.6 ± 0.2 4.2E–02 1.5E–05 6.7E–04
IHPK1 Inositol hexaphosphate kinase 1 D87452 3p21.31 14.1 ± 1.4 5.6 ± 0.2 67% 2.5 ± 0.2 2.4E–02 1.0E–04 1.3E–02
STAT5A Signal transducer and activator of transcription 5A U43185 17q11.2 27.6 ± 4.3 10.4 ± 0.8 78% 2.7 ± 0.4 5.4E–02 2.8E–04
SQSTM1 Sequestosome 1 U46751 5q35 98.8 ± 10.3 40.8 ± 2.5 67% 2.4 ± 0.3 1.6E–02 4.6E–05 4.1E–03
HDGF Hepatoma-derived growth factor BC018991 X 41.6 ± 5.0 17.3 ± 1.6 67% 2.4 ± 0.3 8.6E–02 2.1E–04 3.7E–02
MPP1 Membrane protein, palmitoylated 1 M64925 Xq28 34.9 ± 4.9 15.5 ± 2.1 67% 2.2 ± 0.3 7.4E–01 7.1E–04
RGL2 Ral guanine nucleotide dissociation stimulator-like 2 U68142 6p21.3 15.7 ± 1.2 6.6 ± 0.3 78% 2.4 ± 0.2 4.5E–02 9.1E–06 3.1E–04 2.042
MX1 Myxovirus (influenza virus) resistance 1, interferon-inducible protein p78 M33882 21q22.3 30.1 ± 8.8 8.7 ± 1.0 67% 3.0 ± 1.2 9.3E–01 1.4E–02
RPS6KA1 Ribosomal protein S6 kinase, 90kDa, polypeptide 1 L07597 3 28.8 ± 4.3 11.3 ± 1.1 67% 2.5 ± 0.4 1.2E–01 1.3E–04 1.8E–02
ITPK1 Inositol 1,3,4-triphosphate 5/6 kinase U51336 14q31 49.0 ± 6.3 20.2 ± 1.4 56% 2.4 ± 0.3 5.7E–02 1.5E–04 2.2E–02
ARF3 ADP-ribosylation factor 3 M74491 12q13 54.1 ± 5.9 23.2 ± 2.7 56% 2.3 ± 0.3 5.7E–02 1.6E–04 2.4E–02
FKBP1A FK506 binding protein 1A M34539 20p13 42.8 ± 5.0 17.5 ± 1.3 67% 2.4 ± 0.3 1.9E–02 1.1E–04 1.4E–02
PRKAG1 Protein kinase, AMP-activated, γ 1 non-catalytic subunit U42412 12q12-q14 15.4 ± 2.0 6.7 ± 0.3 67% 2.3 ± 0.3 5.4E–02 1.4E–03
BIRC1 Baculoviral IAP repeat-containing 1 U80017 ,5q12.2-q13.3 12.5 ± 1.5 5.8 ± 0.3 44% 2.1 ± 0.3 7.5E–02 2.3E–04 4.1E–02
ARHGEF1 Rho guanine nucleotide exchange factor 1 U64105 19q13.13 43.6 ± 3.7 19.2 ± 1.4 67% 2.3 ± 0.2 1.0E–02 1.5E–05 6.0E–04
AVPR1B Arginine vasopressin receptor 1B L37112 1q32 14.3 ± 1.4 6.4 ± 0.4 56% 2.2 ± 0.2 1.1E–01 5.8E–05 6.2E–03
DGKZ Diacylglycerol kinase, zeta U51477 11p11.2 32.6 ± 2.9 13.8 ± 0.8 67% 2.4 ± 0.2 2.8E–02 9.1E–06 3.2E–04
PARK7 Parkinson disease (autosomal recessive, early onset) 7 D61380 1p36.33-p36.12 58.4 ± 5.9 26.2 ± 3.0 56% 2.2 ± 0.2 1.3E–01 1.8E–04 2.9E–02
RGS2 G0/G1 switch regulatory gene # 8 L13391 1q31 60.3 ± 12.5 21.4 ± 3.2 67% 2.3 ± 0.8 2.7E–01 8.1E–03
PPM1F Protein phosphatase 1F (PP2C domain containing) D13640 22q11.22 29.0 ± 4.3 11.2 ± 1.0 67% 2.4 ± 0.6 1.7E–01 1.3E–03
ARF5 ADP-ribosylation factor 5 M57567 7q31.3 42.4 ± 6.2 16.2 ± 1.4 78% 2.4 ± 0.5 5.1E–02 4.0E–04
PTK2B PTK2B protein tyrosine kinase 2 beta U43522 8p21.1 13.2 ± 2.2 5.8 ± 0.3 56% 2.3 ± 0.4 5.0E–03 1.5E–03
PIM1 Pim-1 oncogene M54915 6p21.2 54.7 ± 6.4 23.5 ± 1.8 78% 2.1 ± 0.4 1.1E–01 3.4E–04
INPPL1 Inositol polyphosphate phosphatase-like 1 L36818 11q23 19.4 ± 2.8 8.2 ± 0.9 56% 2.4 ± 0.3 1.3E–01 2.1E–04 3.8E–02
PCMT1 Protein-L-isoaspartate (D-aspartate) O-methyltransferase D25547 6q24-q25 13.2 ± 2.3 5.8 ± 0.2 44% 2.3 ± 0.4 1.3E–01 1.4E–03
ARHGAP1 Rho GTPase activating protein 1 U02570 11p12-q12 25.6 ± 3.3 11.8 ± 0.8 56% 2.2 ± 0.3 3.4E–02 6.9E–04
FKBP2 FK506 binding protein 2, 13kDa M75099 11q13.1-q13.3 23.6 ± 3.1 10.8 ± 0.9 56% 2.2 ± 0.3 7.0E–02 9.7E–04
TNIP1 TNFAIP3 interacting protein 1 D30755 5q32-q33.1 29.1 ± 4.0 12.8 ± 0.9 56% 2.3 ± 0.3 5.0E–02 2.2E–04 4.0E–02
IRAK1 Interleukin-1 receptor-associated kinase 1 L76191 Xq28 32.7 ± 2.8 15.5 ± 1.1 56% 2.1 ± 0.2 8.1E–02 2.0E–05 1.0E–03
RHOG Ras homolog gene family, member G (rho G) X61587 11p15.5-p15.4 51.9 ± 8.5 19.7 ± 1.6 78% 2.2 ± 0.7 8.0E–03 4.4E–03
RASSF2 Ras association (RalGDS/AF-6) domain family 2 D79990 20pter-p12.1 29.3 ± 4.5 12.3 ± 1.7 56% 2.1 ± 0.6 4.3E–01 3.7E–03
NEDD8 Neural precursor cell expressed, developmentally down-regulated 8 D23662 14q11.2 53.1 ± 6.8 23.8 ± 2.6 67% 2.0 ± 0.5 1.9E–01 8.6E–04
CAP1 CAP, adenylate cyclase-associated protein 1 L12168 1p34.2 134.7 ± 18.9 58.4 ± 5.4 67% 2.1 ± 0.5 4.2E–02 3.1E–03
ZAP70 Zeta-chain (TCR) associated protein kinase L05148 2q12 36.6 ± 4.3 16.3 ± 1.4 78% 2.0 ± 0.4 4.4E–02 5.1E–04
FKBP8 FK506 binding protein 8 L37033 19p12 27.4 ± 3.7 12.3 ± 1.1 56% 2.0 ± 0.5 1.4E–02 7.6E–04
GRK6 G protein-coupled receptor kinase 6 L16862 5q35 23.4 ± 3.6 10.5 ± 1.0 56% 2.0 ± 0.5 3.0E–02 1.3E–03
MAP2K2 Mitogen-activated protein kinase kinase 2 L11285 7q32 29.1 ± 4.0 12.9 ± 0.7 67% 2.0 ± 0.5 1.5E–02 1.3E–03
PTP4A2 Protein tyrosine phosphatase type IVA, member 2 U14603 1p35 76.6 ± 9.4 35.3 ± 2.6 56% 1.9 ± 0.4 2.0E–02 5.7E–04
RAC1 Ras-related C3 botulinum toxin substrate 1 NM_006908 7p22 20.1 ± 3.4 8.9 ± 0.6 56% 2.0 ± 0.5 3.4E–02 3.2E–03
MX2 Myxovirus (influenza virus) resistance 2 M30818 21q22.3 20.6 ± 3.9 8.7 ± 0.9 56% 1.9 ± 0.7 7.0E–01 8.6E–03
FYB FYN binding protein (FYB-120/130) U93049 5p13.1 5.2 ± 0.7 10.8 ± 1.9 67% −2.3 ± 0.2 4.3E–01 7.1E–03
Structural
MYL9 Myosin, light polypeptide 9, regulatory J02854 20q11.23 17.9 ± 4.7 4.4 ± 0.2 67% 4.1 ± 1.1* 1.4E–01 1.1E–03
PLEC1 Plectin 1, intermediate filament binding protein U53204 8q24 37.8 ± 6.0 9.6 ± 0.9 78% 3.9 ± 0.6 3.0E–02 3.4E–05 2.8E–03
GFAP Glial fibrillary acidic protein S40719 17q21 19.9 ± 3.2 7.2 ± 0.5 67% 2.8 ± 0.4 1.9E–01 3.5E–04
BECN1 Beclin 1 L38932 17q21 40.3 ± 4.0 14.8 ± 1.2 78% 2.7 ± 0.3 5.7E–02 1.6E–05 7.4E–04
MYH9 Myosin, heavy polypeptide 9, non-muscle M31013 22q13.1 149.8 ± 13.7 55.4 ± 4.4 78% 2.7 ± 0.2 5.8E–02 6.7E–06 1.9E–04
KRT1 Keratin 1 M98776 12q12-q13 16.7 ± 2.9 6.2 ± 0.3 67% 2.5 ± 0.6 4.9E–01 3.0E–03
NUMA1 Nuclear mitotic apparatus protein 1 Z14227 11q13 25.7 ± 3.4 9.5 ± 0.6 78% 2.7 ± 0.4 5.5E–02 3.4E–04
PDLIM1 PDZ and LIM domain 1 (elfin) U90878 10q22-q26.3 26.6 ± 2.9 11.0 ± 0.8 56% 2.4 ± 0.3 3.6E–02 2.5E–05 1.6E–03
MYL6 Myosin, light polypeptide 6, alkali, smooth muscle and non-muscle M31212 12 360.2 ± 18.0 138.0 ± 13.6 89% 2.6 ± 0.1 2.4E–02 3.9E–06 8.1E–05
SAFB Scaffold attachment factor B L43631 19p13.3-p13.2 25.4 ± 2.6 10.3 ± 0.7 67% 2.5 ± 0.3 9.9E–02 2.8E–05 1.9E–03
MAPT Microtubule-associated protein tau AH005895 17q21.1 26.3 ± 4.6 10.1 ± 0.8 56% 2.4 ± 0.6 9.5E–02 2.2E–03
TPM3 Tropomyosin 3 BC000771 149.2 ± 16.9 64.5 ± 5.5 67% 2.3 ± 0.3 7.7E–02 1.7E–04 2.9E–02
HSU34301 Nonmuscle myosin heavy chain IIB U34301 17 17.0 ± 2.1 7.4 ± 0.9 56% 2.3 ± 0.3 1.0E–01 1.2E–04 1.7E–02
KNS2 Kinesin 2 60/70kDa L04733 14q32.3 16.1 ± 2.2 6.5 ± 0.4 56% 2.5 ± 0.3 6.5E–02 1.6E–04 2.4E–02
TUBB2C Tubulin, β 2C AK026167 9q34 53.2 ± 4.8 23.9 ± 2.5 67% 2.2 ± 0.2 1.7E–01 3.9E–05 3.2E–03
TUBA3 Tubulin, α 3 X01703 12q12-12q14.3 37.7 ± 5.5 14.5 ± 1.8 67% 2.4 ± 0.5 3.2E–01 9.7E–04
TNNC1 Troponin C, slow M37984 3p21.3-p14.3 11.2 ± 1.2 5.1 ± 0.1 56% 2.2 ± 0.2* 6.7E–02 1.7E–04 2.7E–02
MSN Moesin M69066 Xq11.2-q12 178.8 ± 27.9 72.8 ± 6.9 67% 2.2 ± 0.6 5.4E–02 4.1E–03
Transcription
RELA V-rel reticuloendotheliosis viral oncogene homolog A L19067 11q13 39.8 ± 4.3 11.1 ± 1.1 100% 3.6 ± 0.4 4.1E–03 7.9E–07 4.8E–06 1.964
FOS V-fos FBJ murine osteosarcoma viral oncogene homolog V01512 14q24.3 51.4 ± 14.0 13.9 ± 3.4 67% 3.4 ± 1.1 2.8E–02 4.5E–03
NFE2 Nuclear factor (erythroid-derived 2) S77763 12q13 32.3 ± 4.1 9.5 ± 1.4 78% 3.4 ± 0.4 3.5E–01 1.5E–05 6.1E–04
IRF5 Interferon regulatory factor 5 U51127 7q32 29.0 ± 4.6 10.2 ± 1.0 78% 2.9 ± 0.5 8.4E–02 2.0E–04 3.6E–02
ZNFpT1 Zinc-finger protein X65230 15.2 ± 2.2 5.3 ± 0.1 78% 2.9 ± 0.4 6.4E–02 5.9E–04
SF1 Splicing factor 1 L49380 11q13 46.4 ± 4.3 16.2 ± 0.6 78% 2.9 ± 0.3 3.7E–02 1.5E–05 6.4E–04 2.307
HCFC1 Host cell factor C1 L20010 Xq28 26.0 ± 2.9 8.9 ± 0.5 78% 2.9 ± 0.3 2.7E–02 2.5E–05 1.6E–03
SREBF1 Sterol regulatory element binding transcription factor 1 U00968 17p11.2 25.6 ± 2.1 8.9 ± 0.6 89% 2.9 ± 0.2 6.3E–02 1.6E–06 1.9E–05 2.019
POLR2A Polymerase (RNA) II (DNA directed) polypeptide A X74874 17p13.1 18.9 ± 2.7 6.8 ± 0.5 67% 2.8 ± 0.4 7.3E–02 7.7E–05 8.7E–03
MAZ MYC-associated zinc finger protein M94046 16p11.2 29.7 ± 3.1 10.7 ± 0.6 78% 2.8 ± 0.3 7.9E–03 3.1E–05 2.3E–03
TCFL1 Transcription factor-like 1 D43642 1q21 38.2 ± 5.3 14.3 ± 0.7 67% 2.7 ± 0.4 9.9E–02 4.7E–04
IRF3 Interferon regulatory factor 3 Z56281 19q13.3-q13.4 25.1 ± 1.7 9.5 ± 0.5 78% 2.6 ± 0.2 2.2E–02 9.3E–07 6.5E–06 2.303
BTG2 BTG family, member 2 U72649 1q32 40.3 ± 8.1 14.8 ± 1.1 67% 2.5 ± 0.7 8.1E–02 3.0E–03
VGLL4 Vestigial like 4 D50911 3p25.2 16.4 ± 2.1 6.5 ± 0.3 67% 2.5 ± 0.3 1.3E–01 3.1E–04
RNPC2 RNA-binding region (RNP1, RRM) containing 2 L10910 20q11.23 16.1 ± 2.1 6.4 ± 0.6 67% 2.5 ± 0.3 2.4E–01 4.7E–04
NBL1 Neuroblastoma, suppression of tumorigenicity 1 D28124 1p36.13-p36.11 16.6 ± 1.9 6.8 ± 0.6 67% 2.4 ± 0.3 2.4E–01 1.2E–04 1.6E–02
NCOR2 Nuclear receptor co-repressor 2 U37146 12q24 26.6 ± 2.9 11.2 ± 0.8 67% 2.4 ± 0.3 4.2E–02 5.1E–05 5.0E–03
JUND Jun D proto-oncogene X56681 19p13.2 114.4 ± 9.6 47.5 ± 3.9 89% 2.4 ± 0.2 1.4E–02 3.0E–06 4.8E–05
TRIM28 Tripartite motif-containing 28 U95040 19q13.4 44.0 ± 5.2 17.3 ± 1.2 67% 2.3 ± 0.5 1.4E–02 2.2E–04 4.0E–02
POLR2E Polymerase (RNA) II (DNA directed) polypeptide E D38251 19p13.3 28.4 ± 3.5 11.2 ± 1.1 78% 2.3 ± 0.5 7.7E–02 6.5E–04
BCL6 B-cell CLL/lymphoma 6 (zinc finger protein 51) U00115 3q27 12.0 ± 2.1 5.2 ± 0.2 56% 2.3 ± 0.4 5.3E–02 2.6E–03
PML Promyelocytic leukemia M79462 15q22 10.7 ± 1.3 4.7 ± 0.2 56% 2.3 ± 0.3* 1.5E–02 5.1E–04
CEBPB CCAAT/enhancer binding protein (C/EBP), β X52560 20 80.6 ± 14.7 32.2 ± 2.9 67% 2.2 ± 0.7 3.7E–01 1.3E–02
SFRS11 Splicing factor, arginine/serine-rich 11 M74002 1p31 20.0 ± 2.3 8.4 ± 0.8 78% 2.2 ± 0.5 1.4E–01 3.4E–04
SRF Serum response factor J03161 6p21.1 18.6 ± 2.5 8.3 ± 0.6 56% 2.2 ± 0.3 1.7E–01 3.9E–04
YY1 YY1 transcription factor M77698 14q 13.4 ± 1.5 6.3 ± 0.6 56% 2.1 ± 0.2 2.2E–01 2.8E–04
LYL1 Lymphoblastic leukemia derived sequence 1 M22638 19p13.2 14.2 ± 1.2 6.8 ± 0.3 56% 2.1 ± 0.2 4.2E–02 4.3E–05 3.7E–03
SUPT4H1 Suppressor of Ty 4 homolog 1 U43923 17q21-q23 20.1 ± 3.2 8.9 ± 0.8 56% 2.0 ± 0.5 2.1E–01 2.9E–03
TAF15 TAF15 RNA polymerase II U51334 17q11.1-q11.2 23.9 ± 2.8 11.1 ± 1.0 67% 1.9 ± 0.4 3.9E–02 6.5E–04
Translation
EIF3S9 Eukaryotic translation initiation factor 3, subunit 9 eta U78525 7p22.3 19.6 ± 2.2 9.0 ± 0.7 67% 2.2 ± 0.2 2.3E–02 2.6E–04 4.8E–02
Transport
TCIRG1 T-cell, immune regulator 1, ATPase, H + transporting, lysosomal V0 protein a isoform 3 U45285 11q13.4-q13.5 31.4 ± 3.3 6.7 ± 0.5 100% 4.7 ± 0.5 1.2E–01 1.6E–07 1.6E–07 2.728
TETRAN Tetracycline transporter-like protein L11669 4p16.3 26.8 ± 2.7 6.8 ± 0.5 100% 3.9 ± 0.4 1.2E–01 1.6E–06 1.8E–05 2.404
HD Huntingtin (Huntington disease) L12392 4p16.3 22.3 ± 2.7 6.5 ± 0.3 78% 3.5 ± 0.4 9.1E–02 4.9E–05 4.6E–03 2.082
ATP6AP1 Human mRNA for ORF, Xq terminal portion. D16469 Xq28 31.7 ± 4.2 9.0 ± 0.8 89% 3.5 ± 0.5 6.5E–02 8.8E–06 2.9E–04
GGA3 Golgi associated, γ adaptin ear containing, ARF binding protein 3 D63876 17q25.2 33.9 ± 3.3 10.1 ± 0.6 89% 3.4 ± 0.3 7.1E–02 2.3E–06 3.2E–05 2.236
ATP6V0C ATPase, H + transporting, lysosomal, V0 subunit c M62762 16p13.3 69.7 ± 8.8 21.2 ± 1.9 89% 3.3 ± 0.4 9.8E–02 4.3E–05 3.7E–03
AP2M1 Adaptor-related protein complex 2, mu 1 subunit D63475 3q28 42.6 ± 5.5 14.2 ± 1.3 67% 3.0 ± 0.4 4.1E–02 1.3E–04 1.8E–02
SLC2A3 Solute carrier family 2 (facilitated glucose transporter), member 3 M20681 12p13.3 26.0 ± 2.3 9.3 ± 1.0 89% 2.8 ± 0.2 1.3E–01 9.2E–06 3.3E–04
SLC9A1 Solute carrier family 9 (sodium/hydrogen exchanger), isoform 1 (antiporter, Na + /H +, amiloride sensitive) S68616 1p36.1-p35 19.0 ± 2.4 6.8 ± 0.2 67% 2.8 ± 0.4 4.2E–02 2.3E–04 4.2E–02
SLC11A1 Solute carrier family 11 (proton-coupled divalent metal ion transporters), member 1 D50402 2q35 19.1 ± 3.0 6.8 ± 0.6 78% 2.8 ± 0.4 1.9E–01 4.7E–04
MLC1 Megalencephalic leukoencephalopathy with subcortical cysts 1 D25217 22q13.33 16.1 ± 2.6 5.8 ± 0.5 56% 2.8 ± 0.4* 2.2E–02 5.7E–04
SEC24C FLJ44715 gene product D38555 10q22.3 21.3 ± 2.9 8.0 ± 0.6 67% 2.7 ± 0.4 8.3E–02 3.3E–04
CLTA Clathrin, light polypeptide (Lca) M20471 9p13 73.6 ± 9.1 26.7 ± 1.9 89% 2.8 ± 0.3 4.6E–02 3.8E–05 3.2E–03
AP2B1 Adaptor-related protein complex 2, β 1 subunit M34175 17q11.2-q12 28.2 ± 4.0 10.3 ± 1.2 67% 2.5 ± 0.5 1.7E–01 2.3E–04 4.2E–02
AP1B1 Adaptor-related protein complex 1, β 1 subunit L13939 22q12.2 23.2 ± 3.0 9.5 ± 0.8 67% 2.4 ± 0.3 4.3E–02 6.1E–04
TXN2 Thioredoxin 2 U78678 22q13.1 18.9 ± 2.5 7.5 ± 0.5 67% 2.5 ± 0.3 3.8E–02 2.8E–04
CRIP1 Cysteine-rich protein 1 (intestinal) U09770 7q11.23 35.3 ± 3.9 15.9 ± 1.1 56% 2.2 ± 0.2 9.2E–02 1.4E–04 2.0E–02
NAPA N-ethylmaleimide-sensitive factor attachment protein, alpha U39412 19q13.33 15.0 ± 2.0 6.4 ± 0.4 67% 2.1 ± 0.5 5.5E–02 3.2E–03
Ubiquitin
UBE1 Ubiquitin-activating enzyme E1 M58028 Xp11.23 46.4 ± 6.9 11.3 ± 1.0 89% 4.1 ± 0.6 8.6E–03 3.3E–05 2.5E–03
USP11 Ubiquitin specific protease 11 U44839 Xp11.23 60.2 ± 7.4 18.9 ± 1.1 78% 3.2 ± 0.4 1.2E–02 5.0E–05 4.9E–03
UBC Ubiquitin M26880 12q24.3 198.0 ± 14.3 66.3 ± 10.5 89% 3.0 ± 0.2 2.4E–01 7.8E–06 2.4E–04
UBE1L Ubiquitin-activating enzyme E1-like L13852 3p21 54.8 ± 4.6 18.6 ± 1.3 89% 2.9 ± 0.2 4.7E–02 9.4E–07 7.5E–06 1.966
CUL7 Cullin 7 D38548 6p21.1 14.7 ± 2.0 5.8 ± 0.3 56% 2.5 ± 0.3 6.6E–02 2.2E–04 4.0E–02
RAD23A RAD23 homolog A (S. cerevisiae) D21235 19p13.2 15.6 ± 1.6 6.6 ± 0.3 78% 2.4 ± 0.2 8.8E–02 2.0E–04 3.4E–02
UBE2V1 Homo sapiens UEV-1 BC000468 20q13.2 25.9 ± 3.6 11.2 ± 1.3 67% 2.3 ± 0.3 5.3E–01 4.4E–04
UFD1L Ubiquitin like protein U64444 22q11.21 22.8 ± 2.7 10.1 ± 0.9 67% 2.0 ± 0.4 1.3E–01 5.7E–04
USP4 Ubiquitin specific protease 4 (proto-oncogene) U20657 3p21.3 14.8 ± 1.7 6.6 ± 0.3 67% 2.0 ± 0.4 4.0E–02 5.7E–04
Unknown
LRRC14 Leucine rich repeat containing 14 D25216 8q24.3 31.7 ± 4.1 9.2 ± 1.1 89% 3.4 ± 0.4 5.3E–02 8.8E–06 2.8E–04
WDR42A WD repeat domain 42A U06631 1q22-q23 23.6 ± 2.2 7.4 ± 0.5 89% 3.2 ± 0.3 8.1E–02 3.1E–06 5.6E–05 2.1
C1orf16 Chromosome 1 open reading frame 16 D87437 1q35 19.3 ± 2.2 6.5 ± 0.3 78% 3.0 ± 0.3 4.8E–02 2.8E–05 2.0E–03 2.116
C1orf19 Chromosome 6 open reading frame 9 U89336 6p21.3 41.6 ± 4.1 13.8 ± 1.3 78% 2.8 ± 0.3 7.6E–05 3.9E–06 8.1E–05
KIAA0056 KIAA0056 protein D29954 11q25 17.4 ± 2.8 6.2 ± 0.4 67% 2.6 ± 0.6 2.7E–01 1.5E–03
C21orf2 Chromosome 21 open reading frame 2 U84569 21q22.3 25.2 ± 3.3 9.8 ± 0.9 67% 2.6 ± 0.3 8.5E–02 1.0E–04 1.2E–02
PRCC Papillary renal cell carcinoma X99720 1q21.1 20.1 ± 2.1 7.7 ± 0.6 78% 2.6 ± 0.3 1.4E–01 2.7E–05 1.8E–03
KIAA0226 KIAA0226 gene product D86979 3q29 20.6 ± 2.3 7.8 ± 0.5 78% 2.6 ± 0.3 6.4E–02 5.4E–05 5.5E–03
ARMCX6 Hypothetical protein FLJ20811 L20773 Xq21.33-q22.3 25.6 ± 2.7 10.1 ± 0.9 56% 2.5 ± 0.3 4.6E–02 3.0E–05 2.1E–03
UBAP2L Ubiquitin associated protein 2-like D63478 1q22 13.2 ± 1.9 5.4 ± 0.2 67% 2.5 ± 0.3 5.5E–02 7.3E–04
ARMET Arginine-rich, mutated in early stage tumors M83751 3p21.1 21.2 ± 2.5 8.8 ± 0.5 56% 2.4 ± 0.3 4.9E–02 1.1E–04 1.5E–02
KIAA0174 KIAA0174 gene product D79996 16q22.2 28.6 ± 3.2 12.0 ± 1.3 67% 2.4 ± 0.3 2.1E–01 1.1E–04 1.4E–02
TATDN2 TatD DNase domain containing 2 D86972 3p25.3 13.8 ± 1.5 5.8 ± 0.5 67% 2.4 ± 0.3 1.4E–01 1.0E–04 1.3E–02
PFAAP5 Phosphonoformate immuno-associated protein 5 U50535 13 15.7 ± 1.5 6.8 ± 0.6 67% 2.3 ± 0.2 1.7E–01 7.2E–05 8.0E–03
HSHRTPSN Retrotransposon Z48633 10.6 ± 2.1 4.4 ± 0.2 44% 2.2 ± 0.6* 7.9E–02 6.6E–03
TAGLN2 Transgelin 2 D21261 1q21-q25 278.8 ± 30.1 111.3 ± 10.9 89% 2.2 ± 0.5 1.6E–02 1.3E–03
TRIM26 Tripartite motif-containing 26 U09825 6p21.3 21.4 ± 2.8 9.6 ± 0.6 56% 2.2 ± 0.3 1.4E–01 1.0E–03
NUP188 Nucleoporin 188kDa D79991 9q34.13 13.7 ± 1.3 6.2 ± 0.2 67% 2.2 ± 0.2 4.2E–02 1.6E–04 2.3E–02
FAM53B Family with sequence similarity 53, member B D50930 10q26.2 16.8 ± 2.8 7.2 ± 0.4 56% 2.1 ± 0.5 9.7E–02 3.8E–03
C21orf33 Chromosome 21 open reading frame 33 U53003 21q22.3 12.2 ± 1.1 5.7 ± 0.2 44% 2.1 ± 0.2 6.7E–02 2.1E–05 1.2E–03
CYFIP2 Cytoplasmic FMR1 interacting protein 2 L47738 5q34 29.2 ± 3.9 13.4 ± 1.2 67% 2.0 ± 0.5 2.6E–02 1.4E–03
DXYS155E DNA segment on chromosome X and Y (unique) 155 expressed sequence L03426 Xp22.32, Ypter-p11.2 15.9 ± 2.1 6.7 ± 0.4 78% 2.0 ± 0.5 2.5E–03 1.3E–03
BRD3 Bromodomain containing 3 D26362 9q34 15.9 ± 1.8 7.2 ± 0.6 67% 2.0 ± 0.4 2.6E–01 5.1E–04
FAM50A DNA segment on chromosome X (unique) 9928 expressed sequence D83260 Xq28 16.6 ± 1.7 7.6 ± 0.4 33% 2.0 ± 0.2 6.3E–04 3.0E–05 2.2E–03
NK4 Natural killer cell transcript 4 M59807 16p13.3 116.7 ± 18.5 49.2 ± 6.4 56% 1.9 ± 0.6 6.7E–03 2.8E–03
a

Data are mean ± SE except average ± SE. Genes were filtered based on the average frequency of 10 and average change in expression of at least 2-fold. Fold changes for genes with increased expression are represented as the ratio of RA average frequency/normal average frequency, whereas genes with reduced expression are represented as the negative reciprocal of that ratio. SE was also calculated for the fold change. Average frequency of expression and its SE were calculated for 9 RA and 13 control samples. The results of three separate statistical analyses performed on the data are shown. A Student t test was performed to identify statistically significant differences between samples with a threshold of P < 0.5. Two Welch ANOVA (26) analyses using different multiple testing corrections were performed. The first, performed according to the methods of Benjamini and Hochberg (28) to calculate the FDR, was used with a limit of 0.05. The second, based on Bonferroni (29,30) FWER, was calculated with P value cutoff of <0.05. The rightmost column represents genes that comprise the class prediction analysis. Chromosomal map units shown are based on GenBank information.

*

Genes with an asterisk have control samples with frequencies <5 ppm and are called absent in >50% of samples. Therefore, the fold change calculation may not accurately reflect the actual difference in expression. Annotation was based on GO, Gene, and PubMed to categorize the genes. Genes with different Affymetrix identifiers were not removed from the table.

Unsupervised Clustering

An unsupervised clustering analysis was performed on the 330 genes that passed the initial filtration, based on a hierarchical correlation coefficient algorithm (21). Samples were grouped based on similarity of expression. The resulting dendrogram describes the sample relationships by grouping the RA samples and controls by their expression patterns (Figure 1). Figure 1A depicts a region where expression levels in the RA samples were increased compared with the normal samples. This analysis suggests that there are significant differences in the gene expression of RA and control samples.

Figure 1.

Figure 1

Unsupervised hierarchical cluster analysis of RNA from 9 RA and 13 control PBMC samples. Total RNA samples were analyzed on oligonucleotide arrays as described. In no case were samples pooled. Genes were selected for analysis if they had a present call, a frequency greater than 10 ppm, and two-fold change expression in five of nine RA samples. The expression patterns of 330 genes are displayed in a dendrogram where columns represent each sample and rows represent individual genes. Genes are colored on a gradient (from −10-fold to 10-fold), with those increase in expression relative to the average of the control in red. Those that decrease are in blue, and those with little or no change are in yellow. A, region where expression levels in the RA samples were increased compared with the normal samples.

ANOVA Analysis

To minimize the inclusion of genes not related to the disease state, several statistical approaches were used. The 330 transcripts that passed the initial filtration (Table 2) were subjected to a Student t test and a Welch ANOVA with two multiple testing corrections (22). To control for a proportion of genes that may appear in the analysis by chance, an FDR was calculated set to a threshold of 5%. This analysis defines a proportion of the genes that are expected to occur by chance relative to the total number of transcripts identified; 326 transcripts were called significant with this analysis (Table 2). In addition, the more stringent Bonferroni FWER using a P value cutoff of 0.05 was also performed, with 189 transcripts passing this analysis (Table 2).

Class Prediction

A k-nearest neighbor analysis was performed to identify a gene set that may distinguish the RA samples from normals. The prediction strength was evaluated using the 330 genes shown in Table 2. A list of predictor genes was assembled using the k-nearest neighbor method (26) to organize genes based on normalized expression levels. Cross-validation analyses comparing each sample to the model generated by the remaining samples were used to optimize the analysis parameters. This resulted in a number of neighbors value of 6 with a decision cutoff P value of 0.2 to predict expression patterns in RA vs. controls. Twenty-nine transcripts comprise the prediction gene set. The 29 prediction transcripts were grouped based on a hierarchical correlation to show the relationships (Figure 2).

Figure 2.

Figure 2

Class prediction. Using a class prediction algorithm, a list of genes that most consistently distinguished diseased vs. normal samples was generated. Classification was generated by the k-nearest neighbors algorithm (26). The number of neighbors selected was six, with a decision cutoff for P value ratio of 0.2. The final list was determined by an iterative cross-validation process in which the best combination of number of genes and neighbors was found to derive the most discriminating list. In the cross-validation mode, each sample in turn was set aside as the test article, and the remainder of the samples were used to generate the model, which was then evaluated on the test article. (A) Fold change and P values of the 29 prediction genes. (B) Unsupervised hierarchical cluster analysis of the 29 genes. The expression patterns of 29 genes are displayed in a dendrogram where columns represent each sample and rows represent individual genes. Genes are colored on a gradient (from −10-fold to 10-fold) with those increase in expression relative to the average of the control in red. Those that decrease are in blue, and those with little or no change are in yellow.

Characterization of the RA Disease-Related Genes

The 330 differentially regulated transcripts were categorized into functional groups and are presented as the average fold change of RA frequency compared with that of the controls (Table 2). This analysis clustered the genes into 19 functional classes and highlighted one chromosomal location. Ten genes with increased expression in the RA PBMCs compared with normal controls map to an RA susceptibility locus, 6p21.3 (27) (Table 3). The functional classes are diverse and include genes involved in calcium binding, chaperones, cytokines, transcription, translation, signal transduction, extracellular matrix, integral to plasma membrane, integral to intracellular membrane, mitochondrial, ribosomal, structural, enzymes, and proteases. Many of these 330 genes or gene products are known to be differentially regulated in RA. Twenty-five genes were classified as unknown because they either coded for a hypothetical protein or were identified as an open reading frame of unknown function.

Table 3.

Genes with increased expression in RA compared with normal PBMCs at the RA susceptibility locus 6p21.3

Gene symbol no. Gene name GenBank acc.
MLN Motilin X15393
AGPAT1 1-acylglycerol-3-phosphate O-acyltransferase 1 U56417
HLA-DQA1 Major histocompatibility complex, class II, DQ α 1 M34996
MICB MHC class I chain-related gene B U65416
HLA-DOA Major histocompatibility complex, class II, DO alpha M31525
BRD2 Bromodomain containing 2 X62083
STK19 Serine/threonine kinase 19 BC016916
RGL2 Ral guanine nucleotide dissociation stimulator-like 2 U68142
C1orf19 Chromosome 6 open reading frame 9 U89336
TRIM26 Tripartite motif-containing 26 U09825

The k-nearest neighbor analysis identified genes that may be preferentially regulated in the RA samples. Of the 29 genes identified by the class prediction analysis (Figure 2B) to be expressed in the RA PBMCs compared with the controls, only RELA (NFκB p65) (28), IGF2 (insulin-like growth factor 2) (29)], FTH1 (ferritin heavy chain) (30), and SELP (selectin P) (31) have previously been associated with RA. Furthermore, both NF-κB and selectin P have been used as therapeutic targets in animal models (32,33). INPP5E (inositol polyphosphate-5-phosphatase E), STAB1 (stabilin), AGPAT1 (1-acylglycerol-3-phosphate O-acyltransferase 1), TCIRG1 (T-cell, immune regulator 1, ATPase, H+ transporting, lysosomal V0 protein A isoform 3), HD (Huntingtin), SREBF1 (sterol regulatory element binding factor 1), and IRF3 (interferon regulatory factor 3) are examples of genes that have not previously been associated with RA.

DISCUSSION

In this study, the mRNA levels of 6800 genes were measured in PBMCs from RA patients with active disease and normal individuals. All patients were on DMARD therapy that included methotrexate. Three hundred thirty differentially expressed transcripts were detected in at least 50% of the patients and exhibited a minimum of a two-fold change in expression from normal individuals. A number of genes previously thought to be involved in RA pathogenesis were detected in this study. These include the transcripts for TNF receptor TNFRSF1B (p75) and CCL5 (RANTES). TNFα has a key role in RA, and the expression of mRNA and protein of TNF receptors is increased in RA synovial membranes and sera (3436). In murine models, as well as TNFα transgenic and receptor knockout mice, the pathogenic activity of TNF has been well documented. Furthermore, both the soluble form of the TNF receptor and antibodies against TNF are efficacious in animal models and are effective therapies for RA (4,68,37,38). CCL5 is a chemokine expressed in the serum and synovial joints of patients with RA and is likely to play important roles in recruitment of inflammatory cells (39). A polyclonal antibody to RANTES improved symptoms in animals with adjuvant induced arthritis (40). RNA transcripts encoding proteins from a number of signaling pathways, including NF-κB, were present in increased amounts in individuals with RA, and many of these are targets for therapeutic blockade (41). NF-κB (RELA) has important roles in the production of inflammatory cytokines such as IL-1 and TNF (28). The presence of these known genes in the data set further validates the array data and analysis.

A k-nearest neighbor analysis was applied to the data set to identify genes preferentially expressed in the PBMCs from RA patients compared with controls. Twenty-nine genes were identified. Some of these genes have been previously identified as being differentially regulated in RA and include IGF2 (29), FTH1 (30), and SELP (31). SELP contributes to many inflammatory diseases and has been shown to mediate leukocyte interaction with endothelial cell wall (42). Levels of SELP are increased in the synovial fluid of RA patients (43). In the murine collagen-induced arthritis model, the deletion of SELP resulted in more severe disease compared with wild-type mice (44).

Many genes not previously known as being differently regulated in RA were also identified, for example, TCIRG1 (T-cell, immune regulator 1), INPP5E (inositol polyphosphate-5-phosphatase E), and STAB1 (stabilin). TCIRG1 is a seven-transmembrane, novel T cell protein that plays a role in T cell activation (45). Antibodies to TCIRG1 (TIRC7) prevent human T cell proliferation in vitro, inhibit type I subset–specific IFNγ and IL-2, but not the type II subset cytokine IL-4. A TIRC7 antibody prolonged survival in a rat model of acute kidney allograft rejection (45). TIRC7-null mice have disrupted T and B cell responses in vitro and in vivo, suggesting that TIRC7 may play a role in T and B lymphocyte balance (46).

INPP5E, a member of the inositol polyphosphate 5-phosphatase family, similar to INPP5D (Table 2), regulates PI-3 kinase signal transduction (47). AGPAT1 (1-acylglycerol-3-phosphate O-acyltransferase 1) catalyzes the conversion of lysophosphatidic acid (LPA) to phosphatidic acid (PA). LPA and PA are two phospholipids involved in signal transduction and phospholipid synthesis (48). Overexpression of AGPAT-1 in cell lines leads to the expression of both TNF-α and IL-6 in cells stimulated with IL-1β, suggesting that AGPAT-1 overexpression may amplify cellular signaling responses from cytokines (49).

Interestingly, 10 transcripts, including AGPAT1, differentially regulated in the RA PBMC from this study map to chromosome region 6p21.3, the major histocompatibility (MHC) locus III (27) (Table 3). Many of the genes in the MHCIII region have fundamental roles in a variety of cellular functions and include the inflammatory cytokines TNFα, LTA, LTB, and the advanced glycation end product receptor, RAGE (AGER) (27). Multifactor interactions contribute to the disease process at several levels. One hypothesis is that dys-regulation of genes in a locus could contribute to the etiology of the disease, perhaps through coordinated transcription of regions of a chromosome in response to stress or inflammation. RA is a complex autoimmune disorder, and expression analysis of a larger number of patients may validate this hypothesis.

STAB1 [also known as common lymphatic endothelial and vascular receptor (CLEVER-1 or FEEL-1)] was overexpressed in the RA PBMCs. This gene, identified by the k-nearest neighbor analysis, was expressed in 100% of RA PBMC samples and exhibited the highest fold change in this study (64-fold). Stabilin 1 is a large glycoprotein, multifunction scavenger receptor. Characterized as FEEL-1, this protein demonstrated a role as a scavenger receptor that binds to both advanced glycation end products as well as gram-positive and gram-negative bacteria (50,51). The receptor was shown to be expressed on mononuclear cells, tissue macrophages, and endothelial cells (5052). An antibody to FEEL-1 demonstrated a marked reduction in cell-to-cell interaction in a Matrigel tube formation assay, suggesting a role for the receptor in angiogenesis (50). CLEVER-1 has been demonstrated to be involved in the PMBC transmigration through vascular and lymphatic endothelium (52). The CLEVER-1 gene is encoded by 69 exons, and multiple isoforms are expressed in the endothelium (52). The potential function of CLEVER-1 in RA remains to be elucidated.

Several studies of gene expression in RA have been reported. Devauchelle et al. (53) focused on differences in expression in synovia isolated from RA patients compared with that of synovia from osteoarthritis patients. Watanabe et al. (54) reported on differences in expression between RA and normal synovial fibroblasts, and van der Pouw Kraan et al. (55) identified differences in gene expression in RA synovia, allowing the classification of different disease subtypes. A recent study by Bovin et al. (56), using a 12,000-gene oligonucleotide microarray, examined changes in gene expression between PBMCs from 14 RA patients vs. 7 sex-and age-matched controls, and they identified 25 genes that were discriminative. Although different filter criteria were applied to the data sets present here and the report from Bovin et al. (56), there were nine genes that overlapped between the two studies, including S100A12, NCF4, and GNG10. Of the genes that did not overlap, four were not present on the microarray used in this study, three showed changes but did not meet the strict data filtration criteria, and four were not called present in any of the samples. Another study by Olsen et al. (57), using a 4300-gene cDNA microarray, identified a gene expression signature for early-onset rheumatoid arthritis in PBMCs. In that study, the authors segregated the data based on those with longstanding and early-onset disease. There is some overlap between the Olsen et al. (57) study and the results presented here. Of the 44 genes identified, eight from Olsen, et al. also appeared in the present study. Of the 30 that do not, four were not on the human FL6800 array, 15 were not called present in any of the samples, and the others were not included due to the filtration criteria. In the results presented here, patients were selected from the high disease activity cohort, and during analysis, several filtration criteria were applied to the data set with several statistical analyses and a minimum expression criteria of at least 50% of the patients. These measures ensured that the resulting defined gene signature was as robust as possible.

It must be noted that RA patients possess a broad spectrum of disease severity and time of onset, and the comparisons above serve to highlight the multiple differences in patient selection criteria, study materials, protocols, and data analysis that exist in studies so far. Combining the data from our study with that of others, however, does point to several consistent changes in gene expression that would be useful to investigate further. For example, the increased expression of the RAGE ligand S100A12 has been observed in more than 1 study and, as a result, has highlighted the RAGE pathway as potentially important in RA; it is now subject to further study by our group.

The information from this study can be used in two major ways. First, it allows genes important in the pathogenesis of RA to be identified. These genes can then be investigated in detail to determine their potential roles in disease. Second, the power of DNA microarray profiling, with its ability to monitor the expression of multiple genes simultaneously, may allow the identification of patterns of gene expression associated with RA. This may enable rapid diagnosis of RA and predictions of prognosis, as well as response to, and side effects of, DMARDs. The use of these techniques is most advanced in oncology, where predictions of prognosis can be made for certain cancers (17). This provides clinically useful information that guides decisions about how aggressive a treatment regimen should be for a given patient. There is a marked difference in the clinical features of RA between individuals, and molecular phenotyping (or patient profiling) may identify or characterize different disease subgroups and courses of disease progression.

A weakness of global gene expression analysis techniques lies in identifying the relationship of changes in gene expression to the disease process. Changes in gene expression may either cause a disease process or occur as a consequence of it. The presence of gene expression changes in genes that have been associated with RA validates the data set. However, not all genes are primarily regulated by changes in mRNA levels, with many being subject to posttranscriptional regulation. TNF, the best-validated molecular therapeutic target in RA, does not emerge from this type of analysis. This study examined expression in nine RA patients and identified a set of genes that is preferentially expressed in RA patients compared with controls. Although the data are intriguing, samples from a larger number of patients would aid in a class prediction to determine which genes are most associated with disease state and type of prognosis. It is interesting to note that a recent study of PBMC expression profiles in several autoimmune diseases showed that, whereas all diseases displayed profiles that differed from a normal immune response, not all diseases could be clearly distinguished from each other (58).

Gene expression studies on PBMCs may not exactly represent the situation within the inflamed synovial membranes of RA. RA is a systemic disease, however, and differences in cytokine production and phenotype of PBMCs in RA have been demonstrated (5961). This approach has the advantage of being a rapid and minimally invasive way of obtaining cells from patients. The usefulness of assaying tissue samples in RA is limited by availability and sampling bias due to regional differences in disease activity in synovia. However, if the diagnostic/predictive results of a gene expression profile can be demonstrated, PBMCs are a readily accessible source of cells.

The use of oligonucleotide microarrays enables a broader view of complex inflammatory diseases, such as RA. The simultaneous measurement of multiple mRNA transcripts allows an increased understanding of the complexity of proteins that may be interacting in a disease state rather than focusing on one or two at a time. This study identified 330 mRNA transcripts that were differentially regulated in the PBMCs from RA patients compared with normal volunteers. Having demonstrated that these techniques can be used with PBMCs, the next step involves looking at patterns of gene expression in individuals over time and detailed phenotypic examination of these individuals to determine patterns of gene expression associated with different features of RA.

Acknowledgments

The authors thank Dr. James C. Keith, Jr., for reviewing the manuscript.

Footnotes

Online address: http://www.molmed.org

REFERENCES

  • 1.Markenson JA. Worldwide trends in the socioeconomic impact and long-term prognosis of rheumatoid arthritis. Semin Arthritis Rheum. 1991;21(2 Suppl 1):4–12. doi: 10.1016/0049-0172(91)90046-3. [DOI] [PubMed] [Google Scholar]
  • 2.Wong JB, Ramey DR, Singh G. Long-term morbidity, mortality, and economics of rheumatoid arthritis. Arthritis Rheum. 2001;44(12):2746–9. doi: 10.1002/1529-0131(200112)44:12<2746::aid-art461>3.0.co;2-z. [DOI] [PubMed] [Google Scholar]
  • 3.Guidelines for the management of rheumatoid arthritis: update. Arthritis Rheum. 2002;46:328–46. doi: 10.1002/art.10148. No author. [DOI] [PubMed] [Google Scholar]
  • 4.Feldmann M, Maini RN. Lasker Clinical Medical Research Award. TNF defined as a therapeutic target for rheumatoid arthritis and other autoimmune diseases. Nat Med. 2003;9:1245–50. doi: 10.1038/nm939. [DOI] [PubMed] [Google Scholar]
  • 5.van der Heijde DM, van Leeuwen MA, van Riel PL, van de Putte LB. Radiographic progression on radiographs of hands and feet during the first 3 years of rheumatoid arthritis measured according to Sharp’s method (van der Heijde modification) J Rheumatol. 1995;22:1792–6. [PubMed] [Google Scholar]
  • 6.Bathon JM, et al. A comparison of etanercept and methotrexate in patients with early rheumatoid arthritis. N Engl J Med. 2000;343:1586–93. doi: 10.1056/NEJM200011303432201. [DOI] [PubMed] [Google Scholar]
  • 7.Lipsky PE, et al. Infliximab and methotrexate in the treatment of rheumatoid arthritis: Anti-Tumor Necrosis Factor Trial in Rheumatoid Arthritis with Concomitant Therapy Study Group. N Engl J Med. 2000;343:1594–602. doi: 10.1056/NEJM200011303432202. [DOI] [PubMed] [Google Scholar]
  • 8.Quinn MA, et al. Very early treatment with infliximab in addition to methotrexate in early, poor-prognosis rheumatoid arthritis reduces magnetic resonance imaging evidence of synovitis and damage, with sustained benefit after infliximab withdrawal: results from a twelvemonth randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2005;52:27–35. doi: 10.1002/art.20712. [DOI] [PubMed] [Google Scholar]
  • 9.Combe B, et al. Prognostic factors for radiographic damage in early rheumatoid arthritis: a multiparameter prospective study. Arthritis Rheum. 2001;44:1736–43. doi: 10.1002/1529-0131(200108)44:8<1736::AID-ART308>3.0.CO;2-I. [DOI] [PubMed] [Google Scholar]
  • 10.Dixey J, Solymossy C, Young A. Is it possible to predict radiological damage in early rheumatoid arthritis (RA)? A report on the occurrence, progression, and prognostic factors of radiological erosions over the first 3 years in 866 patients from the Early RA Study (ERAS) J Rheumatol. 2004;69 (Suppl):48–54. [PubMed] [Google Scholar]
  • 11.Lindqvist E, Eberhardt K, Bendtzen K, Heinegard D, Saxne T. Prognostic laboratory markers of joint damage in rheumatoid arthritis. Ann Rheum Dis. 2005;64:196–201. doi: 10.1136/ard.2003.019992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.van Zeben D, Breedveld FC. Prognostic factors in rheumatoid arthritis. J Rheumatol. 1996;44(Suppl):31–3. [PubMed] [Google Scholar]
  • 13.Aune TM, Maas K, Parker J, Moore JH, Olsen NJ. Profiles of gene expression in human autoimmune disease. Cell Biochem Biophys. 2004;40:81–96. doi: 10.1385/CBB:40:2:081. [DOI] [PubMed] [Google Scholar]
  • 14.Mandel M, Gurevich M, Pauzner R, Kaminski N, Achiron A. Autoimmunity gene expression portrait: specific signature that intersects or differentiates between multiple sclerosis and systemic lupus erythematosus. Clin Exp Immunol. 2004;138:164–70. doi: 10.1111/j.1365-2249.2004.02587.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Qing X, Putterman C. Gene expression profiling in the study of the pathogenesis of systemic lupus erythematosus. Autoimmun Rev. 2004;3:505–9. doi: 10.1016/j.autrev.2004.07.001. [DOI] [PubMed] [Google Scholar]
  • 16.Rus V, et al. Gene expression profiling in peripheral blood mononuclear cells from lupus patients with active and inactive disease. Clin Immunol. 2004;112:231–4. doi: 10.1016/j.clim.2004.06.005. [DOI] [PubMed] [Google Scholar]
  • 17.van’t Veer LJ, et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002;415:530–6. doi: 10.1038/415530a. [DOI] [PubMed] [Google Scholar]
  • 18.Arnett FC, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 1988;31:315–24. doi: 10.1002/art.1780310302. [DOI] [PubMed] [Google Scholar]
  • 19.Clancy BM, et al. A gene expression profile for endochondral bone formation: oligonucleotide microarrays establish novel connections between known genes and BMP-2-induced bone formation in mouse quadriceps. Bone. 2003;33:46–s63. doi: 10.1016/s8756-3282(03)00116-9. [DOI] [PubMed] [Google Scholar]
  • 20.Hill AA, Brown EL, Whitley MZ, Tucker-Kellogg G, Hunter CP, Slonim DK. Evaluation of normalization procedures for oligonucleotide array data based on spiked cRNA controls. Genome Biol. 2001;2:RESEARCH0055. doi: 10.1186/gb-2001-2-12-research0055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chiang DY, Brown PO, Eisen MB. Visualizing associations between genome sequences and gene expression data using genome-mean expression profiles. Bioinformatics. 2001;17(Suppl 1):S49–55. doi: 10.1093/bioinformatics/17.suppl_1.s49. [DOI] [PubMed] [Google Scholar]
  • 22.Welch BL. On the comparison of several mean values: an alternative approach. Biometrika. 1951;38:330–6. [Google Scholar]
  • 23.Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Royal Stat Soc B. 1995;57:289–300. [Google Scholar]
  • 24.Bonferroni CE. (1935) Il calcolo delle assicurazioni su gruppi di teste. Studi in onore del Professore Salvatore. Rome. p. 11–60.
  • 25.Bonferroni CE. Teoria statistica delle classi e calcolo delle probabilita. Pubblicazioni del R Istituto Superiore di Scienze Economiche e Commerciali di Firenze. 1936;8:3–62. [Google Scholar]
  • 26.Golub TR, et al. Molecular classification of cancer: class discovery and class prediction by gene expression monitoring. Science. 1999;286:531–7. doi: 10.1126/science.286.5439.531. [DOI] [PubMed] [Google Scholar]
  • 27.Jawaheer D, et al. Screening the genome for rheumatoid arthritis susceptibility genes: a replication study and combined analysis of 512 multicase families. Arthritis Rheum. 2003;48:906–16. doi: 10.1002/art.10989. [DOI] [PubMed] [Google Scholar]
  • 28.Foxwell BM, Bondeson J, Brennan F, Feldmann M. Adenoviral transgene delivery provides an approach to identifying important molecular processes in inflammation: evidence for heterogeneity in the requirement for NFkappaB in tumor necrosis factor production. Ann Rheum Dis. 2000;59(Suppl 1):i54–59. doi: 10.1136/ard.59.suppl_1.i54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Keyszer GM, et al. Detection of insulin-like growth factor I and II in synovial tissue specimens of patients with rheumatoid arthritis and osteoarthritis by in situ hybridization. J Rheumatol. 1995;22:275–81. [PubMed] [Google Scholar]
  • 30.Ota T, Katsuki I. Ferritin subunits in sera and synovial fluids from patients with rheumatoid arthritis. J Rheumatol. 1998;25:2315–8. [PubMed] [Google Scholar]
  • 31.Akin E, Aversa J, Steere AC. Expression of adhesion molecules in synovia of patients with treatment-resistant lyme arthritis. Infect Immun. 2001;69:1774–80. doi: 10.1128/IAI.69.3.1774-1780.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Makarov SS. NF-kappaB in rheumatoid arthritis: a pivotal regulator of inflammation, hyperplasia, and tissue destruction. Arthritis Res. 2001;3:200–6. doi: 10.1186/ar300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sumariwalla PF, Malfait AM, Feldmann M. P-selectin glycoprotein ligand 1 therapy ameliorates established collagen-induced arthritis in DBA/1 mice partly through the suppression of tumor necrosis factor. Clin Exp Immunol. 2004;136:67–75. doi: 10.1111/j.1365-2249.2004.02421.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Brennan FM, Gibbons DL, Mitchell T, Cope AP, Maini RN, Feldmann M. Enhanced expression of tumor necrosis factor receptor mRNA and protein in mononuclear cells isolated from rheumatoid arthritis synovial joints. Eur J Immunol. 1992;22:1907–12. doi: 10.1002/eji.1830220734. [DOI] [PubMed] [Google Scholar]
  • 35.Brennan FM, Gibbons DL, Cope AP, Katsikis P, Maini RN, Feldmann M. TNF inhibitors are produced spontaneously by rheumatoid and osteoarthritic synovial joint cell cultures: evidence of feedback control of TNF action. Scand J Immunol. 1995;42:158–65. doi: 10.1111/j.1365-3083.1995.tb03639.x. [DOI] [PubMed] [Google Scholar]
  • 36.Cope AP, et al. Differential regulation of tumor necrosis factor receptors (TNF-R) by IL-4; upregulation of P55 and P75 TNF-R on synovial joint mononuclear cells. Cytokine. 1993;5:205–12. doi: 10.1016/1043-4666(93)90006-q. [DOI] [PubMed] [Google Scholar]
  • 37.Shealy DJ, et al. Anti-TNF-alpha antibody allows healing of joint damage in polyarthritic transgenic mice. Arthritis Res. 2002;4:R7. doi: 10.1186/ar430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Williams RO, Feldmann M, Maini RN. Anti-tumor necrosis factor ameliorates joint disease in murine collagen-induced arthritis. Proc Natl Acad Sci U S A. 1992;89:9784–8. doi: 10.1073/pnas.89.20.9784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Boiardi L, Macchioni P, Meliconi R, Pulsatelli L, Facchini A, Salvarani C. Relationship between serum RANTES levels and radiological progression in rheumatoid arthritis patients treated with methotrexate. Clin Exp Rheumatol. 1999;17:419–25. [PubMed] [Google Scholar]
  • 40.Barnes DA, et al. Polyclonal antibody directed against human RANTES ameliorates disease in the Lewis rat adjuvant-induced arthritis model. J Clin Invest. 1998;101:2910–9. doi: 10.1172/JCI2172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Firestein GS. NF-kappaB: Holy Grail for rheumatoid arthritis? Arthritis Rheum. 2004;50:2381–6. doi: 10.1002/art.20468. [DOI] [PubMed] [Google Scholar]
  • 42.Tedder TF, Steeber DA, Chen A, Engel P. The selectins: vascular adhesion molecules. FASEB J. 1995;9:866–73. [PubMed] [Google Scholar]
  • 43.Hosaka S, Shah MR, Pope RM, Koch AE. Soluble forms of P-selectin and intercellular adhesion molecule-3 in synovial fluids. Clin Immunol Immunopathol. 1996;78:276–82. doi: 10.1006/clin.1996.0039. [DOI] [PubMed] [Google Scholar]
  • 44.Bullard DC, et al. Acceleration and increased severity of collagen-induced arthritis in P-selectin mutant mice. J Immunol. 1999;163:2844–9. [PubMed] [Google Scholar]
  • 45.Utku N, et al. Prevention of acute allograft rejection by antibody targeting of TIRC7, a novel T cell membrane protein. Immunity. 1998;9:509–18. doi: 10.1016/s1074-7613(00)80634-2. [DOI] [PubMed] [Google Scholar]
  • 46.Utku N, Boerner A, Tomschegg A, Bennai-Sanfourche F, Bulwin GC, Heinemann T, Loehler J, Blumberg RS, Volk HD. TIRC7 deficiency causes in vitro and in vivo augmentation of T and B cell activation and cytokine response. J Immunol. 2004;15:2342–52. doi: 10.4049/jimmunol.173.4.2342. [DOI] [PubMed] [Google Scholar]
  • 47.Tomlinson MG, Heath VL, Turck CW, Watson SP, Weiss A. SHIP family inositol phosphatases interact with and negatively regulate the Tec tyrosine kinase. J Biol Chem. 2004;279:55089–96. doi: 10.1074/jbc.M408141200. [DOI] [PubMed] [Google Scholar]
  • 48.Aguado B, Campbell RD. Characterization of a human lysophosphatidic acid acyltransferase that is encoded by a gene located in the class III region of the human major histocompatibility complex. J Biol Chem. 1998;273:4096–105. doi: 10.1074/jbc.273.7.4096. [DOI] [PubMed] [Google Scholar]
  • 49.West J, et al. Cloning and expression of two human lysophosphatidic acid acyltransferase cDNAs that enhance cytokine-induced signaling responses in cells. DNA Cell Biol. 1997;16:691–701. doi: 10.1089/dna.1997.16.691. [DOI] [PubMed] [Google Scholar]
  • 50.Adachi H, Tsujimoto M. FEEL-1, a novel scavenger receptor with in vitro bacteria-binding and angiogenesis-modulating activities. J Biol Chem. 2002;277:34264–70. doi: 10.1074/jbc.M204277200. [DOI] [PubMed] [Google Scholar]
  • 51.Tamura Y, et al. FEEL-1 and FEEL-2 are endocytic receptors for advanced glycation end products. J Biol Chem. 2003;278:12613–7. doi: 10.1074/jbc.M210211200. [DOI] [PubMed] [Google Scholar]
  • 52.Salmi M, Koskinen K, Henttinen T, Elima K, Jalkanen S. CLEVER-1 mediates lymphocyte transmigration through vascular and lymphatic endothelium. Blood. 2004;104:3849–57. doi: 10.1182/blood-2004-01-0222. [DOI] [PubMed] [Google Scholar]
  • 53.Devauchelle V, et al. DNA microarray allows molecular profiling of rheumatoid arthritis and identification of pathophysiological targets. Genes Immun. 2004;5:597–608. doi: 10.1038/sj.gene.6364132. [DOI] [PubMed] [Google Scholar]
  • 54.Watanabe N, et al. Gene expression profile analysis of rheumatoid synovial fibroblast cultures revealing the overexpression of genes responsible for tumor-like growth of rheumatoid synovium. Biochem Biophys Res Commun. 2002;294:1121–9. doi: 10.1016/S0006-291X(02)00608-3. [DOI] [PubMed] [Google Scholar]
  • 55.van der Pouw Kraan TC, van Gaalen FA, Huizinga TW, Pieterman E, Breedveld FC, Verweij CL. Discovery of distinctive gene expression profiles in rheumatoid synovium using cDNA microarray technology: evidence for the existence of multiple pathways of tissue destruction and repair. Genes Immun. 2003;4:187–96. doi: 10.1038/sj.gene.6363975. [DOI] [PubMed] [Google Scholar]
  • 56.Bovin LF, et al. Blood cell gene expression profiling in rheumatoid arthritis: discriminative genes and effect of rheumatoid factor. Immunol Lett. 2004;93:217–26. doi: 10.1016/j.imlet.2004.03.018. [DOI] [PubMed] [Google Scholar]
  • 57.Olsen N, et al. A gene expression signature for recent onset rheumatoid arthritis in peripheral blood mononuclear cells. Ann Rheum Dis. 2004;63:1387–92. doi: 10.1136/ard.2003.017194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Maas K, et al. Cutting edge: molecular portrait of human autoimmune disease. J Immunol. 2002;169:5–9. doi: 10.4049/jimmunol.169.1.5. [DOI] [PubMed] [Google Scholar]
  • 59.Hirano T, et al. Comparative study of lymphocyte-suppressive potency between prednisolone and methylprednisolone in rheumatoid arthritis. Immunopharmacology. 2000;49:411–7. doi: 10.1016/s0162-3109(00)00263-0. [DOI] [PubMed] [Google Scholar]
  • 60.Schulze-Koops H, Lipsky PE, Kavanaugh AF, Davis LS. Persistent reduction in IL-6 mRNA in peripheral blood mononuclear cells of patients with rheumatoid arthritis after treatment with a monoclonal antibody to CD54 (ICAM-1) Clin Exp Immunol. 1996;106:190–6. doi: 10.1046/j.1365-2249.1996.d01-828.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Schulze-Koops H, Davis LS, Kavanaugh AF, Lipsky PE. Elevated cytokine messenger RNA levels in the peripheral blood of patients with rheumatoid arthritis suggest different degrees of myeloid cell activation. Arthritis Rheum. 1997;40:639–47. doi: 10.1002/art.1780400408. [DOI] [PubMed] [Google Scholar]

Articles from Molecular Medicine are provided here courtesy of The Feinstein Institute for Medical Research at North Shore LIJ

RESOURCES