Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Dec 1.
Published in final edited form as: Schizophr Res. 2012 Oct 9;142(1-3):188–199. doi: 10.1016/j.schres.2012.09.015

Transcriptomic Analysis of Postmortem Brain identifies Dysregulated Splicing Events in Novel Candidate Genes for Schizophrenia

Ori S Cohen 1,2, Sarah Y Mccoy 1,2, Frank A Middleton 2, Sean Bialosuknia 1,2, Yanli Zhang-James 2, Lu Liu 2, Ming T Tsuang 3,4,5, Stephen V Faraone 2, Stephen J Glatt 1,2,*
PMCID: PMC3502694  NIHMSID: NIHMS411006  PMID: 23062752

Abstract

The diverse spatial and temporal expression of alternatively spliced transcript isoforms shapes neurodevelopment and plays a major role in neuronal adaptability. Although alternative splicing is extremely common in the brain, its role in mental illnesses such as schizophrenia has received little attention. To examine this relationship, postmortem brain tissue was obtained from 20 individuals with schizophrenia (SZ) and 20 neuropsychiatrically normal comparison subjects. Gray matter samples were extracted from two brain regions implicated in the disorder: Brodmann area 10 and caudate. Affymetrix Human Gene 1.0 ST arrays were used on four subjects per group to attain an initial profile of differential expression of transcribed elements within and across brain regions in SZ. Numerous genes of interest with altered mRNA transcripts were identified by microarray through the differential expression of particular exons and 3′ untranslated regions (UTRs) between diagnostic groups. Select microarray results—including dysregulation of ENAH exon 11a and CPNE3 3′UTR—were verified by qRTPCR and replicated in the remaining independent sample of 16 SZ patients and 16 normal comparison subjects. These results, if further replicated, clearly illustrate the importance of Identifying transcriptomic variants in expression studies, and implicate novel candidate genes in the disorder.

INTRODUCTION

One method commonly employed to unravel the underlying biology of schizophrenia (SZ) is transcriptomic profiling of postmortem brain tissue (Glatt et al., 2005; Horvath et al., 2011; Mirnics et al., 2006). Most prior transcriptomic studies of the brain in SZ overlooked a potentially important source of phenotypic diversity: the expression of alternatively spliced variants (ASVs). Alternative splicing is a major mechanism by which eukaryotes create enormous proteomic diversity from a smaller-than-expected number of genes. The manner in which elements of a particular gene are spliced has a significant impact on protein function. In fact, the discrete ASVs of some genes have diametrically opposed physiological functions (Clark et al., 2007); thus, traditional discussions about a the function of a given gene may be moot unless a particular ASV of the gene is invoked.

Transcriptome-wide sequence analysis has detected splicing events in up to 95% of multi-exon genes (Pan et al., 2008). Alternative splicing events are tissue-specific, and can be regionally specific within a tissue, including the brain (Twine et al., 2011), resulting in an extremely complex expression profile. These diverse splicing patterns dictate important regulatory decisions in many steps of neuronal development, including axon guidance (Schmucker et al., 2000; Zipursky et al., 2006), cell-fate determination (Dho et al., 1999; Dho et al., 2006; Reugels et al., 2006), and synaptogenesis (Li et al., 2007). Notably, a small handful of candidate genes for SZ have been found to exhibit abnormal splicing patterns in individual regions of postmortem brain (Glatt et al., 2011), including CTNNA2 (Mexal et al., 2008), DISC1 (Nakata et al., 2009), ERBB4 (Law et al., 2007), ESR1 (Weickert et al., 2008), GRM3 (Sartorius et al., 2008), and NRG1 (Tan et al., 2007). It is not clear if these splicing patterns are abnormal only in those specific brain regions or if other regions are similarly affected.

The key technological advance provided by the Affymetrix Human Gene 1.0 ST arrays is measurement of the expression levels of individual exons and UTRs (hereafter collectively referred to as transcribed elements, or TEs). The aim of the current study was to use this technology to compare the expression profiles of TEs across the whole transcriptome in postmortem brain of individuals with SZ and control subjects in two brain regions previously implicated in the disorder (Brodmann Area 10 [BA10] and caudate head) (Ellison-Wright et al., 2008; Goghari, 2010; Yu et al., 2010) in order to identify alternative splicing abnormalities in SZ and to estimate their regional specificity.

METHODS

Samples

Postmortem brain samples were obtained from 20 SZ and 20 neuropsychiatrically normal comparison (NC) subjects in the Harvard Brain Tissue Resource Center. All tissues were collected with the full consent of the family or next of kin of the deceased. A “discovery sample” was constructed of four SZ and four NC subjects matched on age, sex, postmortem interval, and RNA integrity Number (RIN) (Table 1). The SZ discovery sample was selected to include two subjects on antipsychotic medication at the time of death and two who were not, allowing identification and exclusion of some medication effects from further validation steps (we note here that this contrast of small samples did not have adequate power to detect all medication-related effects, but it does provide one basis for filtering out genes that were strongly regulated by medication). Subsequent to verification, select observations from microarray analysis of the discovery sample were tested for replication in an independent, larger, and less homogeneous group of SZ and NC subjects (the “replication sample”).

Table 1.

Demographic and Agonal Characteristics of the SZ and NC Discovery and Replication Samples

Discovery Samples Replication Samples

SZ NC SZ NC
Sample Size: n 4 4 16 16
Sex: % male (n) 100 (4) 100 (4) 37.5 (6) 31.2 (5)
Age: mean years±S.D. 59.0±3.7 58.3±2.1 68.2±22.7 66.5±20.7
Postmortem Interval: mean hours±S.D. 24.9±4.1 24.0±3.5 18.8±7.0 21.9±5.3
RNA Integrity Number (RIN): mean±S.D. 8.0±1.2 7.8±1.3 7.9±0.5 8.3±1.3

No significant differences on any factor were observed between the SZ and NC discovery samples (all p>0.737) or between the SZ and NC replication samples (all p>0.183).

RNA Extraction and Purification

Gray matter tissue samples were dissected from two brain regions (BA10 and CAUD). All brains had a pH between 6 and 7. Samples remained frozen over dry ice until placed in RLT buffer with β-mercaptoethanol and homogenized using Qiashredder columns for subsequent nucleic acid isolation using Qiagen AllPrep DNA/RNA Mini Kits (see Supplemental Methods for further details). Using a Bioanalyzer 2100 and an RNA Nanochip, purity of each sample was determined by the A260:280 ratio, with acceptable values ranging from 1.8–2.2. The quality of each sample was assured by a RIN≥6 and visual confirmation of clear, distinct 28S and 18S rRNA peaks.

Microarray Procedures

Reverse-transcription, hybridization, and scanning were performed according to well-established protocols. Ambion WT Expression Kits were used for amplification and Affymetrix WT Terminal Labeling Kits were used for labeling. Total RNA was reverse-transcribed and hybridized onto GeneChip Human Gene 1.0 ST Arrays (Affymetrix) and scanned on a GeneChip 7G/4C scanner (Affymetrix). The Human Gene 1.0ST Array improves upon previous generations of Affymetrix arrays by probing the entire length of each transcript, rather than the 3′ end only. The Gene 1.0 ST Array interrogates 28,869 well-annotated genes with 764,885 distinct probes, with an average of one probe per exon and 27 probes per transcript. The array has greater than 99 percent coverage of NM sequences present in the November 3, 2006, RefSeq database (Affymetrix, 2007).

Microarray Data Preparation

Partek Genomics Suite software was utilized for all analyses of microarray scan data. Corrections for background signal were made by robust multi-array average (RMA) (Irizarry et al., 2003). The set of eight GeneChips was standardized using quantile normalization, and expression levels of each probe underwent log-2 transformation to yield data distributions more closely approximating normality. Summarization of redundant probesets was obtained by median polish. Probesets with a signal:noise ratio of less than 3.0 were excluded from subsequent analyses (Handran et al., 2002); this led to the exclusion of six probes from the BA10 dataset and seven probes from the CAUD dataset.

Microarray Data Analyses

Primary analyses were designed to detect genes with significantly different TE expression between diagnostic groups, potentially indicating different alternative splicing events between them. SZ and NC groups were compared on mean expression levels of all TEs in each gene on a gene-by-gene basis through ANCOVAs and inspection of interaction terms, as described previously (Glatt et al., 2009; Partek Incorporated, 2008) and in more detail in the Supplemental Methods. The key term for the present analyses was the interaction of TE identity (ID) with diagnostic group, which allowed for the detection of differences in the expression of one or more TEs in a gene between diagnostic groups (Partek Incorporated, 2008). The significance of these interaction terms (one per gene) was judged against a stringent Bonferroni-corrected threshold of p=2.47e−06, and post-hoc F-tests were used to identify the specific dysregulated TE(s) in the genes showing significant interactions.

Genes influenced by a significant interaction of diagnosis and TE ID in one or more brain regions were subjected to the DAVID algorithm (Dennis et al., 2003) to determine if the set was enriched with genes mapping to a particular biological pathway, or genes containing particular protein domains, which might indicate that the corresponding exonic sequences shared by these genes might provide the basis for their targeting by a common alternative-splicing regulator(s).

Reverse Transcription and Quantitative PCR

Quantitative reverse transcription (qRT) PCR was performed to confirm and validate select microarray results. Concentrations of total RNA isolates were adjusted prior to RT, and the same amount of total RNA was used in each reaction (20 ng/ul). QuantiTect RT kits (Qiagen) were used according to the manufacturer’s protocol. Each subject’s cDNA was run by qRTPCR in duplicate for each primer set (Supplemental Tables). Additional details are provided in Supplemental Methods.

Expression of each dysregulated TE was evaluated and compared with a non-dysregulated TE of the same gene. The primer set amplifying the non-dysregulated region of the transcript was considered the “control” primer set. The expression value from the primer set that amplified the dysregulated region (considered the “experimental” primer set) was then compared to that of the control primer set using the ΔΔCT method (Supplemental Methods). TE dysregulation was confirmed using linear regression models with diagnosis, age, sex, PMI, and RIN as independent predictors. The significance of the effect of diagnosis was determined with a 1-tailed p<0.05 for this term after backward removal of clearly non-significant terms (p<0.20) from each regression model.

RESULTS

Discovery Analyses

In BA10 from four SZ subjects and four well-matched NC subjects, 43 transcripts were influenced by a Bonferroni-corrected significant interaction of diagnosis and TE ID, indicative of differential expression of one or some (but not all) TEs between diagnostic groups (Table 2). DAVID analysis found no particular biological pathways or protein domains enriched among these 43 genes.

Table 2.

Transcripts Exhibiting a Bonferroni-Corrected Significant Interaction of Diagnosis (SZ vs. NC) and Transcript Element ID in Brodmann Area 10 (BA10)

Gene Symbol Gene Product Reference Sequence Transcript Cluster ID Probe Sets (n) F p Direction of Dysregulation#
CPNE3 Copine III NM_003909 8147172 21 15.003 6.6e−24
SCIN Scinderin NM_001112706 8131550 22 6.992 5.4e−13
SAMHD1 SAM domain and HD domain 1 NM_015474 8066117 18 8.354 7.6e−13
C4A Complement component 4A (Rodgers blood group) NM_007293 8118409 42 3.830 2.9e−11
IARS Isoleucyl-tRNA synthetase NM_013417 8162313 38 4.012 5.0e−11
CHI3L1 Chitinase 3-like 1 (cartilage glycoprotein-39) NM_001276 7923547 14 9.026 5.7e−11
LPPR4 Plasticity related gene 1 NM_014839 7903214 12 9.775 3.3e−10
C4A Complement component 4A (Rodgers blood group) NM_007293 8118455 42 3.458 9.8e−10
SERPINA3 Serpin peptidase inhibitor, clade A (alpha-1 antiprotein) NM_001085 7976496 9 12.930 1.0e−9
C4A Complement component 4A (Rodgers blood group) NM_007293 8179399 40 3.507 1.5e−9
ALOX5 Arachidonate 5-lipoxygenase NM_000698 7927215 15 7.011 2.6e−9
CSDE1 Cold shock domain containing E1, RNA-binding NM_001007553 7918825 21 5.214 3.5e−9
PPP3CA Protein phosphatase 3 (formerly 2B), catalytic subunit, a NM_000944 8101971 18 5.734 6.3e−9
TLN1 Talin 1 NM_006289 8161056 57 2.791 6.7e−9
IFI16 Interferon, gamma-inducible protein 16 NM_005531 7906400 16 6.209 9.3e−9
RNASET2 Ribonuclease T2 NM_003730 8130768 14 6.923 1.1e−8
ALDH2 Aldehyde dehydrogenase 2 family (mitochondrial) NM_000690 7958784 15 6.330 1.9e−8
CD86 CD86 molecule NM_175862 8082035 10 9.325 2.2e−8
CUX1 Cut-like homeobox 1 NM_181552 8135114 34 3.424 4.5e−8
MAGI2 Membrane associated guanylate kinase, WW and PDZ domain containing 2 NM_012301 8140504 25 4.003 9.1e−8
LAT2 Linker for activation of T cells family, member 2 NM_032464 8133442 16 5.454 1.1e−7
CFDP1 Craniofacial development protein 1 NM_006324 8002865 12 6.858 1.5e−7
RAB8B RAB8B, member RAS oncogene family NM_016530 7984112 11 7.240 2.3e−7
ERCC5 Excision repair cross-complementing 5 NM_000123 7969935 19 4.556 2.8e−7
MYO5A Myosin VA (heavy chain 12, myoxin) NM_000259 7988921 41 2.895 2.9e−7
PTPRC Protein tyrosine phosphatase, receptor type, C NM_002838 7908553 37 3.042 3.0e−7
C5orf15 Chromosome 5 open reading frame 15 NM_020199 8114138 6 14.126 3.9e−7
LPCAT2 Lysophosphatidylcholine acyltransferase 2 NM_017839 7995697 14 5.525 5.4e−7
ENAH Enabled homolog (Drosophila) NM_001008493 7924619 16 4.976 5.5e−7
LAPTM5 Lysosomal protein transmembrane 5 NM_006762 7914270 11 6.802 5.6e−7
UHRF2 Ubiquitin-like with PHD and ring finger domains 2 NM_152896 8154316 16 4.970 5.6e−7
VTI1A Vesicle transport through interaction with t-SNAREs homolog NM_145206 7930524 8 9.380 6.0e−7
RAPGEF4 Rap guanine nucleotide exchange factor (GEF) 4 NM_007023 8046428 32 3.159 7.5e−7
LGALS9C Lectin, galactoside-binding, soluble, 9C NM_001040078 8005458 12 6.102 8.6e−7
CPVL Carboxypeptidase, vitellogenic-like NM_019029 8138805 16 4.779 1.1e−6
RBM33 RNA binding motif protein 33 NM_053043 8137558 7 10.236 1.3e−6
FCER1G Fc fragment of IgE, high affinity I, receptor for; gamma NM_004106 7906720 7 9.985 1.7e−6
ZMYND8 Zinc finger, MYND-type containing 8 NM_183047 8066786 31 3.091 1.8e−6
SAMSN1 SAM domain, SH3 domain and nuclear localization signals 1 NM_022136 8069541 11 6.252 1.8e−6
DOCK8 Dedicator of cytokinesis 8 NM_203447 8153959 52 2.422 2.0e−6
MUM1 Melanoma associated antigen (mutated) 1 NR_024247 8024255 17 4.409 2.0e−6
IGSF6 Immunoglobulin superfamily, member 6 NM_005849 8000184 7 9.837 2.0e−6
ITGB2 Integrin, beta 2 (complement component 3 receptor 3 and 4) NM_000211 8070826 20 3.943 2.1e−6
*

Rows are sorted by p-value in ascending order.

#

of the single most differentially expressed transcribed element per gene.

In CAUD, 31 results surpassed the Bonferroni-adjusted significance threshold (Table 3). These 31 transcripts were significantly enriched with genes sharing common protein domains, suggesting a possible basis for their common exonic dysregulation. The most significantly enriched protein-domain annotations were spectrin repeats (p=5.07e−04) and actinin-type actin-binding domains (p=6.09e−04), both of which were present in the same three transcripts. This ratio (3/31) represents an approximately 80-fold enrichment of both annotations compared to chance expectation. One biological pathway (Agrin in Postsynaptic Differentiation) was also over-represented at a Bonferroni-corrected level of significance (p=0.019). The appearance of two genes in this pathway on a list of 31 transcripts represents an approximately 52-fold enrichment compared to chance expectation.

Table 3.

Transcripts Exhibiting a Bonferroni-Corrected Significant Interaction of Diagnosis (SZ vs. NC) and Transcript Element ID in Caudate Head (CAUD)

Gene Symbol Gene Product Reference Sequence Transcript Cluster ID Probe Sets (n) F p* Direction of Dysregulation#
CPNE3 Copine III NM_003909 8147172 21 18.518 1.0e−27
UTRNa Utrophin NM_007124 8122464 72 4.946 7.1e−26
RXFP2 Relaxin NM_130806 7968389 21 8.816 1.6e−15
DMDa Dystrophin NM_000109 8171921 95 2.540 2.0e−11
GPR98 G protein-coupled receptor 98 NM_032119 8106827 91 2.409 6.6e−10
RGPD6 Ranbp2-like and grip domain containing 6 NM_001123363 8044304 28 4.286 2.8e−9
CIT Citron (rho-interacting, serine) NM_007174 7966878 50 3.008 4.6e−9
SYNE1a Spectrin repeat containing, nuclear envelope 1 NM_182961 8130211 158 1.873 1.3e−8
C6orf174 Chromosome 6 open reading frame 174 NM_001012279 8129392 17 5.689 1.9e−8
Unknown Unknown Unknown 8054557 22 4.447 5.8e−8
NME7 Non-metastatic cells 7 NM_013330 7922137 14 6.201 8.0e−8
CEP192 Centrosomal protein 192Kda NM_032142 8020267 40 3.050 9.7e−8
KIAA0319L KIAA0319-Like NM_024874 7914809 24 4.042 1.3e−7
DGKI Diacylglycerol kinase, iota NM_004717 8143154 33 3.333 1.4e−7
KCNK2 Potassium channel, subfamily k, member 2 NM_001017425 7909730 14 5.970 1.5e−7
MYT1L Myelin transcription factor 1-like NM_015025 8050031 28 3.585 2.3e−7
SEC31A Sec31 Homolog A (S. Cerevisiae) NM_014933 8101376 34 3.193 2.7e−7
HSPA4 Heat shock 70kda protein 4 NM_002154 8108015 24 3.849 3.7e−7
SLC4A1AP solute carrier family 4 (anion exchanger), member 1, adaptor protein NM_018158 8041015 11 6.701 6.9e−7
EPB49 Erythrocyte membrane protein band 4.9 (dematin) NM_001978 8145005 21 4.022 8.3e−7
STXBP2 Syntaxin binding protein 2 NM_006949 8025255 22 3.902 8.4e−7
RABEP1 Rabaptin, rab gtpase binding effector protein 1 NM_004703 8004111 21 4.011 8.8e−7
FBXO44 F-box protein 44 NM_001014765 7897714 13 5.689 8.9e−7
EPRS Glutamyl-prolyl-trna synthetase NM_004446 7924351 34 3.021 1.0e−6
C17orf44 Chromosome 17 open reading frame 44 NR_026951 8012416 6 12.700 1.1e−6
PSMA5 Proteasome (prosome, macropain) subunit, alpha type, 5 NM_002790 7918345 13 5.622 1.1e−6
PSD3 Pleckstrin and sec7 domain containing 3 NM_015310 8149555 18 4.342 1.3e−6
TNKS2 TRF1-interacting ankyrin-related ADP-ribose polymerase 2 NM_025235 7929168 32 3.078 1.4e−6
ADORA3 Adenosine a3 receptor NM_020683 7918533 14 5.180 1.5e−6
VWA3A Von Willebrand factor a domain containing 3a NM_173615 7993898 36 2.837 2.2e−6
OVOS Ovostatin BX647938 7961026 4 22.882 2.3e−6
*

Rows are sorted by p-value in ascending order.

#

of the single most differentially expressed transcribed element per gene.

a

Genes encoding proteins with actinin-type actin-binding domains.

CPNE3, was the lone gene whose modulation by a diagnosis-x-TE ID interaction surpassed Bonferroni-corrected significance in both brain regions; however, 340 additional transcripts were influenced by at least a nominally significant interaction of diagnosis and TE ID in both brain regions (Table 4). This list of 341 transcripts was also significantly enriched with genes with one or more common protein domains, including a 9.4-fold enrichment of genes with actinin-type actin-binding domains (p=8.45e−03). The most significant enrichment was for genes containing one or more fibronectin type-III-like fold (p=7.14e−03), with ten out of 341 genes containing this domain, representing a 2.9-fold enrichment beyond chance expectation. Other protein domains enriched among the genes on this list included calponin-like actin-binding (six genes; 4.6-fold enrichment; p=9.55e−03), peptidase C1A, papain C-terminal (three genes; 11.5-fold enrichment; p=2.68e−02), and histone core (four genes; 5.0-fold enrichment; p=4.50e−02). No biological pathways were enriched at a Bonferroni-corrected level of significance within the list of 341 transcripts.

Table 4.

Transcripts Exhibiting a Significant Interaction of Diagnosis (SZ vs. NC) and Transcript Element ID in both Brodmann Area 10 (BA10) and Caudate (CAUD)

Gene Symbol Gene Product Reference Sequence Transcript Cluster ID Probe Sets (n) BA10 CAUD

F p* F p*
CPNE3 Copine III NM_003909 8147172 21 15.003 6.60e−24 18.518 1.00e−27
CEP192 Centrosomal protein 192kDa NM_032142 8020267 40 2.587 6.20e−06 3.050 9.70e−08
SCIN Scinderin NM_001112706 8131550 22 6.992 5.40e−13 3.055 5.80e−05
OVOS Ovostatin BX647938 7953873 4 12.265 1.30e−04 17.412 1.50e−05
NCKAP1L NCK-associated protein 1-like NM_005337 7955908 34 2.359 1.50e−04 2.642 1.80e−05
LCP1a,c Lymphocyte cytosolic protein 1 (L-plastin) NM_002298 7971461 20 3.062 1.20e−04 3.169 7.20e−05
OVOS Ovostatin BX647938 7961026 4 11.274 2.10e−04 22.882 2.30e−06
KLHL5 Kelch-like 5 (Drosophila) NM_015990 8094625 12 4.170 1.10e−04 3.891 2.40e−04
NME7 Non-metastatic cells 7, protein expressed in (nucleoside) NM_013330 7922137 14 3.348 4.30e−04 6.201 8.00e−08
LAPTM5 Lysosomal protein transmembrane 5 NM_006762 7914270 11 6.802 5.60e−07 3.864 4.50e−04
LAT2 Linker for activation of T cells family, member 2 NM_032464 8133442 16 5.454 1.10e−07 3.064 5.10e−04
ST6GAL1 ST6 beta-galactosamide alpha-2,6-sialyltranferase 1 NM_173216 8084717 8 4.984 3.60e−04 5.319 2.10e−04
APOC2 Apolipoprotein C-II NM_000483 8029551 8 6.543 3.00e−05 4.739 5.40e−04
RPL13A Ribosomal protein L13a NM_012423 8030351 8 4.700 5.80e−04 7.204 1.10e−05
HSD17B4 Hydroxysteroid (17-beta) dehydrogenase 4 NM_000414 8107532 21 2.677 5.10e−04 2.917 1.60e−04
RHOV Ras homolog gene family, member V NM_133639 7987574 9 5.332 8.20e−05 4.281 6.10e−04
NACA Nascent polypeptide-associated complex alpha subunit NM_001113201 7964262 8 4.838 4.60e−04 5.239 2.40e−04
SMOX Spermine oxidase NM_175839 8060745 12 4.805 2.10e−05 3.504 6.80e−04
APOL1 Apolipoprotein L, 1 NM_145343 8072735 8 7.687 5.80e−06 4.299 1.20e−03
EFNA5 Ephrin-A5 NM_001962 8113433 9 6.592 8.80e−06 3.951 1.20e−03
JTB Jumping translocation breakpoint NM_006694 7920409 12 4.304 7.90e−05 3.326 1.10e−03
ADORA3 Adenosine A3 receptor NM_020683 7918533 14 3.019 1.20e−03 5.180 1.50e−06
LEPRb Leptin receptor NM_002303 7902074 27 2.369 6.10e−04 2.347 6.90e−04
CPVL Carboxypeptidase, vitellogenic-like NM_019029 8138805 16 4.779 1.10e−06 2.805 1.30e−03
RNASET2 Ribonuclease T2 NM_003730 8130768 14 6.923 1.10e−08 2.985 1.40e−03
Unknown Unknown Unknown 8179731 17 2.967 5.10e−04 2.800 9.80e−04
NDUFS1 NADH dehydrogenase (ubiquinone) Fe-S protein 1, 75kDa (Na+) NM_005006 8058428 21 2.448 1.50e−03 2.963 1.30e−04
ALDH3A2 Aldehyde dehydrogenase 3 family, member A2 NM_001031806 8005638 16 2.760 1.50e−03 3.459 1.20e−04
HLA-E Major histocompatibility complex, class I, E NM_005516 8117890 9 4.056 9.50e−04 4.130 8.20e−04
TBXAS1 Thromboxane A synthase 1 (platelet) NM_001130966 8136557 18 3.700 1.80e−05 2.567 1.90e−03
DNAJC11 DnaJ (Hsp40) homolog, subfamily C, member 11 NM_018198 7912112 20 2.614 9.00e−04 2.567 1.10e−03
DDRGK1 DDRGK domain containing 1 NM_023935 8064601 11 3.206 2.30e−03 4.249 1.70e−04
IRF8 Interferon regulatory factor 8 NM_002163 7997712 11 5.891 3.90e−06 3.163 2.60e−03
IARS Isoleucyl-tRNA synthetase NM_013417 8162313 38 4.012 5.00e−11 1.888 2.80e−03
SPTLC1 Serine palmitoyltransferase, long chain base subunit 1 NM_006415 8162294 16 2.669 2.20e−03 2.983 6.80e−04
LST1 Leukocyte specific transcript 1 NM_007161 8118149 8 4.695 5.90e−04 3.896 2.40e−03
AKR1CL2 Aldo-keto reductase family 1, member C-like 2 NM_001040177 7925904 13 4.268 4.50e−05 2.834 3.10e−03
PARVGc Parvin, gamma NM_022141 8073682 15 2.889 1.30e−03 2.795 1.90e−03
COL6A2 Collagen, type VI, alpha 2 NM_001849 8069301 30 2.069 2.20e−03 2.184 1.10e−03
CTSSd Cathepsin S NM_004079 7919800 14 3.283 5.30e−04 2.753 2.90e−03
HS6ST2 Heparan sulfate 6-O-sulfotransferase 2 NM_001077188 8175195 10 3.282 3.00e−03 4.068 5.00e−04
CHI3L1 Chitinase 3-like 1 (cartilage glycoprotein-39) NM_001276 7923547 14 9.026 5.70e−11 2.693 3.50e−03
PSD3 Pleckstrin and Sec7 domain containing 3 NM_015310 8149555 18 2.395 3.70e−03 4.342 1.30e−06
TGFBR1 Transforming growth factor, beta receptor 1 NM_004612 8156826 11 4.710 5.80e−05 2.999 3.90e−03
PPHLN1 Periphilin 1 NM_016488 7954940 19 2.470 2.20e−03 2.426 2.10e−03
MS4A7 Membrane-spanning 4-domains, subfamily A, member 7 NM_021201 7940259 19 3.330 5.30e−05 2.304 4.40e−03
FAM118A Family with sequence similarity 118, member A/B NM_001104595 8073752 13 2.812 3.30e−03 3.165 1.20e−03
ITGAV Integrin, alpha V (vitronectin receptor, alpha polypeptide NM_002210 8046861 33 1.917 4.00e−03 2.085 1.30e−03
ERBB4 V-erb-a erythroblastic leukemia viral oncogene homolog 4 NM_005235 8058627 30 2.049 2.50e−03 2.027 2.90e−03
ZC3H14 Zinc finger CCCH-type containing 14 NM_024824 7976101 24 3.043 3.00e−05 2.064 5.60e−03
SYK Spleen tyrosine kinase NM_003177 8156321 15 2.628 3.30e−03 2.680 2.80e−03
RRP1B Ribosomal RNA processing 1 homolog B (S. cerevisiae) NM_015056 8068902 16 2.656 2.30e−03 2.514 3.80e−03
SLC6A18 Solute carrier family 6, member 18 NM_182632 8104281 16 2.565 3.10e−03 2.581 3.00e−03
CIT Citron (rho-interacting, serine) NM_007174 7966878 50 1.649 6.60e−03 3.008 4.60e−09
C3 Complement component 3 NM_000064 8033257 42 2.629 2.50e−06 1.720 6.70e−03
SIK1 Salt-inducible kinase 1 NM_173354 8070665 15 2.408 7.00e−03 3.524 1.50e−04
UHRF2 Ubiquitin-like with PHD and ring finger domains 2 NM_152896 8154316 16 4.970 5.60e−07 2.323 7.60e−03
FCGBP Fc fragment of IgG binding protein NM_003890 8036787 20 2.370 2.70e−03 2.229 5.00e−03
CRHR1 Corticotropin releasing hormone receptor 1 NM_001145146 8007808 17 2.730 1.30e−03 2.312 6.40e−03
ALDOA Aldolase A, fructose-bisphosphate NM_000034 7994737 18 2.354 4.40e−03 2.418 3.40e−03
ATP5SL ATP5S-like NM_018035 8037037 5 4.702 6.00e−03 5.856 2.00e−03
MCM6 Minichromosome maintenance complex component 6 NM_005915 8055426 18 3.304 9.00e−05 2.206 7.90e−03
ENTPD1 Ectonucleoside triphosphate diphosphohydrolase 1 NM_001776 7929511 13 2.696 4.70e−03 2.793 3.50e−03
NCF4 Neutrophil cytosolic factor 4, 40kDa NM_013416 8072744 12 3.347 1.00e−03 2.649 7.20e−03
ERCC8 Excision repair cross-complementing rodent repair deficiency NM_000082 8112285 16 3.321 2.00e−04 2.305 8.00e−03
HNRNPAB Heterogeneous nuclear ribonucleoprotein A NM_031266 8110450 10 2.958 6.30e−03 3.404 2.20e−03
SLC30A10 Solute carrier family 30, member 10 NM_018713 7924342 8 3.763 3.00e−03 3.388 5.90e−03
FYB FYN binding protein (FYB-120) NM_001465 8111739 17 2.763 1.10e−03 2.260 7.80e−03
SCAMP2 Secretory carrier membrane protein 2 NM_005697 7990417 11 4.307 1.50e−04 2.682 8.80e−03
TYROBP TYRO protein tyrosine kinase binding protein NM_003332 8036224 7 5.079 7.30e−04 3.458 8.40e−03
PDIA2 Protein disulfide isomerase family A, member 2 NM_006849 7991815 12 3.062 2.30e−03 2.662 6.90e−03
ARPC1B Actin related protein 2 NM_005720 8134552 11 2.669 9.10e−03 4.392 1.00e−04
CD68 CD68 molecule NM_001251 8004510 10 5.239 4.10e−05 2.791 9.20e−03
H2AFJe H2A histone family, member J NM_177925 7954124 7 3.541 7.40e−03 4.369 2.10e−03
SECISBP2L SECIS binding protein 2-like NM_014701 7988581 19 2.197 6.90e−03 2.415 2.70e−03
RNF7 Ring finger protein 7 NM_014245 8083119 9 3.456 3.20e−03 3.107 6.60e−03
BIRC6 Baculoviral IAP repeat-containing 6 NM_016252 8041283 76 1.561 3.50e−03 1.510 6.30e−03
RTKN2 Rhotekin 2 NM_145307 7933855 16 2.650 2.30e−03 2.314 7.80e−03
IL13RA1b Interleukin 13 receptor, alpha 1 NM_001560 8169580 17 2.254 8.00e−03 2.570 2.40e−03
C1QC Complement component 1, q subcomponent, C chain NM_001114101 7898799 5 13.897 5.30e−06 4.158 1.07e−02
BTF3 Basic transcription factor 3 NM_001037637 8106181 7 3.305 1.08e−02 6.077 1.80e−04
WDR74 WD repeat domain 74 NM_018093 7948881 12 3.360 1.00e−03 2.525 1.01e−02
CD37 CD37 molecule NM_001774 8030277 14 3.460 3.00e−04 2.340 1.09e−02
OSBP Oxysterol binding protein NM_002556 7948379 17 2.940 5.70e−04 2.174 1.07e−02
AVPR2 Arginine vasopressin receptor 2 NM_001146151 8170794 8 3.072 1.05e−02 4.186 1.40e−03
SAFB2 Scaffold attachment factor B2 NM_014649 8032974 21 2.003 1.15e−02 2.652 5.70e−04
SYNPO Synaptopodin NM_007286 8109305 9 2.907 1.00e−02 3.661 2.10e−03
TCIRG1 T-cell, immune regulator 1, ATPase, H+ transporting NM_006019 7941985 21 2.165 5.60e−03 2.111 7.10e−03
RPS28 Ribosomal protein S28 NM_001031 8025395 6 3.525 1.26e−02 7.811 8.30e−05
LGALS9C Lectin, galactoside-binding, soluble, 9C NM_001040078 8005458 12 6.102 8.60e−07 2.430 1.31e−02
CDC42BPA CDC42 binding protein kinase alpha (DMPK-like) NM_003607 7924773 43 2.006 5.80e−04 1.627 1.27e−02
NUDT9 Nudix (nucleoside diphosphate linked moiety X)-type motif NM_024047 8096251 13 3.337 6.90e−04 2.361 1.27e−02
CYBB Cytochrome b-245, beta polypeptide NM_000397 8166730 14 3.664 1.60e−04 2.272 1.35e−02
FGR Gardner-Rasheed feline sarcoma viral (v-fgr) oncogene homolog NM_005248 7914112 14 2.645 4.10e−03 2.380 9.60e−03
GNPTAB N-acetylglucosamine-1-phosphate transferase, alpha and beta NM_024312 7965812 23 1.920 1.30e−02 2.478 8.10e−04
ELK1 ELK1, member of ETS oncogene family NM_001114123 8172345 12 3.579 5.50e−04 2.425 1.33e−02
RGPD1 RANBP2-like and GRIP domain containing 1 NM_001024457 8053622 23 3.369 7.80e−06 1.902 1.41e−02
CYTL1 Cytokine-like 1 NM_018659 8099132 7 3.145 1.39e−02 5.361 4.90e−04
LGALS9B Lectin, galactoside-binding, soluble, 9B NM_001042685 8013450 14 4.636 7.60e−06 2.249 1.45e−02
BSG Basigin (Ok blood group) NM_001728 8023955 12 2.420 1.35e−02 3.196 1.60e−03
PPP2R5B Protein phosphatase 2, regulatory subunit B′, beta isoform NM_006244 7941087 16 2.124 1.53e−02 3.889 2.50e−05
NR1D2 Nuclear receptor subfamily 1, group D, member 2 NM_005126 8078272 11 2.640 9.80e−03 2.856 5.70e−03
ITGB2 Integrin, beta 2 (complement component 3 receptor 3 and 4) NM_000211 8070826 20 3.943 2.10e−06 1.962 1.58e−02
BEX4 Brain expressed, X-linked 4 NM_001080425 8169009 5 5.476 2.80e−03 3.973 1.30e−02
SERPINA1 Serpin peptidase inhibitor, clade A (alpha-1) NM_001002236 7981068 9 6.057 2.20e−05 2.684 1.60e−02
ALOX5 Arachidonate 5-lipoxygenase NM_000698 7927215 15 7.011 2.60e−09 2.161 1.60e−02
MORC2 MORC family CW-type zinc finger 2 NM_014941 8075430 27 2.332 7.60e−04 1.798 1.54e−02
PASK PAS domain containing serine NM_015148 8060205 19 2.946 2.80e−04 1.988 1.62e−02
IFNGR1b Interferon gamma receptor 1 NM_000416 8129861 10 2.933 7.00e−03 2.756 1.00e−02
ACTN1a,c Actinin, alpha 1 NM_001130004 7979824 24 1.978 9.00e−03 1.987 8.20e−03
FAM50B Family with sequence similarity 50, member B NM_012135 8116658 3 6.831 1.00e−02 7.868 6.60e−03
LPAR5 Lysophosphatidic acid receptor 5 NM_020400 7960637 4 5.092 1.00e−02 5.541 7.10e−03
LIPA Lipase A, lysosomal acid, cholesterol esterase NM_001127605 7934920 15 2.567 4.00e−03 2.216 1.34e−02
SAMHD1 SAM domain and HD domain 1 NM_015474 8066117 18 8.354 7.60e−13 2.002 1.76e−02
CD53 CD53 molecule NM_000560 7903893 12 2.329 1.70e−02 3.636 4.70e−04
DDX5 DEAD (Asp-Glu-Ala-Asp) box polypeptide 5 NM_004396 8017634 16 2.088 1.70e−02 3.002 6.40e−04
DCLK1 Doublecortin-like kinase 1 NM_004734 7970954 18 2.446 3.00e−03 2.044 1.50e−02
EPRS Glutamyl-prolyl-tRNA synthetase NM_004446 7924351 34 1.661 1.90e−02 3.021 1.00e−06
HSPA4 Heat shock 70kDa protein 4 NM_002154 8108015 24 1.819 1.87e−02 3.849 3.70e−07
GSTM1 Glutathione S-transferase mu 1 NM_000561 7903765 9 2.603 1.89e−02 6.225 1.70e−05
S100A11 S100 calcium binding protein A11 NM_005620 7920128 6 4.134 5.60e−03 3.481 1.34e−02
ARHGAP26 Rho GTPase activating protein 26 NM_015071 8108873 26 2.376 7.20e−04 1.781 1.86e−02
BAIAP3 BAI1-associated protein 3 NM_003933 7992219 35 1.655 1.81e−02 2.040 1.30e−03
FAM111B Family with sequence similarity 111, member B NM_198947 7940147 5 4.244 9.70e−03 4.211 1.01e−02
LGMN Legumain NM_005606 7980958 11 4.797 4.70e−05 2.357 2.01e−02
CSF2RAb Colony stimulating factor 2 receptor, alpha, low-affinity NM_001161531 8165735 16 2.081 1.80e−02 2.636 2.40e−03
CSF2RAb Colony stimulating factor 2 receptor, alpha, low-affinity NM_001161531 8176306 16 2.081 1.80e−02 2.636 2.40e−03
KDM5B Lysine (K)-specific demethylase 5B NM_006618 7923453 29 1.851 9.00e−03 1.821 1.11e−02
PPP2R2A Protein phosphatase 2 (formerly 2A), regulatory subunit NM_002717 8145440 13 2.204 2.00e−02 3.410 5.60e−04
UTRNa,c Utrophin NM_007124 8122464 72 1.415 2.10e−02 4.946 7.10e−26
BAX BCL2-associated X protein NR_027882 8030158 12 2.659 7.00e−03 2.410 1.39e−02
MUM1 Melanoma associated antigen (mutated) 1 NR_024247 8024255 17 4.409 2.00e−06 1.986 2.15e−02
ZNF509 Zinc finger protein 509 NM_145291 8093829 9 2.619 1.83e−02 3.417 3.50e−03
GABARAPL2 GABA(A) receptor-associated protein-like 2 NM_007285 7997272 8 2.694 2.12e−02 4.776 5.10e−04
CHST15 Carbohydrate (N-acetylgalactosamine 4-sulfate 6-O) NM_015892 7936856 14 2.139 2.05e−02 3.019 1.20e−03
CD247 CD247 molecule NM_198053 7922040 10 3.484 1.90e−03 2.457 2.00e−02
SBF2 SET binding factor 2 NM_030962 7946516 42 1.740 5.80e−03 1.599 1.66e−02
SEC13 SEC13 homolog (S. cerevisiae) NR_024272 8085300 10 3.295 2.90e−03 2.465 1.96e−02
PWP2 PWP2 periodic tryptophan protein homolog (yeast) NM_005049 8069003 22 1.965 1.19e−02 1.986 1.08e−02
ELMOD3 ELMO/CED-12 domain containing 3 NM_001135021 8043131 21 2.138 6.30e−03 1.920 1.66e−02
ZFP82 Zinc finger protein 82 homolog (mouse) NM_133466 8036309 8 2.874 1.52e−02 3.222 8.00e−03
APBB1IP Amyloid beta (A4) precursor protein-binding, family B, m NM_019043 7926786 18 1.969 2.00e−02 2.374 4.10e−03
IGSF6 Immunoglobulin superfamily, member 6 NM_005849 8000184 7 9.837 2.00e−06 2.800 2.44e−02
HDDC2 HD domain containing 2 NM_016063 8129363 11 2.423 1.70e−02 2.736 7.70e−03
GRK1 G protein-coupled receptor kinase 1 NM_002929 7970325 3 12.093 1.30e−03 5.219 2.34e−02
C6orf114 Chromosome 6 open reading frame 114 AF264036 8123981 3 8.242 5.60e−03 5.597 1.92e−02
PEX10 Peroxisomal biogenesis factor 10 NM_153818 7911720 9 2.496 2.37e−02 3.854 1.40e−03
RBM12B RNA binding motif protein 12B NM_203390 8151788 6 3.592 1.15e−02 3.472 1.36e−02
TRPM3 Transient receptor potential cation channel, subfamily M NM_206946 8161654 38 1.574 2.51e−02 2.281 1.30e−04
PHKG2 Phosphorylase kinase, gamma 2 (testis) NM_000294 7994928 10 2.654 1.27e−02 2.651 1.27e−02
C20orf11 Chromosome 20 open reading frame 11 NM_017896 8064007 6 4.198 5.20e−03 3.173 2.04e−02
GFM2 G elongation factor, mitochondrial 2 NM_032380 8112622 23 1.770 2.61e−02 2.695 2.70e−04
RUNX1T1 Runt-related transcription factor 1; translocated to, 1 NM_175634 8151768 15 2.030 2.47e−02 2.813 1.70e−03
CHAF1B Chromatin assembly factor 1, subunit B (p60) NM_005441 8068478 15 2.618 3.40e−03 2.047 2.33e−02
ZNF483 Zinc finger protein 483 NM_133464 8157193 9 2.486 2.42e−02 3.560 2.60e−03
C6orf25 Chromosome 6 open reading frame 25 NM_138277 8178074 5 3.752 1.65e−02 4.142 1.09e−02
TSPY1 Testis specific protein, Y-linked 1 NM_003308 8176524 7 5.534 3.80e−04 2.735 2.71e−02
ZBTB8B Zinc finger and BTB domain containing 8B NM_001145720 7899797 13 2.231 1.86e−02 2.472 9.10e−03
STXBP2 Syntaxin binding protein 2 NM_006949 8025255 22 1.776 2.79e−02 3.902 8.40e−07
ME3 Malic enzyme 3, NADP(+)-dependent, mitochondrial NM_001014811 7950864 18 3.910 7.60e−06 1.876 2.84e−02
HECW2 HECT, C2 and WW domain containing E3 ubiquitin protein NM_020760 8057898 30 1.869 7.60e−03 1.694 2.09e−02
ZNF827 Zinc finger protein 827 NM_178835 8103025 15 2.204 1.39e−02 2.177 1.52e−02
RPS28 Ribosomal protein S28 NM_001031 7942824 5 3.249 2.90e−02 8.974 1.40e−04
FLJ46300 FLJ46300 protein eNST00000341866 7937073 5 5.286 3.40e−03 3.345 2.60e−02
RRAGD Ras-related GTP binding D NM_021244 8128123 9 2.511 2.29e−02 3.100 6.70e−03
BRP44 Brain protein 44 NR_026550 7922095 8 4.073 1.70e−03 2.528 2.90e−02
ANGEL2 Angel homolog 2 (Drosophila) NM_144567 7924190 12 2.190 2.53e−02 2.744 5.50e−03
PRRG4 Proline rich Gla (G-carboxyglutamic acid) 4 (transmembrane) NM_024081 7939150 7 2.888 2.11e−02 3.344 1.01e−02
IL1B Interleukin 1, beta NM_000576 8054722 8 2.764 1.86e−02 2.955 1.30e−02
ADAR Adenosine deaminase, RNA-specific NM_001111 7920531 20 1.803 3.04e−02 2.495 1.50e−03
SYNE2a,c Spectrin repeat containing, nuclear envelope 2 NM_182914 7974920 124 1.461 1.80e−03 1.280 3.02e−02
CXCL16 Chemokine (C-X-C motif) ligand 16 NM_022059 8011713 10 2.254 3.19e−02 4.472 2.10e−04
CSF1 Colony stimulating factor 1 (macrophage) NM_000757 7903786 16 2.060 1.91e−02 2.168 1.31e−02
PTPRE Protein tyrosine phosphatase, receptor type, E NM_006504 7931353 25 1.775 2.10e−02 1.923 1.13e−02
ZNF540 Zinc finger protein 540 NM_152606 8028266 8 2.510 2.99e−02 3.854 2.50e−03
QSOX1 Quiescin Q6 sulfhydryl oxidase 1 NM_002826 7907830 16 1.925 3.05e−02 2.686 2.00e−03
RYR2 Ryanodine receptor 2 (cardiac) NM_001035 7910792 104 1.364 1.49e−02 1.351 1.76e−02
ABCD4 ATP-binding cassette, sub-family D (ALD), member 4 NM_005050 7980115 24 1.983 8.40e−03 1.764 2.44e−02
SPPL2B Signal peptide peptidase-like 2B NM_001077238 8024446 20 1.789 3.22e−02 2.682 6.60e−04
CTSD Cathepsin D NM_001909 7945666 11 2.184 3.11e−02 3.281 1.90e−03
SLC9A6 Solute carrier family 9 (sodium NM_001042537 8170097 19 2.046 1.28e−02 1.933 2.02e−02
ARID1A AT rich interactive domain 1A (SWI-like) NM_006015 7899220 23 1.720 3.27e−02 2.524 6.40e−04
RAPGEF4 Rap guanine nucleotide exchange factor (GEF) 4 NM_007023 8046428 32 3.159 7.50e−07 1.585 3.35e−02
VAPB VAMP (vesicle-associated membrane protein)-associated protein NM_004738 8063620 13 2.075 2.94e−02 2.733 4.20e−03
SLC1A1 Solute carrier family 1 (neuronal) NM_004170 8154135 15 1.932 3.40e−02 4.279 1.20e−05
CIRH1A Cirrhosis, autosomal recessive 1A (cirhin) NM_032830 7996891 16 2.323 7.50e−03 1.966 2.65e−02
NCRNA00169 Non-protein coding RNA 169 NR_026675 7993821 3 4.867 2.83e−02 8.058 6.00e−03
ETF1 Eukaryotic translation termination factor 1 NM_004730 8114443 11 2.412 1.75e−02 2.416 1.73e−02
CDH5 Cadherin 5, type 2 (vascular endothelium) NM_001795 7996264 16 1.984 2.50e−02 2.229 1.05e−02
ACSL4 Acyl-CoA synthetase long-chain family member 4 NM_022977 8174474 19 2.901 3.40e−04 1.788 3.57e−02
LRFN2b Leucine rich repeat and fibronectin type III domain NM_020737 8119390 3 14.282 6.70e−04 4.468 3.55e−02
FLJ43315 Similar to Asparagine synthetase [glutamine-hydrolyzing] BC057848 8077198 5 5.599 2.50e−03 3.117 3.37e−02
RGPD1 RANBP2-like and GRIP domain containing 1 NM_001024457 8043324 22 1.747 3.18e−02 2.158 4.80e−03
CPSF3 Cleavage and polyadenylation specific factor 3, 73kDa NM_016207 8040142 20 1.789 3.21e−02 2.242 4.70e−03
B4GALT1 UDP-Gal:betaGlcNAc beta 1,4- galactosyltransferase, poly NM_001497 8160637 9 3.330 4.20e−03 2.336 3.31e−02
NPTXR Neuronal pentraxin receptor NM_014293 8076169 6 3.733 9.60e−03 2.941 2.82e−02
CYFIP1 Cytoplasmic FMR1 interacting protein 1 NM_014608 7981824 34 2.359 1.50e−04 1.542 3.82e−02
C9orf78 Chromosome 9 open reading frame 78 NM_016520 8164596 10 2.619 1.40e−02 2.362 2.49e−02
KLHL7 Kelch-like 7 (Drosophila) NM_018846 8131815 15 1.979 2.90e−02 2.319 9.40e−03
OCRL Oculocerebrorenal syndrome of Lowe NM_000276 8169811 24 1.664 3.90e−02 2.881 7.30e−05
KPNA2 Karyopherin alpha 2 (RAG cohort 1, importin alpha 1) NM_002266 8019737 13 2.241 1.80e−02 2.193 2.08e−02
RPGR Retinitis pigmentosa GTPase regulator NM_000328 8172056 25 1.648 3.90e−02 2.646 2.00e−04
ABAT 4-aminobutyrate aminotransferase NM_020686 7993126 19 1.988 1.60e−02 1.900 2.30e−02
ZNF852 Zinc finger protein 852 AK296954 8086494 3 6.607 1.20e−02 4.900 2.78e−02
KIAA1324L KIAA1324-like NM_001142749 8140709 20 2.502 1.00e−03 1.747 3.81e−02
ATP4A ATPase, H+ NM_000704 8036110 22 2.147 5.00e−03 1.727 3.46e−02
JAKMIP2 Janus kinase and microtubule interacting protein 2 NM_014790 8114938 25 1.651 3.80e−02 2.268 1.60e−03
PRG2 Plasticity-related gene 2 NM_024888 8032094 11 2.426 1.70e−02 2.305 2.29e−02
ZMYND8 Zinc finger, MYND-type containing 8 NM_183047 8066786 31 3.091 1.80e−06 1.565 3.98e−02
FCGR1B Fc fragment of IgG, high affinity Ib, receptor (CD64) NM_001017986 7919133 5 15.021 2.80e−06 2.970 3.99e−02
PTBP2 Polypyrimidine tract binding protein 2 NM_021190 7903188 14 3.029 1.20e−03 1.931 3.88e−02
PAF1 Paf1, RNA polymerase II associated factor, homolog (S. cerevisiae) NM_019088 8036720 16 3.231 2.80e−04 1.847 3.98e−02
CCDC90B Coiled-coil domain containing 90B NM_021825 7950753 8 2.743 1.90e−02 2.700 2.10e−02
TSPY1 Testis specific protein, Y-linked 1 NM_003308 8176508 8 3.347 6.00e−03 2.437 3.43e−02
CTSZd Cathepsin Z NM_001336 8067279 8 2.353 4.00e−02 4.688 5.90e−04
ZDHHC16 Zinc finger, DHHC-type containing 16 NM_198046 7929634 14 2.485 7.00e−03 1.975 3.41e−02
SMA5 Glucuronidase, beta pseudogene AK289851 8177544 9 2.797 1.30e−02 2.410 2.83e−02
CTPS CTP synthase NM_001905 7900510 20 1.815 2.90e−02 2.020 1.23e−02
TUBB4Q Tubulin, beta polypeptide 4, member Q NM_020040 8021919 4 4.782 1.30e−02 3.797 2.86e−02
ANXA6 Annexin A6 NM_001155 8115234 26 2.174 2.00e−03 1.630 3.93e−02
SMARCA1 SWI/SNF related, matrix associated, actin dependent NM_003069 8174985 30 2.980 5.40e−06 1.568 4.15e−02
TMCO3 Transmembrane and coiled-coil domains 3 NM_017905 7970301 15 2.377 7.70e−03 1.934 3.38e−02
ULK4 Unc-51-like kinase 4 (C. elegans) NM_017886 8086352 15 2.540 4.40e−03 1.899 3.77e−02
ZNF341 Zinc finger protein 341 NM_032819 8061946 11 4.044 2.90e−04 2.061 4.22e−02
TSPAN7 Tetraspanin 7 NM_004615 8166784 10 2.134 4.21e−02 3.962 6.40e−04
TPD52L1 Tumor protein D52-like 1 NM_001003395 8121838 11 3.000 3.90e−03 2.091 3.92e−02
ATP6V0E1 ATPase, H+ transporting, lysosomal 9kDa, V0 subunit e1 NM_003945 8110022 9 2.487 2.41e−02 2.588 1.95e−02
SREBF1 Sterol regulatory element binding transcription factor NM_001005291 8013135 21 1.784 2.97e−02 1.958 1.41e−02
AQP1 Aquaporin 1 (Colton blood group) NM_198098 8132118 11 2.545 1.25e−02 2.176 3.17e−02
DPY19L4 Dpy-19-like 4 (C. elegans) NM_181787 8147375 20 1.731 4.06e−02 2.306 3.60e−03
ZWINT ZW10 interactor NM_032997 7933707 15 2.432 6.40e−03 1.895 3.83e−02
C3orf1 Chromosome 3 open reading frame 1 NM_016589 8081867 10 2.802 9.00e−03 2.203 3.59e−02
CCR10 Chemokine (C-C motif) receptor 10 NM_016602 8015681 3 6.185 1.43e−02 4.721 3.07e−02
ITGA8 Integrin, alpha 8 NM_003638 7932254 30 1.599 3.52e−02 1.823 9.90e−03
BRD7 Bromodomain containing 7 NM_013263 8001350 20 1.704 4.52e−02 3.622 9.10e−06
MPHOSPH9 M-phase phosphoprotein 9 NM_022782 7967386 25 2.763 1.00e−04 1.615 4.52e−02
MKLN1 Muskelin 1, intracellular mediator containing kelch motifs NM_013255 8136259 21 1.724 3.83e−02 2.112 7.10e−03
UGGT1 UDP-glucose glycoprotein glucosyltransferase 1 NM_020120 8045090 43 1.496 3.27e−02 1.617 1.36e−02
ZCCHC2 Zinc finger, CCHC domain containing 2 NM_017742 8021546 16 2.057 1.93e−02 1.961 2.70e−02
STK19 Serine/threonine kinase 19 NR_026717 8118395 12 5.450 4.20e−06 1.965 4.64e−02
STK19 Serine/threonine kinase 19 NR_026717 8178164 12 5.450 4.20e−06 1.965 4.64e−02
CEMP1 Cementum protein 1 NM_001048212 7998817 2 11.348 1.51e−02 7.819 3.13e−02
TRPC4 Transient receptor potential cation channel, subfamily C, NM_016179 7971104 17 1.854 3.46e−02 2.148 1.18e−02
PLA1A Phospholipase A1 member A NM_015900 8081890 12 2.641 7.30e−03 2.029 3.91e−02
PTGS1 Prostaglandin-endoperoxide synthase 1 (prostaglandin G NM_000962 8157650 17 1.773 4.61e−02 2.951 5.40e−04
POLM Polymerase (DNA directed), mu NM_013284 8139281 17 1.960 2.36e−02 1.966 2.32e−02
PPM1D Protein phosphatase 1D magnesium-dependent, delta isoform NM_003620 8008922 10 2.872 7.60e−03 2.164 3.93e−02
COBLL1 COBL-like 1 NM_014900 8056343 15 2.081 2.09e−02 2.014 2.61e−02
CTSL2d Cathepsin L2 NM_001333 8162652 11 2.613 1.05e−02 2.119 3.66e−02
TRIM22 Tripartite motif-containing 22 NM_006074 7938035 11 2.867 5.50e−03 2.063 4.20e−02
FLII Flightless I homolog (Drosophila) NM_002018 8013191 30 2.052 2.50e−03 1.552 4.53e−02
EEF1A2 Eukaryotic translation elongation factor 1 alpha 2 NM_001958 8067652 9 2.183 4.55e−02 3.504 2.90e−03
AGFG1 ArfGAP with FG repeats 1 NM_001135187 8048847 16 1.791 4.80e−02 2.931 8.20e−04
GMDS GDP-mannose 4,6-dehydratase NM_001500 8123562 16 1.804 4.60e−02 2.597 2.80e−03
TMC3 Transmembrane channel-like 3 NM_001080532 7990848 24 1.660 3.94e−02 1.987 9.40e−03
USP18 Ubiquitin specific peptidase 18 NM_017414 8071155 5 2.875 4.45e−02 5.041 4.30e−03
PTPRSb Protein tyrosine phosphatase, receptor type, S NM_002850 8032926 38 2.614 8.10e−06 1.469 4.89e−02
ATP13A2 ATPase type 13A2 NM_022089 7912898 29 1.745 1.71e−02 1.630 3.19e−02
ARX Aristaless related homeobox NM_139058 8171867 6 2.693 3.99e−02 3.754 9.30e−03
AP2A1 Adaptor-related protein complex 2, alpha 1 subunit NM_014203 8030470 27 2.989 1.40e−05 1.569 4.95e−02
LAT Linker for activation of T cells NM_014387 7994541 17 2.383 4.90e−03 1.782 4.47e−02
PLVAP Plasmalemma vesicle associated protein NM_031310 8035297 6 2.663 4.16e−02 3.832 8.40e−03
SLC25A46 Solute carrier family 25, member 46 NM_138773 8107259 10 2.503 1.80e−02 2.251 3.21e−02
DPF1 D4, zinc and double PHD fingers family 1 NM_004647 8036460 12 1.949 4.85e−02 3.183 1.60e−03
PDGFD Platelet derived growth factor D NM_025208 7951351 11 2.718 8.00e−03 2.057 4.27e−02
C1orf175 Chromosome 1 open reading frame 175 NR_026782 7901634 27 1.727 2.23e−02 1.680 2.85e−02
POM121 POM121 membrane glycoprotein (rat) NM_172020 8133275 17 1.804 4.14e−02 2.204 9.60e−03
SUGT1 SGT1, suppressor of G2 allele of SKP1 (S. cerevisiae) NM_001130912 7969271 14 2.474 7.10e−03 1.891 4.39e−02
TSPY1 Testis specific protein, Y-linked 1 NM_003308 8176544 8 2.657 2.28e−02 2.537 2.85e−02
TBC1D21 TBC1 domain family, member 21 NM_153356 7984759 11 2.383 1.88e−02 2.164 3.26e−02
CMTM4 CKLF-like MARVEL transmembrane domain containing 4 NM_181521 8001830 10 2.095 4.60e−02 3.016 5.50e−03
LST1 Leukocyte specific transcript 1 NM_007161 8177988 7 2.367 4.97e−02 4.478 1.70e−03
LST1 Leukocyte specific transcript 1 NM_007161 8179268 7 2.367 4.97e−02 4.478 1.70e−03
OAS2 2′-5′-oligoadenylate synthetase 2, 69/71 kDa NM_002535 7958913 17 1.759 4.84e−02 2.399 4.60e−03
TNS1 Tensin 1 NM_022648 8058869 35 1.578 2.92e−02 1.609 2.41e−02
ECSIT ECSIT homolog (Drosophila) NM_016581 8034286 10 2.653 1.27e−02 2.146 4.09e−02
PCMTD1 Protein-L-isoaspartate (D-aspartate) O-methyltransferase NM_052937 8150714 7 2.696 2.89e−02 2.785 2.50e−02
RBAK RB-associated KRAB zinc finger NM_021163 8131292 3 5.417 2.11e−02 4.573 3.34e−02
RBM33 RNA binding motif protein 33 NM_053043 8137542 15 2.154 1.64e−02 1.896 3.82e−02
GCN1L1 GCN1 general control of amino-acid synthesis 1-like 1 NM_006836 7966938 57 1.620 5.50e−03 1.372 4.91e−02
SSH2 Slingshot homolog 2 (Drosophila) NM_033389 8013965 19 2.277 4.90e−03 1.702 4.97e−02
ZPLD1 Zona pellucida-like domain containing 1 NM_175056 8081407 19 1.806 3.33e−02 1.919 2.13e−02
CKS2 CDC28 protein kinase regulatory subunit 2 NM_001827 8156290 4 3.169 4.96e−02 5.916 5.40e−03
EGFLAMb EGF-like, fibronectin type III and laminin G domains NM_152403 8105013 26 1.853 1.28e−02 1.614 4.25e−02
UROD Uroporphyrinogen decarboxylase NM_000374 7901073 13 2.013 3.52e−02 2.202 2.03e−02
CWH43 Cell wall biogenesis 43 C-terminal homolog (S. cerevisiae) AK300495 8095005 3 4.689 3.13e−02 5.063 2.55e−02
REL V-rel reticuloendotheliosis viral oncogene homolog (avian) NM_002908 8042144 12 1.948 4.86e−02 2.579 8.70e−03
EPB41 Erythrocyte membrane protein band 4.1 (elliptocytosis 1, R) NM_203342 7899534 25 1.605 4.73e−02 1.914 1.05e−02
TSPY1 Testis specific protein, Y-linked 1 NM_003308 8176484 9 2.411 2.83e−02 2.383 3.00e−02
TM2D2 TM2 domain containing 2 NM_031940 8150364 8 2.444 3.39e−02 2.620 2.44e−02
CLSPN Claspin homolog (Xenopus laevis) NM_022111 7914851 26 1.691 2.92e−02 1.690 2.93e−02
CKMT1A Creatine kinase, mitochondrial 1A NM_001015001 7983256 13 1.890 4.98e−02 2.488 8.70e−03
THADA Thyroid adenoma associated NM_022065 8051820 41 1.652 1.21e−02 1.455 4.68e−02
GPR179 G protein-coupled receptor 179 NM_001004334 8014666 12 2.309 1.83e−02 2.014 4.07e−02
PNPLA8 Patatin-like phospholipase domain containing 8 NM_015723 8142307 16 1.784 4.92e−02 2.237 1.03e−02
IGDCC3b Immunoglobulin superfamily, DCC subclass, member 3 NM_004884 7989770 14 2.267 1.37e−02 1.877 4.58e−02
ADAM9 ADAM metallopeptidase domain 9 (meltrin gamma) NM_003816 8146000 23 1.714 3.35e−02 1.769 2.61e−02
PNKP Polynucleotide kinase 3′-phosphatase NM_007254 8038458 18 1.813 3.60e−02 1.924 2.38e−02
IFITM2 Interferon induced transmembrane protein 2 (1–8D) NM_006435 7937330 4 3.691 3.13e−02 3.793 2.87e−02
KCNG2 Potassium voltage-gated channel, subfamily G, member 2 NM_012283 8021900 2 8.880 2.46e−02 7.247 3.60e−02
LHFPL2 Lipoma HMGIC fusion partner-like 2 NM_005779 8112803 3 4.391 3.71e−02 5.200 2.36e−02
PDCD11 Programmed cell death 11 NM_014976 7930226 37 1.615 2.04e−02 1.503 4.15e−02
HIST1H4He Histone cluster 1, H4h NM_003543 8124448 4 4.697 1.36e−02 3.188 4.88e−02
SDCBP Syndecan binding protein (syntenin) NM_005625 8146550 8 2.390 3.75e−02 2.603 2.52e−02
MGC42105 Serine NM_153361 8105146 4 3.781 2.90e−02 3.602 3.38e−02
DIAPH1 Diaphanous homolog 1 (Drosophila) NM_005219 8114658 32 1.522 4.75e−02 1.716 1.56e−02
IL17RA Interleukin 17 receptor A NM_014339 8071069 14 2.100 2.32e−02 1.921 4.00e−02
MYO18A Myosin XVIIIA NM_078471 8013860 45 1.474 3.49e−02 1.502 2.85e−02
ACLY ATP citrate lyase NM_001096 8015460 29 1.687 2.35e−02 1.587 4.00e−02
KCNH3 Potassium voltage-gated channel, subfamily H (eag-related) NM_012284 7955231 16 1.794 4.76e−02 2.090 1.73e−02
CAMTA2 Calmodulin binding transcription activator 2 NM_015099 8011774 22 1.642 4.96e−02 1.908 1.55e−02
CD4 CD4 molecule NM_000616 7953428 13 2.282 1.60e−02 1.896 4.91e−02
PLXNB1 Plexin B1 NM_001130082 8086908 40 1.452 4.98e−02 1.625 1.57e−02
WISP1 WNT1 inducible signaling pathway protein 1 NM_003882 8148435 10 2.379 2.39e−02 2.138 4.17e−02
SLC18A2 Solute carrier family 18 (vesicular monoamine), member 2 NM_003054 7930837 19 1.942 1.95e−02 1.720 4.64e−02
KPNA2 Karyopherin alpha 2 (RAG cohort 1, importin alpha 1) NM_002266 8009417 12 2.332 1.72e−02 1.940 4.96e−02
MAPRE3c Microtubule-associated protein, RP NM_012326 8040742 10 2.330 2.68e−02 2.148 4.07e−02
RPL9 Ribosomal protein L9 NM_001024921 8099887 9 2.281 3.71e−02 2.346 3.24e−02
FOXD4L6 Forkhead box D4-like 6 NM_001085476 8161533 3 4.580 3.33e−02 4.399 3.69e−02
TGM2b Transglutaminase 2 (C polypeptide, protein-glutamine-gamma) NM_004613 8066214 16 1.858 3.83e−02 1.908 3.23e−02
ZDHHC17 Zinc finger, DHHC-type containing 17 NM_015336 7957277 20 1.838 2.63e−02 1.708 4.46e−02
ATP9A ATPase, class II, type 9A NM_006045 8067055 31 1.553 4.25e−02 1.623 2.88e−02
ZNF382 Zinc finger protein 382 NM_032825 8028194 5 3.392 2.46e−02 2.834 4.67e−02
HIST1H3De Histone cluster 1, H3d NM_003530 8124416 6 2.717 3.86e−02 2.824 3.32e−02
SLC25A3 Solute carrier family 25 (mitochondrial carrier) NM_213611 7957746 12 2.217 2.35e−02 1.946 4.88e−02
C12orf56 Chromosome 12 open reading frame 56 NM_001099676 7964687 11 2.185 3.10e−02 2.051 4.32e−02
AEBP1 AE binding protein 1 NM_001129 8132557 22 1.699 3.91e−02 1.714 3.65e−02
TNPO1 Transportin 1 NM_002270 8106122 18 1.748 4.58e−02 1.863 2.99e−02
ACOT2 Acyl-CoA thioesterase 2 NM_006821 7975602 4 3.878 2.66e−02 3.175 4.93e−02
POLR3E Polymerase (RNA) III (DNA directed) polypeptide E (80kDa) NM_018119 7993973 25 1.693 3.14e−02 1.613 4.56e−02
STK32A Serine/threonine kinase NM_001112724 8108981 13 1.971 3.96e−02 1.954 4.16e−02
PARP14 Poly (ADP-ribose) polymerase family, member 14 NM_017554 8082100 17 1.864 3.35e−02 1.755 4.91e−02
FOLR3 Folate receptor 3 (gamma) NM_000804 7942328 3 4.042 4.55e−02 4.389 3.71e−02
HIST1H2BDe Histone cluster 1, H2bd NM_021063 8117382 6 2.783 3.51e−02 2.808 4.82e−02
CRLF3b Cytokine receptor-like factor 3 NM_015986 8014037 9 2.313 3.47e−02 2.141 4.97e−02
MLC1 Megalencephalic leukoencephalopathy with subcortical cysts NM_015166 8076894 14 1.915 4.08e−02 1.881 4.53e−02
DNAJC9 DnaJ (Hsp40) homolog, subfamily C, member 9 NM_015190 7934320 5 2.882 4.42e−02 2.912 4.27e−02
MTSS1 Metastasis suppressor 1 NM_014751 8152764 17 1.780 4.51e−02 1.799 4.21e−02
GLRX3 Glutaredoxin 3 NM_006541 7931393 9 2.197 4.42e−02 2.140 4.98e−02
ACSL5 Acyl-CoA synthetase long-chain family member 5 NM_016234 7930498 25 1.610 4.64e−02 1.602 4.79e−02
NR4A1 Nuclear receptor subfamily 4, group A, member 1 NM_002135 7955589 16 1.787 4.87e−02 1.785 4.90e−02
*

Rows are sorted in ascending order by the average p-value across both brain regions.

a

Genes encoding proteins with actinin-type actin-binding domains

b

Genes encoding proteins with fibronectin type-III-folds

c

Genes encoding proteins with calponin-like actin-binding domains

d

Genes encoding proteins with peptidase C1A, papain C-terminals

e

Genes encoding histone core proteins

Confirmation and Replication Analyses

We selected three genes for confirmation and replication analyses. First was CPNE3, which exhibited a Bonferroni-corrected significant interaction in both BA10 (F=15.00, p=6.6e−24) and CAUD (F=18.52, p=1.0e−27). As shown in Figure 1, the significant difference in expression detected between diagnostic groups by microarray in both brain regions was relegated to the most distal end of the gene’s 3′UTR (panel A: BA10, F=35.89, p=9.7e−4; panel B: CAUD, F=113.56, p=4.0e−5), suggesting that SZ subjects expressed transcripts with relatively shorter 3′UTRs. In the same discovery sample used for the microarray analyses described above, qRTPCR confirmed differences between diagnostic groups in the expression levels of this segment of the 3′UTR of CPNE3 relative to a control region of the gene (upstream in the 3′UTR). This difference attained statistical significance in both CAUD (p=0.001) and BA10 (p=0.041). We next sought to replicate the decreased expression level of the distal 3′UTR segment of CPNE3 (again, relative to the non-differentially expressed proximal 3′UTR segment) in the remaining 16 SZ and 16 NC postmortem brain tissue samples from the Harvard Brain Tissue Resource Center. In this fully independent sample, we successfully replicated a highly significant decrease in 3′UTR expression in BA10 in SZ (p<0.001), and in CAUD SZ samples as well (p=0.034), indicating that this result generalizes across samples and brain regions.

Figure 1. Exonic Expression and Alternative 3′UTR Usage of CPNE3in: A) BA10; and B) CAUD.

Figure 1

Microarray results of differential probe expression of Copine3 (CPNE3) in Brodman Area 10 (A) and Caudate (B) in postmortem brain tissue samples from normal control subjects (NC) (n=4) and individuals with schizophrenia (SZ) (n=4). The interaction of diagnosis and exon ID was highly significant in both brain regions; in fact, it was the only gene for which a Bonferroni-corrected threshold for significance of this term was met. *There was a statistically significant difference in probe-level expression between diagnostic groups at the most distal end of the 3′UTR in both BA10 (p=9.7e-4) and CAUD (p=4.0e-5), indicating that a truncated transcript may be expressed in SZ patients.

The second gene followed-up by qRTPCR was ENAH, which was chosen based on the appearance of exonic dysregulation in the vicinity of a known splice site. Initial analyses of the small discovery sample by microarray showed that ENAH expression levels were influenced by a Bonferroni-corrected significant interaction of diagnosis and TE ID in BA10 (F=4.98, p=5.5e−07) but not in CAUD (F=0.57, p=0.890). As shown in Figure 2, SZ and NC subjects had equivalent levels of TE expression in most gene regions in both BA10 (panel A) and CAUD (panel B); however, there was a significant decrease in exon 11a expression in SZ in BA10 (F=15.96, p=0.007). This precise pattern was recapitulated by qRTPCR, with a significant SZ-associated decrease in ENAH exon 11a expression (relative to a non-dysregulated region of the gene: the boundary of exons 2 and 3) confirmed in BA10 (p=0.037), and no significant difference observed in CAUD (p=0.141) in the discovery sample. When extending this evaluation to the replication sample, the significant dysregulation of ENAH exon 11a was again detected in BA10 in SZ (p=0.035); unexpectedly, we also detected a significant decrease in exon 11a expression in CAUD in the replication sample by qRTPCR (p=0.001).

Figure 2. Exonic Expression and Alternative Splicing of ENAH in: A) BA10; and B) CAUD.

Figure 2

Microarray results of differential exon expression of Enabled Homolog (ENAH) in Brodman Area 10 (A) and Caudate (B) in postmortem brain tissue samples from normal control subjects (NC) (n=4) and individuals with schizophrenia (SZ) (n=4). The interaction of diagnosis and exon ID was significant only in BA10, not CAUD, indicating the possibility of regionally specific differential splice-variant expression between the groups. *There was a statistically significant difference in expression between diagnostic groups at exon 11a in BA10 (p=0.047) but not CAUD (p=0.489), indicating increased expression of the short (11a) isoform in SZ patients in BA10 only.

The last result selected for confirmation by qRTPCR was the up-regulation of KLHL5 exon 10 in SZ. This gene’s expression levels showed highly significant evidence of a diagnosis by TE ID interaction in both BA10 (F=4.17; p=1.1e−4) and CAUD (F=3.89; p=2.4e−4) by microarray. Interestingly, the strongest difference in expression in both brain regions did not involve the use of the known alternate transcription start-sites in the 5′ end of the gene, but was found at exon 10 (BA10: F=5.96, p=0.050; CAUD: F=8.79 p=0.025), where SZ subjects exhibited higher expression on average than NC subjects. Reanalysis of the discovery samples by qRTPCR did not directly verify the microarray results, as expression levels of the exon 10 and 11 boundary (relative to a non-dysregulated region of the gene: the boundary of exons 6 and 7) were not significantly higher in SZ (BA10: p=0.229; CAUD: p=1.000). Yet, when we examined the expression levels of this exon by qRTPCR in the independent replication sample, we again observed a significant increase in exon 10 expression in SZ in BA10 (p=0.011), though not in CAUD (p=1.000). Our inability to consistently verify the precise results observed by microarray in the discovery sample nor confirm them in the replication sample is perhaps not surprising given the relatively lax criteria used in selecting this gene for follow-up study relative to the Bonferroni-corrected significant results that drove our follow-up work on CPNE3 and ENAH. However, further investigation of KLHL5’s transcriptomic profile by direct sequencing or qRTPCR using primers with greater coverage may resolve this uncertainty in the future.

DISCUSSION

The main objective of this study was to assess how common and widespread exonic expression abnormalities are in postmortem brain in SZ. Our work suggests they are not uncommon, and are more complex than initially conceived, involving not only alternative splicing of traditional cassette exons, but also selection of mutually exclusive exons, alternate promoter selection, and 3′UTR constitution. By comparing brain regions linked to the disorder, as well as medicated and unmedicated patients (although in just a small, initial discovery sample), we were able to identify some expression abnormalities that generalized across regions and which may reflect stable traits associated with the disorder. Further work could evaluate whether such expression abnormalities arise from inherited mutations or biological insults acquired early in development. In contrast, most TE expression abnormalities observed in this study were restricted to either BA10 or CAUD. Such regionally specific abnormalities may be less likely attributable to regulatory effects of “splicing quantitative trait loci” (sQTLs) than are ubiquitous expression abnormalities; however, this does not preclude the possibility that sQTLs could be differentially regulated in different cells or brain regions. Such region-specific effects might reflect the diverse developmental influences governing maturation of those brain regions, or their differential sensitivity to schizophrenogenic environmental insults. Alternatively, these region-specific effects may result from differential expression of facilitators or inhibitors of splicing (i.e., alternative splicing regulators) in a similarly region-specific manner. In support of this possibility, we found nominally significant evidence of dysregulation of full-length splicing-regulatory transcripts such as HNRNPH1 (p=0.004), HNRNPH3 (p=0.039), HNRNPC (p=0.040), and SFRS16 (p=0.018) in BA10 but not CAUD in the same samples examined here (unpublished data). The systematic follow-up of our results and evaluation of these contributory possibilities should be a high priority for future work.

Our analyses of microarray-derived TE expression data from a small sample of well-matched SZ and NC samples generated many leads, two of which we validated (either perfectly or partially) using another, more sensitive analytic technique (qRTPCR). These two results also replicated in a larger, more heterogeneous, and fully independent sample of SZ and NC subjects. One of these genes (CPNE3, a calcium-dependent membrane-binding protein that co-localizes with phosphorylated focal adhesion kinase at the leading edge of migrating cells (Heinrich et al., 2010)) has never before been implicated in SZ, and the other (ENAH, an actin-associated protein involved in cytoskeleton remodeling and neuronal projection) was targeted just once among a panel of 18 target genes (Kahler et al., 2008), highlighting the advantage of a transcriptome-wide approach that can generate new candidate genes and hypotheses regarding this complex multifactorial disorder.

Aside from generating new hypotheses, this work validates our prior blood-based biomarker study of TE expression in SZ (Glatt et al., 2009). For example, 44 (28%) of the 156 genes that we previously found to exhibit Bonferroni-corrected significant abnormal expression of a TE in peripheral blood cells in psychotic subjects (SZ plus psychotic bipolar disorder) were also found to have at least nominally significant abnormal TE expression in either BA10 or CAUD in the present study of postmortem brain. Further, eight of those 156 genes (ADAR, ARHGAP26, BIRC6, MAPK14, STXBP2, SYNE2, UTRN, and ZDHHC17) had TEs that were Bonferroni-corrected significant in blood and at least nominally significantly dysregulated in both brain regions, a result very unlikely to occur by chance (binomial test, p<0.0001). This type of convergence suggests two areas for future work; the pursuit of factors that may be capable of disrupting the expression of particular TEs regardless of tissue type, and the validation of blood-based biomarkers for these disorders.

This study also lends support to hypotheses generated by the prior work of others. In particular, the dysregulation of ERBB4 exons we observed in both BA10 and CAUD confirmed prior postmortem work linking particular splice variants of this neuregulin-1 cofactor to risk for the disorder (Kao et al., 2010; Law et al., 2007; Silberberg et al., 2006). We could not confirm in our small microarray sample the differential splicing of other previously observed results for SZ candidate genes, such as CTNNA2, DISC1, ESR1, GRM3, and NRG1; however, because our microarray sample was very small it is possible that these effects may have eluded detection in our sample due primarily to lack of power. This seems particularly likely for DISC1 and NRG1, as we did observe patterns of TE-expression data suggestive of differential splicing of known alternatively spliced TEs of these genes that did not attain statistical significance due to relatively large variance. It is also possible that the prior results are specific for regions of the brain (e.g., BA9) we did not have available for analysis. Yet, several other genes for which functionally distinct and neurodevelopmentally important splice variants are known (including NUMBL, DSCAM, FGF, SNAP25, and Neuroligins 2 and 4X) were also found in our study to have SZ-associated dysregulation of one or more exons in postmortem BA10, but not CAUD, reinforcing the importance of pursuing both ubiquitous and regionally specific alterations in exonic expression.

This study, like all human postmortem studies of brain disorders, is subject to several limitations. Primarily, because postmortem brain tissue from schizophrenia patients is an extremely rare and highly prized commodity, the sample size available for this study was quite small. Our design, which included carving both discovery and replication samples from the same small primary sample, further exacerbated this problem; however, this approach was taken to foster more confidence in those results that did survive replication. This anonymized sample was also not ideal because we were unable to determine the ancestry of each subject, which might relate to either genetically or environmentally mediated differences in TE expression. Thus, if the SZ and NC groups differed systematically in ancestry and if the represented ancestral groups differed systematically in expression levels of particular mRNA isoforms, then it is possible we would have falsely attributed some of these ancestry-related differences to diagnosis. Another limitation is that, because we did not have access to brain tissue from first-episode or prodromal patients, we are unable to rule out the possibility that the observed results are a function of (rather than cause of or contributor to) having SZ, such as treatment, hospitalization, medical and psychiatric comorbidity, and substance use disorders, all of which are more common among SZ patients than NC subjects. In an attempt to control for these factors, we followed-up by qRTPCR only those genes that did not differ in TE expression between the treated and untreated subgroups; however, power to detect such differences was quite low due to the small number of subjects in each group and the fact that all patients had been medicated at some time, even if not at the time of death. Future in vitro studies will be instrumental for validating these splicing abnormalities and more strongly attributing them to sequence variation rather than personal, clinical, agonal, or other factors.

In conclusion, we demonstrated the utility of examining the functional genomic output of the human brain in SZ using TE- and brain-region-specific profiling of expression intensity by microarray. The SZ-associated TE-expression abnormalities validated and replicated in this study would not have been detected using earlier generations of “whole-transcript” expression arrays. Looking back, this might explain in part why prior microarray-based studies of postmortem brain tissue in SZ have not routinely observed identical patterns of transcript dysregulation. Looking ahead, the use of RNA sequencing should facilitate the detection of additional subtle splicing (and other RNA-processing) variations that might characterize particular brain regions (or the whole organism) in SZ. Additional work is needed to extend our results into other samples and other implicated brain regions, as well as to uncover the factors (genetic or otherwise) that influence the observed expression abnormalities in brain and in blood. Ultimately the generation of region-specific transcriptome profiles that include information on the relative abundance of each splice variant may prove essential for gaining a better understanding of the biological basis of SZ and the development of better biomarkers and treatments for the disorder.

Supplementary Material

01
02

Acknowledgments

This work was supported in part by grants R21MH075027 (M.T.T.), P50MH081755-0003 (S.J.G.), and R01MH085521 (S.J.G.) from the U.S. National Institutes of Health, a Young Investigator Award and the Sidney R. Baer, Jr. Prize for Schizophrenia Research (S.J.G.) from NARSAD: The Brain and Behavior Research Fund, and A Research Grant from The Gerber Foundation (S.J.G.). The authors wish to thank Dr. Francine M. Benes, George Tejada, and the staff of the Harvard Brain Tissue Resource Center for providing postmortem brain tissue samples, Dr. George H. Trksak for determining medication status at the time of death of the SZ patients, and those subjects and family members who made this study possible.

The funding source had no role in designing the study, analyzing the data, or interpreting the results.

Footnotes

The authors report no conflicts of interest.

Ori S. Cohen designed and performed the experiments and wrote the manuscript.

Sarah Y. Mccoy, Sean Bialosuknia, Lu Liu, and Yanli Zhang-James assisted with experiments and contributed sections of text to the manuscript.

Frank A. Middleton participated in the design and execution of the neuroanatomical and molecular biological aspects of the experiments.

Ming T. Tsuang, Stephen V. Faraone, & Stephen J. Glatt designed the experiments and wrote and edited portions of the manuscript.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Affymetrix, Inc. Data Sheet: GeneChip® Gene 1.0 ST Array System for Human, Mouse and Rat. 2007. [Google Scholar]
  2. Clark TA, Schweitzer AC, Chen TX, Staples MK, Lu G, Wang H, Williams A, Blume JE. Discovery of tissue-specific exons using comprehensive human exon microarrays. Genome Biology. 2007;8(4):R64. doi: 10.1186/gb-2007-8-4-r64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Dennis G, Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, Lempicki RA. DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biology. 2003;4(5):3. [PubMed] [Google Scholar]
  4. Dho SE, French MB, Woods SA, McGlade CJ. Characterization of four mammalian numb protein isoforms. Identification of cytoplasmic and membrane-associated variants of the phosphotyrosine binding domain. Journal of Biological Chemistry. 1999;274(46):33097–33104. doi: 10.1074/jbc.274.46.33097. [DOI] [PubMed] [Google Scholar]
  5. Dho SE, Trejo J, Siderovski DP, McGlade CJ. Dynamic regulation of mammalian numb by G protein-coupled receptors and protein kinase C activation: Structural determinants of numb association with the cortical membrane. Molecular Biology of the Cell. 2006;17(9):4142–4155. doi: 10.1091/mbc.E06-02-0097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Ellison-Wright I, Glahn DC, Laird AR, Thelen SM, Bullmore E. The anatomy of first-episode and chronic schizophrenia: an anatomical likelihood estimation meta-analysis. Am J Psychiatry. 2008;165(8):1015–1023. doi: 10.1176/appi.ajp.2008.07101562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Glatt SJ, Chandler SD, Bousman CA, Chana G, Lucero GR, Tatro E, May T, Lohr JB, Kremen WS, Everall IP, Tsuang MT. Alternatively Spliced Genes as Biomarkers for Schizophrenia, Bipolar Disorder and Psychosis: A Blood-Based Spliceome-Profiling Exploratory Study. Curr Pharmacogenomics Person Med. 2009;7(3):164–188. doi: 10.2174/1875692110907030164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Glatt SJ, Cohen OS, Faraone SV, Tsuang MT. Dysfunctional gene splicing as a potential contributor to neuropsychiatric disorders. Am J Med Genet B Neuropsychiatr Genet. 2011;156B(4):382–392. doi: 10.1002/ajmg.b.31181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Glatt SJ, Everall IP, Kremen WS, Corbeil J, Sasik R, Khanlou N, Han M, Liew CC, Tsuang MT. Comparative gene expression analysis of blood and brain provides concurrent validation of SELENBP1 up-regulation in schizophrenia. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(43):15533–15538. doi: 10.1073/pnas.0507666102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Goghari VM. Executive functioning-related brain abnormalities associated with the genetic liability for schizophrenia: an activation likelihood estimation meta-analysis. Psychol Med. 2010:1–14. doi: 10.1017/S0033291710001972. [DOI] [PubMed] [Google Scholar]
  11. Handran S, Pickett S, Verdick D. Key Considerations for Accurate Microarray Scanning and Image Analysis. In: Kamberova G, Shah S, editors. DNA Array Image Analysis: Nuts & Bolts. DNA Press, LLC; Salem, MA: 2002. pp. 83–98. [Google Scholar]
  12. Heinrich C, Keller C, Boulay A, Vecchi M, Bianchi M, Sack R, Lienhard S, Duss S, Hofsteenge J, Hynes NE. Copine-III interacts with ErbB2 and promotes tumor cell migration. Oncogene. 2010;29(11):1598–1610. doi: 10.1038/onc.2009.456. [DOI] [PubMed] [Google Scholar]
  13. Horvath S, Janka Z, Mirnics K. Analyzing schizophrenia by DNA microarrays. Biol Psychiatry. 2011;69(2):157–162. doi: 10.1016/j.biopsych.2010.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, Speed TP. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 2003;4(2):249–264. doi: 10.1093/biostatistics/4.2.249. [DOI] [PubMed] [Google Scholar]
  15. Kahler AK, Djurovic S, Kulle B, Jonsson EG, Agartz I, Hall H, Opjordsmoen S, Jakobsen KD, Hansen T, Melle I, Werge T, Steen VM, Andreassen OA. Association analysis of schizophrenia on 18 genes involved in neuronal migration: MDGA1 as a new susceptibility gene. Am J Med Genet B Neuropsychiatr Genet. 2008;147B(7):1089–1100. doi: 10.1002/ajmg.b.30726. [DOI] [PubMed] [Google Scholar]
  16. Kao WT, Wang Y, Kleinman JE, Lipska BK, Hyde TM, Weinberger DR, Law AJ. Common genetic variation in Neuregulin 3 (NRG3) influences risk for schizophrenia and impacts NRG3 expression in human brain. Proc Natl Acad Sci U S A. 2010;107(35):15619–15624. doi: 10.1073/pnas.1005410107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Law AJ, Kleinman JE, Weinberger DR, Weickert CS. Disease-associated intronic variants in the ErbB4 gene are related to altered ErbB4 splice-variant expression in the brain in schizophrenia. Human Molecular Genetics. 2007;16(2):129–141. doi: 10.1093/hmg/ddl449. [DOI] [PubMed] [Google Scholar]
  18. Li Q, Lee JA, Black DL. Neuronal regulation of alternative pre-mRNA splicing. Nature Reviews Neuroscience. 2007;8(11):819–831. doi: 10.1038/nrn2237. [DOI] [PubMed] [Google Scholar]
  19. Mexal S, Berger R, Pearce L, Barton A, Logel J, Adams CE, Ross RG, Freedman R, Leonard S. Regulation of a novel alphaN-catenin splice variant in schizophrenic smokers. American Journal of Medical Genetics B Neuropsychiatric Genetics. 2008;147B(6):759–768. doi: 10.1002/ajmg.b.30679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Mirnics K, Levitt P, Lewis DA. Critical appraisal of DNA microarrays in psychiatric genomics. Biol Psychiatry. 2006;60(2):163–176. doi: 10.1016/j.biopsych.2006.02.003. [DOI] [PubMed] [Google Scholar]
  21. Nakata K, Lipska BK, Hyde TM, Ye T, Newburn EN, Morita Y, Vakkalanka R, Barenboim M, Sei Y, Weinberger DR, Kleinman JE. DISC1 splice variants are upregulated in schizophrenia and associated with risk polymorphisms. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(37):15873–15878. doi: 10.1073/pnas.0903413106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Pan Q, Shai O, Lee LJ, Frey BJ, Blencowe BJ. Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat Genet. 2008;40(12):1413–1415. doi: 10.1038/ng.259. [DOI] [PubMed] [Google Scholar]
  23. Partek Incorporated. Partek Documentation. Partek Incorporated; 2008. [Google Scholar]
  24. Reugels AM, Boggetti B, Scheer N, Campos-Ortega JA. Asymmetric localization of Numb:EGFP in dividing neuroepithelial cells during neurulation in Danio rerio. Developmental Dynamics. 2006;235(4):934–948. doi: 10.1002/dvdy.20699. [DOI] [PubMed] [Google Scholar]
  25. Sartorius LJ, Weinberger DR, Hyde TM, Harrison PJ, Kleinman JE, Lipska BK. Expression of a GRM3 splice variant is increased in the dorsolateral prefrontal cortex of individuals carrying a schizophrenia risk SNP. Neuropsychopharmacology. 2008;33(11):2626–2634. doi: 10.1038/sj.npp.1301669. [DOI] [PubMed] [Google Scholar]
  26. Schmucker D, Clemens JC, Shu H, Worby CA, Xiao J, Muda M, Dixon JE, Zipursky SL. Drosophila Dscam is an axon guidance receptor exhibiting extraordinary molecular diversity. Cell. 2000;101(6):671–684. doi: 10.1016/s0092-8674(00)80878-8. [DOI] [PubMed] [Google Scholar]
  27. Silberberg G, Darvasi A, Pinkas-Kramarski R, Navon R. The involvement of ErbB4 with schizophrenia: association and expression studies. American Journal of Medical Genetics B Neuropsychiatric Genetics. 2006;141B(2):142–148. doi: 10.1002/ajmg.b.30275. [DOI] [PubMed] [Google Scholar]
  28. Tan W, Wang Y, Gold B, Chen J, Dean M, Harrison PJ, Weinberger DR, Law AJ. Molecular cloning of a brain-specific, developmentally regulated neuregulin 1 (NRG1) isoform and identification of a functional promoter variant associated with schizophrenia. Journal of Biological Chemistry. 2007;282(33):24343–24351. doi: 10.1074/jbc.M702953200. [DOI] [PubMed] [Google Scholar]
  29. Twine NA, Janitz K, Wilkins MR, Janitz M. Whole transcriptome sequencing reveals gene expression and splicing differences in brain regions affected by Alzheimer’s disease. PLoS One. 2011;6(1):e16266. doi: 10.1371/journal.pone.0016266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Weickert CS, Miranda-Angulo AL, Wong J, Perlman WR, Ward SE, Radhakrishna V, Straub RE, Weinberger DR, Kleinman JE. Variants in the estrogen receptor alpha gene and its mRNA contribute to risk for schizophrenia. Human Molecular Genetics. 2008;17(15):2293–2309. doi: 10.1093/hmg/ddn130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Yu K, Cheung C, Leung M, Li Q, Chua S, McAlonan G. Are Bipolar Disorder and Schizophrenia Neuroanatomically Distinct? An Anatomical Likelihood Meta-analysis. Front Hum Neurosci. 2010;4:189. doi: 10.3389/fnhum.2010.00189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Zipursky SL, Wojtowicz WM, Hattori D. Got diversity? Wiring the fly brain with Dscam. Trends in Biochemical Science. 2006;31(10):581–588. doi: 10.1016/j.tibs.2006.08.003. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

01
02

RESOURCES