You are on page 1of 12

Archival Report Biological

Psychiatry

An Ultraconserved Brain-Specific Enhancer


Within ADGRL3 (LPHN3) Underpins Attention-
Deficit/Hyperactivity Disorder Susceptibility
Ariel F. Martinez, Yu Abe, Sungkook Hong, Kevin Molyneux, David Yarnell, Heiko Löhr,
Wolfgang Driever, Maria T. Acosta, Mauricio Arcos-Burgos, and Maximilian Muenke

ABSTRACT
BACKGROUND: Genetic factors predispose individuals to attention-deficit/hyperactivity disorder (ADHD). Previous
studies have reported linkage and association to ADHD of gene variants within ADGRL3. In this study, we
functionally analyzed noncoding variants in this gene as likely pathological contributors.
METHODS: In silico, in vitro, and in vivo approaches were used to identify and characterize evolutionary conserved
elements within the ADGRL3 linkage region ( 207 Kb). Family-based genetic analyses of 838 individuals (372
affected and 466 unaffected patients) identified ADHD-associated single nucleotide polymorphisms harbored in
some of these conserved elements. Luciferase assays and zebrafish green fluorescent protein transgenesis tested
conserved elements for transcriptional enhancer activity. Electromobility shift assays were used to verify transcription
factor–binding disruption by ADHD risk alleles.
RESULTS: An ultraconserved element was discovered (evolutionary conserved region 47) that functions as a transcriptional
enhancer. A three-variant ADHD risk haplotype in evolutionary conserved region 47, formed by rs17226398, rs56038622, and
rs2271338, reduced enhancer activity by 40% in neuroblastoma and astrocytoma cells (pBonferroni , .0001). This enhancer
also drove green fluorescent protein expression in the zebrafish brain in a tissue-specific manner, sharing aspects of
endogenous ADGRL3 expression. The rs2271338 risk allele disrupts binding of YY1 transcription factor, an important factor
in the development and function of the central nervous system. Expression quantitative trait loci analysis of postmortem
human brain tissues revealed an association between rs2271338 and reduced ADGRL3 expression in the thalamus.
CONCLUSIONS: These results uncover the first functional evidence of common noncoding variants with potential
implications for the pathology of ADHD.
Keywords: ADGRL3, ADHD, Cis-acting regulatory element, Enhancer, Evolutionary conserved regions, Genetics,
Latrophilin, LPHN3, Zebrafish
http://dx.doi.org/10.1016/j.biopsych.2016.06.026

Attention-deficit/hyperactivity disorder (ADHD) is a complex through 19 of the gene ( 207 Kb). Searching for variants that
heritable trait that affects more children and adolescents than might affect ADGRL3 protein function, Domene et al. (20)
any other psychiatric disorder. Approximately 5.3% of the sequenced the entire coding region of the gene in a small
world’s population is estimated to be affected (1,2). ADHD cohort of subjects with ADHD, but no missense coding
increases the risk of disruptive symptoms, substance use, changes or canonical splice site alterations were found to be
legal problems, and underemployment, which reduces the associated with the disorder. This suggests that intronic
quality of life of ADHD sufferers and their families (3–6). ADHD noncoding variants are the likely pathological contributors.
heritability has been estimated at 76%, suggesting a strong In the present study, we interrogated the ADGRL3 MCR,
genetic component (7). Candidate gene and genome-wide aiming to identify transcriptional enhancer elements with
studies of single nucleotide polymorphism (SNP) and copy potential functional implications for ADHD.
number variants have identified a number of ADHD suscept-
ibility loci (8), but few molecular studies have attempted to
elucidate the functional effects of risk variants. METHODS AND MATERIALS
Common genetic variants within the adhesion G protein–
coupled receptor L3 gene (ADGRL3, also known as latrophilin Subjects
3 or LPHN3) in 4q13.2 are strongly linked and associated with Individuals with and without ADHD were ascertained from
ADHD (9–19). The ADHD-linked region in ADGRL3 (hereafter the metropolitan area of Medellin (Antioquia, Colombia). The
referred to as the minimal critical region [MCR]) spans exons 4 Paisa community is considered a genetic isolate of Caucasian

SEE COMMENTARY ON PAGE

Published by Elsevier Inc on behalf of Society of Biological Psychiatry. 1


ISSN: 0006-3223 Biological Psychiatry ]]], 2016; ]:]]]–]]] www.sobp.org/journal
Biological
Psychiatry Brain Enhancer Associated With ADHD

descent with low admixture with Amerindian and Negroid chromatin state segmentation by a hidden Markov model
ethnicities (21). The cohort consisted of 14 multigenerational (Broad ChromHMM) (26,27), EP300 transcription factor (TF)
families and 125 nuclear families for a total of 838 individuals ChIP-seq (28), and uniform DNAse I hypersensitivity sites (29).
(372 affected and 466 unaffected individuals; 335 children and Additional information on these annotation tracks can be
adolescents [3–16 years of age] and 503 adults [$17 years of found in Supplement 1.
age]). The multigenerational families had an average size of 28
members (range, 9–57 family members) and an average of Luciferase Assays
2.93 generations (range, 2–4 generations). Full details of the The generation of ECR-luciferase constructs, cell culture
clinical, demographic, and genetic ascertainment features and conditions, and transient transfections are described in
the methodology of neuropsychological evaluation have been Supplement 1. After 24 hours of transfection, culture super-
published elsewhere (22–24). The study was reviewed and natants containing secreted luciferases were collected. Luci-
approved by the Institutional Review Board of the National ferase assays were performed using the Pierce Luciferase
Human Genome Research Institute (Protocol 00-HG-0058) Flash Assay kits (Thermo Fisher Scientific, Waltham, MA).
and the University of Antioquia Ethics Committee (Protocol Each ECR-luciferase construct was assayed in triplicate, and
11-13-342). each transfection experiment was repeated at least three
times (n 5 9). Luciferase activity data were analyzed using a
Genetic Statistical Analyses one-way analysis of variance with Bonferroni correction, as
SNP genotyping methods are presented in Supplement 1. implemented in Prism 6 software (GraphPad Software, La
Genotype data were imported into Golden Helix SVS software Jolla, CA).
(version 8.3.1; Golden Helix, Bozeman, MT) for family-based
association testing. Genotype and allelic frequencies were Electromobility Shift Assays
estimated by maximum likelihood. Family-based association Electromobility shift assays were performed using standard
testing analyses using ADHD status as a categorical variable procedures. ECR47 DNA probes were incubated with Myc-
were applied to the entire set of markers that passed quality DDK-tagged recombinant TFs or whole-cell expression lysates
control. We also performed family-based haplotype analyses (Origene, Rockville, MD). For supershift reactions, an anti-DDK
to compare with the results at the marker level. Comorbid (FLAG) monoclonal antibody (Origene) was added.
conditions (e.g., substance use and disruptive symptoms) and
neuropsychological endophenotypes (e.g., Wechsler Intelli- Zebrafish Bioassays
gence Scale for Children [WISC] block design, WISC perform-
ance intelligence quotient [PIQ], WISC full-scale intelligence Zebrafish stocks and manipulations followed the animal care
quotient [FSIQ], “A”-cancellation and vigilance test correct and use protocols used in our Zebrafish Core facility (National
responses [ACVTCR] and omissions [ACVTO], and the Rey– Institutes of Health/National Human Genome Research Insti-
Osterrieth Complex Figure Test [ROCFT copy]) were used as tute). Zebrafish transgenesis and in situ hybridization were
ADHD-interacting variables. We used endophenotype data performed following standard procedures (see Supplement 1
only for 255 children and adolescents (170 affected and 85 for details).
unaffected individuals) with an FSIQ $ 81 and regular school
grades adequate for their age in order to exclude participants RESULTS
with potential learning disabilities.
Because the ADGRL3 genomic region under study is Identification of Potential Enhancer Elements Within
known to be linked to ADHD (11), the null hypothesis of ADGRL3
linkage and no association was tested. Individual genotypes
Using the ECR Browser, we identified highly conserved
inconsistent with Mendelian transmission were excluded from
elements harbored in the ADGRL3 MCR. Although it is
the analyses. All markers were tested for deviations from the
currently well established that the intergenome comparisons
Hardy-Weinberg equilibrium. Allelic tests of association were
of distant species (e.g., humans and fish) are powerful in
applied using a dominant model of inheritance.
identifying critical distant regulatory elements, only 5% of the
genes in the human genome contain a human/fugu noncoding
Identification of Evolutionary Conserved Elements
ECR in their genomic neighborhood (30–32). For that reason,
Within the ADGRL3 MCR and Prediction of an analysis with species more closely related to humans than
Regulatory Function fish is required to identify regulatory elements for many human
We used the Evolutionary Conserved Region (ECR) Browser genes. We therefore used the human/chicken alignment to
(25) to identify highly conserved elements within the select candidate sequences. The human/opossum alignment
ADGRL3 MCR (accession no. NG_033950.2, GRCh37.p13/ (an evolutionarily closer species) was arbitrarily used to give an
hg19 assembly; coordinates, chr4:62,688,419-62,895,842). identity to the conserved elements, thereby naming 51 regions
We looked at DNA sequence conservation across several ECR1 through ECR51 (Supplemental Figure S1 in Supplement
vertebrate species, including human, mouse, chicken, and 1 and Supplemental Table S1 in Supplement 2).
zebrafish, using a sliding window of .150 base pairs long and Given that enhancer activity has been correlated with
.70% identity. To predict transcriptional regulatory function, certain properties of chromatin (33), we gathered p300 binding
we gathered annotation data from three regulation tracks of site, histone mark, and DNase I hypersensitivity site annotation
the University of California, Santa Cruz (UCSC) table browser: data to predict ECR enhancer function (Supplemental Table S1

2 Biological Psychiatry ]]], 2016; ]:]]]–]]] www.sobp.org/journal


Biological
Brain Enhancer Associated With ADHD Psychiatry

Table 1. Association Between Evolutionary Conserved Region Common Variants and Attention-Deficit/Hyperactivity Disorder, Disruptive Symptoms, Substance

ACVTCR, “A”-cancellation and vigilance test correct responses; ACVTO, “A”-cancellation and vigilance test omissions; ADHD, attention-deficit/hyperactivity disorder; CD, conduct
disorder; ECR, evolutionary conserved region; FDR, Benjamini–Hochberg false discovery rate; NS, not significant; ODD, oppositional defiant disorder; ROCFT, Rey–Osterrieth complex figure
.00261 Coding-syn
Coding-syn

.04088 Coding-syn
Function
in Supplement 2). From the list of elements conserved in

.00228 Intronic
.02775 Intronic
.02775 Intronic
.02775 Intronic

Intronic
.00928 Intronic
.02775 Intronic

Intronic
chicken, we excluded those ECRs not predicted to be func-
tional by any of the annotation tracks above and that did not
contain SNPs with a minor allele frequency . 1%, because

Nicotine p Value Alcohol p Value ACVTCR p Value ACVTO p Value ROCFT p Value
FDR
genetic studies predominantly suggest that ADHD risk is

NS

NS
NS
primarily explained by common variants (1,7,8). Following this
approach, the number of candidate ECRs was reduced to 10

.00017 .00015

.00186

.00006 .000004 .00006 .00035


.00098 .01295
.00098 .01295
.00098 .01295
.00098 .01295

.00013 .02180
Raw

NS

NS
NS
(i.e., ECR1, ECR2, ECR4, ECR9, ECR20, ECR22, ECR26,
ECR37, ECR46, and ECR47). Because coding regions can
overlap enhancer sequences (34,35), we did not exclude those

.00373
.00913
FDR

NS
NS
elements containing ADGRL3 exons (i.e., ECR20, ECR26,
ECR37, and ECR47).

.00003
.00046
.00046
.00046
.00046

.00199

.00002
.00548
Raw

NS
NS
Genetic Statistical Analyses of ECR Variants

.00016
.00098
.00098
.00098
.00098

.00376

.00013
.00920
FDR
Supplemental Table S2 in Supplement 1 shows a list of the

NS
NS
common variants harbored in ECR sequences that are pre-
dicted to be functional. We performed family-based associa-

.000004
.00003
.00046
.00046
.00046
.00046

.00201

.00002
.00552
Raw
tion tests in 838 individuals from the same Colombian

NS
NS
multigenerational and nuclear families that previously showed
linkage and association of ADHD to 4q13.2 (11,22). Genotype

.00199
.00199
.00199

.000001 .00001
.00199

.00255

.03866
.00204
FDR
proportions did not significantly depart from the Hardy-

NS

NS
NS
Weinberg equilibrium. SNPs in 6 of the 10 ECRs showed
significant association with ADHD, comorbid disorders, or
.00066
.00066
.00066

.03092
.00066

.00119

.02062

.04748
.00082
Raw

NS
endophenotypes after correction by false discovery rate
(FDR) (Table 1). ECR46, ECR47, and ECR4 showed the highest
association with ADHD (all five variants, pFDR 5 .00219),
.00203
.00203
.00203
.00203

.04241

.04241
.04241
.04241
FDR

NS

NS
NS
followed by ECR26 (both pFDR 5 .00342), ECR2 (variant
rs1868790, pFDR 5 .00988), and ECR37 (variant rs1397548,
pFDR 5 .03173). Interestingly, only variants in ECR46
.00054
.00054
.00054
.00054
.03740
.02262

.02062
.02191
.02223

.04169
Raw

NS
(rs11131352) and ECR47 (rs17226398, rs56038622, and
rs2271338) consistently showed association with disruptive
.00002 .00005 .00020 .00076
.00002 .00005 .00020 .00076
.00002 .00005 .00020 .00076
.00002 .00005 .00020 .00076

.02291 .03819 .01442 .04327


symptoms (i.e., oppositional defiant disorder [all pFDR 5
FDR
CD p Value

NS
NS

NS
NS
NS

NS
.00005] and conduct disorder [all pFDR 5 .00076]; substance
use of alcohol [all pFDR 5 .00199] and nicotine [all pFDR 5
Use, and Neuropsychological Endophenotypes in the Paisa Dataset

.03274
.00155 .00342 .000003 .00003 .02510
.00160 .00342 .000003 .00003 .02641

.01799 .03372 .02892


Raw

NS
NS

.00203]; and endophenotypes ACVTCR [all pFDR 5 .00098],


ACVTO [all pFDR 5 .00098], and ROCFT [all pFDR 5 .02775]).
.00272 .00582
FDR

Variants in ECR1, ECR9, and ECR22 did not show associa-


ODD p Value

NS

NS

tions with ADHD, comorbid conditions, or endophenotypes


and therefore were not considered for functional analysis.
.03921
Raw

NS

None of the variants showed association with WISC block


design, WISC PIQ, or WISC FSIQ. Haplotype analyses for
ADHD affection status are presented in Supplemental Table
ADHD p Value

.00073 .00219
.00073 .00219
.00073 .00219
.00073 .00219
.00063 .00219

.00527 .00988

.01904 .03173
FDR

NS

NS

S3 in Supplement 1.
Given the identical p values for some single-variant
Raw

associations and the physical proximity of variants, we


NS

NS

calculated the linkage disequilibrium (LD) between SNPs. We


used phased genotype data from the 1000 Genomes Project
Allele (Frequency)

for the Northern Europeans from Utah population. For our


G (0.278)

G (0.064)

G (0.059)
C (0.132)

A (0.132)

A (0.427)

A (0.308)
T (0.132)

T (0.132)

T (0.313)
T (0.281)

dataset, we performed an LD pairwise analysis using Golden


Helix SVS software. Variants rs2305339 and rs734644 in
ECR26 were in complete LD (r2 5 .99, D′ 5 1.00, haplotypes
AC/GT [protective/risk]) as were variants rs11131352
test; syn, synonymous.

(i.e., ECR46), rs17226398, rs56038622, and rs2271338


rs11131352
rs17226398
rs56038622

rs10021694

rs73823249
rs2271338

rs2305339
rs1868790

rs1397548
rs1397547
rs734644

(i.e., ECR47) (r2 5 1.00, D′ 5 1.00, haplotypes AGAG/TCTA


Variant

[protective/risk]) (Supplemental Table S4 in Supplement 1).


For initial evaluation in this study, risk alleles in complete
LD were tested in luciferase assays as a haplotype rather
ECR ID
ECR46
ECR47

ECR26

ECR37
ECR4

ECR2

than individually and were compared to the protective


haplotype.

Biological Psychiatry ]]], 2016; ]:]]]–]]] www.sobp.org/journal 3


Biological
Psychiatry Brain Enhancer Associated With ADHD

ECR46 and ECR47 appear as two independent conserved (Figure 1). We compared luciferase activity of ECRs containing the
elements in chickens, but they are part of a single “core” ECR protective versus the risk allele/haplotype. As described above,
($350 base pairs long, $77% identity) (31) in species that are ECR26 and ECR47 contain markers in complete LD; we therefore
evolutionarily closer to humans, such as opossum, mouse, compared haplotypes rather than pairwise combinations of alleles.
and chimpanzee. For that reason, these two sequences were ECR37 and ECR47 were the only elements to stimulate luciferase
evaluated in luciferase assays independently and together. expression. ECR37 showed weak enhancer activity in all cell lines;
however, its ADHD-associated variant rs1397548 did not affect
ECR47 Functions as a Brain-Specific Enhancer enhancer function. ECR47 also showed weak enhancer activity,
We tested the ADHD-associated ECRs for enhancer activity using but only in B35 neuroblastoma and U87 astrocytoma cells,
a dual secreted luciferase reporter assay in four different cell lines suggesting tissue-specific activity. Unlike ECR37, the ECR47 risk

Figure 1. Secreted luciferase assays testing attention-deficit/hyperactivity disorder–associated evolutionary conserved region (ECR) sequences for
enhancer activity. Four different cell lines were transfected with the ECR-luciferase constructs for 24 hours. Luciferase activity was normalized against a
constitutive Gaussia luciferase plasmid and expressed as relative luciferase activity (Cypridina/Gaussia ratio). Results are presented as the stimulation of
luciferase activity above the basal, enhancer-less vector containing only the minimal promoter. Letters in parentheses indicate the alleles or haplotypes being
tested. ECR37 and ECR47 showed weak enhancer activity compared to the strong control enhancers Simian vacuolating virus 40 (SV40) and human Bicore1.
ECR47 risk haplotype CTA decreased enhancer activity in B35 neuroblastoma and U87 astrocytoma cells (pBonferroni , .0001). Statistical differences were
defined using one-way analysis of variance with correction for multiple comparisons. CHO-K1, Chinese hamster ovary K1 cells; P19, mouse embryonic
carcinoma cells.

4 Biological Psychiatry ]]], 2016; ]:]]]–]]] www.sobp.org/journal


Biological
Brain Enhancer Associated With ADHD Psychiatry

haplotype (CTA) reduced luciferase activity by approximately 40% like transcription factor 1 (GRHL1); PAX2; PAX3; YY1 tran-
in both cells lines (pBonferroni , .0001). ECR2, ECR4, ECR26, and scription factor; hypoxia responsive elements; Tax-1/cyclic
ECR46 showed no stimulation of luciferase activity in any of the AMP-responsive element-binding protein 1 (CREB) complex;
cell lines. The activity of core element ECR46/47 was similar to LIM homeobox 3; POU domain, class 3, transcription factor 2
that of ECR47 alone (Supplemental Figure S2 in Supplement 1), (POU3F2); POU domain, class 4, transcription factor 3
indicating that regulatory activity resides on the ECR47 moiety. (POU4F3); POU domain, class 6, transcription factor 2
Subsequently, we evaluated the ability of the human ECR47 (POU6F2); pituitary-specific positive transcription factor 1
sequence to drive green fluorescent protein (GFP) reporter (PIT1); and cone–rod homeobox protein (CRX) (Table 2).
expression in stable transgenic zebrafish lines. A clear GFP signal
was detected in the zebrafish brain that was restricted to the YY1 Binding to ECR47 Is Disrupted by the rs2271338
ventral forebrain, the midbrain, and the hindbrain of embryos 28 to ADHD Risk Allele
30 hours postfertilization (Figure 2A). In contrast with the weak
The binding sites of three important neurodevelopmental TFs
enhancer activity detected in luciferase assays, a strong GFP
were predicted to be disrupted by ECR47 risk allele substitu-
signal was observed in zebrafish. This may be explained by the
tions. PAX2 and YY1 were predicted to be disrupted by
fact that multicopy integration events can occur during trans-
rs2271338 G.A and GRHL1 was predicted to be disrupted
genesis or that ECR47 may function as a developmental
by rs17226398 G.C (Table 2). While GRHL1 and PAX2 did not
enhancer, therefore behaving differently during embryogenesis
bind to their predicted sites (Figure 3B, C), YY1 produced a
compared to differentiated cells in culture. Interestingly, the
clear gel shift when added to a 27–base pair fragment contain-
ECR47-driven GFP expression pattern shared specific aspects
ing the rs2271338 protective allele. YY1 binding was abrogated
of endogenous adgrl3.1 expression in forebrain, midbrain, and
by addition of excess unlabeled protective sequence, but not
hindbrain, but not in telencephalon and retina, as evaluated by
by the sequence containing the risk allele (Figure 3A).
in situ hybridization (Figure 2B, C). Because the zebrafish
We next examined whether YY1 affected endogenous
enhancer detection vector system is not robust enough to allow
ADGRL3 expression. Real-time polymerase chain reaction analy-
for quantitative measurement of enhancer activity in vivo, we did
ses revealed a strong expression of endogenous ADGRL3
not investigate the effect of the ECR47 risk haplotype (CTA) on
messenger RNA (mRNA) in SH-SY5Y neuroblastoma but not in
enhancer function in the zebrafish. A number of variables make it
U87 astrocytoma cells (Supplemental Figure S4A in Supplement 1),
challenging to compare ECR47 risk and protective haplotypes
suggesting that expression of this gene may be specific to the
quantitatively using the zebrafish enhancer detection system,
neuronal lineage in the CNS. After YY1 small interfering RNA
including 1) different copy number integration during zebrafish
transfection, we could not detect any significant changes in
transgenesis; 2) the structural complexity of transgene integration
ADGRL3 mRNA expression in these cell lines (Supplemental
loci in the genome (heterochromatin vs. euchromatin); 3) DNA
Figure S4B in Supplement 1). This result might suggest that
methylation effects; and 4) zebrafish tissue autofluorescence and
the ECR47–YY1 pair is functional only during development or
nonspecific, background GFP expression.
that its spatiotemporal regulation of ADGRL3 expression is
It is important to highlight that ECR47 is also conserved in
complex and cannot be modeled properly outside the native
zebrafish (Supplemental Figure S3 in Supplement 1), which
regulatory circuitry of the brain.
makes it an ultraconserved element that is likely to have an
important biological function.
rs2271338 Is Associated With Decreased ADGRL3
mRNA Expression in the Thalamus
TFs Preferentially Associated With Brain Function
Are Overrepresented in ECR47 Unlike other behavioral disorders, such as schizophrenia and
major depressive disorder, brain tissue from ADHD patients is
Analysis of the TF–binding profiles of ECR47 and 144 in vivo
not readily available. However, SNPs associated with complex
tested brain enhancers from the VISTA Enhancer Browser (36)
diseases are likely to function as expression quantitative trait
(Supplemental Table S5 in Supplement 3) revealed a signifi-
loci, and the tissues of unaffected individuals can be used for
cant overrepresentation of developmental TF families prefer-
gene expression association analyses. Expression quantitative
entially associated with brain tissue, such as distal-less
trait loci analysis of brain tissue from 137 neuropathologically
homeodomain (V$DLXF), NK6 homeobox (V$NKX6), paired
confirmed controls (16–102 years of age) revealed a significant
box (PAX) homeodomain (V$PAXH), and Brn-5 POU (V$BRN5)
association between the rs2271338 AA risk genotype and
TFs, as suggested by the high Z scores (Supplemental Table
reduced ADGRL3 expression in the thalamus (p , .01)
S6 in Supplement 1). Homeobox TFs (V$HBOX), Brn POU
(Supplemental Figure S5 and Supplemental Table S7 in
domain (V$BRNF), cocaine- and amphetamine-regulated tran-
Supplement 1). rs2271338 was either absent or not associated
script 1 (V$CART), and Lim homeodomain (V$LHXF) factors
with ADGRL3 expression in the National Institutes of Health
were also overrepresented, but while they may participate in
Common Fund’s Genotype-Tissue Expression (GTEx), Colum-
neural development and function, they are not preferentially
bia University’s SNPExpress, and the National Heart, Lung,
associated with the central nervous system (CNS).
and Blood Institute’s GRASP databases (Supplement 1).
The potential effects of ECR47 SNP allele substitutions
on TF binding were examined using SNPInspector software
(Genomatix, Munich, Germany). All three ADHD risk alleles DISCUSSION
(CTA) were predicted to produce loss or gain of binding sites Few molecular studies have attempted to explain the molec-
for important neurodevelopmental TFs, such as grainyhead- ular effects of ADHD-associated genetic variants. Studies on

Biological Psychiatry ]]], 2016; ]:]]]–]]] www.sobp.org/journal 5


Biological
Psychiatry Brain Enhancer Associated With ADHD

Figure 2. In vivo enhancer testing and correlation with adgrl3.1 expression in the zebrafish. Evolutionary conserved region 47–driven green fluorescent
protein (GFP) expression was monitored in the brain of transgenic embryos at 28 to 30 hours postfertilization (hpf). (A) Stable transgenic F2 embryo showing
GFP expression restricted to the central nervous system (i.e., the forebrain, midbrain, and hindbrain). (B, C) Expression of adgrl3.1 was detected by in situ
hybridization of embryos at (B) 36 hpf and (C) 48 hpf (top row, left midsagittal and right retina in focus). Evolutionary conserved region 47–driven GFP
expression represents several specific aspects of endogenous adgrl3.1, consistent with the location of neuronal tissue in the developing brain. Endogenous
adgrl3.1 messenger RNA expression is detected in the telencephalon, the ventral forebrain, the midbrain, the hindbrain, and the retina throughout the
analyzed developmental stages. The anterior is to the left in all images, and the dorsal side is up in all lateral views [scale bar 5 100 mm; all images in part (C)
are in the same scale]. FB, forebrain; HB, hindbrain; MB, midbrain; MHB, midbrain–hindbrain boundary; r, rostral; v, ventral.

dopamine transporter (DAT1) (37–40), tryptophan hydroxylase moderate and large effects, but these findings fail to explain
2 (TPH2) (41,42), and T-cadherin (CDH13) (43) have examined the higher incidence of ADHD and larger phenotypic variance
the functional properties of rare missense mutations of observed in populations. Instead, the common disease/

6 Biological Psychiatry ]]], 2016; ]:]]]–]]] www.sobp.org/journal


Biological
Brain Enhancer Associated With ADHD Psychiatry

Table 2. Transcription Factor–Binding Sites Predicted to be Affected by the Attention-Deficit/Hyperactivity Disorder–


Associated Haplotype in Evolutionary Conserved Region 47
Major Minor Allele Predicted TF Family/ Optimized Core Matrix
SNP rsID Allele Allele Change Effect Matrix TF Name Thresholda Strand Similarityb Similarityc
rs17226398 G C G.C Gained V$PAX3/ Pax-3 paired domain protein 0.85 1 1 0.893
PAX3.02
Gained V$HIFF/ Hypoxia-responsive element 0.97 – 1 0.978
HRE.02
Lost V$GRHL/ Grainyhead-like 1 0.86 1 1 0.864
GRHL1.01
Lost V$CP2F/ Transcription factor CP2-like 1 0.87 1 0.815 0.894
TCFCP2L1.01
rs56038622 A T A.T Gained V$CREB/ Tax/CREB complex 0.71 1 0.75 0.739
TAXCREB.02
rs2271338 G A G.A Gained V$LHXF/ LIM-homeodomain 3 0.82 1 1 0.84
LHX3.02
Gained V$BRNF/ Brn-3, POU-IV protein class 0.78 – 0.75 0.815
BRN3.01
Gained V$BRN5/ Retina-derived POU-domain 0.76 1 0.81 0.779
POU6F2.01 factor 1, dimeric
Gained V$BRNF/ POU class 3 homeobox 2 0.82 1 1 0.827
BRN2.04 (POU3F2)
Gained V$OCT1/ Octamer-binding protein 1 0.85 1 0.767 0.854
OCT1.03 (POU2F1)
Gained V$PIT1/ Pituitary transcription factor 1 0.81 1 1 0.821
PIT1.02 (POU1F1)
Gained V$HNF6/ One CUT-homeodomain 0.82 1 1 0.876
OC2.01 protein
Lost V$PAX2/ Zebrafish PAX2 paired domain 0.78 1 0.789 0.784
PAX2.01 protein
Lost V$YY1F/ Yin and Yang 1 repressor 0.94 – 1 0.979
YY1.02
Lost V$BCDF/ Cone–rod homeobox protein 0.94 1 1 0.946
CRX.01
Searches were performed using the SNPInspector function within the Genomatix Software Suite (Genomatix, Munich, Germany). The analyses
are based on the MatInspector and Genomatix libraries of matrix descriptions for transcription factor–binding sites (MatBase).
SNP, single nucleotide polymorphism; TF, transcription factor.
a
At the matrix similarity and optimized threshold, a minimum number of matches is found in nonregulatory test sequences (i.e., with this matrix
similarity, the number of false positive matches is minimized).
b
Core similarity refers to the degree of matching to the “core sequence” of a matrix (i.e., the highest conserved positions of the matrix, usually 4
bases). Maximum core similarity (1.0) is reached when the highest conserved bases of a matrix match exactly in the sequence.
c
Matrix similarity refers to the degree of matching to each sequence position in the matrix. A perfect match to the highest conserved nucleotide
at each position gets a score of 1.00. A “good” match usually has a similarity of . 0.80. Mismatches in highly conserved positions of the matrix
decrease the matrix similarity more than mismatches in less conserved regions.

common variant hypothesis is better supported by a sub- in brain regions implicated in dopaminergic systems (11,45).
stantial number of genetic epidemiological studies, with ADGRL3 endogenous ligand has been identified as fibronectin
common variants accounting for approximately 40% of ADHD leucine rich transmembrane protein 3, a postsynaptic mem-
heritability (7,8). brane protein involved in axon guidance and neuronal cell
ADGRL3 is a strong ADHD candidate gene. ADGRL3 migration during embryonic development (46). More impor-
common variants predispose individuals to ADHD, modulate tantly, the ADGRL3–fibronectin leucine rich transmembrane
brain metabolism, and predict ADHD severity and comorbidity protein 3 synaptic pair regulates excitatory transmission both
with disruptive symptoms and substance use disorder in vitro and in vivo (47). Animal models also support ADGRL3
(11,13,15,16). When combined with other risk factors, ADGRL3 implication in ADHD pathophysiology (48,49).
risk variants improve the prediction of ADHD severity, dysfunc- Using a combination of evolutionary sequence conservation
tional comorbidity, long-term outcome, and response to treat- and regulatory annotation data, we identified candidate
ment with stimulant medication (10–16). Recent work from our sequences within the ADGRL3 MCR with potential regulatory
group also showed the genetic linkage and association of function. Several variants revealed significant association with
ADGRL3 variants to neuropsychological endophenotypes, pro- ADHD, comorbid disorders, or neuropsychological endophe-
viding a more powerful framework for ADHD clinical classifica- notypes, with a four-marker haplotype in the ECR46/ECR47
tion and for the identification of causative genetic variation (44). core element showing the highest level of association across
ADGRL3 encodes a member of the latrophilin subfamily of the board. This result may suggest that ECR46/47 participates
adhesion G protein–coupled receptors that is highly expressed in a common neurobiological pathway to ADHD. Given the

Biological Psychiatry ]]], 2016; ]:]]]–]]] www.sobp.org/journal 7


Biological
Psychiatry Brain Enhancer Associated With ADHD

Figure 3. YY1 transcription factor binding to evolutionary conserved region 47 (ECR47) enhancer is disrupted by the rs2271338 risk allele. (A) A biotin-
labeled DNA fragment containing a rs2271338 attention-deficit/hyperactivity disorder protective allele was incubated with a human embryonic kidney 293
(HEK293) whole cell lysate (WCL) expressing DDK (FLAG)-tagged human YY1 (lanes 2–5 from left to right). A mobility shift in lane 2 indicates protein binding to
the probe, which was abrogated by incubation with molar excess of the unlabeled fragment (lane 3), but not of an unlabeled fragment containing the
rs2271338 risk allele (lane 4). Higher molecular weight shift with the addition of anti-DDK antibody identifies YY1 as the binding factor (lane 5). Lanes 6 to 10
correspond to a known YY1 binding sequence used as positive control (87). Lanes 7 to 10 show protein binding to the control probe, with the anti-DDK
antibody producing a similar supershift, thereby confirming YY1 identity (lane 8). Molar excess of the unlabeled ECR47-protective fragment was capable of
reducing protein binding (lane 9), but the risk fragment was not (lane 10). No binding was detected for grainyhead-like transcription factor 1 (GRHL1) (B) or
paired box 2 (PAX2) (C) transcription factors. (B) Lanes 1 to 5, biotin-labeled fragment containing the rs17226398 protective allele; lanes 6 to 10, GRHL1-
positive control sequence (88). Interestingly, no shift was observed in the positive control except when the antibody was added (lane 8). Apparently, the
addition of the antibody stabilizes the GRHL1–DNA interaction, which otherwise is labile under the experimental conditions used. (C) Left panel, lanes 1 to 5,
same DNA fragment used for YY1 in (A); lanes 6 to 10, PAX2-positive control sequence (89). Neither the protective nor the risk ECR47 fragments affected
PAX2 binding to the positive control probe (lanes 9 and 10). The right panel shows the results for a longer ECR47 probe (Protective 2) extending to the 3ʹ end
of the sequence. PAX2 still did not bind to DNA. Position weight matrices were taken from MotifMap for human hg19 (e.g., YY1 and PAX2) and Drosophila
dm3 (GRHL1) assemblies (90).

complexity of the ADHD phenotype, we must expect complex The genetic association of ADGRL3 with disruptive behav-
genetic interactions within the ADGRL3 locus and with other iors and substance use supports previous observations.
genomic regions, which may explain the different levels of Families with ADHD cluster oppositional defiant disorder,
association observed across ECRs (Table 1). conduct disorder, and substance use disorder (23); children

8 Biological Psychiatry ]]], 2016; ]:]]]–]]] www.sobp.org/journal


Biological
Brain Enhancer Associated With ADHD Psychiatry

diagnosed with ADHD monitored during the transition into preferentially associated with neurodevelopmental processes.
adolescence have higher rates of alcohol, tobacco, and Distal-less (i.e., DLX-1, DLX-2, and DLX-5), BRN5/POU6F1,
psychoactive drug use than unaffected children (50,51); the PAX (i.e., PAX2, PAX3, PAX5, PAX6, PAX7, and PAX8), and
lifetime risk for substance use is approximately 50% in NKX6 (NKX6.2) homeodomain factors are known to play
patients with childhood ADHD that persists into adulthood prominent roles in the development and function of the CNS
(52,53). (66–70). This finding supports our results showing tissue-
Linkage and association of ADGRL3 with neuropsycholog- specific activity of ECR47.
ical endophenotypes was recently reported by Mastronardi The risk allele substitution of rs2271338 disrupts YY1
et al. (44). In agreement with their findings, we show a binding to ECR47. YY1 is a controversial and versatile tran-
significant association of ECR variants with lower ACVTCR scriptional regulator (71); it can either activate or repress gene
scores and higher ACVTO scores. Impaired response inhib- expression depending on the cofactors it recruits (72). The
ition and poor sustained attention, as measured by these two potential function of YY1 in the developing nervous system
tests, are fundamental components of the executive dysfunc- was first suggested by the phenotypic analysis of Yy11/– mice.
tion present in patients with ADHD (54–57). In the same vein, Null Yy1 mice show early embryonic lethality, but a subset of
the inattentive and hyperactive/impulsive motor phenotype Yy11/– mice (20%) display growth retardation and neural
associated with patients with ADHD is expected to affect the tube defects. The brains of Yy11/– mouse embryos show
ability to perform the ROCFT copy test, and therefore the exencephaly, asymmetric structure, and the presence of
significant association with ECR risk variants. However, the pseudoventricles (73). Morpholino-knockdown studies in Xen-
complexity of the multiple cognitive domains assessed by opus reveal similar neurulation defects (74). While a significant
ROCFT (i.e., visuospatial constructional ability, visual memory, reduction of XYY1 protein levels results in early embryonic
and several components of executive function) can confound lethality, a partial depletion results in anteroposterior pattern-
the interpretation of scores, which might explain the lower ing defects and reduction of head structures (74,75). At the
level of significance (58). We did not detect any association molecular level, Xyy1 ablation in Xenopus embryos reveals
with the WISC measures (i.e., block design, PIQ, or FSIQ), downregulation of TFs involved in neural patterning (i.e.,
which might indicate that ADGRL3 variation does not contrib- homeobox genes, engrailed2, otx2, and krox20) and neural
ute to the cognitive deficits evaluated by these particular tests. crest cell specification and migration (i.e., slug and snai1) (76).
Functional testing in vitro identified ECR47 as a transcrip- Studies using other systems have also shown dysregulation of
tional enhancer. ECR47 was active in cultured neurons and neurotransmitter signaling and metabolism genes, such as
astrocytes in a tissue-specific manner, and its function was dynamin-1 and dopamine β-hydroxylase in neurons, and Glast
disrupted by the ADHD risk haplotype defined by the variants glutamate/aspartate transporter in astrocytes (76).
rs17226398, rs56038622, and rs2271338 (CTA) in complete Analysis of the effect of rs2271338 risk allele on ADGRL3
LD. The nonrandom association between these risk alleles expression in cultured human cells and postmortem brain
may suggest evolutionary selective pressure to conserve an tissue showed apparently contradictory results. While YY1
important biological function. Although neurons and astro- downregulation did not affect ADGRL3 expression in neuro-
cytes have distinct transcriptome profiles, they both share a blastoma cells, the rs2271338 risk allele was associated with
common neuroepithelial origin, and over the past two decades reduced expression in the adult thalamus. The thalamus was
it has become clear that astrocytes participate in a wide one of the earliest brain areas considered in the pathophysi-
variety of complex functions in the CNS, including crucial roles ology of ADHD (77) because of its key role in filtering
in synaptic transmission and information processing in neural information and stimulus processing (78). Morphological
circuits (59–61). Evidence also suggests that glial cells may abnormalities of the thalamus (79) and thalamic volume
play a role in the pathogenesis of neuropsychiatric disorders, reduction (80) have been shown previously in children with
including ADHD (62,63). However, additional studies are ADHD. Various methods, including functional connectivity
required to determine whether ECR47 function in astrocytes analysis, have uncovered thalamic abnormalities in patients
is functionally relevant for the pathology of ADHD, because with ADHD (81,82). Morphological abnormalities have also
ADGRL3 expression in astrocytoma cells was low compared been found across a number of cortical regions in children and
to human neuroblastoma cells (Supplemental Figure S4 in adolescents with ADHD (83), supporting the view that cortico-
Supplement 1). striato-thalamo-cortical loops (84) play a key role in the
Strikingly, we also found that ECR47 functions as a brain pathogenesis of ADHD.
enhancer in vivo. ECR47 was able to drive GFP expression in Although additional studies are required to establish the
the zebrafish brain, which is consistent with various aspects of precise role of ECR47, the results presented here suggest an
endogenous adgrl3.1 expression. These results are in concert important neurological function. While brain expression data
with a previous report by Lange et al. (49) showing wide indicate an ADGRL3 expression maximum across fetal and
adgrl3.1 expression in the zebrafish brain at 24, 48, and 72 infant stages (Supplemental Figure S6 in Supplement 1),
hours postfertilization, which suggests that ECR47 activation relatively high expression levels are maintained throughout
may share brain-specific factors with the adgrl3.1 transcrip- life, suggesting that this gene is necessary for proper brain
tional machinery during zebrafish development. function from conception to demise. The lack of effect of YY1
Enhancer elements have signatures that define tissue knockdown on endogenous ADGRL3 expression in differ-
specificity (64,65). Analysis of ECR47 and a large set of entiated cells suggests that ECR47 may only be active during
in vivo–tested brain enhancers revealed a significant over- developmental stages; however, the association between
lapping of binding sites for homeobox TF families that are rs2271338 and reduced ADGRL3 expression levels in the

Biological Psychiatry ]]], 2016; ]:]]]–]]] www.sobp.org/journal 9


Biological
Psychiatry Brain Enhancer Associated With ADHD

adult thalamus hampers our understanding of ADGRL3 spa- 4. American Psychiatric Association (1994): Diagnostic and Statistical
tiotemporal regulation by ECR47–YY1. Elucidation of this Manual of Mental Disorders. Washington, DC: American Psychiatric
genetic interaction will help to decipher the molecular mech- Association.
5. Biederman J, Faraone SV (2004): Attention deficit hyperactivity
anisms underlying ADHD pathogenesis.
disorder: A worldwide concern. J Nerv Ment Dis 192:453–454.
The experimental methodology used in this research could 6. Jain M, Palacio LG, Castellanos FX, Palacio JD, Pineda D, Restrepo
be a paradigm for the evaluation of noncoding risk variants MI, et al. (2007): Attention-deficit/hyperactivity disorder and comorbid
associated with complex traits. disruptive behavior disorders: Evidence of pleiotropy and new sus-
ceptibility loci. Biol Psychiatry 61:1329–1339.
7. Faraone SV, Perlis RH, Doyle AE, Smoller JW, Goralnick JJ, Holmgren
Limitations
MA, et al. (2005): Molecular genetics of attention-deficit/hyperactivity
We tested risk variants only for their effects on transcriptional disorder. Biol Psychiatry 57:1313–1323.
enhancer activity. While other transcriptional regulatory func- 8. Akutagava-Martins GC, Salatino-Oliveira A, Kieling CC, Rohde LA,
tions (e.g., those mediated by silencer and insulator elements) Hutz MH (2013): Genetics of attention-deficit/hyperactivity disorder:
Current findings and future directions. Expert Rev Neurother 13:
were not investigated, functional testing of these types of
435–445.
sequence have proven difficult to design, and there are 9. Arcos-Burgos M, Velez JI, Solomon BD, Muenke M (2012): A common
currently no robust in vivo assays in widespread use (85,86). genetic network underlies substance use disorders and disruptive or
We must also consider the possible effect of risk variants on externalizing disorders. Hum Genet 131:917–929.
ADGRL3 mRNA splicing—especially synonymous changes 10. Ribases M, Ramos-Quiroga JA, Sanchez-Mora C, Bosch R, Richarte
and noncoding variants within 50 base pairs of exon–intron V, Palomar G, et al. (2011): Contribution of LPHN3 to the genetic
susceptibility to ADHD in adulthood: A replication study. Genes Brain
junctions—and on the expression and function of overlapping
Behav 10:149–157.
noncoding RNAs. 11. Arcos-Burgos M, Jain M, Acosta MT, Shively S, Stanescu H, Wallis D,
et al. (2010): A common variant of the latrophilin 3 gene, LPHN3,
confers susceptibility to ADHD and predicts effectiveness of stimulant
ACKNOWLEDGMENTS AND DISCLOSURES medication. Mol Psychiatry 15:1053–1066.
12. Fallgatter AJ, Ehlis AC, Dresler T, Reif A, Jacob CP, Arcos-Burgos M,
This work was supported by intramural resources from the National Human
et al. (2013): Influence of a latrophilin 3 (LPHN3) risk haplotype on
Genome Research Institute of the U.S. National Institutes of Health.
event-related potential measures of cognitive response control in
We thank Paul Kruszka, M.D., for his detailed revision of the manuscript
attention-deficit hyperactivity disorder (ADHD). Eur Neuropsychophar-
and helpful comments.
macol 23:458–468.
This study used the computational capabilities of a demo license to
13. Acosta MT, Velez JI, Bustamante ML, Balog JZ, Arcos-Burgos M,
Genomatix Software (Munich, Germany).
Muenke M (2011): A two-locus genetic interaction between LPHN3
The authors report no biomedical financial interests or potential conflicts
and 11q predicts ADHD severity and long-term outcome. Transl
of interest.
Psychiatry 1:e17.
14. Jain M, Velez JI, Acosta MT, Palacio LG, Balog J, Roessler E, et al.
(2012): A cooperative interaction between LPHN3 and 11q doubles
ARTICLE INFORMATION the risk for ADHD. Mol Psychiatry 17:741–747.
15. Choudhry Z, Sengupta SM, Grizenko N, Fortier ME, Thakur GA,
From the Medical Genetics Branch (AFM, YA, SH, KM, DY, MTA, MA-B,
Bellingham J, et al. (2012): LPHN3 and attention-deficit/hyperactivity
MM), National Human Genome Research Institute, National Institutes of
disorder: Interaction with maternal stress during pregnancy. J Child
Health, Bethesda, Maryland; Institute of Biology I (HL, WD), University of
Psychol Psychiatry 53:892–902.
Freiburg, Freiburg, Germany; and the Genome Biology Department (MA-B),
16. Labbe A, Liu A, Atherton J, Gizenko N, Fortier ME, Sengupta SM, et al.
John Curtin School of Medical Research, The Australian National University,
(2012): Refining psychiatric phenotypes for response to treatment:
Canberra, Australia.
Contribution of LPHN3 in ADHD. Am J Med Genet B Neuropsychiatr
HL is currently affiliated with the Institute for Developmental Biology,
Genet 159B:776–785.
Cologne University, Cologne, Germany.
17. Hwang IW, Lim MH, Kwon HJ, Jin HJ (2015): Association of LPHN3
Address correspondence to Maximilian Muenke, M.D., Medical Genetics
rs6551665 a/g polymorphism with attention deficit and hyperactivity
Branch, National Human Genome Research Institute, National Institutes
disorder in Korean children. Gene 566:68–73.
of Health, 35 Convent Drive, Room 1B-203, Bethesda, MD 20892-3717;
18. Bruxel EM, Salatino-Oliveira A, Akutagava-Martins GC, Tovo-
E-mail: mamuenke@mail.nih.gov.
Rodrigues L, Genro JP, Zeni CP, et al. (2015): LPHN3 and attention-
Received Dec 11, 2015; revised Jun 28, 2016; accepted Jun 30, 2016.
deficit/hyperactivity disorder: A susceptibility and pharmacogenetic
Supplementary material cited in this article is available online at
study. Genes Brain Behav 14:419–427.
http://dx.doi.org/10.1016/j.biopsych.2016.06.026.
19. Song J, Kim SW, Hong HJ, Lee MG, Lee BW, Choi TK, et al. (2014):
Association of SNAP-25, SLC6A2, and LPHN3 with OROS methyl-
phenidate treatment response in attention-deficit/hyperactivity disor-
REFERENCES der. Clin Neuropharmacol 37:136–141.
1. Faraone SV, Asherson P, Banaschewski T, Biederman J, Buitelaar JK, 20. Domene S, Stanescu H, Wallis D, Tinloy B, Pineda DE, Kleta R, et al.
Ramos-Quiroga JA, et al. (2015): Attention-deficit/hyperactivity dis- (2011): Screening of human Lphn3 for variants with a potential impact
order. Nat Rev Dis Primers 1:15020. on ADHD susceptibility. Am J Med Genet B Neuropsychiatr Genet
2. Visser SN, Danielson ML, Bitsko RH, Holbrook JR, Kogan MD, Ghandour 156B:11–18.
RM, et al. (2014): Trends in the parent-report of health care provider- 21. Bravo ML, Valenzuela CY, Arcos-Burgos OM (1996): Polymorphisms
diagnosed and medicated attention-deficit/hyperactivity disorder: United and phyletic relationships of the Paisa community from Antioquia
States, 2003-2011. J Am Acad Child Adolesc Psychiatry 53:34–46; e2. (Colombia). Gene Geogr 10:11–17.
3. Visser SN, Bitsko RH, Danielson ML, Perou R (2010): Increasing 22. Arcos-Burgos M, Castellanos FX, Pineda D, Lopera F, Palacio JD,
prevalence of parent-reported attention-deficit/hyperactivity disorder Palacio LG, et al. (2004): Attention-deficit/hyperactivity disorder in a
among children—United States, 2003 and 2007. MMWR Morb Mortal population isolate: Linkage to loci at 4q13.2, 5q33.3, 11q22, and
Wkly Rep 59:1439–1443. 17p11. Am J Hum Genet 75:998–1014.

10 Biological Psychiatry ]]], 2016; ]:]]]–]]] www.sobp.org/journal


Biological
Brain Enhancer Associated With ADHD Psychiatry

23. Palacio JD, Castellanos FX, Pineda DA, Lopera F, Arcos-Burgos M, 43. Mavroconstanti T, Johansson S, Winge I, Knappskog PM, Haavik J
Quiroz YT, et al. (2004): Attention-deficit/hyperactivity disorder and (2013): Functional properties of rare missense variants of human
comorbidities in 18 Paisa Colombian multigenerational families. J Am CDH13 found in adult attention deficit/hyperactivity disorder (ADHD)
Acad Child Adolesc Psychiatry 43:1506–1515. patients. PLoS One 8:e71445.
24. Pineda DA, Lopera F, Puerta IC, Trujillo-Orrego N, Aguirre-Acevedo 44. Mastronardi CA, Pillai E, Pineda DA, Martinez AF, Lopera F, Velez JI,
DC, Hincapie-Henao L, et al. (2011): Potential cognitive endopheno- et al. (2015): Linkage and association analysis of ADHD endopheno-
types in multigenerational families: Segregating ADHD from a genetic types in extended and multigenerational pedigrees from a genetic
isolate. Atten Defic Hyperact Disord 3:291–299. isolate [published online ahead of print Nov 24]. Mol Psychiatry.
25. Ovcharenko I, Nobrega MA, Loots GG, Stubbs L (2004): ECR Browser: 45. Krain AL, Castellanos FX (2006): Brain development and ADHD. Clin
A tool for visualizing and accessing data from comparisons of multiple Psychol Rev 26:433–444.
vertebrate genomes. Nucleic Acids Res 32:W280–W286. 46. Yamagishi S, Hampel F, Hata K, Del Toro D, Schwark M, Kvachnina E,
26. Ernst J, Kheradpour P, Mikkelsen TS, Shoresh N, Ward LD, Epstein et al. (2011): FLRT2 and FLRT3 act as repulsive guidance cues for
CB, et al. (2011): Mapping and analysis of chromatin state dynamics in Unc5-positive neurons. EMBO J 30:2920–2933.
nine human cell types. Nature 473:43–49. 47. O’Sullivan ML, de Wit J, Savas JN, Comoletti D, Otto-Hitt S, Yates JR
27. Ernst J, Kellis M (2010): Discovery and characterization of chromatin 3rd, et al. (2012): Flrt proteins are endogenous latrophilin ligands and
states for systematic annotation of the human genome. Nat Biotech- regulate excitatory synapse development. Neuron 73:903–910.
nol 28:817–825. 48. Wallis D, Hill DS, Mendez IA, Abbott LC, Finnell RH, Wellman PJ, et al.
28. Wang J, Zhuang J, Iyer S, Lin XY, Greven MC, Kim BH, et al. (2013): (2012): Initial characterization of mice null for Lphn3, a gene implicated
Factorbook.Org: A wiki-based database for transcription factor- in ADHD and addiction. Brain Res 1463:85–92.
binding data generated by the encode consortium. Nucleic Acids 49. Lange M, Norton W, Coolen M, Chaminade M, Merker S, Proft F, et al.
Res 41:D171–D176. (2012): The ADHD-susceptibility gene lphn3.1 modulates dopaminer-
29. Thurman RE, Rynes E, Humbert R, Vierstra J, Maurano MT, Haugen E, gic neuron formation and locomotor activity during zebrafish develop-
et al. (2012): The accessible chromatin landscape of the human ment. Mol Psychiatry 17:946–954.
genome. Nature 489:75–82. 50. Molina BS, Pelham WE Jr (2003): Childhood predictors of adolescent
30. Nobrega MA, Ovcharenko I, Afzal V, Rubin EM (2003): Scanning substance use in a longitudinal study of children with ADHD. J
human gene deserts for long-range enhancers. Science 302:413. Abnorm Psychol 112:497–507.
31. Ovcharenko I, Stubbs L, Loots GG (2004): Interpreting mammalian 51. Molina BS, Hinshaw SP, Eugene Arnold L, Swanson JM, Pelham WE,
evolution using fugu genome comparisons. Genomics 84:890–895. Hechtman L, et al. (2013): Adolescent substance use in the multi-
32. Woolfe A, Goodson M, Goode DK, Snell P, McEwen GK, Vavouri T, modal treatment study of attention-deficit/hyperactivity disorder
et al. (2005): Highly conserved non-coding sequences are associated (ADHD) (MTA) as a function of childhood ADHD, random assignment
with vertebrate development. PLoS Biol 3:e7. to childhood treatments, and subsequent medication. J Am Acad
33. Shlyueva D, Stampfel G, Stark A (2014): Transcriptional enhancers: Child Adolesc Psychiatry 52:250–263.
From properties to genome-wide predictions. Nat Rev Genet 15: 52. Biederman J, Wilens T, Mick E, Milberger S, Spencer TJ, Faraone SV
272–286. (1995): Psychoactive substance use disorders in adults with attention
34. Birnbaum RY, Clowney EJ, Agamy O, Kim MJ, Zhao J, Yamanaka T, deficit hyperactivity disorder (ADHD): Effects of ADHD and psychiatric
et al. (2012): Coding exons function as tissue-specific enhancers of comorbidity. Am J Psychiatry 152:1652–1658.
nearby genes. Genome Res 22:1059–1068. 53. Nogueira M, Bosch R, Valero S, Gomez-Barros N, Palomar G,
35. Birnbaum RY, Patwardhan RP, Kim MJ, Findlay GM, Martin B, Zhao J, Richarte V, et al. (2014): Early-age clinical and developmental features
et al. (2014): Systematic dissection of coding exons at single nucleo- associated to substance use disorders in attention-deficit/hyperac-
tide resolution supports an additional role in cell-specific transcrip- tivity disorder in adults. Compr Psychiatry 55:639–649.
tional regulation. PLoS Genet 10:e1004592. 54. Barkley RA (1997): Behavioral inhibition, sustained attention, and
36. Visel A, Minovitsky S, Dubchak I, Pennacchio LA (2014): Vista executive functions: Constructing a unifying theory of adhd. Psychol
enhancer browser—a database of tissue-specific human enhancers. Bull 121:65–94.
Nucleic Acids Res 35:D88–D92. 55. Castellanos FX, Tannock R (2002): Neuroscience of attention-deficit/
37. Mazei-Robison MS, Bowton E, Holy M, Schmudermaier M, Freiss- hyperactivity disorder: The search for endophenotypes. Nat Rev
muth M, Sitte HH, et al. (2008): Anomalous dopamine release Neurosci 3:617–628.
associated with a human dopamine transporter coding variant. J 56. Liu L, Cheng J, Su Y, Ji N, Gao Q, Li H, et al. (2015): Deficiency
Neurosci 28:7040–7046. of sustained attention in ADHD and its potential genetic
38. Sakrikar D, Mazei-Robison MS, Mergy MA, Richtand NW, Han Q, contributor MAOA [published online ahead of print March 17]. J Atten
Hamilton PJ, et al. (2012): Attention deficit/hyperactivity disorder- Disord.
derived coding variation in the dopamine transporter disrupts 57. Pineda DA, Puerta IC, Aguirre DC, Garcia-Barrera MA, Kamphaus RW
microdomain targeting and trafficking regulation. J Neurosci 32: (2007): The role of neuropsychologic tests in the diagnosis of attention
5385–5397. deficit hyperactivity disorder. Pediatr Neurol 36:373–381.
39. Kovtun O, Sakrikar D, Tomlinson ID, Chang JC, Arzeta-Ferrer X, Blakely 58. Shin MS, Park SY, Park SR, Seol SH, Kwon JS (2006): Clinical and
RD, et al. (2015): Single-quantum-dot tracking reveals altered membrane empirical applications of the Rey-Osterrieth Complex Figure Test. Nat
dynamics of an attention-deficit/hyperactivity-disorder-derived dopamine Protoc 1:892–899.
transporter coding variant. ACS Chem Neurosci 6:526–534. 59. Bayer SA, Altman J (1991): Neocortical development, illustrated ed.
40. Mergy MA, Gowrishankar R, Gresch PJ, Gantz SC, Williams J, Davis New York: Raven Press.
GL, et al. (2014): The rare DAT coding variant Val559 perturbs 60. Shen Q, Wang Y, Dimos JT, Fasano CA, Phoenix TN, Lemischka IR,
DA neuron function, changes behavior, and alters in vivo et al. (2006): The timing of cortical neurogenesis is encoded within
responses to psychostimulants. Proc Natl Acad Sci U S A 111: lineages of individual progenitor cells. Nat Neurosci 9:743–751.
E4779–E4788. 61. Barres BA (2008): The mystery and magic of glia: A perspective on
41. McKinney J, Johansson S, Halmoy A, Dramsdahl M, Winge I, their roles in health and disease. Neuron 60:430–440.
Knappskog PM, et al. (2008): A loss-of-function mutation in trypto- 62. Yamamuro K, Kimoto S, Rosen KM, Kishimoto T, Makinodan M
phan hydroxylase 2 segregating with attention-deficit/hyperactivity (2015): Potential primary roles of glial cells in the mechanisms of
disorder. Mol Psychiatry 13:365–367. psychiatric disorders. Front Cell Neurosci 9:154.
42. McKinney JA, Turel B, Winge I, Knappskog PM, Haavik J (2009): 63. Sandau US, Alderman Z, Corfas G, Ojeda SR, Raber J (2012):
Functional properties of missense variants of human tryptophan Astrocyte-specific disruption of SynCAM1 signaling results in
hydroxylase 2. Hum Mutat 30:787–794. ADHD-like behavioral manifestations. PLoS One 7:e36424.

Biological Psychiatry ]]], 2016; ]:]]]–]]] www.sobp.org/journal 11


Biological
Psychiatry Brain Enhancer Associated With ADHD

64. Erokhin M, Vassetzky Y, Georgiev P, Chetverina D (2015): Eukaryotic 78. Gaudreau JD, Gagnon P (2005): Psychotogenic drugs and delirium
enhancers: Common features, regulation, and participation in dis- pathogenesis: The central role of the thalamus. Med Hypotheses 64:
eases. Cell Mol Life Sci 72:2361–2375. 471–475.
65. Visel A, Blow MJ, Li Z, Zhang T, Akiyama JA, Holt A, et al. (2009): 79. Ivanov I, Bansal R, Hao X, Zhu H, Kellendonk C, Miller L, et al. (2010):
ChIP-seq accurately predicts tissue-specific activity of enhancers. Morphological abnormalities of the thalamus in youths with attention
Nature 457:854–858. deficit hyperactivity disorder. Am J Psychiatry 167:397–408.
66. Andersen B, Schonemann MD, Pearse RV 2nd, Jenne K, Sugarman J, 80. Xia S, Li X, Kimball AE, Kelly MS, Lesser I, Branch C (2012): Thalamic
Rosenfeld MG (1993): Brn-5 is a divergent POU domain factor highly shape and connectivity abnormalities in children with attention-deficit/
expressed in layer IV of the neocortex. J Biol Chem 268:23390–23398. hyperactivity disorder. Psychiatry Res 204:161–167.
67. Cui H, Bulleit RF (1998): Expression of the POU transcription factor 81. Cao X, Cao Q, Long X, Sun L, Sui M, Zhu C, et al. (2009): Abnormal
Brn-5 is an early event in the terminal differentiation of cns neurons. resting-state functional connectivity patterns of the putamen in
J Neurosci Res 52:625–632. medication-naive children with attention deficit hyperactivity disorder.
68. Eisenstat DD, Liu JK, Mione M, Zhong W, Yu G, Anderson SA, et al. Brain Res 1303:195–206.
(1999): DLX-1, DLX-2, and DLX-5 expression define distinct stages of 82. Qiu MG, Ye Z, Li QY, Liu GJ, Xie B, Wang J (2011): Changes of brain
basal forebrain differentiation. J Comp Neurol 414:217–237. structure and function in adhd children. Brain Topogr 24:243–252.
69. Thompson JA, Ziman M (1999): Pax genes during neural development 83. Seidman LJ, Valera EM, Makris N (2005): Structural brain imaging
and their potential role in neuroregeneration. Prog Neurobiol 95: of attention-deficit/hyperactivity disorder. Biol Psychiatry 57:1263–1272.
334–351. 84. Castellanos FX, Sonuga-Barke EJ, Milham MP, Tannock R (2006):
70. Burbach JP, Smidt MP (2006): Molecular programming of stem cells Characterizing cognition in ADHD: Beyond executive dysfunction.
into mesodiencephalic dopaminergic neurons. Trends Neurosci 29: Trends Cogn Sci 10:117–123.
601–603. 85. Petrykowska HM, Vockley CM, Elnitski L (2008): Detection and
71. Deng Z, Cao P, Wan MM, Sui G (2010): Yin Yang 1: A multifaceted characterization of silencers and enhancer-blockers in the greater
protein beyond a transcription factor. Transcription 1:81–84. CFTR locus. Genome Res 18:1238–1246.
72. Yant SR, Zhu W, Millinoff D, Slightom JL, Goodman M, Gumucio DL 86. Bessa J, Tena JJ, de la Calle-Mustienes E, Fernandez-Minan A,
(1995): High affinity YY1 binding motifs: Identification of two core types Naranjo S, Fernandez A, et al. (2009): Zebrafish enhancer detection
(ACAT and CCAT) and distribution of potential binding sites within the (ZED) vector: A new tool to facilitate transgenesis and the functional
human beta globin cluster. Nucleic Acids Res 23:4353–4362. analysis of cis-regulatory regions in zebrafish. Dev Dyn 238:2409–2417.
73. Donohoe ME, Zhang X, McGinnis L, Biggers J, Li E, Shi Y (1999): 87. Kim JD, Hinz AK, Bergmann A, Huang JM, Ovcharenko I, Stubbs L,
Targeted disruption of mouse Yin Yang 1 transcription factor results in et al. (2006): Identification of clustered YY1 binding sites in imprinting
peri-implantation lethality. Mol Cell Biol 19:7237–7244. control regions. Genome Res 16:901–911.
74. Morgan MJ, Woltering JM, In der Rieden PM, Durston AJ, Thiery JP 88. Uv AE, Thompson CR, Bray SJ (1994): The drosophila tissue-specific
(2004): YY1 regulates the neural crest-associated slug gene in factor grainyhead contains novel DNA-binding and dimerization
xenopus laevis. J Biol Chem 279:46826–46834. domains which are conserved in the human protein cp2. Mol Cell
75. Kwon HJ, Chung HM (2003): Yin Yang 1, a vertebrate polycomb group Biol 14:4020–4031.
gene, regulates antero-posterior neural patterning. Biochem Biophys 89. Cross SH, McKie L, West K, Coghill EL, Favor J, Bhattacharya S, et al.
Res Commun 306:1008–1013. (2011): The opdc missense mutation of pax2 has a milder than loss-
76. He Y, Casaccia-Bonnefil P (2008): The Yin and Yang of YY1 in the of-function phenotype. Hum Mol Genet 20:223–234.
nervous system. J Neurochem 106:1493–1502. 90. Daily K, Patel VR, Rigor P, Xie X, Baldi P (2011): Motifmap: Integrative
77. Denhoff E, Laufer MW, Solomons G (1957): Hyperkinetic impulse genome-wide maps of regulatory motif sites for model species. BMC
disorder in children’s behavior problems. Psychosom Med 19:38–49. Bioinformatics 12:495.

12 Biological Psychiatry ]]], 2016; ]:]]]–]]] www.sobp.org/journal

You might also like