ArticlePDF Available

Machine Learning Prediction of ADHD Severity: Association and Linkage to ADGRL3 , DRD4 , and SNAP25

Authors:

Abstract and Figures

Objective To investigate whether single nucleotide polymorphisms (SNPs) in the ADGRL3, DRD4, and SNAP25 genes are associated with and predict ADHD severity in families from a Caribbean community. Method ADHD severity was derived using latent class cluster analysis of DSM-IV symptomatology. Family-based association tests were conducted to detect associations between SNPs and ADHD severity latent phenotypes. Machine learning algorithms were used to build predictive models of ADHD severity based on demographic and genetic data. Results Individuals with ADHD exhibited two seemingly independent latent class severity configurations. SNPs harbored in DRD4, SNAP25, and ADGRL3 showed evidence of linkage and association to symptoms severity and a potential pleiotropic effect on distinct domains of ADHD severity. Predictive models discriminate severe from non-severe ADHD in specific symptom domains. Conclusion This study supports the role of DRD4, SNAP25, and ADGRL3 genes in outlining ADHD severity, and a new prediction framework with potential clinical use.
Content may be subject to copyright.
https://doi.org/10.1177/10870547211015426
Journal of Attention Disorders
1 –19
© The Author(s) 2021
Article reuse guidelines:
sagepub.com/journals-permissions
DOI: 10.1177/10870547211015426
journals.sagepub.com/home/jad
Article
Introduction
ADHD is a neurodevelopmental disorder characterized by a
complex pattern of persistent clinical symptoms of inatten-
tion, hyperactivity, and impulsivity, in association with the
development of chronic functional impairment (Association,
2002a, 2002b, 2013). ADHD affects 8% to 18% of children
and adolescents, and ~1% to 7% of adults worldwide
(Cornejo et al., 2005; Faraone & Biederman, 2005; Pineda
et al., 1999; Polanczyk et al., 2014), and persists into adult-
hood in 40% to 50% of cases (Thapar et al., 1999, 2016).
ADHD is a major risk factor for the appearance of disrup-
tive (externalizing) disorders, including conduct disorder
(CD), oppositional defiant disorder (ODD), and substance
use disorder (SUD) (Molina et al., 2007; Sibley et al., 2011).
Children with ADHD have a higher predisposition to poor
educational achievement, low-income, underemployment,
legal problems, and impaired social relationships (Molina
et al., 2007; Sibley et al., 2011).
A diagnosis of ADHD requires that a specific set of cri-
teria outlined in the Diagnostic Statistical Manual for
Mental Disorders (DSM) are met (Association, 2002a,
2002b, 2013). According to the number of symptoms exhib-
ited by each subject in different environments, and in coher-
ence with a detailed cognitive clinical evaluation and
comparison between previous and current symptoms, each
subject is assorted to ADHD subtypes (Acosta López et al.,
2013; Acosta et al., 2011; Luo et al., 2019; Posner et al.,
2020). These symptoms relate to the presence or absence of
18 items (operational criteria of the DSM), which are regis-
tered using a binary scale (0: absence; 1: presence), and
grouped by domain (inattention symptoms: items 1–9;
hyperactivity/impulsivity symptoms: items 10–18). These
symptoms provide a differential categorical diagnosis into
four subtypes: ADHD of predominantly inattentive type,
1015426JADXXX10.1177/10870547211015426Journal of Attention DisordersCervantes-Henríquez et al.
research-article2021
1Universidad Simón Bolívar, Barranquilla, Colombia
2Universidad del Norte, Barranquilla, Colombia
3National Institutes of Health, Bethesda, MD, USA
4Universidad de Antioquia, Medellín, Colombia
5Universidad del Atlántico, Barranquilla, Colombia
*These authors contributed equally to this work.
Corresponding Authors:
Martha L. Cervantes-Henríquez, Grupo de Neurociencias del Caribe,
Unidad de Neurociencias Cognitivas, Universidad Simón Bolívar, Calle 54
# 59 -189, Sede 1, Bloque C, Barranquilla, Colombia.
Email: cervantesmh@unisimonbolivar.edu.co
Jorge I. Vélez, Universidad del Norte, Km 5 via Puerto Colombia,
Building K, Room 7-3K, Barranquilla, 081007, Colombia.
Email: jvelezv@uninorte.edu.co
Machine Learning Prediction of ADHD
Severity: Association and Linkage to
ADGRL3, DRD4, and SNAP25
Martha L. Cervantes-Henríquez1,2*, Johan E. Acosta-López1, Ariel F. Martinez3,
Mauricio Arcos-Burgos4*, Pedro J. Puentes-Rozo1,5, and Jorge I. Vélez2*
Abstract
Objective: To investigate whether single nucleotide polymorphisms (SNPs) in the ADGRL3, DRD4, and SNAP25 genes
are associated with and predict ADHD severity in families from a Caribbean community. Method: ADHD severity was
derived using latent class cluster analysis of DSM-IV symptomatology. Family-based association tests were conducted to
detect associations between SNPs and ADHD severity latent phenotypes. Machine learning algorithms were used to build
predictive models of ADHD severity based on demographic and genetic data. Results: Individuals with ADHD exhibited
two seemingly independent latent class severity configurations. SNPs harbored in DRD4, SNAP25, and ADGRL3 showed
evidence of linkage and association to symptoms severity and a potential pleiotropic effect on distinct domains of ADHD
severity. Predictive models discriminate severe from non-severe ADHD in specific symptom domains. Conclusion: This
study supports the role of DRD4, SNAP25, and ADGRL3 genes in outlining ADHD severity, and a new prediction framework
with potential clinical use. (J. of Att. Dis. XXXX; XX(X) XX-XX)
Keywords
ADHD severity, machine learning, LPHN3, SNAP25, DRD4.
2 Journal of Attention Disorders 00(0)
ADHD of predominantly hyperactive/impulsive type,
ADHD of combined type, and ADHD unaffected
(Fernandez-Jaen et al., 2018; Schachar et al., 2000).
However, this dichotomous classification approach to
ADHD diagnosis (0: unaffected; 1: affected) excludes the
existence of intermediate spectrum ADHD classes (Cuthbert
& Insel, 2010; Insel, 2014; Larsson et al., 2012) and inexo-
rably defines a minimum-required number of symptoms to
classify individuals (Acosta et al., 2004, 2011; Arcos-
Burgos & Muenke, 2004; Acosta et al., 2011; Cuthbert &
Insel, 2010; Larsson et al., 2012; Lilienfeld & Treadway,
2016).
The use of machine learning (ML) algorithms and multi-
variate analyzes allows for the identification and classifica-
tion of individuals with subtle differences in symptoms.
This is achieved by constructing symptom profiles (i.e.,
indicators) to predict ADHD diagnosis and eventually
resolve ADHD severity (Casey et al., 2014; Insel, 2014;
Kautzky et al., 2020; Tenev et al., 2014; Yasumura et al.,
2017). In addition, these ADHD severity indicators allow
for the construction of more precise and intelligent diagnos-
tic schemes to characterize individuals within extreme
forms of the disorder, either moderate or severe, which con-
fers higher power and resolution in identifying the causes
underpinning ADHD (Tandon et al., 2016).
Recent ADHD studies suggest the presence of subtle but
significantly independent subgroups within ADHD classi-
cal subtypes, mainly characterized by a differential severity
of symptoms and the presence/absence of externalizing
symptoms (i.e., CD and ODD) (Acosta et al., 2004; Andrews
et al., 2009). This system leads to a complex diagnosis
based on severity and persistence (Acosta et al., 2016; Jain
et al., 2011). There is also evidence that ADHD severity is
influenced to some degree by gender, psychiatric comorbid-
ity, family environment, conduct symptoms, and poor anger
management (Ramy et al., 2018). The identification of these
intermediate and extremely hidden classes is important, as
individuals with severe ADHD forms are associated with
poor academic achievement, moderate to poor pharmaco-
logical treatment response (Kotte et al., 2013; Owens &
Jackson, 2017), development of externalizing symptoms
and comorbidities that persist across the life span (Korsgaard
et al., 2016; Lee et al., 2020).
Longitudinal family- and twin-based genetic studies
estimate ADHD heritability at ~75% (Curran et al., 2001;
Faraone & Biederman, 2005), strongly linking genetic fac-
tors to the etiology of ADHD and its comorbidities (Acosta
et al., 2004, 2006; Arcos-Burgos, Castellanos, Pineda, et al.,
2004; Arcos-Burgos, Jain, et al., 2010; Jain et al., 2011;
Puentes-Rozo et al., 2019). Genetic studies have identified
genetic variation in DRD4, DRD5, DAT1, SNAP-25, FGF1,
HTR1B, 5-HTTLPR, SLC6A3, TTC12, NCAM1, and
ADGRL3 to be associated with ADHD and/or ADHD sever-
ity (Acosta et al., 2016, 2011; Arcos-Burgos, Jain, et al.,
2010; Arcos-Burgos & Muenke, 2010; Bruxel et al., 2020;
Faraone & Mick, 2010; Franke et al., 2010; Gizer et al.,
2009; Jain et al., 2011; Kotte et al., 2013; Martinez et al.,
2016; Mastronardi et al., 2016). We showed that variants in
ADGRL3 (also known as LPHN3) interact with gene vari-
ants harbored at chromosome 11q (in a region spanning the
NCAM1-TTC12-ANKK1-DRD2 genes) to dramatically
increase ADHD risk and severity in young children (Acosta
et al., 2016, 2011; Arcos-Burgos, Jain, et al., 2010; Bruxel
et al., 2020; Jain et al., 2011). Most of these associations
have been replicated in samples of Caucasian, Asian, and
Latino descent, but remain to be tested in populations with
a predominantly African ancestry.
In this report, we assess the genetics of ADHD severity
using analytical methods of linkage and association on a
cohort of 113 nuclear families ascertained from a Caribbean
community (Cervantes-Henriquez et al., 2018; Pineda et al.,
2016; Puentes-Rozo et al., 2019). This community has one
of the largest African genetic components in the Caribbean,
Central America and South America (Barragán-Duarte,
2007). We hypothesize that a new phenotypic construct of
ADHD severity based on ML algorithms may reduce intrin-
sic biases associated with its clinical heterogeneity to
enhance the performance of ADHD diagnostic tools. This
approach has potential applications in precision and person-
alized medicine in understudied populations.
Patients and Methods
Patients
During the last 12 years, we prospectively recruited and
clinically characterized 408 individuals (175 [43%] females
and 233 [57%] males; 236 [58%] affected with ADHD)
belonging to 120 nuclear families with at least one child
affected with ADHD (proband) whose members were born
in Barranquilla, Colombia, and its metropolitan area.
Because of the African Diaspora across the Americas dur-
ing the last five centuries, this community exhibits a strong
genetic admixture among aboriginal Amerindian communi-
ties, Spaniards, and Africans (Barragán-Duarte, 2007;
Puentes-Rozo et al., 2019). Admixed populations from
South America and the Caribbean are often grouped into a
single racial construct by the American census, which fails
to grasp the distribution of genetic variation among these
populations in health disparity studies (Tishkoff & Kidd,
2004). This is important as individuals of African ancestry
suffer from a disproportionate burden of morbidity and dis-
ability associated with common chronic diseases (Ezzati
et al., 2008).
The demographic, ascertainment, clinical, and genotyp-
ing data collection methods are reported elsewhere
(Cervantes-Henriquez et al., 2018; Pineda et al., 2016;
Puentes-Rozo et al., 2019). Briefly, 120 nuclear families
Cervantes-Henríquez et al. 3
comprising 408 individuals, ascertained from probands
affected by ADHD, participated in our clinical and genetic
studies of ADHD. Families of size three (n=74; 65.4%),
four (n=33; 29.2%), five (n=4; 3.5%), and six (n=2; 1.8%)
were present. Participant ages ranged between 6 and 60
years (average age=26.6±15.4 years), and 233 (57.1%)
were affected with ADHD. A total of 246 (60.3%) were
adults (aged 18 or older) of whom 97 (39%) were affected
with ADHD (14.5% females) and 149 unaffected (35.7%
females). In children and adolescents (ages 6–18, n = 162),
136 (84%) individuals were affected with ADHD (includ-
ing 34 [25%] females) and 26 (16%) were unaffected (15
[57.6%] females). No children or adults were treated with
medication for ADHD at initial assessment (Cervantes-
Henriquez et al., 2018; Pineda et al., 2016; Puentes-Rozo
et al., 2019). ADHD diagnosis was assessed in all individu-
als using the structured Diagnostic Interview for Children
and Adults (DICA) version IV (Reich, 2000). The DICA-IV
considers the A criterion of the DSM-IV and uses a system-
atic approach to collect clinical information about the
ADHD symptoms exhibited by an individual, and uses a
binary classification (0 = absent; 1 = present) and has been
extensively used by our group and others in genetic studies
of ADHD (Acosta et al., 2008, 2011; Arcos-Burgos,
Castellanos, et al., 2004; Arcos-Burgos, Jain, et al., 2010;
Palacio et al., 2004). Parents or guardians were adminis-
tered the Spanish version of the DICA-IV interview for par-
ents (DICA-IV-P). Parents and teachers of school-age
children also provided behavior rating scales. Adult partici-
pants completed the modified DICA-IV-P disruptive behav-
ior module to retrospectively collect information about the
beginning, severity and duration of their current behaviors
and conduct (Palacio et al., 2004).
In this study, we included 113 out of the 120 nuclear
families from the original cohort; seven families were
excluded because genotyping data were not available
(Puentes-Rozo et al., 2019). The average family size was
3.4 ± 0.65 (74 trios, 33 families with four members, four
families with five members, and two families with six
members). Adult participants signed a written informed
consent and individuals under 18 years of age were signed
for by their parents or legal guardians. Ethics approval was
obtained from the Ethics Committee of Universidad Simón
Bolívar at Barranquilla, Colombia (approval # 00032,
October 13, 2011).
Definition of Severity
Severity of symptoms was defined with unsupervised
machine learning (ML) algorithms aimed at identifying
latent subgroups of individuals with distinctive symptom
profiles based on clinical data. Individuals were classified
as “severe” or “not severe” based on the number of clinical
symptoms that are likely to occur more often within specific
clinical profiles (Acosta et al., 2011). ADHD symptom data
was collected during the clinical assessment stage, where 11
schools were visited (seven of medium socio-economic
stratum). Meetings were held with teachers of children aged
between 6 and 11 years old to explain the objective of the
study. Teachers were asked to identify children about whom
they had concerns that might affect their academic perfor-
mance and/or behavior in the school environment. Screening
for ADHD symptoms was performed using the teachers’
version of a brief questionnaire (checklist) based on the
DSM-IV A criterion questions. This questionnaire has pre-
viously been standardized for Colombian children and ado-
lescents. Possible ADHD cases for further investigation
were selected based on a standard score T = 50 ± 9. Thus,
children who scored T 60 were suspected of having
ADHD per Colombian norms.
Clinical profiles were derived with Latent Class Cluster
Analysis (LCCA) (Vermunt & Magidson, 2002), as imple-
mented in Latent GOLD 4.0 (Statistical Innovations,
Belmont, MA, USA), using clinical symptoms as defined
by the DSM-IV (DSM-IV, 2002) diagnostic criteria and the
DICA interview. LCCA is an unsupervised ML algorithm
widely used to identify subgroups of individuals from
mixed data in which records of variables of different nature
are available. Symptoms were registered using a binary sys-
tem that assessed the presence or absence of 20 clinical
symptoms from a symptom-based questionnaire completed
by all participants (0: absence; 1: presence), which were
further used as indicators in all LCCA models. For children,
questionnaires were answered by the parents or legal guard-
ians. We explored LCCA models, with up to 10 subgroups
for all symptoms (questions 1–20) and those specific to the
inattention (questions 1–10), hyperactivity (questions 11–
16), and impulsivity (questions 17–20) domains separately.
In these models, demographic information such as sex and
age group (children: 4–11 years; adolescents: 12–17 years;
adults >17 years) (Arcos-Burgos et al., 2010) were used as
covariates. Because of the family-based structure of our
data, LCA models were adjusted to account for the non-
independence of individuals within families. The number of
clusters was selected using a likelihood ratio test (LRT) and
their statistical significance was assessed via parametric
bootstrap with B = 500 replicates. Further, individuals are
assigned to each cluster based on the highest posterior prob-
ability of belonging to each cluster according to their symp-
toms’ profile.
To classify individuals as “severe” or “not severe” (i.e.,
the severity phenotype) according to the DSM-IV criteria,
we derived the clinical profile associated with the clusters
identified by LCCA. Individuals within a particular LCCA-
derived cluster were defined as “severe” if the number of
questions with probability of occurrence above 50% was
higher than half of the total number of domain-specific
questions. For example, in the inattention domain, which
4 Journal of Attention Disorders 00(0)
assessed 10 clinical symptoms, a particular cluster (and
therefore all individuals belonging to it) will be classified as
“severe” if at least six questions have an occurrence proba-
bility greater than 50%. From that point on, individuals with
severe symptoms were labeled as “cases” or as “controls”
otherwise.
Demographic Characterization
Measures of central tendency and dispersion were employed
to summarize continuous variables. Frequencies and pro-
portions were estimated for categorical variables. Potential
confounders were controlled with logistic regression, and
odds ratios (OR) with 95% confidence intervals (CI) were
used to summarize results. Unless otherwise stated, statisti-
cal analyzes and plotting were performed in R version 3.6.2
(R Core Team, 2019). The false discovery rate (FDR) was
used to control type I error derived from multiple testing
(Benjamini & Hochberg, 1995; Vélez et al., 2014).
Hierarchical clustering (HC) was used to evaluate group-
ing of symptoms severity. This method, in contrast with the
Spearman correlation coefficient, uses an agglomerative
algorithm that joins the most similar component features,
and sequentially joins the next most similar with the first
two converted to a new combined unit (Venables & Ripley,
2002). Clusters were generated via complete linkage; their
uncertainty was assessed using approximately unbiased
(AU) and bootstrap probability (BP) values as implemented
in the R pvclust (Suzuki et al., 2019) package. The AU
p-values and the BP values were computed by multiscale
and by normal bootstrap resampling, respectively, with the
former being a better approximation to unbiased p-values.
For this analysis, a total of 10,000 bootstrap samples were
generated. Thus, AU p-values > 95% strongly support the
existence of a cluster structure.
Genetic Analysis
DNA extraction and genotyping. DNA extraction and geno-
typing were performed as described elsewhere (Puentes-
Rozo et al., 2019). Briefly, genomic DNA was isolated
from blood samples using the MasterPure® DNA Purifica-
tion Kit (Epicentre Biotechnologies, Chicago, IL, USA)
according to the manufacturer’s protocol. DNA concentra-
tions were measured using a NanoDrop™ 2000 spectropho-
tometer (Thermo Fisher Scientific, Waltham, MA, USA).
Genotyping was performed at the University of Arizona
Genetics Core using the multiplex Sequenom® Technology
on Agena Bioscience’s MassARRAY® MALDI-TOF
instrument.
Family-based association analysis. Genetic association and
linkage analyzes were performed using the generalized fam-
ily-based association test (FBAT) model, which provides a
unified framework for the transmission disequilibrium test
(TDT) (Laird et al., 2000; Spielman et al., 1993). FBAT
accounts for different genetic models, family-based ascer-
tainment designs, complex phenotypes (diseases) architec-
ture, missing parents, and different subtypes of null
hypotheses, (Laird et al., 2000) while being minimally
affected by non-causal SNPs (Brookes et al., 2006). We used
the FBAT model as implemented in the PBAT module of
SNP Variation Suite (SVS) 8.8.3 (Golden Helix, Inc., Boze-
man, MT, USA). Briefly, several genetic models of inheri-
tance were explored (additive, dominant, recessive, and
heterozygous advantage). Haplotype tests for selected com-
binations of phenotypes and markers were applied because
PBAT automatically controls for both the type I error rate
generated by multiple comparisons (Rabinowitz & Laird,
2000) and the problem of genetic stratification (Benjamini &
Hochberg, 1995; Laird et al., 2000; Lange et al., 2004;
Lunetta et al., 2000; Vélez et al., 2014). Another advantage of
the FBAT design is that low genotype call rates in probands
can be compensated by imputation from parental genotypes,
and Mendelian inconsistencies are generally removed from
analyzes (Lange & Laird, 2002a, 2002b). As age and sex are
known to impact ADHD susceptibility (Mowlem et al., 2018;
Oerbeck et al., 2019; Ramtekkar et al., 2010; Skogli et al.,
2013), both variables were included as covariates under the
hypothesis of no linkage and no association. As a result,
inclusion of these covariates substantially improved FBAT
statistical power (Lange & Laird, 2002a, 2002b).
Predictive Genomics Models of Symptoms
Severity
We used ML algorithms to construct predictive models of
symptom severity for different symptom domains (i.e.,
global, inattention, hyperactivity, and impulsivity). The set
of predictors consisted of demographic variables, and
genetic markers. Several ML methods were explored,
including Logistic Regression, Classification and Regression
Tree (CART) (Breiman et al., 1984), Random Forest (RF)
(Breiman, 2001; Satterfield et al., 1974), Support Vector
Machine (SVM) (Cortes & Vapnik, 1995; Salazar et al.,
2012), and Tree Boosting (XGBoost) (Chen & Guestrin,
2016; Chen et al., 2020). The full list of ML algorithms is
provided in Table S1 of the Supplementary Material.
Construction, validation, and testing of these predictive
models were performed using the methods implemented in
the R caret (Kunh, 2020) package. Initially, models were
constructed and tuned with 70% of the data (training data)
using a 10-fold cross-validation procedure with five repeti-
tions. In the construction stage, models were tuned with the
training data set to identify the best combination of param-
eters for each ML algorithm that maximized the model’s
accuracy for predicting symptom severity in a particular
domain. Finally, models were validated using the remaining
Cervantes-Henríquez et al. 5
30% of the data set (testing data) and a measure of balanced
accuracy was derived. This measure was calculated as the
percentage of individuals correctly classified. On average,
the training and testing data sets were composed of 241 and
115 individuals, respectively. Models were assessed using
the Receiver Operating Characteristic (ROC) (Metz, 1978)
curve and the area under the ROC curve (AUC) as perfor-
mance measures. In addition, the sensitivity (Se), specificity
(Sp), correct classification rate (accuracy), positive predic-
tive value (PPV), negative predictive value (NPV), false
discovery rate (FDR), and false positive rate (FPR) perfor-
mance measures, were also calculated.
Results
Severity of Symptoms
Of the 386 patients studied, 224 (58%) were affected by
ADHD. The distribution by sex and age of the 386 individu-
als was as follows: 218 (56.5%) males and 168 (43.5%)
females; 120 (31.1%) were children (6–11 years), 34 (8.8%)
adolescents (12–17 years), and 232 (60.1%) adults (>17
years). We identified six significantly different latent class
clusters using all items of the DSM-IV criteria, three clus-
ters within the inattention domain, three clusters within the
hyperactivity domain, and two clusters within the impulsiv-
ity domain (Table 1 and Figure 1). Fit statistics for LCCA
models, the average posterior probabilities for each cluster
and the distribution of the total number of symptoms by
LCCA-derived cluster are presented in the Supplementary
Material. We identified that the number of DSM-IV symp-
toms differ by LCCA-derived clusters (Figure 1S;
Supplementary Material).
Following our approach, individuals assorted to clusters
2, 3, and 6 based on the full set of items globally exhibit
severe symptoms (n = 167, 43.4%; Figure 1a); individuals
classified in clusters 2 and 3 based on the inattention items
exhibit severe symptoms in this domain (n = 222, 57.5%;
Figure 1b); individuals assorted in cluster 3, based on the
hyperactivity items, were classified as severely affected in
this domain (n = 90, 23.3%; Figure 1c); and individuals
assorted in cluster 2 based on impulsivity items were classi-
fied as severely affected in this domain (n = 142, 36.8%;
Figure 1d). Regardless of the domain, severely affected
clusters are predominantly constituted by male children
with a positive ADHD diagnosis (Table 1). Overall, indi-
viduals exhibiting a complex severity phenotype show
higher number of DSM-IV symptoms (Figure 2S;
Supplementary Material). Further analyzes of age within
LCCA-derived (Figure 3S; Supplementary Material) and
severity phenotype (Figure 4S; Supplementary Material)
indicate that children, adolescents and young adults (age <
25) are severely affected, while the group of not severe indi-
viduals is mostly constituted by adults (age > 25).
Logistic Regression analyzes revealed that individuals
with ADHD are 8.9 times more likely to display a severe
phenotype based on global items (95% CI = 4.9–16.6). As
expected, children and adolescents exhibited the highest
risk of developing global-based severe symptoms (odds
ratio [OR] = 7.3, 95% CI = 4.1–13.4; OR = 4.9, 95% CI
= 2.1–11.7, respectively). Interestingly, 19 (11.4%) indi-
viduals identified as severely affected (Figure 1a) were not
diagnosed with ADHD (six children, two adolescents, and
11 adults; Table S4 and Figure 5S; Supplementary Material).
In the inattention domain, individuals with a positive
ADHD diagnosis were 14.9 times more likely to exhibit
severe inattention than controls, regardless of sex (95% CI
= 8.6–26.5). In addition, children were more likely to have
severe inattention than adults (OR = 2.1, 95% CI = 1.1–
4.1), while adolescents were equally likely (OR = 1.8, 95%
CI = 0.7-4.7). Of the individuals with severe inattention
symptoms (Figure 1b), 35 (15.8%) were not diagnosed with
ADHD (six children, two adolescents, and 27 adults; Table
S4 and Figure 5S; Supplementary Material).
In the hyperactivity domain, individuals with a positive
ADHD diagnosis were 6.9 times more likely to have severe
hyperactivity symptoms than adults, regardless of sex (95%
CI = 3.2–17.7) (Figure 2c). In contrast, adolescents and
adults were equally likely to suffer from severe hyperactiv-
ity symptoms (OR = 1.4, 95% CI = 0.53–3.8). Of individu-
als exhibiting severe inattention symptoms (Figure 1c), 7
(7.7%) were not diagnosed with ADHD (two children, one
adolescent, and four adults; Table S4 and Figure 5S;
Supplementary Material).
In the impulsivity domain, individuals with a positive
ADHD diagnosis were 4.8 times more likely to have severe
inattention symptoms (95% CI = 2.7–8.7). In addition,
children (OR = 4.8, 95% CI = 2.7–8.7), and adolescents
(OR = 2.5, 95% CI = 1.1–5.7) were more likely than adults
to be severely affected in this domain. Of all individuals
with severe impulsivity (Figure 1d), 20 (13.9%) were not
diagnosed with ADHD (four children, four adolescents, and
12 adults; Table S4 and Figure 5S; Supplementary Material).
Differential Clinical Signature on Severity
Hierarchical clustering of the LCCA-derived severity phe-
notypes revealed a differential clinical signature in our
cohort (Figure 2). Our data suggests that individuals diag-
nosed with ADHD manifest severity of symptoms in two
seemingly independent configurations. The first configura-
tion involves a combination of global severity symptoms,
while the second configuration involves symptom severity
ascribed exclusively to the hyperactivity-impulsivity
domain (Figure 2; left). Bootstrap validation suggests that
these two configurations are statistically distinct (strongly
supported by the data) and differ from that manifested by
individuals without an ADHD diagnosis (Figure 2; right).
6
Table 1. Demographic and Clinical Characteristics of Clusters Derived using Latent Class Cluster Analysis Based on DSM-IV Symptomatology.
DSM-IV domain Cluster 1 Cluster 2 Cluster 3 Cluster 4 Cluster 5 Cluster 6
Statistic (df)pGlobal n (%) n (%) n (%) n (%) n (%) n (%)
129 (33.4) 72 (18.7) 57 (14.8) 51 (13.2) 39 (10.1) 38 (9.8)
Number of symptoms (mean ± SD) 1.5 ± 1.5 12.2 ± 2.4 18.4 ± 1.5 6.5 ± 2.2 7.2 ± 1.7 13 ± 2 814.3 (5, 376)a<.000001b
ADHD (no/yes) 111 (86)/18 (14) 9 (12.5)/63 (87.5) 5 (8.8)/52 (91.2) 23 (45.1)/28 (54.9) 9 (23.1)/30 (76.9) 5 (13.2)/33 (86.8) 173.24 (5)c<.000001
Sex (F/M) 70 (54.3)/59 (45.7) 25 (34.7)/47 (65.3) 13 (22.8)/44 (77.2) 28 (54.9)/23 (45.1) 15 (38.5)/24 (61.5) 17 (44.7)/21 (55.3) 21.39 (5)c<.001
Age 153.9 (10)c<.000001
Children 15 (11.6) 42 (58.3) 40 (70.2) 2 (3.9) 8 (20.5) 13 (34.2)
Teens 11 (8.5) 2 (2.8) 6 (10.5) 1 (2) 2 (5.1) 13 (34.2)
Adults 103 (79.8) 28 (38.9) 11 (19.3) 48 (94.1) 29 (74.4) 12 (31.6)
Inattention
Cluster 1 Cluster 2 Cluster 3 Cluster 4 Cluster 5 Cluster 6
Statistic (df)pn (%) n (%) n (%) n (%) n (%) n (%)
164 (42.5) 134 (34.7) 88 (22.8)
Number of symptoms (mean ± SD) 0.9 ± 1.1 5.7 ± 1.5 9.6 ± 0.7 1789.8 (2, 379)a<.00001b
ADHD (no/yes) 127 (77.4)/37 (22.6) 24 (17.9)/110 (82.1) 11 (12.5)/77 (87.5) 147.9 (2)c<.00001
Sex (F/M) 85 (51.8)/79 (48.2) 55 (41)/79 (59) 28 (31.8)/60 (68.2) 9.8 (2)c.00729
Age 69.1 (4)c<.00001
Children 22 (13.4) 49 (36.6) 49 (55.7)
Teens 12 (7.3) 8 (6) 15 (17)
Adults 130 (79.3) 77 (57.5) 24 (27.3)
Hyperactivity
Cluster 1 Cluster 2 Cluster 3 Cluster 4 Cluster 5 Cluster 6
Statistic (df)pn (%) n (%) n (%) n (%) n (%) n (%)
188 (48.7) 108 (28) 90 (23.3)
Number of symptoms (mean ± SD) 2.5 ± 3.1 5.3 ± 3 7.8 ± 2.6 127.97 (2, 379)a<.00001b
ADHD (no/yes) 126 (67)/62 (33) 29 (26.9)/79 (73.1) 7 (7.8)/83 (92.2) 101.8 (2)c<.00001
Sex (F/M) 102 (54.3)/86 (45.7) 42 (38.9)/66 (61.1) 24 (26.7)/66 (73.3) 20.2 (2)c<.00001
Age 73.9 (4)c<.00001
Children 27 (14.4) 35 (32.4) 58 (64.4)
Teens 17 (9) 11 (10.2) 7 (7.8)
Adults 144 (76.6) 62 (57.4) 25 (27.8)
Impulsivity
Cluster 1 Cluster 2 Cluster 3 Cluster 4 Cluster 5 Cluster 6
Statistic (df)pn (%) n (%) n (%) n (%) n (%) n (%)
164 (42.5) 134 (34.7)
Number of symptoms (mean ± SD) 0.6 ± 0.8 2.9 ± 0.9 726.580 (1, 380)a<.00001b
ADHD (no/yes) 143 (58.6)/101 (41.4) 19 (13.4)/123 (86.6) 73.539 (1)c<.00001
Sex (F/M) 121 (49.6)/123 (50.4) 47 (33.1)/95 (66.9) 9.2721 (1)c.002327
Age 90.282 (2)c<.00001
Children 36 (14.8) 84 (59.2)
Teens 20 (8.2) 15 (10.6)
Adults 188 (77) 43 (30.3)
Note. Categories with the higher frequency are shown in bold. See Figure 1 for information. df = degrees of freedom; p = p-value; SD = standard deviation.
aF statistic with (m, n) degrees of freedom.
bCorrected for ADHD diagnosis, age and gender.
c-based test.
Cervantes-Henríquez et al. 7
Figure 1. Profile plots derived using latent class cluster analysis applied to ADHD symptoms derived from DSM-IV criteria. Results
are shown for: (a) all items (Q1–Q20), (b) inattention items (Q1–Q10), (c) hyperactivity items (Q11–Q16) and (d) impulsivity items
(Q17–Q20). The number of individuals per cluster is also shown. Individuals exhibiting a severe phenotype correspond to clusters 2, 3
and 6 in (a), clusters 2 and 3 in (b), cluster 3 in (c), and cluster 2 in (d).
Q1: difficulty to do homework; Q2: concentration problems; Q3: difficulty to listening directly; Q4: insecurity when instructed; Q5: disorganized; Q6:
hates to do homework; Q7: loses objects; Q8: gets distracted by anything; Q9: forgets daily activities; Q10: gets tired, goes from one activity to the
next without finishing; Q11: writhes in his position, moves hands and feet; Q12: difficulty sitting; Q13: frequently running, climbing and jumping; Q14:
has trouble doing something quietly; Q15: does not get tired; continuously moving; Q16: frequently speaks and speaks successively; Q17: answers
questions before they finish asking them; Q18: difficulty waiting for turns; Q19: interrupts conversation and/or games of other children; Q20: performs
dangerous activities.
Both the impulsivity and hyperactivity severity of symp-
toms were different between individuals with ADHD and
controls, with no support for a particular clustering pattern
of severe symptoms in the control group (Figure 2; right).
Markers Conferring Susceptibility to Severe
Symptoms
Table 2 shows the main results of the FBAT analysis. We
found significant linkage and association of either global
or domain-specific severity with markers DRD4-rs916457,
SNAP25-rs362990, ADGRL3-rs2122642, and
ADGRL3-rs10001410. In particular, DRD4-rs916457 and
SNAP25-rs362990 were associated with both global and
inattention symptom severity; ADGRL3-rs2122642,
ADGRL3-rs10001410, and DRD4-rs916457 were found to
be associated with severity of hyperactivity symptoms;
and DRD4-rs916457, ADGRL3-rs2122642, and
SNAP25-rs362990 were associated with the severity of
impulsivity symptoms under different genetic models of
inheritance (Table 2).
Predictive Genomics Framework for Severity of
Symptoms
Figure 3 shows the accuracy of the ML algorithms used to
predict ADHD severity. We found that the SVM with poly-
nomial kernel (svmPoly), linear discriminant analysis (lda),
gradient boosting machine (gbm), and CART (rpart2) ML
algorithms provide the highest accuracy for predicting
global, inattention, hyperactive and impulsivity severity,
respectively (Figure 3a).
ROC analysis indicates that, in all cases, the AUC pro-
vides a moderate ability of the predictive models to discrim-
inate severe from non-severe individuals based on
demographic and genetic data, with accuracy values
8 Journal of Attention Disorders 00(0)
ranging between ~70% and 82% for the derived models
(Table 3 and Figure 3b). These models also provide com-
petitive Se, Sp, PPV, NPV, FDR, and FPR values, which
make them a suitable alternative to diagnose ADHD symp-
tom severity in the clinical setting (Table 3).
Analyzes of variable importance indicate that, overall,
age, sex, and severity-associated SNPs are important pre-
dictors of derived latent class clusters (Figure 3c). Among
those SNPs (Table 2), marker SNAP25-rs362990 is simi-
larly ranked in terms of variable importance for predicting
global, inattention, and hyperactivity symptom severity,
and ranked as least for predicting impulsivity severity
(Figure 3c). Interestingly, ADGRL3-rs10001410 also pro-
duces a similar effect, although to a lesser extent, for pre-
dicting inattention and hyperactivity severity, while marker
ADGRL3-rs2122642 is the third most important predictor
of hyperactivity severity, and the fourth most important to
predict impulsivity severity (Figure 3c). On the other hand,
marker DRD4-rs916457 was identified as the most impor-
tant genetic predictor of impulsivity severity, and ranked
last for predicting the severity of the remaining symptoms
(Figure 3c).
Discussion
ADHD and related behaviors fit better a classification system
based on a continuous spectrum of symptom severity, rather
than in a discrete category with a dichotomous affection status
(i.e., affected and unaffected) (Asherson & Trzaskowski,
2015). Indeed, the severity of ADHD symptoms is
significantly correlated with neuropsychological functioning
(Rajendran et al., 2013) and represents a major risk factor for
major depression disorder and ODD (Tandon et al., 2016).
These aspects might represent important factors to define
ADHD natural history and its implications for the clinical
practice (Acosta et al., 2011; Brown et al., 2017; Fletcher &
Wolfe, 2009; Shaw et al., 2012). Thus, proper and early iden-
tification of individuals at risk for severe ADHD may facili-
tate assessment, diagnosis, treatment, and follow-up
(prediction, prevention and intervention) (Kotte et al., 2013;
Owens & Jackson, 2017). Altogether, these findings highlight
the importance of assessing the severity of symptoms, espe-
cially at young ages, for the definition and selection of appro-
priate therapeutic strategies to reduce ADHD burden.
In this study, a phenotypic classification system based on
the severity of ADHD symptoms was derived from the
identification of latent classes using unsupervised ML algo-
rithms. We argue that this severity phenotype, in conjunc-
tion with ML algorithms, is suitable for and should be
preferred when dissecting causes and building predictive
frameworks, as it reduces global heterogeneity and enhances
more precise, unique and personalized demographic, cul-
tural, clinical, and biological features(Acosta et al., 2008,
2011; Jain et al., 2007; Larsson et al., 2012; Owens &
Jackson, 2017; Pineda et al., 2016). By using different ML,
we were able to build a diagnostic tool for ADHD severity
with the ultimate goal of identifying, based on training data
and a cross-validation strategy, which of them had the best
prediction accuracy. These ML algorithms (Table S1;
Supplementary Material) are particularly useful to construct
Figure 2. Correlation-based dendrograms for the severity of global, inattention, hyperactivity and impulsivity symptoms by ADHD
diagnosis. Approximately unbiased (AU) and bootstrap probability (BP) values are shown in red and green, respectively. AU p-values
greater than 95% suggest that the clustering structure is strongly supported by the data. Node numbers appear in gray. The vertical
dotted line represents the optimal number of clusters for the ADHD group. We identified two different co-occurrence configurations
in ADHD affected individuals (cases), and only one mechanism in unaffected individuals (controls). However, this latter mechanism
was not supported by the data.
Cervantes-Henríquez et al. 9
predictive models when the response variable (i.e., outcome
of interest) is of binary. Considering that individuals are
classified as “severe” or “not severe” based on the number
of symptoms with probability of occurrence above 50%, the
choice of such ML algorithms was justified. In the near
future, those ML best-performing algorithms could easily
be implemented in an intelligent information system that
facilitates the identification of individuals with one or more
ADHD severity patterns.
Our findings suggest the presence of one or more inde-
pendent groups subtly integrated as part of the ADHD phe-
notype that correlate with ADHD symptoms and functional
impairment (Kupper et al., 2012; Theule et al., 2013) ADHD
severity, and parenting problems (Anastopoulos et al., 1992;
Faraone & Larsson, 2019; Graziano et al., 2011; Palacios-
Cruz et al., 2014; Rajendran et al., 2013). In line with previ-
ous studies correlating ADHD diagnosis with long-term
outcome and poorer neuropsychological functioning (Helfer
et al., 2019; Owens & Jackson, 2017; Rajendran et al.,
2013), we found that a positive ADHD diagnosis is a risk
factor for symptoms severity either globally or domain-spe-
cific. We also identified two seemingly independent symp-
toms severity configurations in individuals with ADHD that
differ from those observed in unaffected individuals (Figure
2). Altogether, these findings suggest that symptoms sever-
ity is distinct in individuals with ADHD, but may also have
important implications for those without a positive ADHD
diagnosis. This could be partially explained by the fact that
some individuals are below a well-accepted threshold for
ADHD diagnosis (Larsson et al., 2012).
Previously, we identified that SNPs in the ADGLR3,
DRD4 and SNAP25 and FGF1 genes confer susceptibility
to ADHD in the same families evaluated in this study
(Puentes-Rozo et al., 2019). Here, we explored the associa-
tion between those SNPs and ADHD symptom severity. We
found that markers DRD4-rs916457, SNAP25-rs362990,
ADGRL3-rs2122642, and ADGRL3-rs10001410 are in link-
age and association with the severity of either global or
domain-specific symptoms under different genetic models
of inheritance (Table 2). To the best of our knowledge, this
Table 2. Results of the FBAT Analysis Applied to Symptoms Severity by Domain.
Chr Marker Gene Positiona
FBAT results
Allele
Cohort PFBAT (NIF)
Frequency Additive Dominant Recessive HA
Global
11 rs916457 DRD4 637,014 T 0.050 0.0097 (27) 0.0078 (27)
20 rs362990 SNAP25 10,295,573 A 0.906 0.0495 (55)
Inattention
11 rs916457 DRD4 637,014 T 0.050 0.0067 (27) 0.0053 (27) 0.0339 (27)
C 0.950 0.0067 (27) 0.0053 (27) 0.0339 (27)
4 rs10001410 ADGRL3 61,608,511 C 0.673 0.0067 (66) 0.0265 (54)
A 0.327 0.0067 (66) 0.0265 (54)
20 rs362990 SNAP25 10,295,573 A 0.906 0.0290 (55) 0.0290 (55) 0.0290 (55)
T 0.094 0.0290 (55) 0.0290 (55) 0.0290 (55)
Hyperactivity
4 rs2122642 ADGRL3 61,832,546 T 0.256 0.0004 (55) 0.0010 (45) 0.0339 (55)
C 0.744 0.0004 (55) 0.0010 (45) 0.0339 (55)
4 rs10001410 ADGRL3 61,608,511 C 0.673 0.0183 (66)
A 0.327 0.0183 (66)
11 rs916457 DRD4 637,014 C 0.950 0.0164 (27) 0.0269 (27)
T 0.050 0.0164 (27) 0.0269 (27)
Impulsivity
11 rs916457 DRD4 637,014 C 0.950 0.0043 (27) 0.0059 (27) 0.0253 (27)
T 0.050 0.0043 (27) 0.0059 (27) 0.0253 (27)
4 rs2122642 ADGRL3 61,832,546 T 0.256 0.0060 (55) 0.0221 (45)
C 0.744 0.0060 (55) 0.0221 (45)
20 rs362990 SNAP25 10,295,573 A 0.906 0.0112 (55) 0.0141 (55) 0.0236 (55)
T 0.094 0.0112 (55) 0.0141 (55) 0.0236 (55)
Note. For interpretation purposes, positive p-values indicate susceptibility. Chr=chromosome; HA = heterozygous advantage; NIF = number of infor-
mative families; FBAT = family-based association test.
aUCSC GRCh37/hg19 coordinates.
10 Journal of Attention Disorders 00(0)
is the first report describing an association between genetic
variants within these genes and the severity of ADHD
symptoms in a population of predominantly African
ancestry.
Marker DRD4-rs916457 was previously associated to
ADHD susceptibility in the families evaluated in this study
(Puentes-Rozo et al., 2019) and in families from the Paisa
genetic isolate from Antioquia, Colombia (Arcos-Burgos,
Castellanos, Konecki, et al., 2004). Here, this variant was
found to be associated with the severity of global, hyperac-
tivity and impulsivity symptoms (Table 2), and an impor-
tant predictor of symptom severity in our predictive
genomics framework (Table 3 and Figure 3). The role of the
DRD4 in ADHD etiology has been studied extensively
(DiMaio et al., 2003). Variants in this gene are an important
predictor of preschool aggression and a moderator of family
environmental effects, stressful life events, and inattention
severity among adults with ADHD, as well as parental and
youth ADHD outcomes (Farbiash et al., 2014; Martel et al.,
2011; Nikolas & Momany, 2017; Sanchez-Mora et al.,
2015). Furthermore, DRD4 variants have been associated
with ADHD symptom severity (Tabatabaei et al., 2017;
Tahir et al., 2000; Tovo-Rodrigues et al., 2013) and play a
significant role in the default mode, executive control and
sensorimotor networks in children with ADHD (Qian et al.,
2018). Altogether, these findings support the role of DRD4
not only in the etiology of ADHD, but also in the prediction
of ADHD symptom severity. These results contribute to our
understanding of the genetic basis of ADHD severity in this
understudied population.
SNAP25 has been extensively implicated in the etiology
of ADHD (Barr et al., 2000; Brophy et al., 2002; Feng et al.,
2005; Galvez et al., 2014; Gizer et al., 2009; Herken et al.,
2014; Kim et al., 2007). It plays an important role in the
synaptic function of specific neuronal systems. SNAP25 is
an essential component of the soluble N-ethylmaleimide-
sensitive factor attachment protein receptor (SNARE) com-
plex, which is involved in the exocytotic release of
neurotransmitters during synaptic transmission (Antonucci
et al., 2016). More recently, SNAP25 variants were reported
to be associated with working memory (Wang et al., 2018)
and neuropsychological performance in individuals with
Figure 3. Performance measures for ML algorithms used to construct predictive genomics models for ADHD symptom severity:
(a) Balanced accuracy based on the 10-fold cross validation (CV) procedure. Segments represent 95% confidence intervals. The best
performance was achieved by the Support Vector Machine (SVM) with polynomial kernel (svmPoly), linear discriminant analysis (lda),
gradient boosting machine (gbm) and Classification and Regression Tree (rpart2) ML algorithms when predicting global, inattention,
hyperactivity and impulsivity severity, respectively, (b) ROC curves for the 10-fold CV procedure and the training and testing data
sets. The 10-fold CV procedure was performed using five repetitions for each fold. The training and testing data sets data sets
consisted of 70% (n = 271) and 30% (n = 115) of the data, respectively, and (c) variable importance for the predictive models derived
in our cohort. A brief description of the ML algorithms is provided in the Supplementary Material.
Cervantes-Henríquez et al. 11
ADHD (Kim et al., 2017) Evidence also suggests that
SNAP25 is differentially expressed in the prefrontal cortex
of animal models of ADHD and of children with ADHD
when compared to controls (Li et al., 2009). Having previ-
ously identified that marker SNAP25-rs362990 conferred
susceptibility to ADHD (Puentes-Rozo et al., 2019) and
finding that this same variant is associated with the severity
of inattention and impulsivity symptoms (Table 2) expands
our understating of the role of SNAP25 in ADHD etiology.
Variable importance analyzes revealed that this marker is an
important predictor of severity latent phenotypes when
included in the ML-based predictive framework (Figure 3
and Table 2). The fact that a SNAP25 variant is associated
with impulsivity symptoms severity in our study supports
the findings by Nemeth et al. (2013), who reported an asso-
ciation between impulsivity and a polymorphic microRNA
binding site on the SNAP25 3’UTR region. Furthermore,
considering that our sample exhibits ADHD predominantly
of inattentive and impulsivity component (Cervantes-
Henriquez et al., 2018; Pineda et al., 2016; Puentes-Rozo
et al., 2019), our findings support the role of genetics in
influencing the severity of these symptoms, at least in indi-
viduals from this Caribbean population.
Variant ADGRL3-rs2122642 was previously associated
with increased ADHD susceptibility in our cohort and is
harbored in a susceptibility haplotype formed by markers
rs1565902, rs10001410, and rs2122642 (OR = 1.74,
Ppermuted = 0.021) (Puentes-Rozo et al., 2019). Variants in
ADGRL3, a member of the latrophilin family of adhesion G
protein-coupled receptors (Martinez et al., 2011; Moreno-
Salinas et al., 2019), increase the risk of developing ADHD,
externalizing disorders, including CD, ODD, and SUD, and
Table 3. Performance Measures for ML-Based Predictive Models of ADHD Severity Based on Demographic and Genetic Data.
Measure Procedureb
Severity
Global Inattention Hyperactivity Impulsivity
Confusion
matrixa
10-Fold CV 84/30/32/124 116/41/39/74 21/10/42/197 63/29/38/141
Training 85/32/29/125 119/37/38/77 22/41/9/199 64/37/29/142
Testing 34/16/11/54 51/15/22/27 10/17/8/80 31/11/14/58
Se10-Fold CV 0.724 0.748 0.333 0.624
Training 0.746 0.758 0.710 0.688
Testing 0.756 0.699 0.556 0.689
Sp10-Fold CV 0.805 0.643 0.952 0.829
Training 0.796 0.675 0.829 0.793
Testing 0.771 0.643 0.825 0.841
PPV 10-Fold CV 0.737 0.739 0.677 0.685
Training 0.726 0.763 0.349 0.634
Testing 0.680 0.773 0.370 0.738
NPV 10-Fold CV 0.795 0.655 0.824 0.788
Training 0.812 0.670 0.957 0.830
Testing 0.831 0.551 0.909 0.806
FDR 10-Fold CV 0.263 0.261 0.323 0.315
Training 0.274 0.237 0.651 0.366
Testing 0.320 0.227 0.630 0.262
FPR 10-Fold CV 0.195 0.357 0.048 0.171
Training 0.204 0.325 0.171 0.207
Testing 0.229 0.357 0.175 0.159
Accuracy 10-Fold CV 0.770 0.704 0.807 0.753
Training 0.775 0.723 0.815 0.757
Testing 0.765 0.678 0.783 0.781
AUC 10-Fold CV 0.765 0.696 0.643 0.727
Training 0.771 0.717 0.769 0.741
Testing 0.763 0.671 0.690 0.765
CV = cross-validation; Se = sensitivity; Sp = specificity; PPV = positive predictive value; NPV = negative predictive value; FDR = false discovery rate;
FPR = false positives rate; AUC = area under the ROC curve.
aThe confusion matrix is formatted as i/j/k/l, where i is the number of severe individuals correctly classified, j is the number of severe individuals classi-
fied as non-severe, k corresponds to non-severe individuals classified as severe, and l to the number of non-severe individuals correctly classified.
bThe 10-fold CV procedure was performed using 5 repetitions for each fold based on 70% of the data (training data set; n = 271 on average). Testing
was performed using the remaining 30% (testing data set; n = 115 on average). See methods for more information.
12 Journal of Attention Disorders 00(0)
adverse long-term outcomes (Acosta et al., 2016; Arcos-
Burgos et al., 2010, 2012, 2019; Jain et al., 2007, 2011).
Finding that ADGRL3-rs2122642 confers susceptibility to
more severe hyperactivity and impulsivity symptoms and
that ADGRL3-rs10001410 is associated with an increased
severity of global symptoms (Table 2 and Figure 3) high-
lights the importance of ADGRL3 as a pivotal gene in
ADHD. Although this effect has previously been shown in
the Paisa genetic isolate (Jain et al., 2007), this is the first
time it is reported for a community with a predominantly
African genetic background.
Despite our encouraging results and the possibility of
rapidly applying our findings in the clinical setting, some
limitations need to be acknowledged. First, it is frequent in
this culture to find unmedicated ADHD children.
Consequently, neither children nor adults included in this
study were medicated because they were untreated inciden-
tal cases. Although it may be possible to observe the same
medication pattern in other cultures, this is not necessarily
the norm and imposes some restrictions when extrapolating
our results to the general population. In the same vein, a
second limitation is the representativeness of the sample
across different age groups due to the family-based design
utilized for recruiting the sample. In our case, 94/386 indi-
viduals were children, 34/386 adolescents and 232/386
adults, and age ranged between 6 and 60 years, which indi-
cates that individuals are not uniformly distributed across
age groups. Thirdly, the use of the same assessment meth-
ods across age groups. We clinically assessed all individuals
in our genetic studies of ADHD using a multimodal
approach (Cervantes-Henriquez et al., 2018; Palacio et al.,
2004; Pineda et al., 2016). However, we are aware that
using the same assessment methods across age groups may
not be ideal. Thus, future studies may use different or com-
plementary assessment strategies across age groups, espe-
cially in order groups, as some participants may have
difficulties with episodic memory and hence may not be
able to assess ADHD symptoms retrospectively. A fourth
limitation is the use of related individuals in the LCCA clus-
ter analysis. Even though clustering methods designed for
related individuals exist, LCCA methods for dissecting hid-
den structures was initially proposed for unrelated individu-
als. While we used each individual's family membership to
reduce the impact of individuals' relatedness in the resulting
clusters derived by LCCA, the use of other clustering meth-
ods for identifying complex structures in data with related
individuals is yet to be explored. Finally, our sample is
comprised of nuclear families characterized by a predomi-
nantly African ancestry, which may represent both a limita-
tion and a strength of this study. It would be interesting to
compare the severity patterns in our cohort and those
reported in other populations around the world.
In summary, our study supports the role of the DRD4,
SNAP25, and ADGRL3 genes in the etiology of ADHD
severity in a Caribbean community of predominantly
African ancestry. The identification of variants within these
genes previously reported to confer susceptibility to ADHD
sheds light into the genetic basis of ADHD severity, espe-
cially in this understudied population. We argue that future
genetic studies of ADHD may greatly benefit from our find-
ings. In fact, such studies could use the severity of symp-
toms to elucidate how individuals with symptoms in
different domains—inattention, hyperactivity, impulsiv-
ity—perform neuropsychological tasks (i.e., measures of
working memory, cognitive function, and executive func-
tion) (Jimenez-Figueroa et al., 2017; Pineda-Alhucema
et al., 2018; Suarez et al., 2020), and to determine the con-
tribution to ADHD severity of genetic variants that confer
susceptibility in Caucasian populations (Demontis et al.,
2019). Although these candidate variants have not been rep-
licated in genome-wide analyzes, their utility as risk vari-
ants for ADHD needs further investigation. In addition,
these complex severity phenotypes may be used to identify
at-risk populations using ML approaches (Table 3 and
Figure 3) or genetic markers already identified to confer
susceptibility to ADHD via GWAS. In the latter case, ML
methods (i.e., clustering and dimensionality reduction tech-
niques) may be applied to identify individuals with severe
and subtle forms of ADHD based on clinical information.
Further, such ML-derived phenotypes may be used as the
trait of interest in future genetic association studies either
by evaluating the partial contribution of each genetic marker
or by calculating polygenic risk scores (Du Rietz et al.,
2018). Ultimately, the combination of clinical data, genetic
association analyzes and ML techniques may contribute to
elucidate the molecular basis of ADHD symptomatology
utilizing a multi-omics approach, and the development of
predictive tools with potential clinical application (Tenev
et al., 2014; Vahid et al., 2019).
Acknowledgments
We express our highest sentiment of appreciation to all families
enrolled in this study.
Declaration of Conflicting Interests
The author(s) declared no potential conflicts of interest with
respect to the research, authorship, and/or publication of this
article.
Funding
The author(s) disclosed receipt of the following financial support
for the research, authorship, and/or publication of this article: This
study was financed by COLCIENCIAS, project “Fenotipos
Complejos y Endofenotipos del Trastorno por Déficit de Atención
e Hiperactividad y su Asociación con Genes Mayores y de
Susceptibilidad”, grant 1253-5453-1644, contract RC 384-2011,
awarded to Grupo de Neurociencias del Caribe, Universidad
Simón Bolívar, Barranquilla. M.L.C.H received a PhD scholarship
Cervantes-Henríquez et al. 13
from Universidad Simón Bolívar, Barranquilla; M.L.C.H, J.E.A.L
and J.I.V were partially supported by research grant FOFICO
32101 PE0031 from Universidad del Norte. M.L.C.H is a doctoral
student at Universidad del Norte; some of this work is to be pre-
sented in partial fulfilment of the requirements for the PhD degree.
ORCID iDs
Johan E. Acosta-López https://orcid.org/0000-0002-7330-6356
Jorge I. Vélez https://orcid.org/0000-0002-3146-7899
Supplemental Material
Supplemental material for this article is available online.
References
Acosta López, J., Cervantes-Henríquez, M. L., Jimenez Figueroa,
G., Nuñez Barragan, M., Sanchez Rojas, M., & Puentes Rozo,
P. (2013). Uso de una escala comportamental Wender Utah
para evaluar en retrospectiva trastorno de atención-hiperac-
tividad en adultos de la ciudad de Barranquilla. Universidad
y Salud, 15(1), 45–61.
Acosta, M. T., Arcos-Burgos, M., & Muenke, M. (2004). Attention
deficit/hyperactivity disorder (ADHD): Complex phenotype,
simple genotype? Genetics in Medicine, 6(1), 1–15. https://
doi.org/10.1097/01.gim.0000110413.07490.0b
Acosta, M. T., Castellanos, F. X., Bolton, K. L., Balog, J. Z., Eagen,
P., Nee, L., Jones, J., Palacio, L., Sarampote, C., Russell, H.
F., Berg, K., Arcos-Burgos, M., & Muenke, M. (2008). Latent
class subtyping of attention-deficit/hyperactivity disorder
and comorbid conditions. Journal of American Academy of
Child and Adolescent Psychiatry, 47(7), 797–807. https://doi.
org/10.1097/CHI.0b013e318173f70b
Acosta, M. T., Swanson, J., Stehli, A., Molina, B. S., Team, M. T.
A., Martinez, A. F., Arcos-Burgos, M., & Muenke, M. (2016).
ADGRL3 (LPHN3) variants are associated with a refined
phenotype of ADHD in the MTA study. Molecular Genetics
& Genomic Medicine, 4(5), 540–547. https://doi.org/10.1002/
mgg3.230
Acosta, M. T., Velez, J. I., Bustamante, M. L., Balog, J. Z., Arco-
Burgos, M., & Muenke, M. (2011). A two-locus genetic inter-
action between LPHN3 and 11q predicts ADHD severity and
long-term outcome. Translational Psychiatry, 1(7), e17.
Anastopoulos, A. D., Guevremont, D. C., Shelton, T. L., &
DuPaul, G. J. (1992). Parenting stress among families of
children with attention deficit hyperactivity disorder. Journal
of Abnormal Child Psychology, 20(5), 503–520. http://doi.
org/10.1007/bf00916812
Andrews, G., Pine, D. S., Hobbs, M. J., Anderson, T. M., &
Sunderland, M. (2009). Neurodevelopmental disorders:
Cluster 2 of the proposed meta-structure for DSM-V and
ICD-11. Psychological Medicine, 39(12), 2013–2023. https://
doi.org/:10.1017/S0033291709990274
Antonucci, F., Corradini, I., Fossati, G., Tomasoni, R., Menna,
E., & Matteoli, M. (2016). SNAP-25, a known presynap-
tic protein with emerging postsynaptic functions. Frontiers
in Synaptic Neuroscience, 8, 7. https://doi.org/10.3389/
fnsyn.2016.00007
Arcos-Burgos, M., Castellanos, F. X., Konecki, D., Lopera, F.,
Pineda, D., Palacio, J. D., Rapoport, J. L., Berg, K., Bailey-
Wilson, J., & Muenke, M. (2004). Pedigree disequilibrium
test (PDT) replicates association and linkage between DRD4
and ADHD in multigenerational and extended pedigrees
from a genetic isolate. Molecular Psychiatry, 9(3), 252–259.
https://doi.org/10.1038/sj.mp.4001396
Arcos-Burgos, M., Castellanos, F. X., Pineda, D., Lopera, F.,
Palacio, J. D., Palacio, L. G., Rapoport, J. L., Berg, K.,
Bailey-Wilson, J., & Muenke, M. (2004). Attention-deficit/
hyperactivity disorder in a population isolate: Linkage
to loci at 4q13.2, 5q33.3, 11q22, and 17p11. American
Journal of Human Genetics, 75(6), 998–1014. https://doi.
org/10.1086/426154
Arcos-Burgos, M., Jain, M., Acosta, M. T., Shively, S., Stanescu,
H., Wallis, D., Velez, J. I., Karkera, J., Balog, J., Berg,
K., Kleta, R., Gahl, W. A., Roessler, E., Long, R., Lie, J.,
Pineda, D. A., Londo, A. C., Palacio, J. D., Arbelaez, A., . . .
Muenke, M. (2010). A common variant of the latrophilin 3
gene, LPHN3, confers susceptibility to ADHD and predicts
effectiveness of stimulant medication. Molecular Psychiatry,
15(11), 1053–1066. https://doi.org/10.1038/mp.2010.6
Arcos-Burgos, M., & Muenke, M. (2010). Toward a better under-
standing of ADHD: LPHN3 gene variants and the suscepti-
bility to develop ADHD. Attention Deficit and Hyperactivity
Disorders, 2(3), 139–147. https://doi.org/10.1007/s12402-
010-0030-2
Arcos-Burgos, M., Velez, J. I., Martinez, A. F., Ribases, M.,
Ramos-Quiroga, J. A., Sanchez-Mora, C., Richarte, V.,
Roncero, C., Cormand, B., Fernández-Castillo, N., Casas, M.,
Lopera, F., Pineda, D. A., Palacio, J. D., Acosta-López, J. E.,
Cervantes-Henriquez, M. L., Sánchez-Rojas, M. G., Puentes-
Rozo, P. J., Molina, B. S. G., . . . Muenke, M. (2019).
ADGRL3 (LPHN3) variants predict substance use disorder.
Translational Psychiatry, 9(1), 42. doi:10.1038/s41398-019-
0396-7
Arcos-Burgos, M., Velez, J. I., Solomon, B. D., & Muenke, M.
(2012). A common genetic network underlies substance use
disorders and disruptive or externalizing disorders. Human
Genetics, 131(6), 917–929. https://doi.org/10.1007/s00439-
012-1164-4
Asherson, P., & Trzaskowski, M. (2015). Attention-deficit/hyper-
activity disorder is the extreme and impairing tail of a contin-
uum. Journal of the American Academy of Child Adolescent
Psychiatry, 54(4), 249–250. https://doi.org/10.1016/j.
jaac.2015.01.014
Association, A. P. (2002a). Diagnostic and statistical manual of
mental disorders (DSM-IV) (4th ed.). American Psychiatric
Association.
Association, A. P. (2002b). Manual diagnóstico y estadístico de
los trastornos mentales (DSM-IV-TR) (5th ed.). Masson.
Association, A. P. (2013). Diagnostic and statistical manual of
mental disorders (DSM-V) (5th ed.). American Psychiatric
Association.
Barr, C. L., Feng, Y., Wigg, K., Bloom, S., Roberts, W., Malone,
M., Schachar, R., Tannock, R., & Kennedy, J. L. (2000).
Identification of DNA variants in the SNAP-25 gene and
linkage study of these polymorphisms and attention-deficit
14 Journal of Attention Disorders 00(0)
hyperactivity disorder. Molecular Psychiatry, 5(4), 405-409.
doi:10.1038/sj.mp.4000733
Barragán-Duarte, J. L. (2007). Mapa genético de los colombianos.
UN Periódico.
Benjamini, Y., & Hochberg, Y. (1995). Controlling the false dis-
covery rate: A practical and powerful approach to multiple
testing. Journal of the Royal Statistical Society. Series B
(Methodological), 57(1), 289–300.
Breiman, L. (2001). Random forests. In R. E. Schapire (Ed.),
Machine learning (Vol. 45, pp. 5–32). Kluwer Academic
Publishers.
Breiman, L., Friedman, J. H., Olshen, R. A., & Stone, C. H. (1984).
Classification and regression trees. Wadsworth International
Group, Inc.
Brookes, K., Xu, X., Chen, W., Zhou, K., Neale, B., Lowe, N.,
Anney, R., Franke, B., Gill, M., Ebstein, R., Buitelaar, J.,
Sham, P., Campbell, D., Knight, J., Andreou, P., Altink, M.,
Arnold, R., Boer, F., Buschgens, C., . . . Asherson, P. (2006).
The analysis of 51 genes in DSM-IV combined type attention
deficit hyperactivity disorder: Association signals in DRD4,
DAT1 and 16 other genes. Molecular Psychiatry, 11(10),
934–953. https://doi.org/10.1038/sj.mp.4001869
Brophy, K., Hawi, Z., Kirley, A., Fitzgerald, M., & Gill, M. (2002).
Synaptosomal-associated protein 25 (SNAP-25) and attention
deficit hyperactivity disorder (ADHD): Evidence of linkage
and association in the Irish population. Molecular Psychiatry,
7(8), 913–917. https://doi.org/10.1038/sj.mp.4001092
Brown, N. M., Brown, S. N., Briggs, R. D., German, M.,
Belamarich, P. F., & Oyeku, S. O. (2017). Associations
between adverse childhood experiences and ADHD diagnosis
and severity. Academic Pediatrics, 17(4), 349–355. https://
doi.org/10.1016/j.acap.2016.08.013
Bruxel, E. M., Moreira-Maia, C. R., Akutagava-Martins, G. C.,
Quinn, T. P., Klein, M., Franke, B., Ribasés, M., Rovira,
P., Sánchez-Mora, C., Kappel, D., Mota, N. R., Grevet,
E. H., Bau, C. H. D., Arcos-Burgos, M., Rohde, L. A., &
Hutz, M. H. (2020). Meta-analysis and systematic review of
ADGRL3 (LPHN3) polymorphisms in ADHD susceptibility.
Moleculaar Psychiatry. Advance online publication. https://
doi.org/10.1038/s41380-020-0673-0
Casey, B. J., Oliveri, M. E., & Insel, T. (2014). A neurodevelop-
mental perspective on the research domain criteria (RDoC)
framework. Biological Psychiatry, 76(5), 350–353. https://
doi.org/10.1016/j.biopsych.2014.01.006
Cervantes-Henriquez, M. L., Acosta-Lopez, J. E., Martinez-Banfi,
M. L., Velez, J. I., Mejia-Segura, E., Lozano-Gutierrez,
S. G., Sanchez-Rojas, M., Zurbaran, M. A., Zurek, E. E.,
Arcos-Burgos, M., Pineda, D. A., & Puentes-Rozo, P. J.
(2018). ADHD endophenotypes in Caribbean families.
Journal of Attention Disorders, 24(24), 1087054718763741.
doi:10.1177/1087054718763741
Chen, T., & Guestrin, C. (2016). XGBoost: A scalable tree boost-
ing system [Paper presentation]. 22nd SIGKDD Conference
on Knowledge Discovery and Data Mining. Association
for Computer Machinery (ACM). https://arxiv.org/
abs/1603.02754
Chen, T., He, T., Benesty, M., Khotilovich, V., Tang, Y., Cho, H.,
Chen, K., Mitchell, R., Cano, I., Zhou, T., Li, M., Xie, J., Lin,
M., Geng, Y., & Li, Y. (2020). xgboost: Extreme Gradient
Boosting. R package version 1.0.0.2. https://CRAN.R-project.
org/package=xgboost.
Cornejo, J. W., Osío, O., Sánchez, Y., Carrizosa, J., Sánchez,
G., Grisales, H., Castillo-Parra, H., & Holguín, J. (2005).
Prevalence of attention deficit hyperactivity disorder in
Colombian children and teenagers. Revista de Neurología,
40(12), 716–722.
Cortes, C., & Vapnik, V. (1995). Support-vector networks.
Machine Learning, 20, 273–297. https://doi.org/10.1007/
BF00994018
Curran, S., Mill, J., Sham, P., Rijsdijk, F., Marusic, K., Taylor,
E., & Asherson, P. (2001). QTL association analysis of the
DRD4 exon 3 VNTR polymorphism in a population sample
of children screened with a parent rating scale for ADHD
symptoms. American Journal of Medical Genetics, 105(4),
387–393. https://doi.org/10.1002/ajmg.1366
Cuthbert, B., & Insel, T. (2010). The data of diagnosis: New
approaches to psychiatric classification. Psychiatry, 73(4),
311–314. https://doi.org/10.1521/psyc.2010.73.4.311
Demontis, D., Walters, R. K., Martin, J., Mattheisen, M., Als,
T. D., Agerbo, E., Baldursson, G., Beliveau, R., Bybjerg-
Grauholm, J., Baekvad-Hansen, M., Cerrato, F., Chambert,
K., Churchhouse, C., Dumont, A., Eriksson, N., Gandal,
M., Goldstein, J. I., Grasby, K. L., Grove, J., . . . Neale, B.
M. (2019). Discovery of the first genome-wide significant
risk loci for attention deficit/hyperactivity disorder. Nature
Genetics, 51(1), 63–75. https://doi.org/10.1038/s41588-018-
0269-7
DiMaio, S., Grizenko, N., & Joober, R. (2003). Dopamine genes
and attention-deficit hyperactivity disorder: A review. Journal
of Psychiatry Neuroscience, 28(1), 27–38.
DSM-IV. (2002). Manual diagnóstico y estadístico de los trastor-
nos mentales: Texto revisado. Masson.
Du Rietz, E., Coleman, J., Glanville, K., Choi, S. W., O’Reilly,
P. F., & Kuntsi, J. (2018). Association of polygenic risk
for attention-deficit/hyperactivity disorder with co-occur-
ring traits and disorders. Biological Psychiatry: Cognitive
Neuroscience and Neuroimaging, 3(7), 635–643. https://doi.
org/10.1016/j.bpsc.2017.11.013
Ezzati, M., Friedman, A. B., Kulkarni, S. C., & Murray, C. J.
(2008). The reversal of fortunes: Trends in county mortality
and cross-county mortality disparities in the United States.
PLoS Medicine, 5(4), e66. https://doi.org/10.1371/journal.
pmed.0050066
Faraone, S. V., & Biederman, J. (2005). What is the prevalence of
adult ADHD? Results of a population screen of 966 adults.
Journal of Attention Disorders, 9(2), 384–391. https://doi.
org/10.1177/1087054705281478
Faraone, S. V., & Larsson, H. (2019). Genetics of attention defi-
cit hyperactivity disorder. Molecular Psychiatry, 24(4), 562–
575. https://doi.org/10.1038/s41380-018-0070-0
Faraone, S. V., & Mick, E. (2010). Molecular genetics of atten-
tion deficit hyperactivity disorder. The Psychiatric Clinics
of North America, 33(1), 159–180. https://doi.org/10.1016/j.
psc.2009.12.004
Farbiash, T., Berger, A., Atzaba-Poria, N., & Auerbach, J. G.
(2014). Prediction of preschool aggression from DRD4
Cervantes-Henríquez et al. 15
risk, parental ADHD symptoms, and home chaos. Journal
of Abnormal Child Psychology, 42(3), 489–499. https://doi.
org/10.1007/s10802-013-9791-3
Feng, Y., Crosbie, J., Wigg, K., Pathare, T., Ickowicz, A., Schachar,
R., Tannock, R., Roberts, W., Malone, M., Swanson, J.,
Kennedy, J. L., & Barr, C. L. (2005). The SNAP25 gene as
a susceptibility gene contributing to attention-deficit hyper-
activity disorder. Molecular Psychiatry, 10(11), 998–1005.
https://doi.org/10.1038/sj.mp.4001722
Fernandez-Jaen, A., Martin Fernandez-Mayoralas, D., Fernandez-
Perrone, A. L., Jimenez, A., Albert, J., Lopez-Martin, S.,
Calleja-Perez, B., Tirado, P., & Lopez-Arribas, S. (2018).
[Neurodevelopment and phenocopies of attention deficit
hyperactivity disorder: differential diagnosis]. Revista de
Neurologia, 66(S01), S103–S107.
Fletcher, J., & Wolfe, B. (2009). Long-term consequences of
childhood ADHD on criminal activities. The Journal of
Mental Health Policy and Economics, 12(3), 119–138.
Franke, B., Vasquez, A. A., Johansson, S., Hoogman, M., Romanos,
J., Boreatti-Hummer, A., Heine, M., Jacob, C. P., Lesch, K.
P., Casas, M., Ribasés, M., Bosch, R., Sánchez-Mora, C.,
Barros, N. G., Fernàndez-Castillo, N., Bayes, M., Halmøy,
A., Barone, H., Landaas, E. T., & Reif, A. (2010). Multicenter
analysis of the SLC6A3/DAT1 VNTR haplotype in persis-
tent ADHD suggests differential involvement of the gene in
childhood and persistent ADHD. Neuropsychopharmacology,
35(3), 656–664. https://doi.org/10.1038/npp.2009.170
Galvez, J. M., Forero, D. A., Fonseca, D. J., Mateus, H. E., Talero-
Gutierrez, C., & Velez-van-Meerbeke, A. (2014). Evidence
of association between SNAP25 gene and attention deficit
hyperactivity disorder in a Latin American sample. Attention
Deficit Hyperactive Disorders, 6(1), 19–23. https://doi.
org/10.1007/s12402-013-0123-9
Gizer, I. R., Ficks, C., & Waldman, I. D. (2009). Candidate gene
studies of ADHD: A meta-analytic review. Human Genetics,
126(1), 51–90. https://doi.org/10.1007/s00439-009-0694-x
Graziano, P. A., McNamara, J. P., Geffken, G. R., & Reid, A.
(2011). Severity of children's ADHD symptoms and parenting
stress: A multiple mediation model of self-regulation. Journal
of Abnormal Child Psychology, 39(7), 1073–1083. https://doi.
org/10.1007/s10802-011-9528-0
Helfer, B., Cooper, R. E., Bozhilova, N., Maltezos, S., Kuntsi, J.,
& Asherson, P. (2019). The effects of emotional lability, mind
wandering and sleep quality on ADHD symptom severity in
adults with ADHD. European Psychiatry, 55, 45–51. https://
doi.org/10.1016/j.eurpsy.2018.09.006
Herken, H., Erdal, M. E., Kenar, A. N., Unal, G. A., Cakaloz, B.,
Ay, M. E., Yücel, E., Edgünlü, T. G., & Sengul, C. (2014).
Association of SNAP-25 gene Ddel and Mnll polymorphisms
with adult attention deficit hyperactivity disorder. Psychiatry
Investigation, 11(4), 476–480. https://doi.org/10.4306/
pi.2014.11.4.476
Insel, T. R. (2014). The NIMH Research Domain Criteria
(RDoC) project: Precision medicine for psychiatry. The
American Journal of Psychiatry, 171(4), 395–397. https://doi.
org/10.1176/appi.ajp.2014.14020138
Jain, M., Palacio, L. G., Castellanos, F. X., Palacio, J. D., Pineda,
D., Restrepo, M. I., Muñoz, J. F., Lopera, F., Wallis, D., Berg,
K., Bailey-Wilson, J. E., Arcos-Burgos, M., & Muenke, M.
(2007). Attention-deficit/hyperactivity disorder and comorbid
disruptive behavior disorders: Evidence of pleiotropy and new
susceptibility loci. Biological Psychiatry, 61(12), 1329–1339.
https://doi.org/10.1016/j.biopsych.2006.06.026
Jain, M., Velez, J. I., Acosta, M. T., Palacio, L. G., Balog, J.,
Roessler, E., Pineda, D., Londono, A. C., Palacio, J. D.,
Arbelaez Lopera, F., Elia, J., Hakonarson, H., Seitz, C.,
Freitag, C. M., Palmason, H., Meyer, J., Romanos, M.,
Renner, T., & Muenke, M. (2011). A cooperative inter-
action between LPHN3 and 11q doubles the risk for
ADHD. Molecular Psychiatry, 17(7), 741–747. https://doi.
org/10.1038/mp.2011.59
Jimenez-Figueroa, G., Ardila-Duarte, C., Pineda, D. A., Acosta-
Lopez, J. E., Cervantes-Henriquez, M. L., Pineda-Alhucema,
W., Cervantes-Gutiérrez, J., Quintero-Ibarra., M., Sánchez-
Rojas, M., Vélez, J. I., & Puentes-Rozo, P. J. (2017).
Prepotent response inhibition and reaction times in children
with attention deficit/hyperactivity disorder from a Caribbean
community. Attention Deficit and Hyperactive Disorders,
9(4), 199-211. doi:10.1007/s12402-017-0223-z
Kautzky, A., Vanicek, T., Philippe, C., Kranz, G. S., Wadsak,
W., Mitterhauser, M., Hartmann, A., Hahn, A., Hacker, M.,
Rujescu, D., Kasper, S., & Lanzenberger, R. (2020). Machine
learning classification of ADHD and HC by multimodal sero-
tonergic data. Translational Psychiatry, 10(1), 104. https://
doi.org/10.1038/s41398-020-0781-2
Kim, E., Song, D. H., Kim, N. W., Sohn, I. J., & Cheon, K. A.
(2017). The relationship between the SNAP-25 polymor-
phism and omission errors in Korean children with attention
deficit hyperactivity disorder. Clinical Psychopharmacology
Neuroscience, 15(3), 222–228. https://doi.org/10.9758/
cpn.2017.15.3.222
Kim, J. W., Biederman, J., Arbeitman, L., Fagerness, J., Doyle,
A. E., Petty, C., Perlis, R. H., Purcell, S., Smoller, J. W.,
Faraone, S. V., & Sklar, P. (2007). Investigation of variation
in SNAP-25 and ADHD and relationship to co-morbid major
depressive disorder. American Journal of Medical Genetics
B Neuropsychiatric Genetics, 144B(6), 781–790. https://doi.
org/10.1002/ajmg.b.30522
Korsgaard, H. O., Torgersen, S., Wentzel-Larsen, T., & Ulberg, R.
(2016). Personality disorders and axis I comorbidity in ado-
lescent outpatients with ADHD. BMC Psychiatry, 16, 175.
https://doi.org/10.1186/s12888-016-0871-0
Kotte, A., Faraone, S. V., & Biederman, J. (2013). Association
of genetic risk severity with ADHD clinical characteristics.
American Journal of Medical Genetics B Neuropsychiatric
Genetics, 162B(7), 718–733. https://doi.org/10.1002/
ajmg.b.32171
Kunh, M. (2020). Caret: Classification and regression training.
(Version R package version 6.0-86.) https://CRAN.R-project.
org/package=caret
Kupper, T., Haavik, J., Drexler, H., Ramos-Quiroga, J. A.,
Wermelskirchen, D., Prutz, C., & Schauble, B. (2012). The
negative impact of attention-deficit/hyperactivity disorder on
occupational health in adults and adolescents. International
Archives of Occupational Environmental Health, 85(8), 837–
847. https://doi.org/10.1007/s00420-012-0794-0
16 Journal of Attention Disorders 00(0)
Laird, N. M., Horvath, S., & Xu, X. (2000). Implementing
a unified approach to family-based tests of associa-
tion. Genetic Epidemiology, 19(Suppl 1), S36–S42.
https://doi.org/10.1002/1098-2272(2000)19:1+<::AID-
GEPI6>3.0.CO;2-M
Lange, C., DeMeo, D., Silverman, E. K., Weiss, S. T., & Laird, N.
M. (2004). PBAT: Tools for family-based association stud-
ies. American Journal of Human Genetics, 74(2), 367–369.
https://doi.org/10.1086/381563
Lange, C., & Laird, N. M. (2002a). On a general class of con-
ditional tests for family-based association studies in genet-
ics: The asymptotic distribution, the conditional power, and
optimality considerations. Genetic Epidemiology, 23(2), 165–
180. https://doi.org/10.1002/gepi.209
Lange, C., & Laird, N. M. (2002b). Power calculations for a gen-
eral class of family-based association tests: Dichotomous
traits. American Journal of Human Genetics, 71(3), 575–584.
https://doi.org/10.1086/342406
Larsson, H., Anckarsater, H., Rastam, M., Chang, Z., &
Lichtenstein, P. (2012). Childhood attention-deficit hyper-
activity disorder as an extreme of a continuous trait: A
quantitative genetic study of 8,500 twin pairs. Journal of
Child Psychology and Psychiatry, 53(1), 73–80. https://doi.
org/10.1111/j.1469-7610.2011.02467.x
Lee, T., Park, K. J., Lee, H. J., & Kim, H. W. (2020). Clinical and
neuropsychological characteristics of ADHD according to
DSM-5 age-of-onset criterion in Korean children and adoles-
cents. Journal of Attention Disorders, 24(1), 20–28. https://
doi.org/10.1177/1087054716684378
Li, Q., Wong, J. H., Lu, G., Antonio, G. E., Yeung, D. K., Ng,
T. B., Forster, L. E., & Yew, D. T. (2009). Gene expression
of synaptosomal-associated protein 25 (SNAP-25) in the pre-
frontal cortex of the spontaneously hypertensive rat (SHR).
Biochimica et Biophysica Acta, 1792(8), 766–776. https://doi.
org/10.1016/j.bbadis.2009.05.006
Lilienfeld, S. O., & Treadway, M. T. (2016). Clashing diagnos-
tic approaches: DSM-ICD versus RDoC. Annual Review of
Clinical Psychology, 12, 435–463. https://doi.org/10.1146/
annurev-clinpsy-021815-093122
Lunetta, K. L., Faraone, S. V., Biederman, J., & Laird, N. M.
(2000). Family-based tests of association and linkage that
use unaffected sibs, covariates, and interactions. American
Journal of Human Genetics, 66(2), 605–614. https://doi.
org/10.1086/302782
Luo, Y., Weibman, D., Halperin, J. M., & Li, X. (2019). A review
of heterogeneity in attention deficit/hyperactivity disorder
(ADHD). Frontiers in Human Neuroscience, 13, 42. https://
doi.org/10.3389/fnhum.2019.00042
Martel, M. M., Nikolas, M., Jernigan, K., Friderici, K., Waldman,
I., & Nigg, J. T. (2011). The dopamine receptor D4 gene
(DRD4) moderates family environmental effects on ADHD.
Journal of Abnormal Child Psychology, 39(1), 1–10. https://
doi.org/10.1007/s10802-010-9439-5
Martinez, A. F., Abe, Y., Hong, S., Molyneux, K., Yarnell, D.,
Lohr, H., Driever, W., Acosta, M. T., Arcos-Burgos, M.,
& Muenke, M. (2016). An ultraconserved brain-specific
enhancer within ADGRL3 (LPHN3) underpins attention-
deficit/hyperactivity disorder susceptibility. Biological
Psychiatry, 80(12), 943–954. https://doi.org/10.1016/j.bio-
psych.2016.06.026
Martinez, A. F., Muenke, M., & Arcos-Burgos, M. (2011). From
the black widow spider to human behavior: Latrophilins, a
relatively unknown class of G protein-coupled receptors,
are implicated in psychiatric disorders. American Journal
of Medical Genetics B Neuropsychiatric Genetics, 156B(1),
1–10. https://doi.org/10.1002/ajmg.b.31137
Mastronardi, C. A., Pillai, E., Pineda, D. A., Martinez, A. F.,
Lopera, F., Velez, J. I., Palacio, J. D., Patel, H., Easteal, S.,
Acosta, M. T., Castellanos, F. X., Muenke, M., & Arcos-
Burgos, M. (2016). Linkage and association analysis of
ADHD endophenotypes in extended and multigenerational
pedigrees from a genetic isolate. Molecular Psychiatry,
21(10), 1434–1440. https://doi.org/10.1038/mp.2015.172
Metz, C. E. (1978). Basic principles of ROC analysis. Seminars
in Nuclear Medicine, 8(4), 283–298. https://doi.org/10.1016/
s0001-2998(78)80014-2
Molina, B. S., Pelham, W. E., Gnagy, E. M., Thompson, A. L., &
Marshal, M. P. (2007). Attention-deficit/hyperactivity disor-
der risk for heavy drinking and alcohol use disorder is age spe-
cific. Alcoholism, Clinical and Experimental Research, 31(4),
643–654. https://doi.org/10.1111/j.1530-0277.2007.00349.x
Moreno-Salinas, A. L., Avila-Zozaya, M., Ugalde-Silva, P.,
Hernandez-Guzman, D. A., Missirlis, F., & Boucard, A. A.
(2019). Latrophilins: A neuro-centric view of an evolutionary
conserved adhesion G protein-coupled receptor subfamily.
Frontiers in Neuroscience, 13, 700. https://doi.org/10.3389/
fnins.2019.00700
Mowlem, F. D., Rosenqvist, M. A., Martin, J., Lichtenstein,
P., Asherson, P., & Larsson, H. (2018). Sex differences in
predicting ADHD clinical diagnosis and pharmacological
treatment. European Child & Adolescent Psychiatry, 28(4),
481–489. https://doi.org/10.1007/s00787-018-1211-3
Nemeth, N., Kovacs-Nagy, R., Szekely, A., Sasvari-Szekely, M.,
& Ronai, Z. (2013). Association of impulsivity and poly-
morphic microRNA-641 target sites in the SNAP-25 gene.
PLoS One, 8(12), e84207. https://doi.org/10.1371/journal.
pone.0084207
Nikolas, M. A., & Momany, A. M. (2017). DRD4 variants mod-
erate the impact of parental characteristics on child atten-
tion-deficit hyperactivity disorder: Exploratory evidence
from a multiplex family design. Journal of Abnormal Child
Psychology, 45(3), 429–442. https://doi.org/10.1007/s10802-
017-0264-y
Oerbeck, B., Overgaard, K., Pripp, A. H., Aase, H., Reichborn-
Kjennerud, T., & Zeiner, P. (2019). Adult ADHD symp-
toms and satisfaction with life: Does age and sex matter?
Journal of Attention Disorders, 23(1), 3–11. https://doi.
org/10.1177/1087054715612257
Owens, J., & Jackson, H. (2017). Attention-deficit/hyperactivity
disorder severity, diagnosis, & later academic achievement
in a national sample. Social Science Research, 61, 251–265.
https://doi.org/10.1016/j.ssresearch.2016.06.018
Palacio, J. D., Castellanos, F. X., Pineda, D. A., Lopera, F.,
Arcos-Burgos, M., Quiroz, Y. T., Henao, G. C., Puerta, I. C.,
Ramírez, D. L., Rapoport, J. L., Bailey-Wilson, J., Berg, K., &
Muenke, M. (2004). Attention-deficit/hyperactivity disorder
Cervantes-Henríquez et al. 17
and comorbidities in 18 Paisa Colombian multigenerational
families. Journal of American Academy of Child Adolescent
Psychiatry, 43(12), 1506–1515. https://doi.org10.1097/01.
chi.0000142279.79805.dc
Palacios-Cruz, L., Arias-Caballero, A., Ulloa, R. E., González-
Reyna, N., Mayer-Villa, P., Feria, M., Sosa, L., de La Pena, F.
R., Cabrera-Lagunes, A., Fragosa, A., Sepulveda, A., Cruz, C.
S., Peralta, P. C., Perez, J. C., Lara-Muñoz, M. C., & Benjet,
C. (2014). Adversidad psicosocial, psicopatología y funcio-
namiento en hermanos adolescentes en alto riesgo (HAR)
con y sin trastorno por déficit de atención con hiperactividad
(TDAH). Salud Mental, 37(6), 10.
Pineda, D., Acosta López, J., Cervantes-Henríquez, M. L.,
Jimenez-Figueroa, G. S.-R. M., Pineda-Alhucema, W., Mejía-
Segura, E., & Puentes-Rozo, P. J. (2016). Conglomerados de
clases latentes en 408 miembros de 120 familias nucleares de
Barranquilla con un caso índice afectado de trastorno de aten-
ción hiperactividad (TDAH). Acta Neurológica Colombiana,
32(4), 275–284.
Pineda, D. A., Henao, G. C., Puerta, I. C., Mejía-Mag, S. E.,
Gómez, L. F., Miranda, M. L., Rosselli, M., Ardila, A.,
Restrepo, M. A., & Murrelle, L. (1999). The usefulness of a
short questionnaire for the diagnosis of attention deficiency.
Revista de Neurología, 28(4), 365–372.
Pineda-Alhucema, W., Aristizabal, E., Escudero-Cabarcas, J.,
Acosta-Lopez, J. E., & Velez, J. I. (2018). Executive func-
tion and theory of mind in children with ADHD: A systematic
review. Neuropsychology Review, 28(3), 341–358. https://
doi.org/10.1007/s11065-018-9381-9
Polanczyk, G. V., Willcutt, E. G., Salum, G. A., Kieling, C., &
Rohde, L. A. (2014). ADHD prevalence estimates across
three decades: An updated systematic review and meta-
regression analysis. International Journal of Epidemiology,
43(2), 434–442. https://doi.org/10.1093/ije/dyt261
Posner, J., Polanczyk, G. V., & Sonuga-Barke, E. (2020).
Attention-deficit hyperactivity disorder. Lancet, 395(10222),
450–462. https://doi.org/10.1016/S0140-6736(19)33004-1
Puentes-Rozo, P. J., Acosta-Lopez, J. E., Cervantes-Henriquez, M.
L., Martinez-Banfi, M. L., Mejia-Segura, E., Sanchez-Rojas,
M., Anaya-Romero, M. E., Acosta-Hoyos, A., Garcia-Llinas,
G. A., Mastronardi, C. A., Pineda, D. A., Castellanos, F. X.,
Arcos-Burgos, M., & Velez, J. I. (2019). Genetic variation
underpinning ADHD risk in a Caribbean community. Cells,
8(8), 907. https://doi.org/10.3390/cells8080907
Qian, A., Tao, J., Wang, X., Liu, H., Ji, L., Yang, C., Ye, Q.,
Chen, C., Li, J., Cheng, J., Wang, M., & Zhao, K. (2018).
Effects of the 2-repeat allele of the DRD4 gene on neural net-
works associated with the prefrontal cortex in children with
ADHD. Frontiers in Human Neuroscience, 12, 279. https://
doi.org/10.3389/fnhum.2018.00279
R Core Team. (2019). R: A language and environment for sta-
tistical computing. R Foundation for Statistical Computing.
http://www.R-project.org
Rabinowitz, D., & Laird, N. (2000). A unified approach to
adjusting association tests for population admixture with
arbitrary pedigree structure and arbitrary missing marker
information. Human Heredity, 50(4), 211–223. https://doi.
org/10.1159/000022918
Rajendran, K., Rindskopf, D., O’Neill, S., Marks, D. J., Nomura,
Y., & Halperin, J. M. (2013). Neuropsychological function-
ing and severity of ADHD in early childhood: A four-year
cross-lagged study. Journal of Abnormal Psychology, 122(4),
1179–1188. https://doi.org/10.1037/a0034237
Ramtekkar, U. P., Reiersen, A. M., Todorov, A. A., & Todd, R.
D. (2010). Sex and age differences in attention-deficit/hyper-
activity disorder symptoms and diagnoses: Implications for
DSM-V and ICD-11. Journal of the American Academy of
Child and Adolescent Psychiatry, 49(3), 217–228 e211–213.
Ramy, H., El Sheikh, M., Sultan, M., Bassim, R., Eid, M., Ali,
R., & El Missiry, M. (2018). Risk factors influencing sever-
ity of attention deficit hyperactivity disorder in a sample
of preparatory school students in Cairo. Clinical Child
Psychology and Psychiatry, 23(4), 542–555. https://doi.
org/10.1177/1359104518755218
Reich, W. (2000). Diagnostic interview for children and ado-
lescents (DICA). Journal of the American Academy of
Child and Adolescent Psychiatry, 39(1), 59–66. https://doi.
org/10.1097/00004583-200001000-00017
Salazar, D. A., Vélez, J. I., & Salazar, J. C. (2012). Comparison
between SVM and logistic regression: Which one is better
to discriminate? Revista Colombiana de Estadistica, 35(2),
223–237.
Sanchez-Mora, C., Richarte, V., Garcia-Martinez, I., Pagerols, M.,
Corrales, M., Bosch, R., Vidal, R., Viladevall, L., Casas, M.,
Cormand, B., Ramos-Quiroga, J. A., & Ribases, M. (2015).
Dopamine receptor DRD4 gene and stressful life events in
persistent attention deficit hyperactivity disorder. American
Journal of Medical Genetics B Neuropsychiatric Genetics,
168(6), 480–491. https://doi.org/10.1002/ajmg.b.32340
Satterfield, J. H., Cantwell, D. P., & Satterfield, B. T. (1974).
Pathophysiology of the hyperactive child syndrome. Archives
of General Psychiatry, 31(6), 839–844.
Schachar, R., Mota, V. L., Logan, G. D., Tannock, R., & Klim,
P. (2000). Confirmation of an inhibitory control defi-
cit in attention-deficit/hyperactivity disorder. Journal of
Abnormal Child Psychology, 28(3), 227–235. https://doi.
org/10.1023/a:1005140103162
Shaw, M., Hodgkins, P., Caci, H., Young, S., Kahle, J., Woods, A.
G., & Arnold, L. E. (2012). A systematic review and analysis
of long-term outcomes in attention deficit hyperactivity disor-
der: Effects of treatment and non-treatment. BMC Medicine,
10, 99. https://doi.org/10.1186/1741-7015-10-99
Sibley, M. H., Pelham, W. E., Molina, B. S., Gnagy, E. M.,
Waschbusch, D. A., Biswas, A., MacLean, M. G., Babinski,
D. E., & Karch, K. M. (2011). The delinquency outcomes
of boys with ADHD with and without comorbidity. Journal
of Abnormal Child Psychology, 39(1), 21–32. https://doi.
org/10.1007/s10802-010-9443-9
Skogli, E. W., Teicher, M. H., Andersen, P. N., Hovik, K. T., & Oie,
M. (2013). ADHD in girls and boys—Gender differences in
co-existing symptoms and executive function measures. BMC
Psychiatry, 13, 298. https://doi.org/10.1186/1471-244X-13-298
Spielman, R. S., McGinnis, R. E., & Ewens, W. J. (1993).
Transmission test for linkage disequilibrium: The insulin
gene region and insulin-dependent diabetes mellitus (IDDM).
American Journal of Human Genetics, 52(3), 506–516.
18 Journal of Attention Disorders 00(0)
Suarez, I., De Los ReyesAragon, C., Diaz, E., Iglesias, T., Barcelo,
E., Velez, J. I., & Casini, L. (2020). How is temporal process-
ing affected in children with attention-deficit/hyperactivity
disorder? Developmental Neuropsychology, 45(4), 246–261.
https://doi.org/10.1080/87565641.2020.1764566
Suzuki, R., Terada, Y., & Shimodaira, H. (2019). Pvclust:
Hierarchical clustering with p-values via multiscale boot-
strap resampling. R package version 2.2-0. https://CRAN.R-
project.org/package=pvclust.
Tabatabaei, S. M., Amiri, S., Faghfouri, S., Noorazar, S. G.,
AbdollahiFakhim, S., & Fakhari, A. (2017). DRD4 gene poly-
morphisms as a risk factor for children with attention deficit
hyperactivity disorder in Iranian population. International
Scholarly Research Notices, 2017, 2494537. https://doi.
org/10.1155/2017/2494537
Tahir, E., Yazgan, Y., Cirakoglu, B., Ozbay, F., Waldman, I., &
Asherson, P. J. (2000). Association and linkage of DRD4 and
DRD5 with attention deficit hyperactivity disorder (ADHD)
in a sample of Turkish children. Molecular Psychiatry, 5(4),
396–404. https://doi.org/10.1038/sj.mp.4000744
Tandon, M., Tillman, R., Agrawal, A., & Luby, J. (2016).
Trajectories of ADHD severity over 10 years from childhood
into adulthood. Attention Deficit and Hyperactivity Disorders,
8(3), 121–130. https://doi.org/10.1007/s12402-016-0191-8
Tenev, A., Markovska-Simoska, S., Kocarev, L., Pop-Jordanov,
J., Muller, A., & Candrian, G. (2014). Machine learning
approach for classification of ADHD adults. International
Journal of Psychophysiology, 93(1), 162–166. https://doi.
org/10.1016/j.ijpsycho.2013.01.008
Thapar, A., Holmes, J., Poulton, K., & Harrington, R. (1999).
Genetic basis of attention deficit and hyperactivity. British
Journal of Psychiatry, 174, 105–111. https://doi.org/10.1192/
bjp.174.2.105
Thapar, A., Martin, J., Mick, E., Arias Vasquez, A., Langley,
K., Scherer, S. W., Schachar, R., Crosbie, J., Williams, N.,
Franke, B., Elia, J., Glessner, J., Hakonarson, H., Owen, M.
J., Faraone, S. V., O’Donovan, M. C., & Holmans, P. (2016).
Psychiatric gene discoveries shape evidence on ADHD’s
biology. Molecular Psychiatry, 21(9), 1202–1207. https://doi.
org/10.1038/mp.2015.163
Theule, J., Tannock, R., & Jenkins, J. M. (2013). Parenting stress
in families of children with ADHD: A meta-analysis. Journal
of Emotional and Behavioral Disorders, 21(1), 15.
Tishkoff, S. A., & Kidd, K. K. (2004). Implications of biogeog-
raphy of human populations for ‘race’ and medicine. Nature
Genetics, 36(11 Suppl), S21–S27. https://doi.org/10.1038/
ng1438
Tovo-Rodrigues, L., Rohde, L. A., Menezes, A. M., Polanczyk,
G. V., Kieling, C., Genro, J. P., Anselmi, L., & Hutz, M. H.
(2013). DRD4 rare variants in attention-deficit/hyperactivity
disorder (ADHD): Further evidence from a birth cohort study.
PLoS One, 8(12), e85164. https://doi.org/10.1371/journal.
pone.0085164
Vahid, A., Bluschke, A., Roessner, V., Stober, S., & Beste, C.
(2019). Deep learning based on event-related EEG differenti-
ates children with ADHD from healthy controls. Journal of
Clinical Medicine, 8(7). https://doi.org/10.3390/jcm8071055
Vélez, J. I., Correa, J. C., & Arcos-Burgos, M. (2014). A new
method for detecting significant p-values with applications
to genetic data. Revista Colombiana de Estadistica, 37(1),
67–76.
Venables, W. N., & Ripley, B. D. (2002). Modern applied statis-
tics with S (4th ed.). Springer
Vermunt, J., & Magidson, J. (2002). Latent class cluster analysis
In J. Haagenars & A. L. McCutcheon (Eds.), Applied latent
class analysis. Cambridge University Press.
Wang, C., Yang, B., Fang, D., Zeng, H., Chen, X., Peng, G.,
Cheng, Q., & Liang, G. (2018). The impact of SNAP25 on
brain functional connectivity density and working memory
in ADHD. Biological Psychology, 138, 35–40. https://doi.
org/10.1016/j.biopsycho.2018.08.005
Yasumura, A., Omori, M., Fukuda, A., Takahashi, J., Yasumura,
Y., Nakagawa, E., Koike, T., Yamashita, Y., Miyajima, T.,
Koeda, T., Aihara, M., Tachimori, H., & Inagaki, M. (2017).
Applied machine learning method to predict children with
ADHD using prefrontal cortex activity: A multicenter study in
Japan. Journal of Attention Disorders, 24, 1087054717740632.
https://doi.org/10.1177/1087054717740632
Author Biographies
Martha L. Cervantes-Henríquez is a psychologist and MSc in
Genetics from Universidad Simón Bolívar, Barranquilla, member
of the Grupo de Neurociencias del Caribe and Research Professor
at Psychology Department from the same institution. PhD student
in Biomedical Sciences at Universidad del Norte, Barranquilla,
Colombia. Her current research includes the genetics of ADHD,
and the development and application of bioinformatics tools to
underpin the genetic causes of this neuropsychiatric disorder.
Johan E. Acosta-López is a psychologist, MSc in
Neuropsychology and PhD student in Neuropsychology and
Cognitive Sciences at Universidad Maimonides in Buenos Aires,
Argentina. Currently, he is the Coordinator of the Cognitive
Neurosciences Unit and Research Professor at the Psychology
Department of Universidad Simón Bolívar, Barranquilla. He has a
vast experience in clinical neuropsychology and is currently con-
ducting research studies on the neuropsychology profiles and
genetic components of mild cognitive impairment, substance use
disorders and ADHD.
Ariel F. Martinez is a research biologist at the Medical Genetics
Branch, National Human Genome Research Institute, National
Institutes of Health (Bethesda, MD) with 10 years of experience
working on human developmental diseases, both Mendelian and
complex. Dr. Martinez earned his Ph.D. in Biochemistry and
Systems Biology from the George Washington University in
Washington, D.C., United States. He is an Active Candidate of the
American Board of Medical Genetics and Genomics in the spe-
cialty of Clinical Molecular Genetics. His current work involves
genetic studies of holoprosencephaly (HPE) and ADHD, and
genetic testing of patients with HPE and Muenke syndrome. His
research is focused on understanding the molecular mechanisms
underlying disease pathogenicity.
Mauricio Arcos-Burgos is a full professor at the Department of
Psychiatry, Institute of Medical Research, Universidad de
Antioquia, Medellín, Colombia. He received his MD degree from
Universidad del Cauca, MSc from University of Antioquia, a PhD
in Genetics from Universidad de Chile and a PhD in Clinical
Cervantes-Henríquez et al. 19
Genetics from the NIH-Johns Hopkins University Partnership
Program. He has a vast experience in clinical and basic research,
management, administration, teaching, publishing, consulting,
and supervision in human genetics, clinical genetics, genetic epi-
demiology, population genetics, and evolution. His current
research aims at identifying genetic variations, biomarkers, prog-
nostic assays, and eventual personalised treatment options for dis-
eases that have a genetic background, such as rheumatologic dis-
eases, ADHD, Alzheimer's disease, cleft lip with or without cleft
palate, major depressive disorder, and obesity.
Pedro J. Puentes-Rozo is a psychologist with a specialisation in
Clinical Neuropsychology, a MSc in Neuropsychology and a PhD
in Psychology. He is a Research Professor at Universidad Simón
Bolívar, Barranquilla, and Universidad del Atlántico, Barranquilla,
and the Director of the Grupo de Neurociencias del Caribe in both
institutions. His research interests include Cognitive Neuroscience
and genetics of ADHD.
Jorge I. Vélez is an assistant professor at the Department of
Industrial Engineering of Universidad del Norte, Barranquilla. He
earned a BSc in Industrial Engineering and a MSc in Statistics
from the National University of Colombia at Medellín, and a PhD
in Medical Sciences (Genomics and Precision Medicine) from the
Australian National University in Canberra, Australia. He is inter-
ested in the development and application of Machine Learning,
bioinformatic and statistical genetic models/tools to lead to a bet-
ter understanding of the aetiology of complex human diseases as
well as the identification of key genetic factors that confer suscep-
tibility to ADHD, Alzheimer’s disease and Cancer.
... Finally, with the help of computational analysis, such as machine learning algorithms, SNPs of different genes (ADGRL3, DRD4, and SNAP25) have been used to predict symptoms severity in ADHD. In this study, polymorphisms of genes involved in dopamine circuitry (DRD4-rs916457), synaptic plasticity and working memory (SNAP25-rs362990), and hyperactivity and impulsivity symptoms (ADGRL3-rs2122642, ADGRL3-rs10001410) have been associated with the severity of ADHD under different genetic models of inheritance [54]. Another study performed a machine learning analysis to evaluate positron emission tomography imaging and genetic predictors involved in the serotonergic system. ...
Article
Full-text available
This article explores the complex relationship between genetics and cognition, specifically examining the impact of genetic variants, particularly single nucleotide polymorphisms (SNPs), on cognitive functions and the development of neuropsychiatric disorders. Focusing on neurotransmitter regulation within the prefrontal cortex’s dopaminergic circuits, this study emphasizes the role of genes like COMT, PRODH, and DRD in shaping executive functions and influencing conditions such as ADHD and schizophrenia. Additionally, it explores the significance of genetic factors in neurodevelopmental disorders, emphasizing the need for early identification to guide appropriate therapeutic interventions. This article also investigates polymorphisms in the transsulfuration pathway, revealing their association with cognitive impairment diseases. Computational analyses, including machine learning algorithms, are highlighted for their potential in predicting symptom severity in ADHD based on genetic variations. In conclusion, this article underscores the intricate interplay of genetic and environmental factors in shaping cognitive outcomes, providing valuable insights for tailored treatments and a more comprehensive understanding of neuropsychiatric conditions.
... [21] SNPs in SNAP25, DRD4, and ADGRL3 were studied to see whether they were associated with ADHD symptoms in Caribbean families. [22] A reduced-order model is created using Galperin fuzzification and the Euler-Lagrange principle. Galperin's approach is used to calculate the van der Pol wake oscillation coefficients using a five-mode approximation. ...
Conference Paper
Full-text available
ADHD, a neurodevelopmental disorder characterized by hyperactivity, inattention, and impulsivity, has many detrimental impacts and is out of proportion to age. ADHD causes executive failure and emotional instability, which can lower academic performance. We propose a machine learning and artificial intelligence-driven approach to diagnose and early detect this disease and assist ADHD medicine. SVM, logistic regression, XGBoost, AdaBoost, and two deep learning models were applied to our dataset (ANN and CNN). Our ANN model had 99% accuracy in dependability, expandability, and generalizability. We plan to use our machine learning technology to enhance ADHD diagnosis and treatment for everyone.
... A subsequent study found another polymorphism associated with ADHD, the rs362549 [140]. SNAP-25 polymorphisms influence ADHD severity [141,142]. The impact of the MnlI on symptom intensity in ADHD is the most well-known. ...
Article
Full-text available
Attention deficit hyperactivity disorder (ADHD) is one of the most common neurodevelopmental disorders, although the aetiology of ADHD is not yet understood. One proposed theory for developing ADHD is N-methyl-D-aspartate receptors (NMDARs) dysfunction. NMDARs are involved in regulating synaptic plasticity and memory function in the brain. Abnormal expression or polymorphism of some genes associated with ADHD results in NMDAR dysfunction. Correspondingly, NMDAR malfunction in animal models results in ADHD-like symptoms, such as impulsivity and hyperactivity. Currently, there are no drugs for ADHD that specifically target NMDARs. However, NMDAR-stabilizing drugs have shown promise in improving ADHD symptoms with fewer side effects than the currently most widely used psychostimulant in ADHD treatment, methylphenidate. In this review, we outline the molecular and genetic basis of NMDAR malfunction and how it affects the course of ADHD. We also present new therapeutic options related to treating ADHD by targeting NMDAR.
... Other studies have used random forest and convolutional neural networks to study genetic predictors of ADHD and have revealed that the gene regions GRM1, GRM8, and EPHA5 are important predictors of ADHD [123,124]. Using multiple machine learning algorithms, a recent study reported that age and sex were significant predictors in genetic information-based classification models [125]. In addition, gene regions SNAP25, ADGRL3, and DRD4 significantly contributed to the prediction of inattentive, hyperactive, or impulsive symptoms. ...
Article
Full-text available
Attention-deficit/hyperactivity disorder (ADHD) is a highly prevalent and heterogeneous neurodevelopmental disorder in children and has a high chance of persisting in adulthood. The development of individualized, efficient, and reliable treatment strategies is limited by the lack of understanding of the underlying neural mechanisms. Diverging and inconsistent findings from existing studies suggest that ADHD may be simultaneously associated with multivariate factors across cognitive, genetic, and biological domains. Machine learning algorithms are more capable of detecting complex interactions between multiple variables than conventional statistical methods. Here we present a narrative review of the existing machine learning studies that have contributed to understanding mechanisms underlying ADHD with a focus on behavioral and neurocognitive problems, neurobiological measures including genetic data, structural magnetic resonance imaging (MRI), task-based and resting-state functional MRI (fMRI), electroencephalogram, and functional near-infrared spectroscopy, and prevention and treatment strategies. Implications of machine learning models in ADHD research are discussed. Although increasing evidence suggests that machine learning has potential in studying ADHD, extra precautions are still required when designing machine learning strategies considering the limitations of interpretability and generalization.
... Several fields have proved the efficiency of applying regular as well as federated ML [16] to predict certain real-world answers. According to authors in [11], a major application of ML would be in the field of medicine and healthcare, such as predicting certain human diseases, traits, or syndromes [17]. Furthermore, in [18], the authors tackle the issue of medical data scarcity and sensitivity, by proposing an additional level of privacy protection in medical imaging data through the use of federated privacy-preserving artificial intelligence. ...
Preprint
Full-text available
Autism Spectrum Disorder (ASD) is a neuro-developmental syndrome resulting from alterations in the embryological brain before birth. This disorder distinguishes its patients by special socially restricted and repetitive behavior in addition to specific behavioral traits. Hence, this would possibly deteriorate their social behavior among other individuals, as well as their overall interaction within their community. Moreover, medical research has proved that ASD also affects the facial characteristics of its patients, making the syndrome recognizable from distinctive signs within an individual's face. Given that as a motivation behind our work, we propose a novel privacy-preserving federated learning scheme to predict ASD in a certain individual based on their behavioral and facial features, embedding a merging process of both data features through facial feature extraction while respecting patient data privacy. After training behavioral and facial image data on federated machine learning models, promising results are achieved, with 70\% accuracy for the prediction of ASD according to behavioral traits in a federated learning environment, and a 62\% accuracy is reached for the prediction of ASD given an image of the patient's face. Then, we test the behavior of regular as well as federated ML on our merged data, behavioral and facial, where a 65\% accuracy is achieved with the regular logistic regression model and 63\% accuracy with the federated learning model.
... e decreased expression of BDNF can cause significant changes in DA neuron synapse morphology and dysfunction, leading to a decrease in DA content in the brain. is impact may be related to the low activation of the BDNF/TrkB signaling pathway, which inhibits the function of the SNARE complex and affects presynaptic membrane DA vesicle circulation and transmitter release [57,58]. e phospholipase Cc was recruited and activated by phosphorylation of Tyr816 residue for promoting neuronal survival and implicating neurite outgrowth and synaptic plasticity [59]. ...
Article
Full-text available
Attention deficit hyperactivity disorder (ADHD) is a common childhood neurodevelopmental disorder. It may impact the cognitive and social functions throughout childhood and determine adult outcomes. Dopamine (DA) deficiency theory is the pathogenesis of ADHD that is recognized by most international literature. Existing studies have shown that DA deficiency is caused by the abnormal function of the DA transporter and an imbalance in the DA receptor functionality. Recent clinical and experimental studies have found that the brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) signaling pathway acts a pivotal part in DA vesicle circulation and ADHD pathogenesis. An Shen Ding Zhi Ling (ASDZL) is a traditional Chinese medicine (TCM) prescription, which was widely prescribed to treat ADHD in Jiangsu, China, but its therapeutic mechanism is unclear. Therefore, we constructed a spontaneously hypertensive rat (SHR) model to explain its mechanism. SHRs were randomly assigned to four groups: SHR model group (vehicle), methylphenidate hydrochloride group (MPH), ASDZL group, and 7,8-dihydroxyflavone group (7,8-DHF). At the same time, the above groups were given continuous medication for four weeks. The results show that ASDZL, MPH, and 7,8-DHF group could significantly improve the spatial memory of SHRs in the Morris water maze tests. ASDZL increased the levels of BDNF, TrkB, p75 neurotrophin receptor (p75), C-Jun N-terminal kinases 1 (JNK1), and nuclear factor kappa B (NF-κB) in the prefrontal cortex (PFC) and hippocampus synaptosome of SHRs. The results of this study suggest that ASDZL can relieve the symptoms of ADHD in SHRs by regulating the balance between the BDNF/TrkB signaling pathway (promoting vesicle circulation) and the BDNF/P75/JNK1/NF-κB signaling pathway (inhibiting vesicle circulation) within the PFC and hippocampus synaptosome to increase the DA concentration in the synaptic cleft. The BDNF/TrkB signal pathway within the PFC and hippocampus synaptosome was activated by 7,8-DHF to increase DA concentration in the synaptic cleft. Whether 7,8-DHF can activate or inhibit the BDNF/P75 signaling pathway remains unclear.
Article
Full-text available
There have been increasing efforts to develop prediction models supporting personalised detection, prediction, or treatment of ADHD. We overviewed the current status of prediction science in ADHD by: (1) systematically reviewing and appraising available prediction models; (2) quantitatively assessing factors impacting the performance of published models. We did a PRISMA/CHARMS/TRIPOD-compliant systematic review (PROSPERO: CRD42023387502), searching, until 20/12/2023, studies reporting internally and/or externally validated diagnostic/prognostic/treatment-response prediction models in ADHD. Using meta-regressions, we explored the impact of factors affecting the area under the curve (AUC) of the models. We assessed the study risk of bias with the Prediction Model Risk of Bias Assessment Tool (PROBAST). From 7764 identified records, 100 prediction models were included (88% diagnostic, 5% prognostic, and 7% treatment-response). Of these, 96% and 7% were internally and externally validated, respectively. None was implemented in clinical practice. Only 8% of the models were deemed at low risk of bias; 67% were considered at high risk of bias. Clinical, neuroimaging, and cognitive predictors were used in 35%, 31%, and 27% of the studies, respectively. The performance of ADHD prediction models was increased in those models including, compared to those models not including, clinical predictors (β = 6.54, p = 0.007). Type of validation, age range, type of model, number of predictors, study quality, and other type of predictors did not alter the AUC. Several prediction models have been developed to support the diagnosis of ADHD. However, efforts to predict outcomes or treatment response have been limited, and none of the available models is ready for implementation into clinical practice. The use of clinical predictors, which may be combined with other type of predictors, seems to improve the performance of the models. A new generation of research should address these gaps by conducting high quality, replicable, and externally validated models, followed by implementation research.
Article
Full-text available
Attention deficit/hyperactivity disorder (ADHD) is the most common childhood neurodevelopmental disorder. Single nucleotide polymorphisms (SNPs) in the Adhesion G Protein-Coupled Receptor L3 (ADGRL3) gene are associated with increased susceptibility to developing ADHD worldwide. However, the effect of ADGRL3 non-synonymous SNPs (nsSNPs) on the ADGRL3 protein function is vastly unknown. Using several bioinformatics tools to evaluate the impact of mutations, we found that nsSNPs rs35106420, rs61747658, and rs734644, previously reported to be associated and in linkage with ADHD in disparate populations from the world over, are predicted as pathogenic variants. Docking analysis of rs35106420, harbored in the ADGLR3-hormone receptor domain (HRM, a common extracellular domain of the secretin-like GPCRs family), showed that HRM interacts with the Glucose-dependent insulinotropic polypeptide (GIP), part of the incretin hormones family. GIP has been linked to the pathogenesis of diabetes mellitus, and our analyses suggest a potential link to ADHD. Overall, the comprehensive application of bioinformatics tools showed that functional mutations in the ADGLR3 gene disrupt the standard and wild ADGRL3 structure, most likely affecting its metabolic regulation. Further in vitro experiments are granted to evaluate these in silico predictions of the ADGRL3-GIP interaction and dissect the complexity underlying the development of ADHD.
Article
Full-text available
Background: Attention-deficit/hyperactive disorder (ADHD) is a neurodevelopmental disorder that commonly occurs in children with a prevalence ranging from 3.4 to 7.2%. It profoundly affects academic achievement, well-being, and social interactions. As a result, this disorder is of high cost to both individuals and society. Despite the availability of knowledge regarding the mechanisms of ADHD, the pathogenesis is not clear, hence, the existence of many challenges especially in making correct early diagnosis and provision of accurate management. Objectives: We aimed to review the pathogenic pathways of ADHD in children. The major focus was to provide an update on the reported etiologies in humans, animal models, modulators, therapies, mechanisms, epigenetic changes, and the interaction between genetic and environmental factors. Methods: References for this review were identified through a systematic search in PubMed by using special keywords for all years until January 2022. Results: Several genes have been reported to associate with ADHD: DRD1, DRD2, DRD4, DAT1, TPH2, HTR1A, HTR1B, SLC6A4, HTR2A, DBH, NET1, ADRA2A, ADRA2C, CHRNA4, CHRNA7, GAD1, GRM1, GRM5, GRM7, GRM8, TARBP1, ADGRL3, FGF1, MAOA, BDNF, SNAP25, STX1A, ATXN7, and SORCS2. Some of these genes have evidence both from human beings and animal models, while others have evidence in either humans or animal models only. Notably, most of these animal models are knockout and do not generate the genetic alteration of the patients. Besides, some of the gene polymorphisms reported differ according to the ethnic groups. The majority of the available animal models are related to the dopaminergic pathway. Epigenetic changes including SUMOylation, methylation, and acetylation have been reported in genes related to the dopaminergic pathway. Conclusion: The dopaminergic pathway remains to be crucial in the pathogenesis of ADHD. It can be affected by environmental factors and other pathways. Nevertheless, it is still unclear how environmental factors relate to all neurotransmitter pathways; thus, more studies are needed. Although several genes have been related to ADHD, there are few animal model studies on the majority of the genes, and they do not generate the genetic alteration of the patients. More animal models and epigenetic studies are required.
Article
Full-text available
Serotonin neurotransmission may impact the etiology and pathology of attention-deficit and hyperactivity disorder (ADHD), partly mediated through single nucleotide polymorphisms (SNPs). We propose a multivariate, genetic and positron emission tomography (PET) imaging classification model for ADHD and healthy controls (HC). Sixteen patients with ADHD and 22 HC were scanned by PET to measure serotonin transporter (SERT‘) binding potential with [11C]DASB. All subjects were genotyped for thirty SNPs within the HTR1A, HTR1B, HTR2A and TPH2 genes. Cortical and subcortical regions of interest (ROI) were defined and random forest (RF) machine learning was used for feature selection and classification in a five-fold cross-validation model with ten repeats. Variable selection highlighted the ROI posterior cingulate gyrus, cuneus, precuneus, pre-, para- and postcentral gyri as well as the SNPs HTR2A rs1328684 and rs6311 and HTR1B rs130058 as most discriminative between ADHD and HC status. The mean accuracy for the validation sets across repeats was 0.82 (±0.09) with balanced sensitivity and specificity of 0.75 and 0.86, respectively. With a prediction accuracy above 0.8, the findings underlying the proposed model advocate the relevance of the SERT as well as the HTR1B and HTR2A genes in ADHD and hint towards disease-specific effects. Regarding the high rates of comorbidities and difficult differential diagnosis especially for ADHD, a reliable computer-aided diagnostic tool for disorders anchored in the serotonergic system will support clinical decisions.
Article
Full-text available
The gene encoding adhesion G protein-coupled receptor L3 (ADGRL3, also referred to as latrophilin 3 or LPHN3) has been associated with ADHD susceptibility in independent ADHD samples. We conducted a systematic review and a comprehensive meta-analysis to summarize the associations between the most studied ADGRL3 polymorphisms (rs6551665, rs1947274, rs1947275, and rs2345039) and both childhood and adulthood ADHD. Eight association studies (seven published and one unpublished) fulfilled criteria for inclusion in our meta-analysis. We also incorporated GWAS data for ADGRL3. In order to avoid overlapping samples, we started with summary statistics from GWAS samples and then added data from gene association studies. The results of our meta-analysis suggest an effect of ADGRL3 variants on ADHD susceptibility in children (n = 8724/14,644 cases/controls and 1893 families): rs6551665 A allele (Z score = −2.701; p = 0.0069); rs1947274 A allele (Z score = −2.033; p = 0.0421); rs1947275 T allele (Z score = 2.339; p = 0.0978); and rs2345039 C allele (Z score = 3.806; p = 0.0026). Heterogeneity was found in analyses for three SNPs (rs6551665, rs1947274, and rs2345039). In adults, results were not significant (n = 6532 cases/15,874 controls): rs6551665 A allele (Z score = 2.005; p = 0.0450); rs1947274 A allele (Z score = 2.179; p = 0.0293); rs1947275 T allele (Z score = −0.822; p = 0.4109); and rs2345039 C allele (Z score = −1.544; p = 0.1226). Heterogeneity was found just for rs6551665. In addition, funnel plots did not suggest publication biases. Consistent with ADGRL3’s role in early neurodevelopment, our findings suggest that the gene is predominantly associated with childhood ADHD.
Article
Full-text available
Attention Deficit Hyperactivity Disorder (ADHD) is a highly heritable and prevalent neurodevelopmental disorder that frequently persists into adulthood. Strong evidence from genetic studies indicates that single nucleotide polymorphisms (SNPs) harboured in the ADGRL3 (LPHN3), SNAP25, FGF1, DRD4, and SLC6A2 genes are associated with ADHD. We genotyped 26 SNPs harboured in genes previously reported to be associated with ADHD and evaluated their potential association in 386 individuals belonging to 113 nuclear families from a Caribbean community in Barranquilla, Colombia, using family-based association tests. SNPs rs362990-SNAP25 (T allele; p = 2.46 × 10−4), rs2282794-FGF1 (A allele; p = 1.33 × 10−2), rs2122642-ADGRL3 (C allele, p = 3.5 × 10−2), and ADGRL3 haplotype CCC (markers rs1565902-rs10001410-rs2122642, OR = 1.74, Ppermuted = 0.021) were significantly associated with ADHD. Our results confirm the susceptibility to ADHD conferred by SNAP25, FGF1, and ADGRL3 variants in a community with a significant African American component, and provide evidence supporting the existence of specific patterns of genetic stratification underpinning the susceptibility to ADHD. Knowledge of population genetics is crucial to define risk and predict susceptibility to disease.
Article
Full-text available
Attention Deficit Hyperactivity Disorder (ADHD) is one of the most prevalent neuropsychiatric disorders in childhood and adolescence and its diagnosis is based on clinical interviews, symptom questionnaires, and neuropsychological testing. Much research effort has been undertaken to evaluate the usefulness of neurophysiological (EEG) data to aid this diagnostic process. In the current study, we applied deep learning methods on event-related EEG data to examine whether it is possible to distinguish ADHD patients from healthy controls using purely neurophysiological measures. The same was done to distinguish between ADHD subtypes. The results show that the applied deep learning model ("EEGNet") was able to distinguish between both ADHD subtypes and healthy controls with an accuracy of up to 83%. However, a significant fraction of individuals could not be classified correctly. It is shown that neurophysiological processes indicating attentional selection associated with superior parietal cortical areas were the most important for that. Using the applied deep learning method, it was not possible to distinguish ADHD subtypes from each other. This is the first study showing that deep learning methods applied to EEG data are able to dissociate between ADHD patients and healthy controls. The results show that the applied method reflects a promising means to support clinical diagnosis in ADHD. However, more work needs to be done to increase the reliability of the taken approach.
Article
Full-text available
The adhesion G protein-coupled receptors latrophilins have been in the limelight for more than 20 years since their discovery as calcium-independent receptors for α-latrotoxin, a spider venom toxin with potent activity directed at neurotransmitter release from a variety of synapse types. Latrophilins are highly expressed in the nervous system. Although a substantial amount of studies has been conducted to describe the role of latrophilins in the toxin-mediated action, the recent identification of endogenous ligands for these receptors helped confirm their function as mediators of adhesion events. Here we hypothesize a role for latrophilins in inter-neuronal contacts and the formation of neuronal networks and we review the most recent information on their role in neurons. We explore molecular, cellular and behavioral aspects related to latrophilin adhesion function in mice, zebrafish, Drosophila melanogaster and Caenorhabditis elegans, in physiological and pathophysiological conditions, including autism spectrum, bipolar, attention deficit and hyperactivity and substance use disorders.
Article
We compared the performance of children with ADHD and typically developing children on two temporal tasks, a bisection task and a reproduction task, in auditory and visual modalities. Children with ADHD presented a larger variability when performing auditory and visual temporal tasks. Moreover, they overestimated the durations in bisection tasks and under-produced duration intervals in the visual reproduction task. In the context of the pacemaker-accumulator model, these results suggest that temporal deficits might result from a dysfunction in the switch and/or memory impairment. ARTICLE HISTORY
Article
Attention-deficit hyperactivity disorder (ADHD), like other psychiatric disorders, represents an evolving construct that has been refined and developed over the past several decades in response to research into its clinical nature and structure. The clinical presentation and course of the disorder have been extensively characterised. Efficacious medication-based treatments are available and widely used, often alongside complementary psychosocial approaches. However, their effectiveness has been questioned because they might not address the broader clinical needs of many individuals with ADHD, especially over the longer term. Non-pharmacological approaches to treatment have proven less effective than previously thought, whereas scientific and clinical studies are starting to fundamentally challenge current conceptions of the causes of ADHD in ways that might have the potential to alter clinical approaches in the future. In view of this, we first provide an account of the diagnosis, epidemiology, and treatment of ADHD from the perspective of both the fifth edition of the Diagnostic and Statistical Manual of Mental Disorders and the eleventh edition of the International Classification of Diseases. Second, we review the progress in our understanding of the causes and pathophysiology of ADHD on the basis of science over the past decade or so. Finally, using these discoveries, we explore some of the key challenges to both the current models and the treatment of ADHD, and the ways in which these findings can promote new perspectives.