Summary
ADHD is a common chronic neurodevelopmental disorder and is characterized by persistent inattention, hyperactivity, impulsivity and are often accompanied by learning and memory impairment. Great evidence has shown that learning and memory impairment of ADHD plays an important role in its executive function deficits, which seriously affects the development of academic, cognitive and daily social skills and will cause a serious burden on families and society. With the increasing attention paid to learning and memory impairment in ADHD, relevant research is gradually increasing. In this article, we will present the current research results of learning and memory impairment in ADHD from the following aspects. Firstly, the animal models of ADHD, which display the core symptoms of ADHD as well as with learning and memory impairment. Secondly, the molecular mechanism of has explored, including some neurotransmitters, receptors, RNAs, etc. Thirdly, the susceptibility gene of ADHD related to the learning and impairment in order to have a more comprehensive understanding of the pathogenesis.
Keywords: Learning and memory, ADHD, Review
Introduction
Attention-deficit/hyperactivity disorder (ADHD) is a common chronic neurodevelopmental disorder that affects from 3 % to 7 % of children and adolescents worldwide [1]. ADHD is characterized by persistent inattention, hyperactivity, and impulsivity, which are often accompanied by learning and memory impairments [2]. Working memory is an important part of executive function, which serves as both the storage workstation of long-term memory and the basis for advanced cognitive functions including learning, language, and understanding [3,4]. Therefore, working memory deficits are likely to contribute to learning and cognitive processing problems [5].
With research advances, increasing evidence shows that ADHD-related learning and memory impairments play an important role in the disorder’s characteristic executive function deficits, which seriously impact academic and cognitive development, and daily social skills, also causing serious burden for families and society [6–8]. There is also ample evidence suggesting that adolescents and adults with ADHD continue to experience obstacles from processing speed, distraction, and learning and memory abilities [9]. Use of methylphenidate (MPH), an effective medicine for treating attention, impulsivity, and cognitive function, has also increased among adult patients [10,11].
With gradual recognition of the importance of learning and memory impairments, researchers have more deeply, and across many fields, explored the mechanism of this in ADHD. For example, dopamine (DA), serotonin (5-HT), N-methyl-D-aspartate (NMDA), and up- or down-regulation of upstream molecules involved in related pathways, all impact expression of a series of proteins and affect physiological activities [8,12–14]. ADHD is a multifactorial disorder, related to both genetic and environmental factors [15]. International twin studies on children with ADHD have described a hereditary range of 71–90 %; though they have not identified exact genetic profiles, scientists are attempting to find the best gene–trait correlation through research investigating candidate genes, genome-wide association, and copy number variants [16,17].
To develop a more comprehensive understanding of the pathogenesis of learning and memory defects in ADHD, we present the current research on this topic from several perspectives. First, we describe the current animal models of ADHD, which display both the core disorder symptoms and learning and memory impairments. Second, we explore the molecular mechanisms, including involvement of neurotransmitters, receptors, and RNAs. Third, we discuss genetic susceptibility in ADHD, specifically related to learning and memory impairments.
Animal models of ADHD with learning and memory impairments
Most studies to date on the mechanism of learning and memory impairments in ADHD have been conducted at the non-human animal level; therefore, selecting an appropriate animal model is important [18]. Three factors in establishing a good animal model are important: face, construct, and predictive validities [19]. Regan et al. [19] asserted that although there is presently no rodent model that captures all ADHD characteristics, several show promise. Herein, we describe several commonly used ADHD animal models, with learning and memory impairments, in addition to inattention, hyperactivity, and impulsivity (Table 1).
Table 1.
Animal models of ADHD displaying inattention hyperactivity, impulsivity as well as learning and WM impairment
Model | Face validity | Construct Validity | Predictive validity | Limitations | Others | References |
---|---|---|---|---|---|---|
SHR | inattention hyperactivity, impulsivity, learning, and memory impairment | disturbances in dopamine, norepinephrine and glutamate function is parallel with the evidence of ADHD patients | MPH and APH can attenuate ADHD like behaviors. | No gender differences | The control of WKY | [20–27] |
DAT-KO mouse | inattention hyperactivity, impulsivity, learning, and memory impairment | DAT changes found in ADHD patients | MPH can improve hyperactivity and learning disorders | DAT is not absence but reduced in ADHD | ADHD including growth delay and death before maturity | [28–31] |
LPHN3-KO mouse and rat | inattention hyperactivity, impulsivity, learning, and memory impairment | Variants found in ADHD patients | amphetamine, attenuates hyperactivity and activity | New model needs more data | - | [32–36] |
NDRG2-KO mouse | inattention hyperactivity, impulsivity, learning, and memory impairment | SNP in NDRG2 risked to ADHD | not established treatment of | NDRG2 rescued hyperactivity but routine MPH not effective | new idea for the pathogenesis of ADHD | [37–38] |
Spontaneously hypertensive and Wistar Kyoto rats
In ADHD research, spontaneously hypertensive rats (SHR), and the control Wistar Kyoto (WKY) rats, are the most widely used animal model. SHR are hyperactive, impulsive, inattentive, and have learning and memory deficits compared with WKY [20]. Effective first-line ADHD drugs can attenuate ADHD-like behaviors in SHR [21]. Additional evidence has shown that the SHR displays disturbances in DA, norepinephrine (NE), and glutamate functions, consistent with evidence of defects on the neural circuits required for learning and memory formation in patients with ADHD [22–24]. The Campden behavior test has shown that total motor activity, average speed, and maximum speed are higher for SHR than for WKY [25]. And in the eight-arm radial maze, there is a clear tendency for errors, indicating hyperactivity, impulsivity, and learning and working memory deficits in SHR [26]. SHR also exhibits dysfunction of the NE system related to movement, learning, and cognition [24]. In addition, with ADHD drug treatment, SHR open field test average speed, time spent in the central area, and number of center visits can be reduced, while Morris water maze (MWM) number of platform penetrations and target quadrant residence time can be increased, indicating improved learning and memory [27]. This cumulative evidence supports SHR as a relatively ideal ADHD animal model.
Dopamine transporter knockout mice
DA transporter (DAT) knockout (KO) mice, which lack the gene encoding DAT, exhibit hyperactivity and spatial memory deficits in new environments. Cliff avoidance shows this animal model to be hyperactive and have impaired attention, response inhibition, learning, and memory [28]. MPH can improve hyperactivity and learning ability in DAT-KO mice by augmenting DA levels in the PFC [29]. In these mice, decreased DA receptor D2 may affect long-term potentiation (LTP), leading to decreased synaptic strength and impaired associative learning [30]. However, DAT-KO mice can also show unpredictable phenomenon related to impaired DAT function, such as growth delay and death before maturity [31].
Latrophilin-3 knockout mice and rats
In recent years, latrophilin-3 (LPHN3), which plays a role in regulating neuroplasticity, has attracted extensive research activity [32]. Among Turkish population, Researchers have found that LPHN3 rs6551665 and rs1947274 polymorphisms are significantly associated with ADHD and LPHN3 rs6551665 polymorphism may be related to poor response to treatment in ADHD [33]. Animal research has also shown that LPHN3 KO (LPHN3-KO) mice have a reduced response to amphetamine compared with baseline. In the open field test, these mice are hyperactive; in the continuous performance test, they have increased premature responses, indicating impulsivity; and in the MWM, they exhibit spatial deficits and memory impairments [34,35]. Studies exploring the mechanism of this have found that it may be related to the downregulation of DA receptor D1 and upregulation of DAT [35,36]. In addition, Fallgatter et al. examined the effects of LPHN3 haplotype on neural activity, using a visual task to find that patients with ADHD and two copies of the LPHN3 risk haplotype have more errors compared with those with at least one LPHN3 non-risk haplotype [35,36]. These results indicate that LPHN3 is closely related to ADHD-based learning and memory impairments. Thus, although the LPHN3-KO mouse and rat models may become valuable ADHD animal models, more in-depth research is needed.
N-myc downstream-regulated gene 2-knockout mice
There is evidence that the N-myc downstream-regulated gene 2 (NDRG2) plays a role in the patho-genesis of ADHD [37]. Chinese children heterozygous for rs1998848, a single nucleotide polymorphism (SNP) in NDRG2, had higher risk of ADHD compared with children homozygous for rs1998848 [38]. In the open field test, NDRG2-KO mice have a significantly increased total distance and numbers of lines crossed. In the 5-choice serial reaction time task (5-CSRTT), NDRG2-KO mice exhibit display altered attention and impulsivity. In a novel object recognition test, NDRG2-KO mice display impaired memory [38]. Moreover, NDRG2 treatment can rescued hyperactivity in NDRG2-KO mice, but routine MPH is ineffective [38]. This cumulative evidence indicates that NDRG2 deficiency can result in an ADHD phenotype, which is different from the DA deficit hypothesis. This provides a novel direction for evaluating the pathogenesis of ADHD.
Molecular biology of learning and memory impairments in ADHD
DA system
DA system dysfunction is the most classic pathogenesis of ADHD. Rhodes et al. found that MPH, the first-line drug treatment for ADHD, can increase synaptic DA concentration by inhibiting DAT action, to achieve therapeutic effects and improve working memory task performance among children with ADHD [39]. At present, the two most well-studied candidate functional genes are DAT gene DAT1 and DA receptor D4 (DRD4), which are also associated with ADHD learning and memory disability.
DAT1 has a polymorphic variable number tandem repeat (VNTR), with its 10-repeat (10R) and nine-repeat (9R) alleles occurring frequently [40]. Scientists have shown that ADHD carriers of 9R have higher working memory-related activity compared with controls [41]. Chi-Yung et al. found significant diagnosis × genotype interactions associated with working memory level, preliminarily suggesting that DAT1 VNTR polymorphism may modulate working memory-related activity among children with ADHD [42]. In addition, when Brown et al. studied the relations among DAT1, working memory, and adult ADHD and the results showed 9R is related to ADHD and marginal DAT1 is associated with task-related suppressions [43].
Li et al. examined DA receptor levels in the PFC of SHR and compared learning and memory abilities with WKY [44], finding that DRD4, but not other DA receptors, was significantly downregulated in SHR. Considering that there were no other gene marker differences, they thought this memory performance difference may be closely associated with DRD4. Yin et al. used ABT-724, a D4R-selective agonist, to find that SHR displayed spatial learning, hyperactivity, and non-selective attention impairments compared with controls, and that ABT-724 treatment can alleviate both hyperactivity and spatial learning impairment in SHR [45]. In addition, most investigators assert that the 7-repeat allele of the DRD4 receptor gene is a risk factor for ADHD, and that children with ADHD who have this gene have lower verbal intelligence, operational intelligence, and working memory abilities compared with those without this gene [46]. However, other studies have found no significant association between this gene and working memory performance [47]. Therefore, more research may be needed.
5-HT
Serotonergic system disruption has also been implicated in ADHD [48]. Research on both animal models and adults with psychiatric diagnoses have shown that the neurotransmitter 5-HT is closely associated with learning and memory processes [49]. Previous studies have shown that moderate doses of MPH, can enhance learning and memory abilities, while higher doses have the opposite result [50]. Salma et al. further revealed that animals administered moderate MPH doses, and showing improved performance, had higher 5-HT metabolism, while those administered higher doses had impaired memory and downregulated 5-HT receptor expression [51]. However, in youth with ADHD, Zepf et al. found no association between 5-HT expression and verbal declarative memory [52]. Therefore, more research concerning the association between 5-HT and ADHD-related learning and memory impairments is warranted, especially in adults with ADHD.
Glutamate
Researchers have also identified glutamatergic dysfunction in ADHD pathogenesis [53]. There is a marked increase in glutamate in some brain areas with ADHD, and glutamate level has been associated with the ADHD symptoms of hyperactivity and impulsivity [54,55]. The glial glutamate transporter GLT1 plays an important role in glutamatergic neurotransmission. Hiraoka et al. found GLT1 knocko(GLT1-KO) mice exhibit ADHD symptoms of hyperactivity, impulsivity, and impaired memory [56]. Assessing the excitatory synaptic function and analyzing NMDA receptors in an ADHD animal model and control, Shikana et al. found that the former has impaired cortical excitatory synapses, based on NMDA receptor dysfunction [57]. Others have also found that MPH, the medicine for treating ADHD, can enhance NMDA receptor-mediated excitatory postsynaptic currents and improve PFC-mediated memory and attention [58]. To explore the mechanism for this, Kawade et al. studied caspase-3 and phosphorylated Akt, which is often used to cause a ADHD-like condition, finding that working memory errors were greatly increased and NMDA receptor expression dramatically changed [59]. Moreover, Jensen et al. found that transmission in hippocampal CA3-to-CA1 synapses of SHR were significantly reduced, and that NMDAR-containing NR2B subunits contributed substantially to LTP induction, thus leading to learning and memory dysfunction [13].
Acetylcholine receptors
Increasing evidence shows that acetylcholine receptors (nAChRs) also play a role in ADHD. Neuronal nAChR agonists improve cognitive function and increase expressions of DA, 5-HT, and glutamate [60,61]. In animal models, nicotinic ABT-418, a nAChR agonist, can significantly improve SHR memory, and ABT-418 treatment greatly improves expressions of cortical α4 and β2 nAChR subunits, and hippocampal α4 subunit. Moreover, different doses of Moreover, in an animal model, Jeong et al. found that treadmill exercise can alleviate spatial learning deficits by enhancing BDNF and TrkB expressions in the SHR [62,63]
Neurotrophic factors and synaptic proteins
Brain-derived neurotrophic factor (BDNF), a neurotrophin nerve growth factor family member, is involved in neuronal survival and differentiation, neurotransmitter modulation, and neuronal plasticity, which can significantly affect learning and memory [64]. Corominas-Roso et al. found that serum BDNF is altered in patients with ADHD, and that when treated with medications that increase DA levels, levels of BDNF and its TrkB receptor are increased in some brain areas [14]. Liang-Jen et al. showed that BDNF is associated with sex-specific ADHD susceptibility; its expression in boys with ADHD is higher than in controls, while levels in girls with ADHD are lower compared with controls. Boys with higher BDNF expression also performed worse on the Wechsler Intelligence Scale for Children, 4th edition, including the working memory test [65]. Moreover, in an animal model, Jeong et al. found that treadmill exercise can alleviate spatial learning deficits by enhancing BDNF and TrkB expressions in the SHR [62].
Neurexins are highly polymorphic presynaptic cell-adhesion molecules that play critical roles in establishing and maintaining synaptic connections. Neurexin 1 (NRXN1) has been described in neuro-developmental disorders, including autism spectrum disorder and ADHD [66]. Zhang et al. injected intracerebroventricular HBAD-r-NRXN1 virus, showing that NRXN1 overexpression improved learning and memory performance of the SHR, and that expression of synapse-related hippocampal genes was consistent with changes in water maze learning ability. Furthermore, through RNA-seq sequencing, they preliminarily found that the mechanism may be related to the influence of the 5-hydroxytryptamine receptor [67].
Homer proteins, localized at the postsynaptic density of glutamatergic excitatory synapses, play a crucial role in cognitive function [68]. Hong et al. found that hippocampal levels of Homer 1a and 2a/b mRNA/protein are significantly lower in SHR compared with WKY rats. SHRs treated with MPH had higher learning and memory abilities, and levels of hippocampal Homer 1a and Homer 2a/b were up-regulated [69]. This indicates that Homer protein, Homer 1a, and Homer 2a/b may be involved in the mechanism of ADHD learning and memory deficits, though further research should be performed.
Risk genes
Cadherin 13 (CDH13) has been associated with impulsivity and hyperactivity in ADHD [70] (Table 2). Ziegler et al. revealed that a common genetic variation of CDH13 has an important impact on neural processing during working memory tasks [71]. Kiser et al. found that CDH13-deficient mice have cognitive impairments, highlighting that CDH13 is related to memory formation and cognitive flexibility. In addition, CDH13 KO mice have enhanced inhibitory driving force of pyramidal neurons in hippocampal CA1, resulting in excita-tory/inhibitory changes closely related to learning and memory processes [72]. Furthermore, Kiser et al. found that Cdh13-deficient mice have cognitive impairments, emphasizing that CDH13 is related to memory formation and cognitive flexibility [73]. Thus, CDH13 may contribute to symptomatic core dysfunctions of social and cognitive impairments in ADHD.
Table 2.
Some risk genes associated with ADHD and learning and WM impairment
Gene | Description | Findings | References |
---|---|---|---|
DAT1 | responsible for rapid uptake of DA from the synaptic cleft | modulate WM-related brain activity among ADHD children | [40–43] |
DRD4 | highly expressed in central nervous system and regulates signal transduction of nerve cell | 7-repeat allele of D4 receptor gene associated with worse working memory performance | [44–47] |
CDH13 | Crucial for neuro- developmental processes | impacts neural processing during working memory tasks | [70–73] |
GIT1 | widely expressed throughout the brain and related to intellectual disability genes | loss of GIT1 leads to cognitive dysfunctions in mice | [75–77] |
DCDC2 | a member of the doublecortin family related to abnormal neuronal migration | both heterozygous and homozygous mutations of Dcdc2 result in persistent visuo-spatial memory deficits, as well as visual discrimination and long-term memory deficits | [78–80] |
ATXN1 | interacts with large protein complexes, binds RNA, and is thought to be involved in transcriptional repression | ATXN1–CIC in forebrain leads to learning and memory deficits in mice | [81–84] |
COMT | important in dopaminergic neurotransmission. | numerous studies have also pointed that this COMT SNP is related to the executive function of ADHD but some meta-analysis revealed no association | [85–90] |
Gastrointestinal tract (GIT) proteins are widely expressed throughout the brain and largely related to genes influencing intellectual disability [74]. With advanced single molecule RNA sequencing, McCafrey et al. identified GIT1 as one of several differentially expressed genes between those with ADHD and control [75]. An intronic single-nucleotide polymorphism in GIT1, the minor allele of which causes reduced GIT1 expression, shows a strong association with ADHD susceptibility in humans. Martyn et al. found that GIT1 knockdown mice display learning and memory impairments; their exploration of the mechanism for this showed that GIT1 loss can reduce hippocampal synapse density and structural plasticity, leading to cognitive dysfunction [76]. Meanwhile, Won et al. found amphetamine can normalize the impaired memory of GIT1-deficient mice [77]. In summary, GIT1 plays a role in learning and memory impairments in ADHD.
As a common childhood central nervous system disorder, ADHD is often comorbid with other dysfunctions [78]. Studies have implicated doublecortin domain containing protein 2 (DCDC2), the reading disabilities risk gene, which is associated with hyperactivity, inattention, and impulsivity [79] (Table 2). And DCDC2 mutations can lead to long-term memory deficits [80]. Thus, further research exploring the role of DCDC2 in ADHD learning and memory impairments would be valuable.
Studies has already suggested a possible function of ataxin-1 (ATXN1) in learning and memory processes [81]. Ten years ago, Rizzi et al. found that ATXN1 and other SNPs in areas 6p25-21.2 and 14q11.2-12 are related to intelligence in ADHD [82]. In addition, a study of five individuals with de novo heterozygous truncating mutations in capicua (CIC) who share similar clinical features, including intellectual disability, ADHD, and autism spectrum disorder, indicated that ATXN1 capicua (ATXN1-CIC) may be closely related to some neurobehavioral phenotypes, including learning and memory deficits [83]. Hsiang-Chih et al. discovered that in mice, specific deletion of forebrain ATXN1-CIC leads to hyperactivity, learning and memory deficits, and abnormal maturation and maintenance of upper layer cortical neurons [83,84] (Table 2).
As catechol-O-methyltransferase (COMT) is involved in synaptic DA catabolism, many studies have focused on its role in ADHD [85–87]. Gothelf et al. found that low COMTVal158Met allele activity can increase ADHD risk [86]. COMT is not only related to ADHD behavior, it may also be associated with cortical thickness and surface area [88]. Numerous studies have also shown that this COMT SNP is related to executive function in ADHD [89]. However, a recent meta-analysis revealed no significant association between COMTVal158Met and ADHD [90] (Table 2). Therefore, more research should be performed to explore its role.
Conclusion
Impaired learning and memory in ADHD often seriously affects individual’s cognitive, academic, social skills, and other development, and may represent a heavy burden on families and society. However, the mechanism of learning and memory impairments in ADHD is as yet unclear, and effective clinical treatment methods are lacking. Herein, we described several aspects of the research progress related to learning and memory impairments in ADHD, to contribute a more comprehensive understanding of this disorder.
Acknowledgements
This work was supported by the Youth Program of the National Natural Science Foundation of China (No.82101618) and the Key Discipline of Maternal and Child Health in Jiangsu Province (FXK201705).
Footnotes
Conflict of Interest: There is no conflict of interest.
References
- 1.Andersen PN, Egeland J, Øie M. Learning and memory impairments in children and adolescents with attention-deficit/hyperactivity disorder. J Learn Disabil. 2013;46:453–460. doi: 10.1177/0022219412437040. [DOI] [PubMed] [Google Scholar]
- 2.Austerman J. ADHD and behavioral disorders: Assessment, management, and an update from DSM-5. Cleve Clin J Med. 2015;82:S2–7. doi: 10.3949/ccjm.82.s1.01. [DOI] [PubMed] [Google Scholar]
- 3.D’esposito M, Detre JA, Alsop DC, Shin RK, Atlas S, Grossman M. The neural basis of the central executive system of working memory. Nature. 1995;378:279–281. doi: 10.1038/378279a0. [DOI] [PubMed] [Google Scholar]
- 4.Baddeley A. Working memory and language: an overview. J Commun Disord. 2003;36:189–208. doi: 10.1016/S0021-9924(03)00019-4. [DOI] [PubMed] [Google Scholar]
- 5.Cohen R, Cohen-Kroitoru B, Halevy A, Aharoni S, Aizenberg I, Shuper A. Handwriting in children with Attention Deficient Hyperactive Disorder: role of graphology. BMC Pediatr. 2019;19:484. doi: 10.1186/s12887-019-1854-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Tibu F, Sheridan MA, Mclaughlin KA, Nelson CA, Fox NA, Zeanah CH. Disruptions of working memory and inhibition mediate the association between exposure to institutionalization and symptoms of attention deficit hyperactivity disorder. Psychol Med. 2016;46:529–541. doi: 10.1017/S0033291715002020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Tibu F, Sheridan MA, Mclaughlin KA, Nelson CA, Fox NA, Zeanah CH. Reduced working memory mediates the link between early institutional rearing and symptoms of ADHD at 12 Years. Front Psychol. 2016;7:1850. doi: 10.3389/fpsyg.2016.01850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Willcutt EG, Doyle AE, Nigg JT, Faraone SV, Pennington BF. Validity of the executive function theory of attention-deficit/hyperactivity disorder: a meta-analytic review. Biol Psychiatry. 2005;57:1336–1346. doi: 10.1016/j.biopsych.2005.02.006. [DOI] [PubMed] [Google Scholar]
- 9.Phillips MS, Bing-Canar H, Shields AN, Cerny B, Chang F, Wisinger AM, Leib SI, et al. Assessment of learning and memory impairments in adults with predominately inattentive versus combined presentation attention-deficit/hyperactivity disorder. Appl Neuropsychol Adult. 2023:1–10. doi: 10.1080/23279095.2023.2169887. [DOI] [PubMed] [Google Scholar]
- 10.Peterson K, Mcdonagh MS, Fu R. Comparative benefits and harms of competing medications for adults with attention-deficit hyperactivity disorder: a systematic review and indirect comparison meta-analysis. Psychopharmacology (Berl) 2008;197:1–11. doi: 10.1007/s00213-007-0996-4. [DOI] [PubMed] [Google Scholar]
- 11.Quinn D. Does chirality matter? pharmacodynamics of enantiomers of methylphenidate in patients with attention-deficit/hyperactivity disorder. J Clin Psychopharmacol. 2008;28:S62–S66. doi: 10.1097/JCP.0b013e3181744aa6. [DOI] [PubMed] [Google Scholar]
- 12.Li J, Wang Y, Zhou R, Zhang H, Yang L, Wang B, Khan S, et al. Serotonin 5-HT1B receptor gene and attention deficit hyperactivity disorder in Chinese Han subjects. Am J Med Genet B Neuropsychiatr Genet. 2005;132b:59–63. doi: 10.1002/ajmg.b.30075. [DOI] [PubMed] [Google Scholar]
- 13.Jensen V, Rinholm JE, Johansen TJ, Medin T, Storm-Mathisen J, Sagvolden T, Hvalby O, et al. N-methyl-D-aspartate receptor subunit dysfunction at hippocampal glutamatergic synapses in an animal model of attention-deficit/hyperactivity disorder. Neuroscience. 2009;158:353–364. doi: 10.1016/j.neuroscience.2008.05.016. [DOI] [PubMed] [Google Scholar]
- 14.Corominas-Roso M, Ramos-Quiroga JA, Ribases M, Sanchez-Mora C, Palomar G, Valero S, Bosch R, et al. Decreased serum levels of brain-derived neurotrophic factor in adults with attention-deficit hyperactivity disorder. Int J Neuropsychopharmacol. 2013;16:1267–1275. doi: 10.1017/S1461145712001629. [DOI] [PubMed] [Google Scholar]
- 15.Eilertsen EM, Gjerde LC, Kendler KS, Røysamb E, Aggen SH, Gustavson K, Reichborn-Kjennerud T, et al. Development of ADHD symptoms in preschool children: Genetic and environmental contributions. Dev Psychopathol. 2019;31:1299–1305. doi: 10.1017/S0954579418000731. [DOI] [PubMed] [Google Scholar]
- 16.Kian N, Samieefar N, Rezaei N. Prenatal risk factors and genetic causes of ADHD in children. World J Pediatr. 2022;18:308–319. doi: 10.1007/s12519-022-00524-6. [DOI] [PubMed] [Google Scholar]
- 17.Faraone SV, Larsson H. Genetics of attention deficit hyperactivity disorder. Mol Psychiatry. 2019;24:562–575. doi: 10.1038/s41380-018-0070-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ochozková A, Mihalčíková L, Yamamotová A, Šlamberová R. Can prenatal methamphetamine exposure be considered a good animal model for ADHD? Physiol Res. 2021;70:S431–s440. doi: 10.33549/physiolres.934815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Regan SL, Williams MT, Vorhees CV. Review of rodent models of attention deficit hyperactivity disorder. Neurosci Biobehav Rev. 2022;132:621–637. doi: 10.1016/j.neubiorev.2021.11.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Meneses A, Perez-Garcia G, Ponce-Lopez T, Tellez R, Gallegos-Cari A, Castillo C. Spontaneously hypertensive rat (SHR) as an animal model for ADHD: a short overview. Rev Neurosci. 2011;22:365–371. doi: 10.1515/rns.2011.024. [DOI] [PubMed] [Google Scholar]
- 21.Kishikawa Y, Kawahara Y, Yamada M, Kaneko F, Kawahara H, Nishi A. The spontaneously hypertensive rat/Izm (SHR/Izm) shows attention deficit/hyperactivity disorder-like behaviors but without impulsive behavior: therapeutic implications of low-dose methylphenidate. Behav Brain Res. 2014;274:235–242. doi: 10.1016/j.bbr.2014.08.026. [DOI] [PubMed] [Google Scholar]
- 22.Oades RD, Sadile AG, Sagvolden T, Viggiano D, Zuddas A, Devoto P, Aase H, et al. The control of responsiveness in ADHD by catecholamines: evidence for dopaminergic, noradrenergic and interactive roles. Dev Sci. 2005;8:122–31. doi: 10.1111/j.1467-7687.2005.00399.x. [DOI] [PubMed] [Google Scholar]
- 23.Miller EM, Pomerleau F, Huettl P, Gerhardt GA, Glaser PE. Aberrant glutamate signaling in the prefrontal cortex and striatum of the spontaneously hypertensive rat model of attention-deficit/hyperactivity disorder. Psychopharmacology (Berl) 2014;231:3019–29. doi: 10.1007/s00213-014-3479-4. [DOI] [PubMed] [Google Scholar]
- 24.Sterley TL, Howells FM, Russell VA. Genetically determined differences in noradrenergic function: The spontaneously hypertensive rat model. Brain Res. 2016;1641:291–305. doi: 10.1016/j.brainres.2015.11.019. [DOI] [PubMed] [Google Scholar]
- 25.Fasmer OB, Johansen EB. Patterns of motor activity in spontaneously hypertensive rats compared to Wistar Kyoto rats. Behav Brain Funct. 2016;12:32. doi: 10.1186/s12993-016-0117-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Nakamura-Palacios EM, Caldas CK, Fiorini A, Chagas KD, Chagas KN, Vasquez EC. Deficits of spatial learning and working memory in spontaneously hypertensive rats. Behav Brain Res. 1996;74:217–227. doi: 10.1016/0166-4328(95)00165-4. [DOI] [PubMed] [Google Scholar]
- 27.Yuan H, Ni X, Zheng M, Han X, Song Y, Yu M. Effect of catalpol on behavior and neurodevelopment in an ADHD rat model. Biomed Pharmacother. 2019;118:109033. doi: 10.1016/j.biopha.2019.109033. [DOI] [PubMed] [Google Scholar]
- 28.Mereu M, Contarini G, Buonaguro EF, Latte G, Managò F, Iasevoli F, De Bartolomeis A, et al. Dopamine transporter (DAT) genetic hypofunction in mice produces alterations consistent with ADHD but not schizophrenia or bipolar disorder. Neuropharmacology. 2017;121:179–194. doi: 10.1016/j.neuropharm.2017.04.037. [DOI] [PubMed] [Google Scholar]
- 29.Takamatsu Y, Hagino Y, Sato A, Takahashi T, Nagasawa SY, Kubo Y, Mizuguchi M, et al. Improvement of learning and increase in dopamine level in the frontal cortex by methylphenidate in mice lacking dopamine transporter. Curr Mol Med. 2015;15:245–52. doi: 10.2174/1566524015666150330144018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zhu X, Li T, Peng S, Ma X, Chen X, Zhang X. Maternal deprivation-caused behavioral abnormalities in adult rats relate to a non-methylation-regulated D2 receptor levels in the nucleus accumbens. Behav Brain Res. 2010;209:281–288. doi: 10.1016/j.bbr.2010.02.005. [DOI] [PubMed] [Google Scholar]
- 31.Morice E, Denis C, Giros B, Nosten-Bertrand M. Phenotypic expression of the targeted null-mutation in the dopamine transporter gene varies as a function of the genetic background. Eur J Neurosci. 2004;20:120–126. doi: 10.1111/j.1460-9568.2004.03465.x. [DOI] [PubMed] [Google Scholar]
- 32.Meza-Aguilar DG, Boucard AA. Latrophilins updated. Biomol Concepts. 2014;5:457–478. doi: 10.1515/bmc-2014-0032. [DOI] [PubMed] [Google Scholar]
- 33.Özaslan A, Güney E, Ergün MA, Okur İ, Yapar D. CDH13 and LPHN3 gene polymorphisms in attention-deficit/hyperactivity disorder: their relation to clinical characteristics. J Mol Neurosci. 2021;71:394–408. doi: 10.1007/s12031-020-01662-0. [DOI] [PubMed] [Google Scholar]
- 34.Regan SL, Pitzer EM, Hufgard JR, Sugimoto C, Williams MT, Vorhees CV. A novel role for the ADHD risk gene latrophilin-3 in learning and memory in Lphn3 knockout rats. Neurobiol Dis. 2021;158:105456. doi: 10.1016/j.nbd.2021.105456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Regan SL, Hufgard JR, Pitzer EM, Sugimoto C, Hu YC, Williams MT, Vorhees CV. Knockout of latrophilin-3 in Sprague-Dawley rats causes hyperactivity, hyper-reactivity, under-response to amphetamine, and disrupted dopamine markers. Neurobiol Dis. 2019;130:104494. doi: 10.1016/j.nbd.2019.104494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Fallgatter AJ, Ehlis AC, Dresler T, Reif A, Jacob CP, Arcos-Burgos M, Muenke M, et al. Influence of a latrophilin 3 (LPHN3) risk haplotype on event-related potential measures of cognitive response control in attention-deficit hyperactivity disorder (ADHD) Eur Neuropsychopharmacol. 2013;23:458–468. doi: 10.1016/j.euroneuro.2012.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Li X, Wu X, Luo P, Xiong L. Astrocyte-specific NDRG2 gene: functions in the brain and neurological diseases. Cell Mol Life Sci. 2020;77:2461–2472. doi: 10.1007/s00018-019-03406-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Li Y, Yin A, Sun X, Zhang M, Zhang J, Wang P, Xie R, et al. Deficiency of tumor suppressor NDRG2 leads to attention deficit and hyperactive behavior. J Clin Invest. 2017;127:4270–4284. doi: 10.1172/JCI94455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Rhodes SM, Coghill DR, Matthews K. Methylphenidate restores visual memory, but not working memory function in attention deficit-hyperkinetic disorder. Psychopharmacology (Berl) 2004;175:319–330. doi: 10.1007/s00213-004-1833-7. [DOI] [PubMed] [Google Scholar]
- 40.Kang AM, Palmatier MA, Kidd KK. Global variation of a 40-bp VNTR in the 3′-untranslated region of the dopamine transporter gene (SLC6A3) Biol Psychiatry. 1999;46:151–160. doi: 10.1016/S0006-3223(99)00101-8. [DOI] [PubMed] [Google Scholar]
- 41.Zuschlag ZD, Compean E, Nietert P, Lauzon S, Hamner M, Wang Z. Dopamine transporter (DAT1) gene in combat veterans with PTSD: A case-control study. Psychiatry Res. 2021;298:113801. doi: 10.1016/j.psychres.2021.113801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bacanlı A, Unsel-Bolat G, Suren S, Yazıcı KU, Callı C, Aygunes Jafari D, Kosova B, et al. Effects of the dopamine transporter gene on neuroimaging findings in different attention deficit hyperactivity disorder presentations. Brain Imaging Behav. 2021;15:1103–1114. doi: 10.1007/s11682-020-00437-w. [DOI] [PubMed] [Google Scholar]
- 43.Brown AB, Biederman J, Valera E, Makris N, Doyle A, Whitfield-Gabrieli S, Mick E, et al. Relationship of DAT1 and adult ADHD to task-positive and task-negative working memory networks. Psychiatry Res. 2011;193:7–16. doi: 10.1016/j.pscychresns.2011.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Li Q, Lu G, Antonio GE, Mak YT, Rudd JA, Fan M, Yew DT. The usefulness of the spontaneously hypertensive rat to model attention-deficit/hyperactivity disorder (ADHD) may be explained by the differential expression of dopamine-related genes in the brain. Neurochem Int. 2007;50:848–857. doi: 10.1016/j.neuint.2007.02.005. [DOI] [PubMed] [Google Scholar]
- 45.Yin P, Cao AH, Yu L, Guo LJ, Sun RP, Lei GF. ABT-724 alleviated hyperactivity and spatial learning impairment in the spontaneously hypertensive rat model of attention-deficit/hyperactivity disorder. Neurosci Lett. 2014;580:142–146. doi: 10.1016/j.neulet.2014.08.008. [DOI] [PubMed] [Google Scholar]
- 46.Loo SK, Rich EC, Ishii J, Mcgough J, Mccracken J, Nelson S, Smalley SL. Cognitive functioning in affected sibling pairs with ADHD: familial clustering and dopamine genes. J Child Psychol Psychiatry. 2008;49:950–957. doi: 10.1111/j.1469-7610.2008.01928.x. [DOI] [PubMed] [Google Scholar]
- 47.Brookes KJ, Xu X, Chen CK, Huang YS, Wu YY, Asherson P. No evidence for the association of DRD4 with ADHD in a Taiwanese population within-family study. BMC Med Genet. 2005;6:31. doi: 10.1186/1471-2350-6-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Schaefer TL, Vorhees CV, Williams MT. Mouse plasmacytoma-expressed transcript 1 knock out induced 5-HT disruption results in a lack of cognitive deficits and an anxiety phenotype complicated by hypoactivity and defensiveness. Neuroscience. 2009;164:1431–1443. doi: 10.1016/j.neuroscience.2009.09.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.González-Burgos I, Feria-Velasco A. Serotonin/dopamine interaction in memory formation. Prog Brain Res. 2008;172:603–623. doi: 10.1016/S0079-6123(08)00928-X. [DOI] [PubMed] [Google Scholar]
- 50.Salman T, Nawaz S, Waraich RS, Haleem DJ. Repeated administration of methylphenidate produces reinforcement and downregulates 5-HT-1A receptor expression in the nucleus accumbens. Life Sci. 2019;218:139–146. doi: 10.1016/j.lfs.2018.12.046. [DOI] [PubMed] [Google Scholar]
- 51.Salman T, Afroz R, Nawaz S, Mahmood K, Haleem DJ, Zarina S. Differential effects of memory enhancing and impairing doses of methylphenidate on serotonin metabolism and 5-HT1A, GABA, glutamate receptor expression in the rat prefrontal cortex. Biochimie. 2021;191:51–61. doi: 10.1016/j.biochi.2021.08.009. [DOI] [PubMed] [Google Scholar]
- 52.Zepf FD, Landgraf M, Biskup CS, Dahmen B, Poustka F, Wöckel L, Stadler C. No effect of acute tryptophan depletion on verbal declarative memory in young persons with ADHD. Acta Psychiatr Scand. 2013;128:133–141. doi: 10.1111/acps.12089. [DOI] [PubMed] [Google Scholar]
- 53.Huang X, Wang M, Zhang Q, Chen X, Wu J. The role of glutamate receptors in attention-deficit/hyperactivity disorder: From physiology to disease. Am J Med Genet B Neuropsychiatr Genet. 2019;180:272–286. doi: 10.1002/ajmg.b.32726. [DOI] [PubMed] [Google Scholar]
- 54.Maltezos S, Horder J, Coghlan S, Skirrow C, O’gorman R, Lavender TJ, Mendez MA, et al. Glutamate/glutamine and neuronal integrity in adults with ADHD: a proton MRS study. Transl Psychiatry. 2014;4:e373. doi: 10.1038/tp.2014.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Bauer J, Werner A, Kohl W, Kugel H, Shushakova A, Pedersen A, Ohrmann P. Hyperactivity and impulsivity in adult attention-deficit/hyperactivity disorder is related to glutamatergic dysfunction in the anterior cingulate cortex. World J Biol Psychiatry. 2018;19:538–546. doi: 10.1080/15622975.2016.1262060. [DOI] [PubMed] [Google Scholar]
- 56.Hiraoka Y, Sugiyama K, Nagaoka D, Tsutsui-Kimura I, Tanaka KF, Tanaka K. Mice with reduced glutamate transporter GLT1 expression exhibit behaviors related to attention-deficit/hyperactivity disorder. Biochem Biophys Res Commun. 2021;567:161–165. doi: 10.1016/j.bbrc.2021.06.057. [DOI] [PubMed] [Google Scholar]
- 57.Shikanai H, Oshima N, Kawashima H, Kimura SI, Hiraide S, Togashi H, Iizuka K, et al. N-methyl-d-aspartate receptor dysfunction in the prefrontal cortex of stroke-prone spontaneously hypertensive rat/Ezo as a rat model of attention deficit/hyperactivity disorder. Neuropsychopharmacol Rep. 2018;38:61–66. doi: 10.1002/npr2.12007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Cheng J, Xiong Z, Duffney LJ, Wei J, Liu A, Liu S, Chen GJ, et al. Methylphenidate exerts dose-dependent effects on glutamate receptors and behaviors. Biol Psychiatry. 2014;76:953–962. doi: 10.1016/j.biopsych.2014.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kawade HM, Borkar CD, Shambharkar AS, Singh O, Singru PS, Subhedar NK, Kokare DM. Intracellular mechanisms and behavioral changes in mouse model of attention deficit hyperactivity disorder: Importance of age-specific NMDA receptor blockade. Pharmacol Biochem Behav. 2020;188:172830. doi: 10.1016/j.pbb.2019.172830. [DOI] [PubMed] [Google Scholar]
- 60.Wilens TE, Decker MW. Neuronal nicotinic receptor agonists for the treatment of attention-deficit/hyperactivity disorder: focus on cognition. Biochem Pharmacol. 2007;74:1212–1223. doi: 10.1016/j.bcp.2007.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Guo T, Yang C, Guo L, Liu K. A comparative study of the effects of ABT-418 and methylphenidate on spatial memory in an animal model of ADHD. Neurosci Lett. 2012;528:11–15. doi: 10.1016/j.neulet.2012.08.068. [DOI] [PubMed] [Google Scholar]
- 62.Jeong HI, Ji ES, Kim SH, Kim TW, Baek SB, Choi SW. Treadmill exercise improves spatial learning ability by enhancing brain-derived neurotrophic factor expression in the attention-deficit/hyperactivity disorder rats. J Exerc Rehabil. 2014;10:162–167. doi: 10.12965/jer.140111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wilens TE, Verlinden MH, Adler LA, Wozniak PJ, West SA. ABT-089, a neuronal nicotinic receptor partial agonist, for the treatment of attention-deficit/hyperactivity disorder in adults: results of a pilot study. Biol Psychiatry. 2006;59:1065–1070. doi: 10.1016/j.biopsych.2005.10.029. [DOI] [PubMed] [Google Scholar]
- 64.Lu B, Nagappan G, Lu Y. BDNF and synaptic plasticity, cognitive function, and dysfunction. Handb Exp Pharmacol. 2014;220:223–250. doi: 10.1007/978-3-642-45106-5_9. [DOI] [PubMed] [Google Scholar]
- 65.Wang LJ, Wu CC, Lee MJ, Chou MC, Lee SY, Chou WJ. Peripheral brain-derived neurotrophic factor and contactin-1 levels in patients with attention-deficit/hyperactivity disorder. J Clin Med. 2019:8. doi: 10.3390/jcm8091366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Kasem E, Kurihara T, Tabuchi K. Neurexins and neuropsychiatric disorders. Neurosci Res. 2018;127:53–60. doi: 10.1016/j.neures.2017.10.012. [DOI] [PubMed] [Google Scholar]
- 67.Zhang S, Wu D, Xu Q, You L, Zhu J, Wang J, Liu Z, et al. The protective effect and potential mechanism of NRXN1 on learning and memory in ADHD rat models. Exp Neurol. 2021;344:113806. doi: 10.1016/j.expneurol.2021.113806. [DOI] [PubMed] [Google Scholar]
- 68.Luo P, Li X, Fei Z, Poon W. Scaffold protein Homer 1: implications for neurological diseases. Neurochem Int. 2012;61:731–738. doi: 10.1016/j.neuint.2012.06.014. [DOI] [PubMed] [Google Scholar]
- 69.Hong Q, Wang YP, Zhang M, Pan XQ, Guo M, Li F, Tong ML, et al. Homer expression in the hippocampus of an animal model of attention-deficit/hyperactivity disorder. Mol Med Rep. 2011;4:705–712. doi: 10.3892/mmr.2011.479. [DOI] [PubMed] [Google Scholar]
- 70.Salatino-Oliveira A, Genro JP, Polanczyk G, Zeni C, Schmitz M, Kieling C, Anselmi L, et al. Cadherin-13 gene is associated with hyperactive/impulsive symptoms in attention/deficit hyperactivity disorder. Am J Med Genet B Neuropsychiatr Genet. 2015;168b:162–169. doi: 10.1002/ajmg.b.32293. [DOI] [PubMed] [Google Scholar]
- 71.Ziegler GC, Ehlis AC, Weber H, Vitale MR, Zöller JEM, Ku HP, Schiele MA, et al. A common CDH13 variant is associated with low agreeableness and neural responses to working memory tasks in ADHD. Genes (Basel) 2021:12. doi: 10.3390/genes12091356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Rivero O, Selten MM, Sich S, Popp S, Bacmeister L, Amendola E, Negwer M, et al. Cadherin-13, a risk gene for ADHD and comorbid disorders, impacts GABAergic function in hippocampus and cognition. Transl Psychiatry. 2015;5:e655. doi: 10.1038/tp.2015.152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Forero A, Rivero O, Wäldchen S, Ku HP, Kiser DP, Gärtner Y, Pennington LS, et al. Cadherin-13 deficiency increases dorsal raphe 5-ht neuron density and prefrontal cortex innervation in the mouse brain. Front Cell Neurosci. 2017;11:307. doi: 10.3389/fncel.2017.00307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Shin EY, Lee CS, Cho TG, Kim YG, Song S, Juhnn YS, Park SC, et al. betaPak-interacting exchange factor-mediated Rac1 activation requires smgGDS guanine nucleotide exchange factor in basic fibroblast growth factor-induced neurite outgrowth. J Biol Chem. 2006;281:35954–64. doi: 10.1074/jbc.M602399200. [DOI] [PubMed] [Google Scholar]
- 75.Mccaffrey TA, St Laurent G, 3rd, Shtokalo D, Antonets D, Vyatkin Y, Jones D, Battison E, et al. Biomarker discovery in attention deficit hyperactivity disorder: RNA sequencing of whole blood in discordant twin and case-controlled cohorts. BMC Med Genomics. 2020;13:160. doi: 10.1186/s12920-020-00808-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Martyn AC, Toth K, Schmalzigaug R, Hedrick NG, Rodriguiz RM, Yasuda R, Wetsel WC, et al. GIT1 regulates synaptic structural plasticity underlying learning. PLoS One. 2018;13:e0194350. doi: 10.1371/journal.pone.0194350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Won H, Mah W, Kim E, Kim JW, Hahm EK, Kim MH, Cho S, et al. GIT1 is associated with ADHD in humans and ADHD-like behaviors in mice. Nat Med. 2011;17:566–572. doi: 10.1038/nm.2330. [DOI] [PubMed] [Google Scholar]
- 78.Couto JM, Gomez L, Wigg K, Ickowicz A, Pathare T, Malone M, Kennedy JL, et al. Association of attention-deficit/hyperactivity disorder with a candidate region for reading disabilities on chromosome 6p. Biol Psychiatry. 2009;66:368–75. doi: 10.1016/j.biopsych.2009.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Willcutt EG, Pennington BF, Smith SD, Cardon LR, Gayán J, Knopik VS, Olson RK, et al. Quantitative trait locus for reading disability on chromosome 6p is pleiotropic for attention-deficit/hyperactivity disorder. Am J Med Genet. 2002;114:260–268. doi: 10.1002/ajmg.10205. [DOI] [PubMed] [Google Scholar]
- 80.Gabel LA, Marin I, Loturco JJ, Che A, Murphy C, Manglani M, Kass S. Mutation of the dyslexia-associated gene Dcdc2 impairs LTM and visuo-spatial performance in mice. Genes Brain Behav. 2011;10:868–875. doi: 10.1111/j.1601-183X.2011.00727.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Matilla A, Roberson ED, Banfi S, Morales J, Armstrong DL, Burright EN, Orr HT, et al. Mice lacking ataxin-1 display learning deficits and decreased hippocampal paired-pulse facilitation. J Neurosci. 1998;18:5508–5516. doi: 10.1523/JNEUROSCI.18-14-05508.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Rizzi TS, Arias-Vasquez A, Rommelse N, Kuntsi J, Anney R, Asherson P, Buitelaar J, et al. The ATXN1 and TRIM31 genes are related to intelligence in an ADHD background: evidence from a large collaborative study totaling 4,963 subjects. Am J Med Genet B Neuropsychiatr Genet. 2011;156:145–157. doi: 10.1002/ajmg.b.31149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Lu HC, Tan Q, Rousseaux MW, Wang W, Kim JY, Richman R, Wan YW, et al. Disruption of the ATXN1-CIC complex causes a spectrum of neurobehavioral phenotypes in mice and humans. Nat Genet. 2017;49:527–536. doi: 10.1038/ng.3808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Celestino-Soper PB, Skinner C, Schroer R, Eng P, Shenai J, Nowaczyk MM, Terespolsky D, et al. Deletions in chromosome 6p22.3-p24.3, including ATXN1, are associated with developmental delay and autism spectrum disorders. Mol Cytogenet. 2012;5:17. doi: 10.1186/1755-8166-5-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Laatikainen LM, Sharp T, Harrison PJ, Tunbridge EM. Sexually dimorphic effects of catechol-O-methyltransferase (COMT) inhibition on dopamine metabolism in multiple brain regions. PLoS One. 2013;8:e61839. doi: 10.1371/journal.pone.0061839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Gothelf D, Michaelovsky E, Frisch A, Zohar AH, Presburger G, Burg M, Aviram-Goldring A, et al. Association of the low-activity COMT 158Met allele with ADHD and OCD in subjects with velocardiofacial syndrome. Int J Neuropsychopharmacol. 2007;10:301–308. doi: 10.1017/S1461145706006699. [DOI] [PubMed] [Google Scholar]
- 87.Eisenberg J, Mei-Tal G, Steinberg A, Tartakovsky E, Zohar A, Gritsenko I, Nemanov L, et al. Haplotype relative risk study of catechol-O-methyltransferase (COMT) and attention deficit hyperactivity disorder (ADHD): association of the high-enzyme activity Val allele with ADHD impulsive-hyperactive phenotype. Am J Med Genet. 1999;88:497–502. doi: 10.1002/(SICI)1096-8628(19991015)88:5<497::AID-AJMG12>3.0.CO;2-F. [DOI] [PubMed] [Google Scholar]
- 88.Jung M, Mizuno Y, Fujisawa TX, Takiguchi S, Kong J, Kosaka H, Tomoda A. The effects of COMT polymorphism on cortical thickness and surface area abnormalities in children with ADHD. Cereb Cortex. 2019;29:3902–3911. doi: 10.1093/cercor/bhy269. [DOI] [PubMed] [Google Scholar]
- 89.Jin J, Liu L, Gao Q, Chan RC, Li H, Chen Y, Wang Y, et al. The divergent impact of COMT Val158Met on executive function in children with and without attention-deficit/hyperactivity disorder. Genes Brain Behav. 2016;15:271–279. doi: 10.1111/gbb.12270. [DOI] [PubMed] [Google Scholar]
- 90.Kang P, Luo L, Peng X, Wang Y. Association of Val158Met polymorphism in COMT gene with attention-deficit hyperactive disorder: An updated meta-analysis. Medicine (Baltimore) 2020;99:e23400. doi: 10.1097/MD.0000000000023400. [DOI] [PMC free article] [PubMed] [Google Scholar]