Home Random Page


CATEGORIES:

BiologyChemistryConstructionCultureEcologyEconomyElectronicsFinanceGeographyHistoryInformaticsLawMathematicsMechanicsMedicineOtherPedagogyPhilosophyPhysicsPolicyPsychologySociologySportTourism






Externalizing behaviors

As mentioned previously in the ADHD section, ADHD is often accompanied by ODD starting in early childhood, and by CD diagnosed in late childhood – adolescence or by APD diagnosed after age 18 (Biederman, 2005). According to DSM-IV (American Psychiatric Association, 1994), ODD is characterized by negativistic, hostile, and defiant behaviors (e.g., actively defying or refusing to comply with adults requests or rules, deliberately annoying people, blaming others for his or her mistakes or misbehavior). Whereas CD symptoms include aggressive and destructive behaviors (e.g., cruel behavior toward people or animals, destruction of property, stealing), and violations of rules. After the age of 18, CD may develop into APD, which is characterized by the above mentioned aggressive and destructive behaviors with an apparent lack of remorse. APD symptoms can also include recurring difficulties with the law, irresponsible work behavior, and abusive relationships. It has been shown that childhood ADHD predicts adult substance abuse, but it has also been demonstrated that early intervention and treatment in ADHD can prevent later illicit substance abuse during adolescence (Faraone and Wilens, 2003). Externalizing disorders, such as CD, APD, and substance abuse showed 80% heritability in a large-scale twin study conducted among 542 families (the Minnesota Twin Family Study, Hicks et al., 2004). Using a quantitative genetic model, the same workgroup could detect a gene × environment interaction in the development of externalizing disorders showing that genetic factors exhibited a greater effect under environmental adversity (Hicks et al., 2009). Using candidate gene analyses, a reproducible gene × environment interaction involving the MAOA uVNTR and childhood adversities has been shown for antisocial behaviors in independent association studies (reviewed by Thapar et al., 2007).

In a large Dutch family study a rare mutation in the X chromosome indicated that MAOA is involved in aggressive behavior, because aggressive and violent behavior was observed in MAOA deficient men (Brunner et al., 1993). Later, an animal study supported this observation as mice lacking the MAOA enzyme also exhibited enhanced aggression during adulthood (Cases et al., 1995). Since the gene encoding MAOA is localized on the X chromosome, robust effects are usually observed in males. In addition, it is not recommended to include heterozygote females in the analyses because of the potential inactivation of one of the X chromosomes; therefore, most association studies included only male subjects. The first study reporting gene × environment interaction regarding the MAOA uVNTR and childhood maltreatment showed that males with the low-activity MAOA genotype in the severe maltreatment group were more likely to develop CD in adolescence, had more APD symptoms at age 26, and were convicted for violent offence more frequently compared to those maltreated subjects with the high-activity MAOA genotype (Caspi et al., 2002). Although some subsequent studies reported negative findings concerning the interaction between the MAOA genotype and childhood maltreatment, the results of a meta-analysis supported the original finding (Taylor and Kim-Cohen, 2007). A recent study suggested that this interactive effect of MAOA and childhood maltreatment can be observed only at moderate levels of trauma exposure, because extreme levels of trauma appear to overshadow the effects of MAOA genotype (Weder et al., 2009). The underlying neural mechanisms responsible for the MAOA findings have been localized in the limbic system, as low-activity MAOA genotype men had a reduced limbic volume and a higher amygdala responsiveness during emotional arousal (Meyer-Lindenberg et al., 2006).



MAOA metabolizes serotonin and norepinephrine in addition to dopamine, thus the MAOA genetic findings could be attributed to other monoamine theories. For purely dopaminergic genes, the picture is less clear: three longitudinal, general population-based studies and three clinical sample-based studies investigating adolescents with ADHD or CD resulted in contradictory findings for the DAT1 3′ UTR VNTR. The 9-repeat allele or the 9/10 genotype (vs. 10/10) was associated with higher levels of externalizing symptoms (Young et al., 2002, Barkley et al., 2006), but this finding was not supported by later studies using either community-based samples (Jorm et al., 2001; Caspi et al., 2008) or clinical samples (Caspi et al., 2008; Schulz-Heik et al., 2008). One possible explanation for these contradictory findings may be the distinct underlying factors for violent, physically aggressive behaviors, and for non-aggressive, rule-breaking antisocial behaviors. This explanation was presented by Burt and Mikolajewski (2008), who reported that more rule-breaking behavior was observed in male undergraduates with the DAT1 10/10 genotype than those without this genotype. At the dopamine receptor genes, a gene × gene interaction was described in connection to adolescent CD problems and adult antisocial behaviors in a large, population-based study: males with the DRD2 A1-allele and DRD4 7-repeat allele showed more CD symptoms (Beaver et al., 2007).

In terms of the COMT Val158Met polymorphism, CD and APD were studied in the general population (birth cohorts) and among subjects diagnosed with ADHD. The Val/Val genotype was repeatedly associated with more CD and aggressive symptoms among children with ADHD (Caspi et al., 2008; Monuteaux et al., 2009; DeYoung et al., 2010) but not among children without ADHD (Caspi et al., 2008). In addition to the association of CD symptoms and the COMT Val/Val genotype in ADHD children, there was a gene × environment interaction described: children with the Val/Val genotype were more susceptible to the adverse effects of prenatal risk as indexed by lower birth weight (Thapar et al., 2007). Other gene × environment interaction findings in connection to antisocial behavior among ADHD children were also reported in the Cardiff ADHD genetic study, for example between DAT1 and DRD5 polymorphisms and maternal smoking during pregnancy (Thapar et al., 2007). Future analyses from independent studies are needed to support these findings.

Novelty seeking

One possible dimensional approach to impulsive traits and substance use disorders is the assessment of risk-taking behaviors (defined as high levels of risk-taking, exploration, and novelty or sensation seeking). These behaviors are especially characteristic of adolescence (Romer, 2010), and it would be useful to conduct a longitudinal study related to genetic vulnerability; however, most of the genetic association studies are carried out in young adult (student) populations. Several studies have shown that individuals exhibiting high novelty or sensation seeking personality traits are at an increased risk for using drugs of abuse (Bardo et al., 1996; Zuckerman and Kuhlman, 2000). Studies have consistently shown that polysubstance abusers have particularly high levels of impulsivity and sensation seeking (Conway et al., 2003). In most of the prevailing personality models, one main dimension is novelty seeking or impulsivity or extraversion, which are generally referred to as approach-related traits (Munafo et al., 2008). Novelty seeking is measured using the Tridimensional Personality Questionnaire (TPQ) or the Temperament and Character Inventory (TCI). Extraversion is one of the main dimensions of the Neuroticism Extraversion Openness (NEO) Personality Inventory and the Eysenck Personality Inventory or Questionnaire (EPI or EPQ). Where as the Karolinska Scales of Personality (KSP) has an impulsivity scale. Association studies typically use only one questionnaire, however, dimensions measuring the same construct are highly correlated. Therefore, comparison of genetic findings is possible with the assumption that all of these interrelated traits reflect a common underlying neurobiological motivational mechanism. Because the psychobiological model of TPQ/TCI provides a well-established theoretical framework for an association between the dopamine system and novelty seeking temperament dimension (Cloninger et al., 1993), the original novelty seeking scores or transformed scores from other impulsivity scales are usually reported in dopaminergic genetic association studies.

A substantial genetic component of personality traits has been demonstrated in twin and adoption studies. For example, a large Australian twin study reported heritability for TCI temperament scales that ranged from 30% to 40% (Gillespie et al., 2003). To investigate the specific genetic factors, several candidate gene studies have been conducted, initiated by the groundbreaking association of novelty seeking and the DRD4 7-repeat (or long) allele (Benjamin et al., 1996; Ebstein et al., 1996). Studies aiming to replicate this association resulted in contradictory findings, probably because the indicated genetic factor has a small or modest effect size (Ebstein et al., 2000). Therefore, increasing the size of the studied population and/or utilizing published results for meta-analyses may help to settle the contradictions. Based on a recent meta-analysis, the DRD4 VNTR does not seem to have a significant effect on novelty seeking, whereas the −521 C/T (rs1800955) SNP probably affects this trait (Munafo et al., 2008). The DRD4 −521 T-allele carriers had lower scores on approach-related trait scales; interestingly, a significant association could be observed for novelty seeking (TCI/TPQ) and impulsivity (KSP) but not for extraversion (NEO or EPI/EPQ). This finding leads to questioning the basic assumption that these traits assess the same neurobiological mechanism. An early meta-analysis of the other dopaminergic polymorphisms did not show any significant effect for DRD2 or DAT1 polymorphisms, only the DRD3 Ser9Gly SNP was associated with approach-related traits (Munafo et al., 2003). It is important to mention that the social environment might have a significant and possibly interacting effect. In a longitudinal study, for example, higher novelty seeking scores in the DRD2 A1-allele carriers were observed only if participants had a negative, punitive environment in childhood, and the DRD2 genotype had no effect under more favorable conditions (Keltikangas-Järvinen et al., 2009).

There are only a few MAOA and COMT related studies, and their meta-analyses have not yet been carried out. Interesting findings have been reported in regard to the COMT Val158Met polymorphism. A three-way interaction was observed between COMT, DRD4, and the serotonin transporter polymorphism (5-HTTLPR) on novelty seeking. In the absence of the short 5-HTTLPR allele together with the presence of the high-activity Val/Val COMT genotype, the novelty seeking scores were higher in the DRD4 7+ individuals in two independent populations (Benjamin et al., 2000; Strobel et al., 2003). In terms of solely the COMT genetic effect, the results are not in agreement with one another. The low-activity Met/Met genotype was associated with a high level of novelty seeking (Golimbet et al., 2007), whereas the high-activity Val/Val homozygotes were reported to have higher extraversion scores (Reuter and Hennig, 2005).

A number of studies assessed personality traits as part of the genetic investigations of substance use disorders. For example, higher novelty seeking score was associated with the DRD4 7-repeat allele among heavy-drinking college students (Ray et al., 2009) and in adolescent boys from a high-risk community sample (Laucht et al., 2007). In addition, two preliminary studies were conducted among methamphetamine dependents. Consistent with previous findings on drug users, methamphetamine users had higher novelty seeking scores than controls in both samples. Within the patient groups, a subgroup of the DRD2 A1-allele carriers (Han et al., 2008) or COMT Met-allele carriers (Hosak et al., 2006) had the highest novelty seeking scores. The COMT genetic finding was supported in opiate dependence by our laboratory. We reported increased novelty seeking scores in the presence of the Met-allele among 117 heroin-dependent subjects that participated in a methadone substitution program (Demetrovics et al., 2010).

The self-report questionnaires have strong limitations in measuring human personality dimensions. To better understand the genetic underpinnings of personality, novel and complex experimental paradigms are suggested for further studies, such as broader personality phenotypes (e.g., altruism and pro-social behavior) or objectively measurable endophenotypes from neurophysiological tests (e.g., event-related potentials and prepulse inhibition) or from computer games (Ebstein, 2006). The latter approach has been successfully applied recently for testing a reward-related endophenotype by the Iowa Gambling Task. The DRD4 long allele carriers had higher novelty seeking scores and showed elevated levels of risk-taking behavior (Roussos et al., 2009). Also, emotional processes were assessed in a subset of subjects by startle reactivity. The DRD4 long allele was associated with constricted emotional responses. Another gambling test study measuring event-related potentials did not report a DRD4 −521 C/T SNP effect (unfortunately, the analysis of the DRD4 VNTR was not reported) but showed an increased amplitude of the medial frontal negativity (a larger difference between the gain and loss conditions) in COMT Val/Val homozygotes compared to Met/Met homozygotes. The Val/Val group also exhibited greater beta activity during gain trials compared to the Met/Met group (Marco-Pallares et al., 2009). If these genetic findings are replicated, the objective phenotypes that characterize risk-taking behaviors could be helpful in genetic association studies and diagnostic processes. Animal models for risk-taking behaviors are numerous, and they would be useful to elucidate the underlying neurobiological mechanisms of impulsive decision-making. Adolescent rats, like their human counterparts, exhibit increased risk-taking and novelty seeking behaviors and drug use (Laviola et al., 2003). Therefore, animal studies can be used to assess this vulnerable period of development.

Go to:

CONCLUSIONS

We would like to emphasize that genetic association studies do not aim to find good or bad gene variants. Identifying the genetic factors in the background of heritable disorders can help us to better understand the underlying neural mechanisms of the disorder and the related behaviors (e.g., ADHD and impulsive behaviors). Psychiatric disorders can be seen as extremes of certain behaviors. In many cases, a small portion of these extreme behaviors are evolutionarily beneficial in human populations, because they maintain diversity. For example, impulsive risk-taking behavior in a minority of the population can aid survival (Williams and Taylor, 2006). Therefore, gene variants that have been indicated as risk factors in certain psychiatric disorders might have favorable effects under special circumstances.

Since single genetic factors have only small effects on complex inheritance disorders and traits, refining the studied phenotype is necessary. Although heterogeneous disorder categories have been continually replaced by quantitative trait measures in psychiatric genetic studies during the last decade, current parent-rated symptom scales and self-report questionnaires still do not provide the best phenotypes. Objectively measured endophenotypes, such as event-related potentials, fMRI signals that mirror regional brain activity, reaction time measures, or error rates on standardized computerized neurocognitive tests would help us to decipher specific genetic effects. Based on the presented dopaminergic genetic data (summarized in Table 1), we can conclude that dopamine D2 receptors in the subcortical brain regions may be important in reward-related associative learning and behavioral inhibition, as DRD2 gene variants resulting in reduced expression (A1-, B1-, and 957 C-allele) are repeatedly linked to substance abuse and impulsive phenotypes. The widely investigated DRD4 exon 3 VNTR has an influence on PFC-related executive functioning; however, recent results also point out the importance of DRD4 promoter variants. The COMT Val158Met SNP affects cognitive functions by influencing cortical dopamine level. This polymorphism is likely the best example of the good vs. the bad effect of a specific gene variant. The approximate 50–50% frequency of the Val- and Met-alleles of this functional polymorphism shows that both variants can have advantageous effects. It has been proposed that the Val-variant is associated with better cognitive flexibility, whereas the Met-variant has been reported to lead to an advantage on memory and attention tasks which require stability (Bilder et al., 2004). The dopamine transporter is a key component of dopamine transmission in the basal ganglia, and therefore, it is important in inhibitory control. However, specific DAT1 gene variants with convincing functional effects have not yet been confirmed. The low-activity allele of the MAOA uVNTR has been shown to be related to aggressive traits. This association could be explained by dopaminergic or serotonergic pathways because this enzyme converts monoamines (dopamine, norepinephrine, and serotonin). The MAOA effect shows the importance of gene × environment interactions. As it can be noticed in recent review papers (Grisham et al., 2008; Nigg et al., 2010), the environmental factors which have been convincingly replicated as risk factors in childhood-onset psychiatric disorders, such as low birth weight and physical or sexual abuse, are not specific to these disorders. Finding specific genetic factors that make individuals vulnerable or resilient to harmful environmental factors is the prevailing approach in present psychiatric genetic research. It is important to note that newer hypotheses refer to the MAOA uVNTR and to the other most commonly researched dopaminergic and serotonergic polymorphisms, i.e., the DRD4 VNTR and the 5-HTTLPR, as plasticity and not vulnerability factors, arguing that the indicated gene variants make individuals more sensitive to environmental influences (to both positive and negative influences) (Belsky et al., 2009). Early adverse environments might exert their effects via epigenetic modulations (Meaney, 2010), which should be taken into account when trying to understand the biological processes leading to disorders and maladaptive behaviors. Longitudinal population-based studies collecting environmental data and animal studies controlling for specific environmental factors would help shed light on the important steps in the development of psychopathologies.

Table 1

Dopaminergic genetic risk factors of psychiatric disorders and related traits.

Go to:

Highlights

  • description of functional polymorphisms in the dopaminergic genes
  • dopaminergic genetic findings of psychiatric disorders in adolescence, e.g., ADHD
  • genetic findings of quantitative traits, like impulsive and externalizing behaviors
  • genetic findings of objective endophenotypes, like attentional performance

Go to:

Acknowledgments

This work was supported by the NIH R03 TW007656 Fogarty International Research grant awarded to Maria Sasvari-Szekely and by the Hungarian fund OTKA F67784, awarded to Zsofia Nemoda. We thank Krisztina Lakatos for valuable discussions.

Go to:

Footnotes

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

Go to:

References

  1. Abramowitz JS, Taylor S, McKay D. Obsessive-compulsive disorder. Lancet. 2009;374:491–499. [PubMed]
  2. Adriani W, Boyer F, Gioiosa L, Macri S, Dreyer JL, Laviola G. Increased impulsive behavior and risk proneness following lentivirus-mediated dopamine transporter over-expression in rats’ nucleus accumbens. Neuroscience. 2009;159:47–58. [PubMed]
  3. Agrawal A, Lynskey MT. Candidate genes for cannabis use disorders: findings, challenges and directions. Addiction. 2009;104:518–532. [PMC free article] [PubMed]
  4. Albanese V, Biguet NF, Kiefer H, Bayard E, Mallet J, Meloni R. Quantitative effects on gene silencing by allelic variation at a tetranucleotide microsatellite. Hum Mol Genet. 2001;10:1785–1792. [PubMed]
  5. Albin RL, Mink JW. Recent advances in Tourette syndrome research. Trends Neurosci. 2006;29:175–182. [PubMed]
  6. Almasy L, Blangero J. Endophenotypes as quantitative risk factors for psychiatric disease: rationale and study design. Am J Med Genet. 2001;105:42–44. [PubMed]
  7. American Psychiatric Association. Diagnostic and statistical manual of mental disorders. American Psychiatric Press; Washington, DC: 1994.
  8. Andrews G, Goldberg DP, Krueger RF, Carpenter WT, Hyman SE, Sachdev P, Pine DS. Exploring the feasibility of a meta-structure for DSM-V and ICD-11: could it improve utility and validity? Psychol Med. 2009;39:1993–2000. [PubMed]
  9. Apud JA, Mattay V, Chen J, Kolachana BS, Callicott JH, Rasetti R, Alce G, Iudicello JE, Akbar N, Egan MF, Goldberg TE, Weinberger DR. Tolcapone improves cognition and cortical information processing in normal human subjects. Neuropsychopharmacology. 2007;32:1011–1020. [PubMed]
  10. Arinami T, Gao M, Hamaguchi H, Toru M. A functional polymorphism in the promoter region of the dopamine D2 receptor gene is associated with schizophrenia. Hum Mol Genet. 1997;6:577–582. [PubMed]
  11. Arnsten AF. Stress signalling pathways that impair prefrontal cortex structure and function. Nat Rev Neurosci. 2009;10:410–422. [PMC free article] [PubMed]
  12. Asghari V, Sanyal S, Buchwaldt S, Paterson A, Jovanovic V, Van Tol HH. Modulation of intracellular cyclic AMP levels by different human dopamine D4 receptor variants. J Neurochem. 1995;65:1157–1165. [PubMed]
  13. Asherson P, Brookes K, Franke B, Chen W, Gill M, Ebstein RP, Buitelaar J, Banaschewski T, Sonuga-Barke E, Eisenberg J, Manor I, Miranda A, Oades RD, Roeyers H, Rothenberger A, Sergeant J, Steinhausen HC, Faraone SV. Confirmation that a specific haplotype of the dopamine transporter gene is associated with combined-type ADHD. Am J Psychiatry. 2007;164:674–677. [PubMed]
  14. Balciuniene J, Emilsson L, Oreland L, Pettersson U, Jazin E. Investigation of the functional effect of monoamine oxidase polymorphisms in human brain. Hum Genet. 2002;110:1–7. [PubMed]
  15. Bardo MT, Donohew RL, Harrington NG. Psychobiology of novelty seeking and drug seeking behavior. Behav Brain Res. 1996;77:23–43. [PubMed]
  16. Barkley RA, Smith KM, Fischer M, Navia B. An examination of the behavioral and neuropsychological correlates of three ADHD candidate gene polymorphisms (DRD4 7+, DBH TaqI A2, and DAT1 40 bp VNTR) in hyperactive and normal children followed to adulthood. Am J Med Genet B Neuropsychiatr Genet. 2006;141B:487–498. [PMC free article] [PubMed]
  17. Barnett JH, Scoriels L, Munafo MR. Meta-analysis of the cognitive effects of the catechol-O-methyltransferase gene Val158/108Met polymorphism. Biol Psychiatry. 2008;64:137–144. [PubMed]
  18. Barnett JH, Heron J, Goldman D, Jones PB, Xu K. Effects of catechol-O-methyltransferase on normal variation in the cognitive function of children. Am J Psychiatry. 2009;166:909–916. [PMC free article] [PubMed]
  19. Barr CL, Feng Y, Wigg KG, Schachar R, Tannock R, Roberts W, Malone M, Kennedy JL. 5′-untranslated region of the dopamine D4 receptor gene and attention-deficit hyperactivity disorder. Am J Med Genet. 2001;105:84–90. [PubMed]
  20. Batel P, Houchi H, Daoust M, Ramoz N, Naassila M, Gorwood P. A haplotype of the DRD1 gene is associated with alcohol dependence. Alcohol Clin Exp Res. 2008;32:567–572. [PubMed]
  21. Beaver KM, Wright JP, DeLisi M, Walsh A, Vaughn MG, Boisvert D, Vaske J. A gene × gene interaction between DRD2 and DRD4 is associated with conduct disorder and antisocial behavior in males. Behav Brain Funct. 2007;3:30. [PMC free article] [PubMed]
  22. Bellgrove MA, Mattingley JB. Molecular genetics of attention. Ann N Y Acad Sci. 2008;1129:200–212. [PubMed]
  23. Belsky J, Jonassaint C, Pluess M, Stanton M, Brummett B, Williams R. Vulnerability genes or plasticity genes? Mol Psychiatry. 2009;14:746–754. [PMC free article] [PubMed]
  24. Benjamin J, Li L, Patterson C, Greenberg BD, Murphy DL, Hamer DH. Population and familial association between the D4 dopamine receptor gene and measures of Novelty Seeking. Nat Genet. 1996;12:81–84. [PubMed]
  25. Benjamin J, Osher Y, Lichtenberg P, Bachner-Melman R, Gritsenko I, Kotler M, Belmaker RH, Valsky V, Drendel M, Ebstein RP. An interaction between the catechol O-methyltransferase and serotonin transporter promoter region polymorphisms contributes to tridimensional personality questionnaire persistence scores in normal subjects. Neuropsychobiology. 2000;41:48–53. [PubMed]
  26. Bentivoglio M, Morelli M. Neural wiring in the basal ganglia. In: Dunnett SB, Bentivoglio M, Björklund A, Hökfelt T, editors. Dopamine. Elsevier; Amsterdam: 2005. pp. 38–44.
  27. Biederman J. Attention-deficit/hyperactivity disorder: a selective overview. Biol Psychiatry. 2005;57:1215–1220. [PubMed]
  28. Biederman J, Petty CR, Ten Haagen KS, Small J, Doyle AE, Spencer T, Mick E, Monuteaux MC, Smoller JW, Faraone SV. Effect of candidate gene polymorphisms on the course of attention deficit hyperactivity disorder. Psychiatry Res. 2009;170:199–203. [PMC free article] [PubMed]
  29. Bilder RM, Volavka J, Lachman HM, Grace AA. The catechol-O-methyltransferase polymorphism: relations to the tonic-phasic dopamine hypothesis and neuropsychiatric phenotypes. Neuropsychopharmacology. 2004;29:1943–1961. [PubMed]
  30. Björklund A, Dunnett SB. Dopamine neuron systems in the brain: an update. Trends Neurosci. 2007;30:194–202. [PubMed]
  31. Bloch MH, Landeros-Weisenberger A, Sen S, Dombrowski P, Kelmendi B, Coric V, Pittenger C, Leckman JF. Association of the serotonin transporter polymorphism and obsessive-compulsive disorder: systematic review. Am J Med Genet B Neuropsychiatr Genet. 2008;147B:850–858. [PubMed]
  32. Bousman CA, Glatt SJ, Everall IP, Tsuang MT. Genetic association studies of methamphetamine use disorders: A systematic review and synthesis. Am J Med Genet B Neuropsychiatr Genet. 2009;150B:1025–1049. [PubMed]
  33. Brookes KJ, Neale BM, Sugden K, Khan N, Asherson P, D’Souza UM. Relationship between VNTR polymorphisms of the human dopamine transporter gene and expression in post-mortem midbrain tissue. Am J Med Genet B Neuropsychiatr Genet. 2007;144B:1070–1078. [PubMed]
  34. Brunner HG, Nelen M, Breakefield XO, Ropers HH, van Oost BA. Abnormal behavior associated with a point mutation in the structural gene for monoamine oxidase A. Science. 1993;262:578–580. [PubMed]
  35. Burmeister M, McInnis MG, Zollner S. Psychiatric genetics: progress amid controversy. Nat Rev Genet. 2008;9:527–540. [PubMed]
  36. Burt SA, Mikolajewski AJ. Preliminary evidence that specific candidate genes are associated with adolescent-onset antisocial behavior. Aggress Behav. 2008;34:437–445. [PubMed]
  37. Butcher LM, Davis OS, Craig IW, Plomin R. Genome-wide quantitative trait locus association scan of general cognitive ability using pooled DNA and 500K single nucleotide polymorphism microarrays. Genes Brain Behav. 2008;7:435–446. [PMC free article] [PubMed]
  38. Bymaster FP, Katner JS, Nelson DL, Hemrick-Luecke SK, Threlkeld PG, Heiligenstein JH, Morin SM, Gehlert DR, Perry KW. Atomoxetine increases extracellular levels of norepinephrine and dopamine in prefrontal cortex of rat: a potential mechanism for efficacy in attention deficit/hyperactivity disorder. Neuropsychopharmacology. 2002;27:699–711. [PubMed]
  39. Camarena B, Loyzaga C, Aguilar A, Weissbecker K, Nicolini H. Association study between the dopamine receptor D(4) gene and obsessive-compulsive disorder. Eur Neuropsychopharmacol. 2007;17:406–409. [PubMed]
  40. Carrasco X, Rothhammer P, Moraga M, Henriquez H, Chakraborty R, Aboitiz F, Rothhammer F. Genotypic interaction between DRD4 and DAT1 loci is a high risk factor for attention-deficit/hyperactivity disorder in Chilean families. Am J Med Genet B Neuropsychiatr Genet. 2006;141B:51–54. [PubMed]
  41. Carrel L, Willard HF. X-inactivation profile reveals extensive variability in X-linked gene expression in females. Nature. 2005;434:400–404. [PubMed]
  42. Cases O, Seif I, Grimsby J, Gaspar P, Chen K, Pournin S, Muller U, Aguet M, Babinet C, Shih JC, De Maeyer E. Aggressive behavior and altered amounts of brain serotonin and norepinephrine in mice lacking MAOA. Science. 1995;268:1763–1766. [PMC free article] [PubMed]
  43. Casey BJ, Durston S, Fossella JA. Evidence for a mechanistic model of cognitive control. Clinical Neuroscience Research. 2001;1:267–282.
  44. Caspi A, Langley K, Milne B, Moffitt TE, O’Donovan M, Owen MJ, Polo Tomas M, Poulton R, Rutter M, Taylor A, Williams B, Thapar A. A replicated molecular genetic basis for subtyping antisocial behavior in children with attention-deficit/hyperactivity disorder. Arch Gen Psychiatry. 2008;65:203–210. [PubMed]
  45. Caspi A, McClay J, Moffitt TE, Mill J, Martin J, Craig IW, Taylor A, Poulton R. Role of genotype in the cycle of violence in maltreated children. Science. 2002;297:851–854. [PubMed]
  46. Castellanos FX, Tannock R. Neuroscience of attention-deficit/hyperactivity disorder: the search for endophenotypes. Nat Rev Neurosci. 2002;3:617–628. [PubMed]
  47. Chen J, Lipska BK, Halim N, Ma QD, Matsumoto M, Melhem S, Kolachana BS, Hyde TM, Herman MM, Apud J, Egan MF, Kleinman JE, Weinberger DR. Functional analysis of genetic variation in catechol-O-methyltransferase (COMT): effects on mRNA, protein, and enzyme activity in postmortem human brain. Am J Hum Genet. 2004;75:807–821. [PMC free article] [PubMed]
  48. Cho SC, Kim JW, Kim BN, Hwang JW, Park M, Kim SA, Cho DY, Yoo HJ, Chung US, Son JW, Park TW. No evidence of an association between norepinephrine transporter gene polymorphisms and attention deficit hyperactivity disorder: a family-based and case-control association study in a Korean sample. Neuropsychobiology. 2008;57:131–138. [PubMed]
  49. Cichon S, Craddock N, Daly M, Faraone SV, Gejman PV, Kelsoe J, Lehner T, Levinson DF, Moran A, Sklar P, Sullivan PF. Genomewide association studies: history, rationale, and prospects for psychiatric disorders. Am J Psychiatry. 2009;166:540–556. [PubMed]
  50. Cloninger CR, Svrakic DM, Przybeck TR. A psychobiological model of temperament and character. Arch Gen Psychiatry. 1993;50:975–990. [PubMed]
  51. Comings DE, Blum K. Reward deficiency syndrome: genetic aspects of behavioral disorders. Prog Brain Res. 2000;126:325–341. [PubMed]
  52. Comings DE, Gonzalez N, Wu S, Gade R, Muhleman D, Saucier G, Johnson P, Verde R, Rosenthal RJ, Lesieur HR, Rugle LJ, Miller WB, MacMurray JP. Studies of the 48 bp repeat polymorphism of the DRD4 gene in impulsive, compulsive, addictive behaviors: Tourette syndrome, ADHD, pathological gambling, and substance abuse. Am J Med Genet. 1999;88:358–368. [PubMed]
  53. Comings DE, Wu S, Chiu C, Ring RH, Gade R, Ahn C, MacMurray JP, Dietz G, Muhleman D. Polygenic inheritance of Tourette syndrome, stuttering, attention deficit hyperactivity, conduct, and oppositional defiant disorder: the additive and subtractive effect of the three dopaminergic genes--DRD2, D beta H, and DAT1. Am J Med Genet. 1996;67:264–288. [PubMed]
  54. Congdon E, Canli T. A neurogenetic approach to impulsivity. J Pers. 2008;76:1447–1484. [PMC free article] [PubMed]
  55. Congdon E, Constable RT, Lesch KP, Canli T. Influence of SLC6A3 and COMT variation on neural activation during response inhibition. Biol Psychol. 2009;81:144–152. [PMC free article] [PubMed]
  56. Congdon E, Lesch KP, Canli T. Analysis of DRD4 and DAT polymorphisms and behavioral inhibition in healthy adults: implications for impulsivity. Am J Med Genet B Neuropsychiatr Genet. 2008;147B:27–32. [PubMed]
  57. Conner BT, Hellemann GS, Ritchie TL, Noble EP. Genetic, personality, and environmental predictors of drug use in adolescents. J Subst Abuse Treat. 2010;38:178–190. [PubMed]
  58. Conway KP, Kane RJ, Ball SA, Poling JC, Rounsaville BJ. Personality, substance of choice, and polysubstance involvement among substance dependent patients. Drug Alcohol Depend. 2003;71:65–75. [PubMed]
  59. Cravchik A, Gejman PV. Functional analysis of the human D5 dopamine receptor missense and nonsense variants: differences in dopamine binding affinities. Pharmacogenetics. 1999;9:199–206. [PubMed]
  60. Cravchik A, Sibley DR, Gejman PV. Functional analysis of the human D2 dopamine receptor missense variants. J Biol Chem. 1996;271:26013–26017. [PubMed]
  61. Cruz C, Camarena B, King N, Paez F, Sidenberg D, de la Fuente JR, Nicolini H. Increased prevalence of the seven-repeat variant of the dopamine D4 receptor gene in patients with obsessive-compulsive disorder with tics. Neurosci Lett. 1997;231:1–4. [PubMed]
  62. Curran S, Mill J, Sham P, Rijsdijk F, Marusic K, Taylor E, Asherson P. 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. Am J Med Genet. 2001;105:387–393. [PubMed]
  63. D’Souza UM, Russ C, Tahir E, Mill J, McGuffin P, Asherson PJ, Craig IW. Functional effects of a tandem duplication polymorphism in the 5′flanking region of the DRD4 gene. Biol Psychiatry. 2004;56:691–697. [PubMed]
  64. Deckert J, Catalano M, Syagailo YV, Bosi M, Okladnova O, Di Bella D, Nothen MM, Maffei P, Franke P, Fritze J, Maier W, Propping P, Beckmann H, Bellodi L, Lesch KP. Excess of high activity monoamine oxidase A gene promoter alleles in female patients with panic disorder. Hum Mol Genet. 1999;8:621–624. [PubMed]
  65. Demetrovics Z, Varga G, Szekely A, Vereczkei A, Csorba J, Balazs H, Hoffman K, Sasvari-Szekely M, Barta C. Association between Novelty Seeking of opiate-dependent patients and the catechol-O-methyltransferase Val(158)Met polymorphism. Compr Psychiatry. 2010;51:510–515. [PubMed]
  66. Denney RM, Koch H, Craig IW. Association between monoamine oxidase A activity in human male skin fibroblasts and genotype of the MAOA promoter-associated variable number tandem repeat. Hum Genet. 1999;105:542–551. [PubMed]
  67. DeYoung CG, Getchell M, Koposov RA, Yrigollen CM, Haeffel GJ, af Klinteberg B, Oreland L, Ruchkin VV, Pakstis AJ, Grigorenko EL. Variation in the catechol-O-methyltransferase Val 158 Met polymorphism associated with conduct disorder and ADHD symptoms, among adolescent male delinquents. Psychiatr Genet. 2010;20:20–24. [PMC free article] [PubMed]
  68. Diaz-Anzaldua A, Joober R, Riviere JB, Dion Y, Lesperance P, Richer F, Chouinard S, Rouleau GA. Tourette syndrome and dopaminergic genes: a family-based association study in the French Canadian founder population. Mol Psychiatry. 2004;9:272–277. [PubMed]
  69. Dickinson D, Elvevag B. Genes, cognition and brain through a COMT lens. Neuroscience. 2009;164:72–87. [PMC free article] [PubMed]
  70. Ding YC, Chi HC, Grady DL, Morishima A, Kidd JR, Kidd KK, Flodman P, Spence MA, Schuck S, Swanson JM, Zhang YP, Moyzis RK. Evidence of positive selection acting at the human dopamine receptor D4 gene locus. Proc Natl Acad Sci U S A. 2002;99:309–314. [PMC free article] [PubMed]
  71. Dlugos AM, Hamidovic A, Palmer AA, de Wit H. Further evidence of association between amphetamine response and SLC6A2 gene variants. Psychopharmacology (Berl) 2009;206:501–511. [PubMed]
  72. Durston S, de Zeeuw P, Staal WG. Imaging genetics in ADHD: a focus on cognitive control. Neurosci Biobehav Rev. 2009;33:674–689. [PubMed]
  73. Durston S, Fossella JA, Mulder MJ, Casey BJ, Ziermans TB, Vessaz MN, Van Engeland H. Dopamine transporter genotype conveys familial risk of attention-deficit/hyperactivity disorder through striatal activation. J Am Acad Child Adolesc Psychiatry. 2008;47:61–67. [PubMed]
  74. Ebstein RP. The molecular genetic architecture of human personality: beyond self-report questionnaires. Mol Psychiatry. 2006;11:427–445. [PubMed]
  75. Ebstein RP, Benjamin J, Belmaker RH. Personality and polymorphisms of genes involved in aminergic neurotransmission. Eur J Pharmacol. 2000;410:205–214. [PubMed]
  76. Ebstein RP, Novick O, Umansky R, Priel B, Osher Y, Blaine D, Bennett ER, Nemanov L, Katz M, Belmaker RH. Dopamine D4 receptor (D4DR) exon III polymorphism associated with the human personality trait of Novelty Seeking. Nat Genet. 1996;12:78–80. [PubMed]
  77. Egan MF, Goldberg TE, Kolachana BS, Callicott JH, Mazzanti CM, Straub RE, Goldman D, Weinberger DR. Effect of COMT Val108/158 Met genotype on frontal lobe function and risk for schizophrenia. Proc Natl Acad Sci U S A. 2001;98:6917–6922. [PMC free article] [PubMed]
  78. Eisenberg DT, Mackillop J, Modi M, Beauchemin J, Dang D, Lisman SA, Lum JK, Wilson DS. Examining impulsivity as an endophenotype using a behavioral approach: a DRD2 TaqI A and DRD4 48-bp VNTR association study. Behav Brain Funct. 2007;3:2. [PMC free article] [PubMed]
  79. Ernst M, Romeo RD, Andersen SL. Neurobiology of the development of motivated behaviors in adolescence: a window into a neural systems model. Pharmacol Biochem Behav. 2009;93:199–211. [PubMed]
  80. Evenden JL. Varieties of impulsivity. Psychopharmacology (Berl) 1999;146:348–361. [PubMed]
  81. Fan J, Wu Y, Fossella JA, Posner MI. Assessing the heritability of attentional networks. BMC Neurosci. 2001:2–14. [PMC free article] [PubMed]
  82. Faraone SV. Genetics of adult attention-deficit/hyperactivity disorder. Psychiatr Clin North Am. 2004;27:303–321. [PubMed]
  83. Faraone SV, Perlis RH, Doyle AE, Smoller JW, Goralnick JJ, Holmgren MA, Sklar P. Molecular genetics of attention-deficit/hyperactivity disorder. Biol Psychiatry. 2005;57:1313–1323. [PubMed]
  84. Faraone SV, Wilens T. Does stimulant treatment lead to substance use disorders? J Clin Psychiatry. 2003;64(Suppl 11):9–13. [PubMed]
  85. Floresco SB, Magyar O. Mesocortical dopamine modulation of executive functions: beyond working memory. Psychopharmacology (Berl) 2006;188:567–585. [PubMed]
  86. Forbes EE, Brown SM, Kimak M, Ferrell RE, Manuck SB, Hariri AR. Genetic variation in components of dopamine neurotransmission impacts ventral striatal reactivity associated with impulsivity. Mol Psychiatry. 2009;14:60–70. [PMC free article] [PubMed]
  87. Fossella J, Sommer T, Fan J, Wu Y, Swanson JM, Pfaff DW, Posner MI. Assessing the molecular genetics of attention networks. BMC Neurosci. 2002;3:14. [PMC free article] [PubMed]
  88. Fraeyman NH, Vermis K. Cellular activity of dopamine DA2 receptor splice variants and mutants. Pharmacol Res. 2003;48:535–540. [PubMed]
  89. Franke B, Neale BM, Faraone SV. Genome-wide association studies in ADHD. Hum Genet. 2009;126:13–50. [PMC free article] [PubMed]
  90. Franke B, Vasquez AA, Johansson S, Hoogman M, Romanos J, Boreatti-Hummer A, Heine M, Jacob CP, Lesch KP, Casas M, Ribases M, Bosch R, Sanchez-Mora C, Gomez-Barros N, Fernandez-Castillo N, Bayes M, Halmoy A, Halleland H, Landaas ET, Fasmer OB, Knappskog PM, Heister AJ, Kiemeney LA, Kooij JJ, Boonstra AM, Kan CC, Asherson P, Faraone SV, Buitelaar JK, Haavik J, Cormand B, Ramos-Quiroga JA, Reif A. Multicenter analysis of the SLC6A3/DAT1 VNTR haplotype in persistent ADHD suggests differential involvement of the gene in childhood and persistent ADHD. Neuropsychopharmacology. 2010;35:656–664. [PMC free article] [PubMed]
  91. Fuke S, Suo S, Takahashi N, Koike H, Sasagawa N, Ishiura S. The VNTR polymorphism of the human dopamine transporter (DAT1) gene affects gene expression. Pharmacogenomics J. 2001;1:152–156. [PubMed]
  92. Gainetdinov RR, Jones SR, Caron MG. Functional hyperdopaminergia in dopamine transporter knock-out mice. Biol Psychiatry. 1999;46:303–311. [PubMed]
  93. Garrido E, Palomo T, Ponce G, Garcia-Consuegra I, Jimenez-Arriero MA, Hoenicka J. The ANKK1 Protein Associated with Addictions has Nuclear and Cytoplasmic Localization and Shows a Differential Response of Ala239Thr to Apomorphine. Neurotox Res 2010 [PubMed]
  94. Gelernter J, Kranzler HR. Genetics of alcohol dependence. Hum Genet. 2009;126:91–99. [PMC free article] [PubMed]
  95. Giakoumaki SG, Roussos P, Bitsios P. Improvement of prepulse inhibition and executive function by the COMT inhibitor tolcapone depends on COMT Val158Met polymorphism. Neuropsychopharmacology. 2008;33:3058–3068. [PubMed]
  96. Gillespie NA, Zhu G, Neale MC, Heath AC, Martin NG. Direction of causation modeling between cross-sectional measures of parenting and psychological distress in female twins. Behav Genet. 2003;33:383–396. [PubMed]
  97. Gizer IR, Ficks C, Waldman ID. Candidate gene studies of ADHD: a meta-analytic review. Hum Genet. 2009;126:51–90. [PubMed]
  98. Glatt SJ, Faraone SV, Tsuang MT. Psychiatric Genetics: A Primer. In: Smoller JW, Shiedly BR, Tsuang MT, editors. Psychiatric Genetics: Applications in Clinical Practice. American Psychiatric Publishing; Washington, D.C: 2008. pp. 3–27.
  99. Golimbet VE, Alfimova MV, Gritsenko IK, Ebstein RP. Relationship between dopamine system genes and extraversion and novelty seeking. Neurosci Behav Physiol. 2007;37:601–606. [PubMed]
  100. Goodman WK, Price LH, Rasmussen SA, Mazure C, Fleischmann RL, Hill CL, Heninger GR, Charney DS. The Yale-Brown Obsessive Compulsive Scale. I. Development, use, and reliability. Arch Gen Psychiatry. 1989;46:1006–1011. [PubMed]
  101. Grados MA. The genetics of obsessive-compulsive disorder and Tourette syndrome: an epidemiological and pathway-based approach for gene discovery. J Am Acad Child Adolesc Psychiatry. 2010;49:810–819. e811–812. [PubMed]
  102. Grice DE, Leckman JF, Pauls DL, Kurlan R, Kidd KK, Pakstis AJ, Chang FM, Buxbaum JD, Cohen DJ, Gelernter J. Linkage disequilibrium between an allele at the dopamine D4 receptor locus and Tourette syndrome, by the transmission-disequilibrium test. Am J Hum Genet. 1996;59:644–652. [PMC free article] [PubMed]
  103. Grisham JR, Anderson TM, Sachdev PS. Genetic and environmental influences on obsessive-compulsive disorder. Eur Arch Psychiatry Clin Neurosci. 2008;258:107–116. [PubMed]
  104. Guindalini C, Howard M, Haddley K, Laranjeira R, Collier D, Ammar N, Craig I, O’Gara C, Bubb VJ, Greenwood T, Kelsoe J, Asherson P, Murray RM, Castelo A, Quinn JP, Vallada H, Breen G. A dopamine transporter gene functional variant associated with cocaine abuse in a Brazilian sample. Proc Natl Acad Sci U S A. 2006;103:4552–4557. [PMC free article] [PubMed]
  105. Han DH, Yoon SJ, Sung YH, Lee YS, Kee BS, Lyoo IK, Renshaw PF, Cho SC. A preliminary study: novelty seeking, frontal executive function, and dopamine receptor (D2) TaqI A gene polymorphism in patients with methamphetamine dependence. Compr Psychiatry. 2008;49:387–392. [PubMed]
  106. Happe F, Ronald A, Plomin R. Time to give up on a single explanation for autism. Nat Neurosci. 2006;9:1218–1220. [PubMed]
  107. Hebebrand J, Nothen MM, Ziegler A, Klug B, Neidt H, Eggermann K, Lehmkuhl G, Poustka F, Schmidt MH, Propping P, Remschmidt H. Nonreplication of linkage disequilibrium between the dopamine D4 receptor locus and Tourette syndrome. Am J Hum Genet. 1997;61:238–239. [PMC free article] [PubMed]
  108. Hellstrand M, Danielsen EA, Steen VM, Ekman A, Eriksson E, Nilsson CL. The ser9gly SNP in the dopamine D3 receptor causes a shift from cAMP related to PGE2 related signal transduction mechanisms in transfected CHO cells. J Med Genet. 2004;41:867–871. [PMC free article] [PubMed]
  109. Hemmings SM, Kinnear CJ, Lochner C, Niehaus DJ, Knowles JA, Moolman-Smook JC, Corfield VA, Stein DJ. Early- versus late-onset obsessive-compulsive disorder: investigating genetic and clinical correlates. Psychiatry Res. 2004;128:175–182. [PubMed]
  110. Hendriks RW, Chen ZY, Hinds H, Schuurman RK, Craig IW. An X chromosome inactivation assay based on differential methylation of a CpG island coupled to a VNTR polymorphism at the 5′ end of the monoamine oxidase A gene. Hum Mol Genet. 1992;1:662. [PubMed]
  111. Hicks BM, Krueger RF, Iacono WG, McGue M, Patrick CJ. Family transmission and heritability of externalizing disorders: a twin-family study. Arch Gen Psychiatry. 2004;61:922–928. [PubMed]
  112. Hicks BM, South SC, Dirago AC, Iacono WG, McGue M. Environmental adversity and increasing genetic risk for externalizing disorders. Arch Gen Psychiatry. 2009;66:640–648. [PMC free article] [PubMed]
  113. Hirvonen MM, Laakso A, Någren K, Rinne JO, Pohjalainen T, Hietala J. C957T polymorphism of dopamine D2 receptor gene affects striatal DRD2 in vivo availability by changing the receptor affinity. Synapse. 2009a;63:907–912. [PubMed]
  114. Hirvonen MM, Lumme V, Hirvonen J, Pesonen U, Någren K, Vahlberg T, Scheinin H, Hietala J. C957T polymorphism of the human dopamine D2 receptor gene predicts extrastriatal dopamine receptor availability in vivo. Prog Neuropsychopharmacol Biol Psychiatry. 2009b;33:630–636. [PubMed]
  115. Ho MK, Tyndale RF. Overview of the pharmacogenomics of cigarette smoking. Pharmacogenomics J. 2007;7:81–98. [PubMed]
  116. Hoenicka J, Quinones-Lombrana A, Espana-Serrano L, Alvira-Botero X, Kremer L, Perez-Gonzalez R, Rodriguez-Jimenez R, Jimenez-Arriero MA, Ponce G, Palomo T. The ANKK1 gene associated with addictions is expressed in astroglial cells and upregulated by apomorphine. Biol Psychiatry. 2010;67:3–11. [PubMed]
  117. Hosak L, Libiger J, Cizek J, Beranek M, Cermakova E. The COMT Val158Met polymorphism is associated with novelty seeking in Czech methamphetamine abusers: preliminary results. Neuro Endocrinol Lett. 2006;27:799–802. [PubMed]
  118. Housley DJ, Nikolas M, Venta PJ, Jernigan KA, Waldman ID, Nigg JT, Friderici KH. SNP discovery and haplotype analysis in the segmentally duplicated DRD5 coding region. Ann Hum Genet. 2009;73:274–282. [PMC free article] [PubMed]
  119. Huang SY, Lu RB, Ma KH, Shy MJ, Lin WW. Norepinephrine transporter polymorphisms T-182C and G1287A are not associated with alcohol dependence and its clinical subgroups. Drug Alcohol Depend. 2008;92:20–26. [PubMed]
  120. Huang W, Li MD. Differential allelic expression of dopamine D1 receptor gene (DRD1) is modulated by microRNA miR-504. Biol Psychiatry. 2009;65:702–705. [PMC free article] [PubMed]
  121. Huang W, Ma JZ, Payne TJ, Beuten J, Dupont RT, Li MD. Significant association of DRD1 with nicotine dependence. Hum Genet. 2008;123:133–140. [PubMed]
  122. Huang W, Payne TJ, Ma JZ, Beuten J, Dupont RT, Inohara N, Li MD. Significant association of ANKK1 and detection of a functional polymorphism with nicotine dependence in an African-American sample. Neuropsychopharmacology. 2009;34:319–330. [PubMed]
  123. Hudziak JJ, Faraone SV. The new genetics in child psychiatry. J Am Acad Child Adolesc Psychiatry. 2010;49:729–735. [PubMed]
  124. Jeanneteau F, Funalot B, Jankovic J, Deng H, Lagarde JP, Lucotte G, Sokoloff P. A functional variant of the dopamine D3 receptor is associated with risk and age-at-onset of essential tremor. Proc Natl Acad Sci U S A. 2006;103:10753–10758. [PMC free article] [PubMed]
  125. Johnson KA, Kelly SP, Robertson IH, Barry E, Mulligan A, Daly M, Lambert D, McDonnell C, Connor TJ, Hawi Z, Gill M, Bellgrove MA. Absence of the 7-repeat variant of the DRD4 VNTR is associated with drifting sustained attention in children with ADHD but not in controls. Am J Med Genet B Neuropsychiatr Genet. 2008;147B:927–937. [PubMed]
  126. Jonsson EG, Nothen MM, Grunhage F, Farde L, Nakashima Y, Propping P, Sedvall GC. Polymorphisms in the dopamine D2 receptor gene and their relationships to striatal dopamine receptor density of healthy volunteers. Mol Psychiatry. 1999;4:290–296. [PubMed]
  127. Jorm AF, Prior M, Sanson A, Smart D, Zhang Y, Easteal S. Association of a polymorphism of the dopamine transporter gene with externalizing behavior problems and associated temperament traits: a longitudinal study from infancy to the mid-teens. Am J Med Genet. 2001;105:346–350. [PubMed]
  128. Käenmäki M, Tammimäki A, Myöhänen T, Pakarinen K, Amberg C, Karayiorgou M, Gogos JA, Männistö PT. Quantitative role of COMT in dopamine clearance in the prefrontal cortex of freely moving mice. J Neurochem. 2010;114:1745–1755. [PubMed]
  129. Katerberg H, Cath DC, Denys DA, Heutink P, Polman A, van Nieuwerburgh FC, Deforce DL, Bochdanovits Z, van Balkom AJ, den Boer JA. The role of the COMT Val(158)Met polymorphism in the phenotypic expression of obsessive-compulsive disorder. Am J Med Genet B Neuropsychiatr Genet. 2010;153B:167–176. [PubMed]
  130. Keltikangas-Järvinen L, Pulkki-Raback L, Elovainio M, Raitakari OT, Viikari J, Lehtimäki T. DRD2 C32806T modifies the effect of child-rearing environment on adulthood novelty seeking. Am J Med Genet B Neuropsychiatr Genet. 2009;150B:389–394. [PubMed]
  131. Kendler KS, Jacobson KC, Prescott CA, Neale MC. Specificity of genetic and environmental risk factors for use and abuse/dependence of cannabis, cocaine, hallucinogens, sedatives, stimulants, and opiates in male twins. Am J Psychiatry. 2003;160:687–695. [PubMed]
  132. Kereszturi E, Kiraly O, Barta C, Molnar N, Sasvari-Szekely M, Csapo Z. No direct effect of the −521 C/T polymorphism in the human dopamine D4 receptor gene promoter on transcriptional activity. BMC Mol Biol. 2006;7:18. [PMC free article] [PubMed]
  133. Kereszturi E, Kiraly O, Csapo Z, Tarnok Z, Gadoros J, Sasvari-Szekely M, Nemoda Z. Association between the 120-bp duplication of the dopamine D4 receptor gene and attention deficit hyperactivity disorder: genetic and molecular analyses. Am J Med Genet B Neuropsychiatr Genet. 2007;144B:231–236. [PubMed]
  134. Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62:593–602. [PubMed]
  135. Khan ZU, Gutierrez A, Martin R, Penafiel A, Rivera A, De La Calle A. Differential regional and cellular distribution of dopamine D2-like receptors: an immunocytochemical study of subtype-specific antibodies in rat and human brain. J Comp Neurol. 1998;402:353–371. [PubMed]
  136. Kieling C, Roman T, Doyle AE, Hutz MH, Rohde LA. Association between DRD4 gene and performance of children with ADHD in a test of sustained attention. Biol Psychiatry. 2006;60:1163–1165. [PubMed]
  137. Kim CH, Hahn MK, Joung Y, Anderson SL, Steele AH, Mazei-Robinson MS, Gizer I, Teicher MH, Cohen BM, Robertson D, Waldman ID, Blakely RD, Kim KS. A polymorphism in the norepinephrine transporter gene alters promoter activity and is associated with attention-deficit hyperactivity disorder. Proc Natl Acad Sci U S A. 2006;103:19164–19169. [PMC free article] [PubMed]
  138. Kollins SH, Anastopoulos AD, Lachiewicz AM, FitzGerald D, Morrissey-Kane E, Garrett ME, Keatts SL, Ashley-Koch AE. SNPs in dopamine D2 receptor gene (DRD2) and norepinephrine transporter gene (NET) are associated with continuous performance task (CPT) phenotypes in ADHD children and their families. Am J Med Genet B Neuropsychiatr Genet. 2008;147B:1580–1588. [PMC free article] [PubMed]
  139. Koob GF, Volkow ND. Neurocircuitry of addiction. Neuropsychopharmacology. 2010;35:217–238. [PMC free article] [PubMed]
  140. Kramer UM, Rojo N, Schule R, Cunillera T, Schols L, Marco-Pallares J, Cucurell D, Camara E, Rodriguez-Fornells A, Munte TF. ADHD candidate gene (DRD4 exon III) affects inhibitory control in a healthy sample. BMC Neurosci. 2009;10:150. [PMC free article] [PubMed]
  141. Kraschewski A, Reese J, Anghelescu I, Winterer G, Schmidt LG, Gallinat J, Finckh U, Rommelspacher H, Wernicke C. Association of the dopamine D2 receptor gene with alcohol dependence: haplotypes and subgroups of alcoholics as key factors for understanding receptor function. Pharmacogenet Genomics. 2009;19:513–527. [PubMed]
  142. Krause KH, Dresel SH, Krause J, Kung HF, Tatsch K. Increased striatal dopamine transporter in adult patients with attention deficit hyperactivity disorder: effects of methylphenidate as measured by single photon emission computed tomography. Neurosci Lett. 2000;285:107–110. [PubMed]
  143. Krause KH, Dresel SH, Krause J, la Fougere C, Ackenheil M. The dopamine transporter and neuroimaging in attention deficit hyperactivity disorder. Neurosci Biobehav Rev. 2003;27:605–613. [PubMed]
  144. Lachman HM. Does COMT val158met affect behavioral phenotypes: yes, no, maybe? Neuropsychopharmacology. 2008;33:3027–3029. [PubMed]
  145. Lachman HM, Papolos DF, Saito T, Yu YM, Szumlanski CL, Weinshilboum RM. Human catechol-O-methyltransferase pharmacogenetics: description of a functional polymorphism and its potential application to neuropsychiatric disorders. Pharmacogenetics. 1996;6:243–250. [PubMed]
  146. Lanau F, Zenner MT, Civelli O, Hartman DS. Epinephrine and norepinephrine act as potent agonists at the recombinant human dopamine D4 receptor. J Neurochem. 1997;68:804–812. [PubMed]
  147. Lander E, Kruglyak L. Genetic dissection of complex traits: guidelines for interpreting and reporting linkage results. Nat Genet. 1995;11:241–247. [PubMed]
  148. Lasky-Su J, Lange C, Biederman J, Tsuang M, Doyle AE, Smoller JW, Laird N, Faraone S. Family-based association analysis of a statistically derived quantitative traits for ADHD reveal an association in DRD4 with inattentive symptoms in ADHD individuals. Am J Med Genet B Neuropsychiatr Genet. 2008;147B:100–106. [PubMed]
  149. Laucht M, Becker K, Blomeyer D, Schmidt MH. Novelty seeking involved in mediating the association between the dopamine D4 receptor gene exon III polymorphism and heavy drinking in male adolescents: results from a high-risk community sample. Biol Psychiatry. 2007;61:87–92. [PubMed]
  150. Laucht M, Becker K, Frank J, Schmidt MH, Esser G, Treutlein J, Skowronek MH, Schumann G. Genetic variation in dopamine pathways differentially associated with smoking progression in adolescence. J Am Acad Child Adolesc Psychiatry. 2008;47:673–681. [PubMed]
  151. Laviola G, Macri S, Morley-Fletcher S, Adriani W. Risk-taking behavior in adolescent mice: psychobiological determinants and early epigenetic influence. Neurosci Biobehav Rev. 2003;27:19–31. [PubMed]
  152. Lee CC, Chou IC, Tsai CH, Wang TR, Li TC, Tsai FJ. Dopamine receptor D2 gene polymorphisms are associated in Taiwanese children with Tourette syndrome. Pediatr Neurol. 2005;33:272–276. [PubMed]
  153. Le Foll B, Gallo A, Strat YL, Lu L, Gorwood P. Genetics of dopamine receptors and drug addiction: a comprehensive review. Behavioural Pharmacology. 2009;20:1–17. [PubMed]
  154. Lewis DA, Melchitzky DS, Sesack SR, Whitehead RE, Auh S, Sampson A. Dopamine transporter immunoreactivity in monkey cerebral cortex: regional, laminar, and ultrastructural localization. J Comp Neurol. 2001;432:119–136. [PubMed]
  155. Li D, Sham PC, Owen MJ, He L. Meta-analysis shows significant association between dopamine system genes and attention deficit hyperactivity disorder (ADHD) Hum Mol Genet. 2006;15:2276–2284. [PubMed]
  156. Lochner C, Hemmings SM, Kinnear CJ, Nel D, Hemmings SM, Seedat S, Moolman-Smook JC, Stein DJ. Cluster analysis of obsessive-compulsive symptomatology: identifying obsessive-compulsive disorder subtypes. Isr J Psychiatry Relat Sci. 2008;45:164–176. [PubMed]
  157. 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. [PubMed]
  158. Lotta T, Vidgren J, Tilgmann C, Ulmanen I, Melen K, Julkunen I, Taskinen J. Kinetics of human soluble and membrane-bound catechol O-methyltransferase: a revised mechanism and description of the thermolabile variant of the enzyme. Biochemistry. 1995;34:4202–4210. [PubMed]
  159. Lowe N, Kirley A, Mullins C, Fitzgerald M, Gill M, Hawi Z. Multiple marker analysis at the promoter region of the DRD4 gene and ADHD: evidence of linkage and association with the SNP -616. Am J Med Genet B Neuropsychiatr Genet. 2004;131B:33–37. [PubMed]
  160. Lundstrom K, Turpin MP. Proposed schizophrenia-related gene polymorphism: expression of the Ser9Gly mutant human dopamine D3 receptor with the Semliki Forest virus system. Biochem Biophys Res Commun. 1996;225:1068–1072. [PubMed]
  161. Madras BK, Miller GM, Fischman AJ. The dopamine transporter and attention-deficit/hyperactivity disorder. Biol Psychiatry. 2005;57:1397–1409. [PubMed]
  162. Männistö PT, Kaakkola S. Catechol-O-methyltransferase (COMT): biochemistry, molecular biology, pharmacology, and clinical efficacy of the new selective COMT inhibitors. Pharmacol Rev. 1999;51:593–628. [PubMed]
  163. Manor I, Corbex M, Eisenberg J, Gritsenkso I, Bachner-Melman R, Tyano S, Ebstein RP. Association of the dopamine D5 receptor with attention deficit hyperactivity disorder (ADHD) and scores on a continuous performance test (TOVA) Am J Med Genet B Neuropsychiatr Genet. 2004;127B:73–77. [PubMed]
  164. Manor I, Tyano S, Eisenberg J, Bachner-Melman R, Kotler M, Ebstein RP. The short DRD4 repeats confer risk to attention deficit hyperactivity disorder in a family-based design and impair performance on a continuous performance test (TOVA) Mol Psychiatry. 2002a;7:790–794. [PubMed]
  165. Manor I, Tyano S, Mel E, Eisenberg J, Bachner-Melman R, Kotler M, Ebstein RP. Family-based and association studies of monoamine oxidase A and attention deficit hyperactivity disorder (ADHD): preferential transmission of the long promoter-region repeat and its association with impaired performance on a continuous performance test (TOVA) Mol Psychiatry. 2002b;7:626–632. [PubMed]
  166. Marco-Pallares J, Cucurell D, Cunillera T, Kramer UM, Camara E, Nager W, Bauer P, Schule R, Schols L, Munte TF, Rodriguez-Fornells A. Genetic variability in the dopamine system (dopamine receptor D4, catechol-O-methyltransferase) modulates neurophysiological responses to gains and losses. Biol Psychiatry. 2009;66:154–161. [PubMed]
  167. Marco R, Miranda A, Schlotz W, Melia A, Mulligan A, Muller U, Andreou P, Butler L, Christiansen H, Gabriels I, Medad S, Albrecht B, Uebel H, Asherson P, Banaschewski T, Gill M, Kuntsi J, Mulas F, Oades R, Roeyers H, Steinhausen HC, Rothenberger A, Faraone SV, Sonuga-Barke EJ. Delay and reward choice in ADHD: an experimental test of the role of delay aversion. Neuropsychology. 2009;23:367–380. [PubMed]
  168. Matsumoto M, Weickert CS, Akil M, Lipska BK, Hyde TM, Herman MM, Kleinman JE, Weinberger DR. Catechol O-methyltransferase mRNA expression in human and rat brain: evidence for a role in cortical neuronal function. Neuroscience. 2003;116:127–137. [PubMed]
  169. Mattay VS, Goldberg TE, Fera F, Hariri AR, Tessitore A, Egan MF, Kolachana B, Callicott JH, Weinberger DR. Catechol O-methyltransferase val158-met genotype and individual variation in the brain response to amphetamine. Proc Natl Acad Sci U S A. 2003;100:6186–6191. [PMC free article] [PubMed]
  170. McGeary J. The DRD4 exon 3 VNTR polymorphism and addiction-related phenotypes: a review. Pharmacol Biochem Behav. 2009;93:222–229. [PMC free article] [PubMed]
  171. Meaney MJ. Epigenetics and the biological definition of gene × environment interactions. Child Dev. 2010;81:41–79. [PubMed]
  172. Meyer-Lindenberg A, Buckholtz JW, Kolachana B, ARH, Pezawas L, Blasi G, Wabnitz A, Honea R, Verchinski B, Callicott JH, Egan M, Mattay V, Weinberger DR. Neural mechanisms of genetic risk for impulsivity and violence in humans. Proc Natl Acad Sci U S A. 2006;103:6269–6274. [PMC free article] [PubMed]
  173. Mill J, Asherson P, Browes C, D’Souza U, Craig I. Expression of the dopamine transporter gene is regulated by the 3′ UTR VNTR: Evidence from brain and lymphocytes using quantitative RT-PCR. Am J Med Genet. 2002a;114:975–979. [PubMed]
  174. Mill J, Asherson P, Craig I, D’Souza UM. Transient expression analysis of allelic variants of a VNTR in the dopamine transporter gene (DAT1) BMC Genet. 2005a;6:3. [PMC free article] [PubMed]
  175. Mill J, Caspi A, Williams BS, Craig I, Taylor A, Polo-Tomas M, Berridge CW, Poulton R, Moffitt TE. Prediction of heterogeneity in intelligence and adult prognosis by genetic polymorphisms in the dopamine system among children with attention-deficit/hyperactivity disorder: evidence from 2 birth cohorts. Arch Gen Psychiatry. 2006;63:462–469. [PubMed]
  176. Mill J, Fisher N, Curran S, Richards S, Taylor E, Asherson P. Polymorphisms in the dopamine D4 receptor gene and attention-deficit hyperactivity disorder. Neuroreport. 2003;14:1463–1466. [PubMed]
  177. Mill J, Xu X, Ronald A, Curran S, Price T, Knight J, Craig I, Sham P, Plomin R, Asherson P. Quantitative trait locus analysis of candidate gene alleles associated with attention deficit hyperactivity disorder (ADHD) in five genes: DRD4, DAT1, DRD5, SNAP-25, and 5HT1B. Am J Med Genet B Neuropsychiatr Genet. 2005b;133B:68–73. [PubMed]
  178. Mill JS, Caspi A, McClay J, Sugden K, Purcell S, Asherson P, Craig I, McGuffin P, Braithwaite A, Poulton R, Moffitt TE. The dopamine D4 receptor and the hyperactivity phenotype: a developmental-epidemiological study. Mol Psychiatry. 2002b;7:383–391. [PubMed]
  179. Miller GM, Madras BK. Polymorphisms in the 3′-untranslated region of human and monkey dopamine transporter genes affect reporter gene expression. Mol Psychiatry. 2002;7:44–55. [PubMed]
  180. Missale C, Nash SR, Robinson SW, Jaber M, Caron MG. Dopamine receptors: from structure to function. Physiol Rev. 1998;78:189–225. [PubMed]
  181. Monuteaux MC, Biederman J, Doyle AE, Mick E, Faraone SV. Genetic risk for conduct disorder symptom subtypes in an ADHD sample: specificity to aggressive symptoms. J Am Acad Child Adolesc Psychiatry. 2009;48:757–764. [PubMed]
  182. Moron JA, Brockington A, Wise RA, Rocha BA, Hope BT. Dopamine uptake through the norepinephrine transporter in brain regions with low levels of the dopamine transporter: evidence from knock-out mouse lines. J Neurosci. 2002;22:389–395. [PubMed]
  183. Munafo MR, Clark TG, Moore LR, Payne E, Walton R, Flint J. Genetic polymorphisms and personality in healthy adults: a systematic review and meta-analysis. Mol Psychiatry. 2003;8:471–484. [PubMed]
  184. Munafo MR, Timpson NJ, David SP, Ebrahim S, Lawlor DA. Association of the DRD2 gene Taq1A polymorphism and smoking behavior: a meta-analysis and new data. Nicotine Tob Res. 2009;11:64–76. [PMC free article] [PubMed]
  185. Munafo MR, Yalcin B, Willis-Owen SA, Flint J. Association of the dopamine D4 receptor (DRD4) gene and approach-related personality traits: meta-analysis and new data. Biol Psychiatry. 2008;63:197–206. [PubMed]
  186. Nackley AG, Shabalina SA, Tchivileva IE, Satterfield K, Korchynskyi O, Makarov SS, Maixner W, Diatchenko L. Human catechol-O-methyltransferase haplotypes modulate protein expression by altering mRNA secondary structure. Science. 2006;314:1930–1933. [PubMed]
  187. Nambu A, Tokuno H, Takada M. Functional significance of the cortico-subthalamo-pallidal ‘hyperdirect’ pathway. Neurosci Res. 200

    Date: 2016-01-03; view: 1045


    <== previous page | next page ==>
    Impulsive behaviors | ΐβςξασρϋ ξςεχερςβεννϋε θ ρςπΰν ΡΝΓ
doclecture.net - lectures - 2014-2024 year. Copyright infringement or personal data (0.01 sec.)