Epidemiology and Risk Factors-- Gene/Environment Interaction

| categories: genetics

Contents

1 Epidemiology and Risk Factors

The etiology of attention-deficit/hyperactivity disorder (ADHD) is unclear. However, it is well accepted that ADHD has both genetic and environmental risk factors [313]. ADHD is likely not a single pathophysiological entity, which suggests complex and multiple etiologies. Multiple genetic and environmental factors may act in concert, which gives rise to a spectrum of neurobiological liability [10] which manifests as a set of ADHD symptoms. Estimates of heritability from many familial and twin studies range from 0.5 to 0.9, average 0.75; which, taken together with a five-fold increased risk in first-degree relatives, strongly implies a genetic component [38101330]. Environmental risk factors such as obstetric complications, family conflict, and lead exposure have also shown positive, yet small, increases in risk for ADHD.

2 Genetic Risk Factors

No single gene abnormality reliably predicts ADHD, but consistent association between several genes regulating dopamine (DA) transport and signaling have been implicated (these have been the genes most studied by candidate gene association studies because of prevalent DA hypotheses concerning the pathophysiology of ADHD [1937] and the mechanisms of the most popular drugs used to treat ADHD [323436] ). Genome-wide association studies have failed to report any associations that are significant after correction for multiple testing [10]. Cortese et al. [8] estimates that a genome-wide association study would have to use sample sizes between 10,000 to 20,000 subjects in order to accurately detect novel genes involved in ADHD with risk factors similar to those seen in candidate gene association studies. Candidate gene association studies have shown positive, but small correlations with the dopamine receptor D4 gene (DRD4), dopamine receptor D5 gene (DRD5), dopamine transporter gene (SLC6A3), dopamine β-hydroxylase (DβH), synaptosomal-associated protein of 25 kDa gene (SNAP25), serotonin transporter gene (SLC6A4), and the serotonin 1B receptor gene (HTR1B) [3], which is in line with the multifactorial polygenic model in which a plethora of genes each confer a small but real risk to the disorder [8]. It is easy to imagine that combinations of risk alleles impacting dopaminergic signaling could produce an extreme hypodopaminergic state that would lead to poor cognitive function [30].

The SLC6A3 and DRD4 genes show variation amongst the population based on a variable number of tandem repeats where the nucleotide or base pair sequence is repeated a different number of times in different alleles of the gene. A 40 base pair variable tandem nucleotide repeat in the 3 untranslated region of the human DAT1 gene (SLC6A3 [7]) and a 48 base pair DA D4 receptor 7-repeat allele [17] have both been implicated in ADHD, and have been the most consistently positive across multiple candidate gene association studies [13142029], which is not always common [69].

Dopamine receptor genes linked to ADHD The DA D4 receptor is prevalently expressed in frontal and subcortical networks that are implicated in the pathophysiology of ADHD [3]. DRD4 association studies have assayed a variant known as the exon-III 7-repeat allele (16 amino acid repeat in the 3rd intracellular loop of the receptor that couples the receptor to pre- or post-synaptic G-protein effectors [30]), which while it shows a positive association with ADHD, may paradoxically be protective, as the DRD4 7-repeat allele produces an in vitro blunted response to dopamine, but subjects with the DRD4 7-repeat allele and ADHD tend to have better clinical and academic outcomes than those with ADHD and the DRD4 4-repeat allele more common in the general population [31730]. Candidate gene studies of the DA D5 receptor, which is expressed in the hippocampus, and especially the dentate gyrus [16], have shown positive correlation with ADHD with a repeated sequence near the transcription start site [3].

Although DA D1 and D2 receptors are thought to be the major effectors of stimulant-induced alterations in impulsive, addictive, and compulsive behavior [33], it is unsettled as to whether variants of these full-length genes are associated with ADHD [27].

Dopamine transporter gene linked to ADHD When pooled in meta-analysis, studies of the SLC6A3 10-repeat sequence in the 3 untranslated region (the 3 untranslated region of mRNA often contains regulatory sequences that can influence post-transcriptional gene expression) estimate an increased odds ratio of 1.13 for ADHD [37]. It may be possible that different variants of the dopamine transporter (DAT) have an altered sensitivity to endogenous DA. At present, no such variations in the coding sequence of the DAT are known, nor are they separable pharmacologically [27].

Norepinephrine synthesis linked to ADHD Dopamine β-hydroxylase is the main enzyme and rate-limiting step in converting DA to norepinephrine (NE). Meta-analysis of multiple family studies suggest a significant association with ADHD and the 5Taq1 polymorphism of the DβH gene [3].

Serotonin signaling linked to ADHD Both the serotonin transporter gene (SLC6A4) and the serotonin 1B receptor gene (HTR1B) have been implicated in ADHD. A functional variant of the SLC6A4 has a meta-analysis adjusted odds ratio of 1.31 for ADHD. Many studies of the HTR1B have also been positive, with a non-functional marker showing an odds ratio of 1.44 [3].

Synaptic transmission linked to ADHD SNAP25 is a neuron-specific protein involved in synaptic vesicle transport, fusion, and release. A meta-analysis of several studies assessing the association between the gene and ADHD found an odds ratio of 1.19. SNAP25 knockout mice also show spontaneous hyperactivity which can be reduced with stimulant drugs [3].

Negative candidate gene results Candidate gene association studies have also been done on several genes that have provided negative or equivocal results. Two genes involved in monoamine degredation, catechol-O-methyl-transferase (COMT) and monoamine oxidase (MAO), have shown negative results, as well as the norepinephrine transporter gene (SLC6A2) and the norepinephrine 1C, 2A, and 2C receptors [3].

3 Environmental Risk Factors

Environmental factors play a critical role in the etiology of ADHD. One of the best evidences supporting this point comes from the genetic twin studies, which show that the risk of ADHD in monozygotic twins is much lower than 100 % [8]. Many studies have indicated that complications during pregnancy and delivery could be associated with childhood ADHD. The complications that are associated with later ADHD tend to include chronic exposures to the fetus, and complications that lead to hypoxia rather than acute events [3].

Prenatal risk factors Maternal alcohol use during pregnancy leads to behavioral, cognitive, and learning problems that could present as ADHD. Prenatal alcohol exposure is known to induce brain structural anomalies, and children exposed to prenatal alcohol are more hyperactive, disruptive, impulsive, and are at increased risk of a range of psychiatric disorders [10]. Studies of ADHD children show an increased likelihood of having been exposed to alcohol as a fetus [3].

There is also a well documented, almost 3-fold increased risk for the disorder in children whose mothers smoked during pregnancy [324]. Exposure of the fetus to nicotine can damage the development of the brain at critical time points as nicotinic receptors modulate dopaminergic activity [111231], and animal studies have also shown a correlation between chronic nicotine exposure of pregnant dams and hyperactive offspring [3].

Perinatal risk factors Low birthweight (< 2500g) and premature birth (< 37weeks gestation) increase the risk for ADHD [231822]. Very low birthweight children show a 2-fold increase in the proportion of those with ADHD vs. average birthweight counterparts [10]. Interestingly, prenatal tobacco exposure can result in both low birthweight and preterm birth [15]. In a huge study from the Danish longitudinal birth registers, Linnet et al. [21] found a rate ratio for hyperkinetic disorder (the International Statistical Classification of Diseases and Related Health Problems (ICD) analogue of the Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV)’s ADHD) of 1.7 for 34 to 37 weeks gestation and rate ratio 2.7 for less than 34 weeks gestation. Likewise, low birth weight for children born at term gave a rate ratio of 1.9 for birthweight of 1500 g to 2499 g and 1.5 for birthweight 2500 g to 2999 g [21]. In a similar study, Mick et al. [23] reported that ADHD cases were 3.1 times more likely than controls to be born under 2500 g.

Postnatal and childhood risk factors Malnutrition and dietary deficiency have been proposed, and iron deficiency has been implicated in some cases of ADHD, however further evidence is necessary to firmly establish a role [10].

Lead exposure Lead exposure has been implicated in the pathophysiology of ADHD, although most children with ADHD do not show lead contamination and many children with high lead exposure do not become affected with the disorder [3]. Nigg et al. [26] suggests that low-level lead exposure, below the US Environmental Protection Agency (EPA) limits, may represent a hidden major effect on ADHD incidence. A population sample, Braun et al. [4] reported that very low levels of lead were associated with ADHD, even with lead exposures below 5 μg/dL, which highlights the importance of a gene–environment interaction model, as these levels of lead exposure are common in the US population [5].

4 Gene–Environment Interactions

Molecular genetics supports the association of ADHD with several DA signaling related genes, and environmental effects suggests increased risks for ADHD related to maternal smoking, exposure to low levels of lead, premature birth or low birth weight, and other factors that alter fetal development with lasting or possibly permanent effects on attention and behavior [30]. It is noteworthy that individual genes or environmental risk factors carry quite modest risk, and also that some of the environmental risk factors associated with ADHD (e.g. low lead exposure) are quite common in the general population [30]. More complex models of the etiology of ADHD which incorporate gene–environment interactions are being studied. However, the multitude of possible etiologies of gene–environment combinations add complexity, and would require enormous sample sizes for adequate evaluation [30]. Neuman et al. [25] demonstrated that smoking during pregnancy is associated with the ADHD combined subtype in children with the ‘susceptible’ DRD4 and dopamine transporter gene (SLC6A3) gene variants. Also, a significant interaction between SLC6A3 genotype and prenatal smoke exposure was found in males: Men with prenatal smoke exposure and homozygous for the SLC6A3 10-repeat allele had higher hyperactivity/impulsivity than males from all other groups [1]. Through unknown mechanisms, abnormal DAT density is a common feature among subjects with ADHD [28].

Acronyms

ADHD
attention-deficit/hyperactivity disorder
CDC
Center for Disease Control and Prevention
COMT
catechol-O-methyl-transferase
DAT
dopamine transporter
DA
dopamine
DbH
dopamine β-hydroxylase
DRD4
dopamine receptor D4 gene
DRD5
dopamine receptor D5 gene
DSM-IV
Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition
EPA
US Environmental Protection Agency
HTR1B
serotonin 1B receptor gene
ICD
International Statistical Classification of Diseases and Related Health Problems
MAO
monoamine oxidase
NE
norepinephrine
SLC6A2
norepinephrine transporter gene
SLC6A3
dopamine transporter gene
SLC6A4
serotonin transporter gene
SNAP25
synaptosomal-associated protein of 25 kDa gene

References

[1]    K. Becker, M. El-Faddagh, M. H. Schmidt, G. Esser, and M. Laucht. Interaction of dopamine transporter genotype with prenatal smoke exposure on adhd symptoms. J Pediatr, 152(2):263–9, Feb 2008. doi: 10.1016/j.jpeds.2007.07.004.

[2]    A. T. Bhutta, M. A. Cleves, P. H. Casey, M. M. Cradock, and K. J. S. Anand. Cognitive and behavioral outcomes of school-aged children who were born preterm: a meta-analysis. JAMA, 288(6):728–37, Aug 2002.

[3]    J. Biederman and S. V. Faraone. Attention-deficit hyperactivity disorder. The Lancet, 366(9481):237 – 248, 2005. ISSN 0140-6736. doi: 10.1016/S0140-_6736(05)66915-_2. URL http://www.sciencedirect.com/science/article/pii/S0140673605669152.

[4]    J. M. Braun, R. S. Kahn, T. Froehlich, P. Auinger, and B. P. Lanphear. Exposures to environmental toxicants and attention deficit hyperactivity disorder in u.s. children. Environ Health Perspect, 114(12): 1904–9, Dec 2006.

[5]    Center for Disease Control and Prevention (CDC). Blood lead levels–united states, 1999-2002. MMWR. Morbidity and mortality weekly report, 54(20):513, 2005.

[6]    D. A. Collier, S. Curran, and P. Asherson. Mission: not impossible? candidate gene studies in child psychiatric disorders. Mol Psychiatry, 5(5): 457–60, Sep 2000.

[7]    E. H. Cook, Jr, M. A. Stein, M. D. Krasowski, N. J. Cox, D. M. Olkon, J. E. Kieffer, and B. L. Leventhal. Association of attention-deficit disorder and the dopamine transporter gene. Am J Hum Genet, 56(4): 993–8, Apr 1995.

[8]    S. Cortese, S. V. Faraone, and J. Sergeant. Misunderstandings of the genetics and neurobiology of adhd: moving beyond anachronisms. Am J Med Genet B Neuropsychiatr Genet, 156B(5):513–6, Jul 2011. doi: 10.1002/ajmg.b.31207.

[9]    R. R. Crowe. Candidate genes in psychiatry: an epidemiological perspective. Am J Med Genet, 48(2):74–7, Jul 1993. doi: 10.1002/ajmg. 1320480203.

[10]    P. Curatolo, E. D’Agati, and R. Moavero. The neurobiological basis of adhd. Ital J Pediatr, 36(1):79, 2010. doi: 10.1186/1824-_7288-_36-_79.

[11]    J. A. Dani, D. Ji, and F.-M. Zhou. Synaptic plasticity and nicotine addiction. Neuron, 31(3):349–352, 2001.

[12]    J. A. Dani, D. Jenson, J. I. Broussard, and M. De Biasi. Neurophysiology of nicotine addiction. Journal of addiction research & therapy, 1(1), 2011.

[13]    S. DiMaio, N. Grizenko, and R. Joober. Dopamine genes and attention-deficit hyperactivity disorder: a review. J Psychiatry Neurosci, 28(1):27–38, Jan 2003.

[14]    S. V. Faraone, A. E. Doyle, E. Mick, and J. Biederman. Meta-analysis of the association between the 7-repeat allele of the dopamine d(4) receptor gene and attention deficit hyperactivity disorder. Am J Psychiatry, 158(7): 1052–7, Jul 2001.

[15]    J. J. Jaakkola, N. Jaakkola, and K. Zahlsen. Fetal growth and length of gestation in relation to prenatal exposure to environmental tobacco smoke assessed by hair nicotine concentration. Environ Health Perspect, 109(6):557–61, Jun 2001.

[16]    Z. U. Khan, L. Mrzljak, A. Gutierrez, A. de la Calle, and P. S. Goldman-Rakic. Prominence of the dopamine d2 short isoform in dopaminergic pathways. Proc Natl Acad Sci U S A, 95(13):7731–6, Jun 1998.

[17]    G. J. LaHoste, J. M. Swanson, S. B. Wigal, C. Glabe, T. Wigal, N. King, and J. L. Kennedy. Dopamine d4 receptor gene polymorphism is associated with attention deficit hyperactivity disorder. Mol Psychiatry, 1 (2):121–4, May 1996.

[18]    J. Lahti, K. Räikkönen, E. Kajantie, K. Heinonen, A.-K. Pesonen, A.-L. Järvenpää, and T. Strandberg. Small body size at birth and behavioural symptoms of adhd in children aged five to six years. J Child Psychol Psychiatry, 47(11):1167–74, Nov 2006. doi: 10.1111/j.1469-_7610. 2006.01661.x.

[19]    F. Levy. The dopamine theory of attention deficit hyperactivity disorder (adhd). Aust N Z J Psychiatry, 25(2):277–83, Jun 1991.

[20]    J. Li, Y. Wang, R. Zhou, H. Zhang, L. Yang, B. Wang, and S. V. Faraone. Association between polymorphisms in serotonin 2c receptor gene and attention-deficit/hyperactivity disorder in han chinese subjects. Neurosci Lett, 407(2):107–11, Oct 2006. doi: 10.1016/j.neulet.2006.08.022.

[21]    K. M. Linnet, K. Wisborg, C. Obel, N. J. Secher, P. H. Thomsen, E. Agerbo, and T. B. Henriksen. Smoking during pregnancy and the risk for hyperkinetic disorder in offspring. Pediatrics, 116(2):462–7, Aug 2005. doi: 10.1542/peds.2004-_2054.

[22]    K. M. Linnet, K. Wisborg, E. Agerbo, N. J. Secher, P. H. Thomsen, and T. B. Henriksen. Gestational age, birth weight, and the risk of hyperkinetic disorder. Arch Dis Child, 91(8):655–60, Aug 2006. doi: 10.1136/adc.2005.088872.

[23]    E. Mick, J. Biederman, J. Prince, M. J. Fischer, and S. V. Faraone. Impact of low birth weight on attention-deficit hyperactivity disorder. J Dev Behav Pediatr, 23(1):16–22, Feb 2002.

[24]    S. Milberger, J. Biederman, S. V. Faraone, L. Chen, and J. Jones. Is maternal smoking during pregnancy a risk factor for attention deficit hyperactivity disorder in children? Am J Psychiatry, 153(9):1138–42, Sep 1996.

[25]    R. J. Neuman, E. Lobos, W. Reich, C. A. Henderson, L.-W. Sun, and R. D. Todd. Prenatal smoking exposure and dopaminergic genotypes interact to cause a severe adhd subtype. Biol Psychiatry, 61(12):1320–8, Jun 2007. doi: 10.1016/j.biopsych.2006.08.049.

[26]    J. T. Nigg, M. M. Wong, M. M. Martel, J. M. Jester, L. I. Puttler, J. M. Glass, K. M. Adams, H. E. Fitzgerald, and R. A. Zucker. Poor response inhibition as a predictor of problem drinking and illicit drug use in adolescents at risk for alcoholism and other substance use disorders. J Am Acad Child Adolesc Psychiatry, 45(4):468–75, Apr 2006. doi: 10.1097/01.chi.0000199028.76452.a9.

[27]    P. Seeman and B. K. Madras. Anti-hyperactivity medication: methylphenidate and amphetamine. Mol Psychiatry, 3:386–396, 1998.

[28]    T. J. Spencer, J. Biederman, B. K. Madras, S. V. Faraone, D. D. Dougherty, A. A. Bonab, and A. J. Fischman. In vivo neuroreceptor imaging in attention-deficit/hyperactivity disorder: a focus on the dopamine transporter. Biol Psychiatry, 57(11):1293–300, Jun 2005. doi: 10.1016/j.biopsych.2005.03.036.

[29]    J. M. Swanson, P. Flodman, J. Kennedy, M. A. Spence, R. Moyzis, S. Schuck, M. Murias, J. Moriarity, C. Barr, M. Smith, and M. Posner. Dopamine genes and adhd. Neurosci Biobehav Rev, 24(1):21–5, Jan 2000.

[30]    J. M. Swanson, M. Kinsbourne, J. Nigg, B. Lanphear, G. A. Stefanatos, N. Volkow, E. Taylor, B. J. Casey, F. X. Castellanos, and P. D. Wadhwa. Etiologic subtypes of attention-deficit/hyperactivity disorder: brain imaging, molecular genetic and environmental factors and the dopamine hypothesis. Neuropsychol Rev, 17(1):39–59, Mar 2007. doi: 10.1007/s11065-_007-_9019-_9.

[31]    J. Tang and J. A. Dani. Dopamine enables in vivo synaptic plasticity associated with the addictive drug nicotine. Neuron, 63 (5):673–682, Sep 2009. doi: 10.1016/j.neuron.2009.07.025. URL http://dx.doi.org/10.1016/j.neuron.2009.07.025.

[32]    N. D. Volkow, Y. S. Ding, J. S. Fowler, G. J. Wang, J. Logan, J. S. Gatley, S. Dewey, C. Ashby, J. Liebermann, and R. Hitzemann. Is methylphenidate like cocaine? Studies on their pharmacokinetics and distribution in the human brain. Arch. Gen. Psychiatry, 52(6):456–463, Jun 1995.

[33]    N. D. Volkow, G. J. Wang, M. W. Fischman, R. W. Foltin, J. S. Fowler, N. N. Abumrad, S. Vitkun, J. Logan, S. J. Gatley, N. Pappas, R. Hitzemann, and C. E. Shea. Relationship between subjective effects of cocaine and dopamine transporter occupancy. Nature, 386(6627):827–30, Apr 1997. doi: 10.1038/386827a0.

[34]    N. D. Volkow, G. J. Wang, J. S. Fowler, S. J. Gatley, J. Logan, Y. S. Ding, R. Hitzemann, and N. Pappas. Dopamine transporter occupancies in the human brain induced by therapeutic doses of oral methylphenidate. Am J Psychiatry, 155(10):1325–1331, Oct 1998.

[35]    N. D. Volkow, G. Wang, J. S. Fowler, J. Logan, M. Gerasimov, L. Maynard, Y. Ding, S. J. Gatley, A. Gifford, and D. Franceschi. Therapeutic doses of oral methylphenidate significantly increase extracellular dopamine in the human brain. J Neurosci, 21(2): RC121, Jan 2001.

[36]    N. D. Volkow, J. S. Fowler, G.-J. Wang, and R. Z. Goldstein. Role of dopamine, the frontal cortex and memory circuits in drug addiction: insight from imaging studies. Neurobiol Learn Mem, 78(3):610–24, Nov 2002.

[37]    P. H. Wender, R. S. Epstein, I. J. Kopin, and E. K. Gordon. Urinary monoamine metabolites in children with minimal brain dysfunction. Am J Psychiatry, 127(10):1411–5, Apr 1971.