Entry - *126455 - SOLUTE CARRIER FAMILY 6 (NEUROTRANSMITTER TRANSPORTER, DOPAMINE), MEMBER 3; SLC6A3 - OMIM
 
* 126455

SOLUTE CARRIER FAMILY 6 (NEUROTRANSMITTER TRANSPORTER, DOPAMINE), MEMBER 3; SLC6A3


Alternative titles; symbols

DOPAMINE TRANSPORTER; DAT
DAT1


HGNC Approved Gene Symbol: SLC6A3

Cytogenetic location: 5p15.33     Genomic coordinates (GRCh38): 5:1,392,794-1,445,440 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
5p15.33 {Nicotine dependence, protection against} 188890 3
Parkinsonism-dystonia, infantile, 1 613135 AR 3

TEXT

Description

The dopamine transporter (DAT), which is encoded by the SLC6A3 gene, mediates the active reuptake of dopamine from the synapse and is a principal regulator of dopaminergic neurotransmission. The SLC6A3 gene has been implicated in human disorders such as parkinsonism, Tourette syndrome, and substance abuse (Vandenbergh et al., 1992).


Cloning and Expression

Vandenbergh et al. (1992) cloned cDNAs encoding the entire coding sequence and 3-prime untranslated sequence of the human DAT1 gene.

Kurian et al. (2009) noted that SLC6A3 contains 12 transmembrane domains with helically unwound regions in the first and sixth domains. Extracellular and intracellular loops include helical portions E2, E3, E4A, E4B, I1, and I5. Mature, glycosylated DAT has a molecular mass of 85 kD, whereas immature nonglycosylated DAT has a molecular mass of 55 kD and does not transport dopamine as efficiently as the mature protein.

Navaroli et al. (2011) reported that DAT has an endocytic signal near the C terminus that modulates both basal and protein kinase C (PKC; see 176960)-enhanced DAT internalization. Immunohistochemical analysis revealed that rat Dat localized to plasma membrane foci that corresponded to both lipid rafts and non-raft microdomains in PC12 pheochromocytoma cells.


Gene Function

By yeast 2-hybrid analysis of a human substantia nigra cDNA library, Navaroli et al. (2011) found that the small GTPase RIN (RIT2; 609592) interacted with the isolated C-terminal endocytic signal of human DAT. Coimmunoprecipitation and protein pull-down assays with PC12 cells that were transfected with human DAT revealed direct interaction between DAT and Rin. Rin did not interact with other members of the SLC6 transporter family. DAT colocalized with Rin at plasma membrane foci, predominantly at lipid rafts, in transfected PC12 cells. Phorbol ester-induced PKC activation enhanced DAT-Rin interaction, followed by their dissociation and DAT internalization into endocytic vesicles. These effects did not occur with a DAT mutant lacking the C-terminal endocytic signal or with a Rin mutant lacking GTPase activity. Knockdown of Rin via short hairpin RNA completely inhibited PKC-induced DAT internalization.


Gene Structure

The SLC6A3 gene contains 15 exons spanning approximately 60 kb (Vandenbergh et al., 1992).


Mapping

By in situ hybridization and by PCR amplification of rodent/human somatic cell hybrid DNAs, Vandenbergh et al. (1992) and Giros et al. (1992) mapped the SLC6A3 gene to chromosome 5p15.3.

Lossie et al. (1994) mapped the mouse Dat1 homolog to chromosome 15 by linkage analysis of an interspecific backcross.

Gelernter et al. (1995) demonstrated close linkage between the SLC6A3 gene and several markers previously mapped to distal chromosome 5p.


Biochemical Features

Crystal Structure

Zhou et al. (2007) determined the crystal structure at 2.9 angstroms of the bacterial leucine transporter (LeuT), a homolog of SERT (182138), NET (163970), and DAT, in complex with leucine and the antidepressant desipramine. Desipramine binds at the inner end of the extracellular cavity of the transporter and is held in place by a hairpin loop and by a salt bridge. This binding site is separated from the leucine-binding site by the extracellular gate of the transporter. By directly locking the gate, desipramine prevents conformational changes and blocks substrate transport. Mutagenesis experiments on human SERT and DAT indicate that both the desipramine-binding site and its inhibition mechanism are probably conserved in the human neurotransmitter transporters.


Molecular Genetics

Vandenbergh et al. (1992) identified a 40-bp variable-number tandem repeat (VNTR) polymorphism in the 3-prime untranslated region of the DAT1 gene with repeat copy numbers ranging from 3 to 11.

Byerley et al. (1993) demonstrated a VNTR polymorphism related to the DAT1 gene. Doucette-Stamm et al. (1995) established allele frequencies for the 40-bp VNTR polymorphism. Differences were found between black Americans and Caucasians or Hispanics, but there were no differences between Caucasians and Hispanics. A previously unreported allele was detected in all 3 populations.

Association with Attention-Deficit Hyperactivity Disorder

Attention-deficit hyperactivity disorder (ADHD; 143465) is presumably heritable in some cases. Some ADHD patients respond to medications that inhibit the dopamine transporter, including methylphenidate, amphetamine, pemoline, and bupropion. Cook et al. (1995) used the haplotype-based haplotype relative risk (HHRR) method to test for association between a VNTR polymorphism at the DAT1 locus and 49 cases of ADHD. Eight cases of undifferentiated attention-deficit disorder (UADD) were also studied. All cases were studied in trios composed of father, mother, and affected offspring. HHRR analysis revealed significant association between ADHD/UADD and the 480-bp DAT1 allele. When cases of UADD were dropped from the analysis, similar results were found. Cook et al. (1995) suggested that molecular analysis of DAT1 may identify mutations that increase susceptibility to this disorder and that biochemical analysis of such mutations may lead to development of more effective therapeutic interventions.

Gill et al. (1997) found a significant association between ADHD and the 480-bp DAT1 VNTR allele. Gill et al. (1997) reported concordance rates for ADHD in monozygotic and dizygotic twins of 81% and 29%, respectively.

Waldman et al. (1998) used 4 analytic strategies to examine the association and linkage of the DAT1 gene with ADHD. The study group comprised 122 children referred to psychiatric clinics for behavioral and learning problems that included but were not limited to ADHD, as well as their parents and sibs. Within-family analyses of linkage disequilibrium, using the transmission disequilibrium test (TDT), confirmed the 480-bp DAT1 allele as the high-risk allele. In between-family association analyses, levels of hyperactive-impulsive symptoms but not inattentive symptoms were related to the number of DAT1 high-risk alleles. Sibs discordant for the number of DAT1 high-risk alleles differed markedly in their levels of both hyperactive-impulsive and inattentive symptoms, such that the sibs with the higher number of high-risk alleles had much higher symptom levels. Within-family analyses of linkage disequilibrium, using the TDT, suggested association and linkage of ADHD with DAT1 and that this relationship was especially strong with the combined but not the inattentive subtype. The findings were believed to represent one of the first replicated relations of a candidate gene and a psychiatric disorder in children.

Kahn et al. (2003) found a significant association between homozygosity for the 480-bp DAT allele and hyperactivity-impulsivity and oppositional behaviors only in the presence of maternal prenatal smoking. They emphasized the importance of incorporating environmental cofactors in genetic studies of ADHD.

Langley et al. (2005) tested the DAT1 3-prime VNTR and 3 putative DAT1 promoter SNPs for association with ADHD in 263 parent-proband trios using family-based association methods. No evidence of association with any of the promoter region SNPs or the VNTR was seen. Haplotype analysis was also nonsignificant, and no association was found between the VNTR and response to stimulant medications. By case-control analysis of the VNTR in 263 cases and 287 controls, the 10-repeat allele showed no significant association compared to all other alleles combined.

Feng et al. (2005) investigated whether the DAT1 3-prime UTR contributed to ADHD by genotyping DNA variants around the VNTR region in a sample of 178 ADHD families. Variants included an MspI polymorphism (rs27072), a DraI T-C transition reported to influence DAT1 expression levels, and a BstUI polymorphism (rs3863145), in addition to the VNTR. They found an association between the G allele of the MspI SNP and ADHD (P = 0.009) but not with alleles of the VNTR polymorphism or the BstUI polymorphism, and they did not observe the DraI SNP. Feng et al. (2005) screened the VNTR region by direct sequencing to determine if there were additional variants within the repeats that could account for association with ADHD and found no variation in the VNTR region for either the 10- or 9-repeat alleles in the probands screened.

In both a Taiwanese ADHD sample and an English ADHD sample, Brookes et al. (2006) identified association with the DAT1 3-prime UTR VNTR and a novel DAT1 intron 8 repeat polymorphism. A risk haplotype composed of the 2 repeat polymorphisms was also associated with ADHD in both populations, and the authors found that the risk haplotype showed significant interactions with maternal use of alcohol during pregnancy in the English sample.

Hebebrand et al. (2006) found no evidence of association between ADHD and the DAT1 VNTR in 102 German families with 2 or more offspring who fulfilled diagnostic criteria for ADHD.

Cheuk et al. (2006) studied the association between the DAT1 VNTR and ADHD in Chinese children (64 ADHD cases, including 52 boys and 12 girls, their families, and 64 sex-matched normal controls) in Hong Kong. Patients were diagnosed with combined, hyperactive-impulsive, or inattentive subtypes of ADHD. The 10-repeat allele (92.6%) and the 10/10 (85.2%) repeat genotype were most prevalent. Both family-based and case-control analyses showed no association between the DAT1 polymorphisms and ADHD.

Animal studies suggest that the development of substance dependence (e.g., alcoholism) is associated with dopaminergic activity in striatum and the limbic system (Tiihonen et al., 1995). Several genetic studies had indicated that allele A1 at the DRD2 (126450) locus is associated with both density of dopamine receptor-2 and alcoholism. Tiihonen et al. (1995) studied striatal dopamine reuptake sites in 19 healthy controls, 19 habitually impulsive violent alcoholics, and 10 nonviolent alcoholics with single photon emission computed tomography (SPECT) using iodine-123-labeled beta-CIT as a tracer. Blind quantitative analysis revealed that the striatal dopamine transporter density was markedly lower in nonviolent alcoholics than in healthy controls (P less than 0.001), while violent alcoholics had slightly higher dopamine transporter densities than controls (P less than 0.10). Goldman (1995) suggested that the next step would be to look for inherited variants of the DAT1 gene that may influence vulnerability to alcoholism and other psychiatric diseases.

Association with Tourette Syndrome

In a large Canadian Mennonite pedigree segregating for Tourette syndrome (137580) (Kurlan et al., 1986), Gelernter et al. (1995) excluded linkage to SLC6A3. They cautiously pointed out, however, that these results did not exclude a role for the dopamine transporter in influencing risk for Tourette syndrome in combination with other loci.

Association with Cigarette Smoking

Dopaminergic genes are likely candidates for heritable influences on cigarette smoking (188890). Lerman et al. (1999) and Sabol et al. (1999) reported associations between allele 9 of a DAT1 polymorphism, SLC6A3*9 (126455.0001), and smoking status. While Lerman et al. (1999) reported association of the SLC6A3*9 allele with lack of smoking, late initiation of smoking, and length of quitting attempts, Sabol et al. (1999) reported that the significant association between SLC6A3*9 and smoking status was due to an effect on cessation rather than initiation. The SLC6A3*9 polymorphism was also associated with low scores for novelty seeking, which was the most significant personality correlate of smoking cessation.

Association with Bipolar Disorder

Greenwood et al. (2001) reported evidence for an association between DAT1 and bipolar disorder (MAFD1; 125480) in a sample of 50 parent-proband trios. Using the transmission disequilibrium test (TDT), they showed an association between a haplotype composed of 5 SNPs in the 3-prime region of the DAT1 gene, exon 9 through exon 15, and bipolar disorder (allele-wise TDT empirical P = 0.001; genotype-wise TDT empirical P = 0.0004). Greenwood et al. (2006) analyzed a total of 22 SNPs in the 50 previously studied parent-proband trios and an independent set of 70 parent-proband trios. Using TDT analysis, an intron 8 SNP and an intron 13 SNP were found to be moderately associated with bipolar disorder, each in 1 of the 2 independent samples. Analysis of haplotypes of all 22 SNPs in sliding windows of 5 adjacent SNPs revealed an association to the region near intron 7 and 8 in both samples (empirical P values of 0.002 and 0.001, respectively, for the same window). The haplotype block structure observed by Greenwood et al. (2001) was confirmed in this new sample with the greater resolution allowing for discrimination of a third haplotype block in the middle of the gene.

Infantile Parkinsonism-Dystonia 1

By linkage analysis followed by candidate gene sequencing of a consanguineous Pakistani family with infantile parkinsonism-dystonia (PKDYS1; 613135), Kurian et al. (2009) identified a homozygous mutation (L368Q; 126455.0002) in the SLC6A3 gene. A similarly affected individual from a second family had a different homozygous mutation (P395L; 126455.0003). In vitro functional expression studies showed that both mutant proteins had no dopamine uptake activity.

In 8 unrelated patients with dopamine transporter deficiency syndrome, Kurian et al. (2011) identified homozygous or compound heterozygous mutations in the SLC6A3 gene (see, e.g., 126455.0005-126455.0007). None of the patients shared a mutation, suggesting the absence of mutational hotspots. In vitro functional expression studies in HEK293 cells showed that the mutations caused a loss of transporter function and decreased expression of the normal protein. All patients presented in early infancy with a complex motor disorder involving both hypo- and hyperkinetic movements. There was no effective treatment, and several patients died in the teenage years.

By homozygosity mapping followed by exome sequencing of a Mennonite family in which 2 sisters had infantile parkinsonism-dystonia, Puffenberger et al. (2012) identified a homozygous splice site mutation in the SLC6A3 gene (126455.0004).


Animal Model

Giros et al. (1996) found that the disruption of the mouse Dat1 gene results in spontaneous hyperlocomotion despite major adaptive changes such as decreases in neurotransmitter and receptor levels. In homozygous-null mice, dopamine persisted at least 100 times longer in the extracellular space, providing a biochemical explanation of the hyperdopaminergic phenotype and demonstrating the critical role of DAT1 in regulating neurotransmission. The authors noted that the dopamine transporter is an obligatory target of cocaine and amphetamine, as demonstrated by the fact that these psychostimulants had no effect on locomotor activity or dopamine release and uptake in mice lacking the transporter. Giros et al. (1996) noted that there are similarities between the hyperdopaminergic phenotype of the knockout mice and some of the positive symptoms of patients with schizophrenia. The authors also postulated that specific blockade of DAT1 with high-affinity inhibitors may be beneficial in illnesses such as Parkinson disease (see 168600), where the effective levels of dopamine are markedly reduced.

Gainetdinov et al. (1999) continued the studies of Giros et al. (1992), which demonstrated mice lacking the gene encoding the plasma membrane dopamine transporter have elevated dopaminergic tone and are hyperactive. They found that this activity was exacerbated by exposure to a novel environment. Additionally, these mice were impaired in spatial cognitive function, and they showed a decrease in locomotion in response to psychostimulants. This paradoxical calming effect of psychostimulants depended on serotonergic neurotransmission, and emphasized the importance of a relative balance of the serotonin and dopamine systems for normal motor activity. The parallels between the DAT knockout mice and individuals with ADHD suggested that common mechanisms may underlie some of their behaviors and responses to psychostimulants.

Sotnikova et al. (2006) developed a novel acute mouse model of severe dopamine deficiency using Dat-null mice and pharmacologic inhibition of tyrosine hydroxylase. Dopamine-deficient Dat-null mice (DDD) demonstrated severe akinesia, rigidity, tremor, and ptosis, similar to behaviors observed in patients with Parkinson disease. Interestingly, DDD mice were able to swim in water, indicating that certain movements and conditions can occur independently of dopamine. Dopamine agonists such as L-DOPA temporarily restored locomotion in DDD mice, and amphetamine derivatives showed effectiveness in reducing motor abnormalities in DDD mice. Sotnikova et al. (2006) noted that the DDD mouse model provides a unique opportunity to screen potential therapeutic agents for the treatment of Parkinson disease.

Using an in silico search, followed by PCR, Hejjas et al. (2007) identified VNTR polymorphisms in genes of the dopaminergic system in 4 dog breeds and European gray wolves. Polymorphisms of the DRD4 (126452), DBH (609312), and DAT genes were associated with attention deficit, but not activity-impulsivity, in Belgian Tervuerens, a breed that had almost all genetic variants identified.


ALLELIC VARIANTS ( 7 Selected Examples):

.0001 NICOTINE DEPENDENCE, PROTECTION AGAINST

SLC6A3, 9-REPEAT VNTR
  
RCV000018247

Lerman et al. (1999) reported associations between allele 9 of the SLC6A3 gene (SLC6A3*9), a polymorphism containing 9 repeats of the 40-basepair 3-prime VNTR, and lack of smoking, late initiation of smoking, and length of quitting attempts (see 188890). The association with smoking risk was modified by the DRD2 (126450) genotype, resulting in a 50% reduction in smoking risk for individuals carrying both SLC6A3*9 and DRD2-A2. Sabol et al. (1999) extended this study by examining both smoking behavior and personality traits of 1,107 individuals in a diverse population of nonsmokers, current smokers, and former smokers. A significant association between SLC6A3*9 and smoking status was confirmed and was due to an effect on cessation rather than initiation. The SLC6A3*9 polymorphism was also associated with low scores for novelty seeking, which was the most significant personality correlate of smoking cessation. Sabol et al. (1999) hypothesized that individuals carrying the SLC6A3*9 polymorphism have altered dopamine transmission, which reduces their need for novelty and reward by external stimuli, including cigarettes. Sabol et al. (1999) found that individuals carrying the SLC6A3*9 allele were 1.5-fold more likely to have quit smoking than were individuals lacking this polymorphism. The results supported the finding of Lerman et al. (1999) of an association between SLC6A3 and length of previous cessation attempts in current smokers.


.0002 PARKINSONISM-DYSTONIA, INFANTILE, 1

SLC6A3, LEU368GLN
  
RCV000018249

In 2 affected members of a consanguineous Pakistani family with infantile parkinsonism-dystonia (PKDYS1; 613135), Kurian et al. (2009) identified a homozygous 1103T-A transversion in exon 8 of the SLC6A3 gene, resulting in a leu368-to-gln (L368Q) substitution in a highly conserved residue in transmembrane domain-7 toward the exterior of the protein surface. In vitro functional expression assays in HEK293 cells showed that L368Q-mutant DAT had no dopamine reuptake activity. Binding affinity for a cocaine analog was near normal, but the potency of dopamine in inhibiting cocaine analog binding was greatly reduced in the L368Q mutant, indicating reduced dopamine binding affinity. The mutant protein also resulted in a profound reduction in mature glycosylated DAT, which likely impaired transport function. The mutation was not identified in 544 alleles from Asian individuals or in 438 alleles from individuals of mixed European descent.


.0003 PARKINSONISM-DYSTONIA, INFANTILE, 1

SLC6A3, PRO395LEU
  
RCV000018250

In a patient of European descent, born of consanguineous parents, with infantile parkinsonism-dystonia (PKDYS1; 613135), Kurian et al. (2009) identified a homozygous 1184C-T transition in exon 9 of the SLC6A3 gene, resulting in a pro395-to-leu (P395L) substitution in a highly conserved residue of the E4B loop close to transmembrane domain-8. In vitro functional expression assays in HEK293 cells showed that the P395L-mutant protein had no dopamine reuptake activity. Binding affinity for a cocaine analog was near normal, and the potency of dopamine in inhibiting cocaine analog binding was near normal in the P395L mutant, suggesting no impact on dopamine binding affinity. The mutant protein did result in a profound reduction in mature glycosylated DAT, which likely impaired transport function. The mutation was not identified in 544 alleles from Asian individuals or in 438 alleles from individuals of mixed European descent.


.0004 PARKINSONISM-DYSTONIA, INFANTILE, 1

SLC6A3, IVS9DS, G-T, +1
  
RCV000022531...

In 2 Mennonite sisters with infantile parkinsonism-dystonia (PKDYS1; 613135), Puffenberger et al. (2012) identified a homozygous G-to-T transversion in intron 9 of the SLC6A3 gene. The mutation was found by homozygosity mapping followed by exome sequencing of the candidate region. No carriers of this mutation were found among 201 Mennonite control samples. The proband developed irritability and feeding difficulties soon after birth, followed by generalized rigidity and dystonia during early infancy. She had impaired motor development and severe rigid parkinsonism by late childhood. She could not speak or use her hands to communicate, and it was difficult to assess cognitive function or thought content. Brain structure was normal. Cerebrospinal fluid showed increased homovanillic acid (HVA). A similarly affected sister had died.


.0005 PARKINSONISM-DYSTONIA, INFANTILE, 1

SLC6A3, IVS7DS, G-A, +1
  
RCV000083262...

In a patient, born of consanguineous Turkish parents, with infantile parkinsonism-dystonia (PKDYS1; 613135), Kurian et al. (2011) identified a homozygous G-to-A transition in intron 7 of the SLC6A3 gene (c.1031+1G-A), predicted to cause aberrant splicing.


.0006 PARKINSONISM-DYSTONIA, INFANTILE, 1

SLC6A3, LEU224PRO
  
RCV000083263

In a patient of mixed European descent, born of consanguineous parents, with PKDYS (PKDYS1; 613135), Kurian et al. (2011) identified a homozygous c.671T-C transition in exon 5 of the SLC6A3 gene, resulting in a leu224-to-pro (L224P) substitution at a highly conserved residue in mammalian species. In vitro functional expression studies of the mutation in HEK293 cells showed nonspecific uptake for the mutant transporter, and Western blot analysis indicated deficiency of the mature protein. These findings were consistent with a loss of function.


.0007 PARKINSONISM-DYSTONIA, INFANTILE, 1

SLC6A3, ARG521TRP
  
RCV000083264...

In a patient of mixed European descent, born of consanguineous parents, with PKDYS (PKDYS1; 613135), Kurian et al. (2011) identified a homozygous c.1561C-T transition in exon 12 of the SLC6A3 gene, resulting in an arg521-to-trp (R521W) substitution at a highly conserved residue. In vitro functional expression studies of the mutation in HEK293 cells showed decreased dopamine uptake (27% of wildtype), and Western blot analysis indicated deficiency of the mature protein. These findings were consistent with a loss of function.


REFERENCES

  1. Brookes, K.-J., Mill, J., Guindalini, C., Curran, S., Xu, X., Knight, J., Chen, C.-K., Huang, Y.-S., Sethna, V., Taylor, E., Chen, W., Breen, G., Asherson, P. A common haplotype of the dopamine transporter gene associated with attention-deficit/hyperactivity disorder and interacting with maternal use of alcohol during pregnancy. Arch. Gen. Psychiat. 63: 74-81, 2006. [PubMed: 16389200, related citations] [Full Text]

  2. Byerley, W., Hoff, M., Holik, J., Caron, M. G., Giros, B. VNTR polymorphism for the human dopamine transporter gene (DAT1). Hum. Molec. Genet. 2: 335, 1993. [PubMed: 8098980, related citations] [Full Text]

  3. Cheuk, D. K. L., Li, S. Y. H., Wong, V. No association between VNTR polymorphisms of dopamine transporter gene and attention deficit hyperactivity disorder in Chinese children. Am. J. Med. Genet. 141B: 123-125, 2006. [PubMed: 16402340, related citations] [Full Text]

  4. Cook, E. H., Jr., Stein, M. A., Krasowski, M. D., Cox, N. J., Olkon, D. M., Kieffer, J. E., Leventhal, B. L. Association of attention-deficit disorder and the dopamine transporter gene. Am. J. Hum. Genet. 56: 993-998, 1995. [PubMed: 7717410, related citations]

  5. Doucette-Stamm, L., Blakely, D. J., Tian, J., Mockus, S., Mao, J. Population genetic study of the human dopamine transporter gene (DAT1). Genet. Epidemiol. 12: 303-308, 1995. [PubMed: 7557351, related citations] [Full Text]

  6. Feng, Y., Wigg, K. G., Makkar, R., Ickowicz, A., Pathare, T., Tannock, R., Roberts, W., Malone, M., Kennedy, J. L., Schachar, R., Barr, C. L. Sequence variation in the 3-prime-untranslated region of the dopamine transporter gene and attention-deficit hyperactivity disorder (ADHD). Am. J. Med. Genet. 139B: 1-6, 2005. [PubMed: 16082693, related citations] [Full Text]

  7. Gainetdinov, R. R., Wetsel, W. C., Jones, S. R., Levin, E. D., Jaber, M., Caron, M. G. Role of serotonin in the paradoxical calming effect of psychostimulants on hyperactivity. Science 283: 397-401, 1999. [PubMed: 9888856, related citations] [Full Text]

  8. Gelernter, J., Vandenbergh, D., Kruger, S. D., Pauls, D. L., Kurlan, R., Pakstis, A. J., Kidd, K. K., Uhl, G. The dopamine transporter protein gene (SLC6A3): primary linkage mapping and linkage studies in Tourette syndrome. Genomics 30: 459-463, 1995. [PubMed: 8825631, related citations] [Full Text]

  9. Gill, M., Daly, G., Heron, S., Hawi, Z., Fitgerald, M. Confirmation of association between attention deficit hyperactivity disorder and a dopamine transporter polymorphism. Molec. Psychiat. 2: 311-313, 1997. [PubMed: 9246671, related citations] [Full Text]

  10. Giros, B., El Mestikawy, S., Godinot, N., Zheng, K., Han, H., Yang-Feng, T., Caron, M. G. Cloning, pharmacological characterization, and chromosome assignment of the human dopamine transporter. Molec. Pharm. 42: 383-390, 1992. [PubMed: 1406597, related citations]

  11. Giros, B., Jaber, M., Jones, S. R., Wightman, R. M., Caron, M. G. Hyperlocomotion and indifference to cocaine and amphetamine in mice lacking the dopamine receptor. Nature 379: 606-612, 1996. [PubMed: 8628395, related citations] [Full Text]

  12. Goldman, D. Dopamine transporter, alcoholism and other diseases. Nature Med. 1: 624-625, 1995. [PubMed: 7585136, related citations] [Full Text]

  13. Greenwood, T. A., Alexander, M., Keck, P. E., McElroy, S., Sadovnick, A. D., Remick, R. A., Kelsoe, J. R. Evidence for linkage disequilibrium between the dopamine transporter and bipolar disorder. Am. J. Med. Genet. 105: 145-151, 2001. [PubMed: 11304827, related citations] [Full Text]

  14. Greenwood, T. A., Schork, N. J., Eskin, E., Kelsoe, J. R. Identification of additional variants within the human dopamine transporter gene provides further evidence for an association with bipolar disorder in two independent samples. Molec. Psychiat. 11: 125-133, 2006. [PubMed: 16261167, related citations] [Full Text]

  15. Hebebrand, J., Dempfle, A., Saar, K., Thiele, H., Herpertz-Dahlmann, B., Linder, M., Kiefl, H., Remschmidt, H., Hemminger, U., Warnke, A., Knolker, U., Heiser, P., Friedel, S., Hinney, A., Schafer, H., Nurnberg, P., Konrad, K. A genome-wide scan for attention-deficit/hyperactivity disorder in 155 German sib-pairs. Molec. Psychiat. 11: 196-205, 2006. [PubMed: 16222334, related citations] [Full Text]

  16. Hejjas, K., Vas, J., Kubinyi, E., Sasvari-Szekely, M., Miklosi, A., Ronai, Z. Novel repeat polymorphisms of the dopaminergic neurotransmitter genes among dogs and wolves. Mammalian Genome 18: 871-879, 2007. [PubMed: 18049838, related citations] [Full Text]

  17. Kahn, R. S., Khoury, J., Nichols, W. C., Lanphear, B. P. Role of dopamine transporter genotype and maternal prenatal smoking in childhood hyperactive-impulsive, inattentive, and oppositional behaviors. J. Pediat. 143: 104-110, 2003. [PubMed: 12915833, related citations] [Full Text]

  18. Kurian, M. A., Li, Y., Zhen, J., Meyer, E., Hai, N., Christen, H.-J., Hoffmann, G. F., Jardine, P., von Moers, A., Mordekar, S. R., O'Callaghan, F., Wassmer, E., and 10 others. Clinical and molecular characterisation of hereditary dopamine transporter deficiency syndrome: an observational cohort and experimental study. Lancet Neurol. 10: 54-62, 2011. [PubMed: 21112253, images, related citations] [Full Text]

  19. Kurian, M. A., Zhen, J., Cheng, S.-Y., Li, Y., Mordekar, S. R., Jardine, P., Morgan, N. V., Meyer, E., Tee, L., Pasha, S., Wassmer, E., Heales, S. J. R., Gissen, P., Reith, M. E. A., Maher, E. R. Homozygous loss-of-function mutations in the gene encoding the dopamine transporter are associated with infantile parkinsonism-dystonia. J. Clin. Invest. 119: 1595-1603, 2009. [PubMed: 19478460, images, related citations] [Full Text]

  20. Kurlan, R., Behr, J., Medved, L., Shoulson, I., Pauls, D., Kidd, J. R., Kidd, K. K. Familial Tourette's syndrome: report of a large pedigree and potential for linkage analysis. Neurology 36: 772-776, 1986. [PubMed: 3458031, related citations] [Full Text]

  21. Langley, K., Turic, D., Peirce, T. R., Mills, S., Van Den Bree, M. B., Owen, M. J., O'Donovan, M. C., Thapar, A. No support for association between the dopamine transporter (DAT1) gene and ADHD. Am. J. Med. Genet. 139B: 7-10, 2005. [PubMed: 16082688, related citations] [Full Text]

  22. Lerman, C., Caporaso, N. E., Audrain, J., Main, D., Bowman, E. D., Lockshin, B., Boyd, N. R., Shields, P. G. Evidence suggesting the role of specific genetic factors in cigarette smoking. Health Psychol. 18: 14-20, 1999. [PubMed: 9925041, related citations] [Full Text]

  23. Lossie, A. C., Vandenbergh, D. J., Uhl, G. R., Camper, S. A. Localization of the dopamine transporter gene, Dat1, on mouse chromosome 13. Mammalian Genome 5: 117-118, 1994. [PubMed: 8180472, related citations] [Full Text]

  24. Navaroli, D. M., Stevens, Z. H., Uzelac, Z., Gabriel, L., King, M. J., Lifshitz, L. M., Sitte, H. H., Melikian, H. E. The plasma membrane-associated GTPase Rin interacts with the dopamine transporter and is required for protein kinase C-regulated dopamine transporter trafficking. J. Neurosci. 31: 13758-13770, 2011. [PubMed: 21957239, images, related citations] [Full Text]

  25. Puffenberger, E. G., Jinks, R. N., Sougnez, C., Cibulskis, K., Willert, R. A., Achilly, N. P., Cassidy, R. P., Fiorentini, C. J., Heiken, K. F., Lawrence, J. J., Mahoney, M. H., Miller, C. J., and 13 others. Genetic mapping and exome sequencing identify variants associated with five novel diseases. PLoS One 7: e28936, 2012. Note: Electronic Article. [PubMed: 22279524, images, related citations] [Full Text]

  26. Sabol, S. Z., Nelson, M. L., Fisher, C., Gunzerath, L., Brody, C. L., Hu, S., Sirota, L. A., Marcus, S. E., Greenberg, B. D., Lucas, F. R., IV, Benjamin, J., Murphy, D. L., Hamer, D. H. A genetic association for cigarette smoking behavior. Health Psychol. 18: 7-13, 1999. [PubMed: 9925040, related citations] [Full Text]

  27. Sotnikova, T. D., Caron, M. G., Gainetdinov, R. R. DDD mice, a novel acute mouse model of Parkinson's disease. Neurology 67 (suppl. 2): S12-S17, 2006. [PubMed: 17030735, related citations] [Full Text]

  28. Tiihonen, J., Kuikka, J., Bergstrom, K., Hakola, P., Karhu, J., Ryynanen, O.-P., Fohr, J. Altered striatal dopamine re-uptake site densities in habitually violent and non-violent alcoholics. Nature Med. 1: 654-657, 1995. [PubMed: 7585146, related citations] [Full Text]

  29. Vandenbergh, D. J., Persico, A. M., Hawkins, A. L., Griffin, C. A., Li, X., Jabs, E. W., Uhl, G. R. Human dopamine transporter gene (DAT1) maps to chromosome 5p15.3 and displays a VNTR. Genomics 14: 1104-1106, 1992. [PubMed: 1478653, related citations] [Full Text]

  30. Vandenbergh, D. J., Persico, A. M., Uhl, G. R. A human dopamine transporter cDNA predicts reduced glycosylation, displays a novel repetitive element and provides racially-dimorphic TaqI RFLPs. Molec. Brain Res. 15: 161-166, 1992. [PubMed: 1359373, related citations] [Full Text]

  31. Waldman, I. D., Rowe, D. C., Abramowitz, A., Kozel, S. T., Mohr, J. H., Sherman, S. L., Cleveland, H. H., Sanders, M. L., Gard, J. M. C., Stever, C. Association and linkage of the dopamine transporter gene and attention-deficit hyperactivity disorder in children: heterogeneity owing to diagnostic subtype and severity. Am. J. Hum. Genet. 63: 1767-1776, 1998. [PubMed: 9837830, related citations] [Full Text]

  32. Zhou, Z., Zhen, J., Karpowich, N. K., Goetz, R. M., Law, C. J., Reith, M. E. A., Wang, D.-N. LeuT-desipramine structure reveals how antidepressants block neurotransmitter reuptake. Science 317: 1390-1393, 2007. [PubMed: 17690258, images, related citations] [Full Text]


Patricia A. Hartz - updated : 1/20/2015
Cassandra L. Kniffin - updated : 1/29/2014
Cassandra L. Kniffin - updated : 2/27/2012
Cassandra L. Kniffin - updated : 11/23/2009
Patricia A. Hartz - updated : 10/24/2008
Ada Hamosh - updated : 10/15/2007
Cassandra L. Kniffin - updated : 9/10/2007
John Logan Black, III - updated : 8/4/2006
John Logan Black, III - updated : 6/2/2006
John Logan Black, III - updated : 5/17/2006
John Logan Black, III - updated : 5/12/2006
Natalie E. Krasikov - updated : 3/29/2004
Victor A. McKusick - updated : 3/11/1999
Victor A. McKusick - updated : 1/14/1999
Victor A. McKusick - updated : 12/18/1998
Victor A. McKusick - updated : 4/29/1998
Victor A. McKusick - updated : 8/13/1997
Creation Date:
Victor A. McKusick : 1/8/1993
carol : 01/08/2020
carol : 07/09/2018
ckniffin : 07/05/2018
carol : 06/05/2017
carol : 06/02/2017
carol : 02/25/2015
mgross : 1/26/2015
mcolton : 1/20/2015
mcolton : 2/21/2014
mgross : 2/10/2014
ckniffin : 1/29/2014
carol : 3/5/2012
terry : 3/2/2012
ckniffin : 2/27/2012
terry : 12/16/2009
wwang : 12/11/2009
wwang : 12/11/2009
ckniffin : 11/23/2009
carol : 9/4/2009
wwang : 4/16/2009
mgross : 10/24/2008
mgross : 10/24/2008
carol : 7/25/2008
alopez : 10/26/2007
terry : 10/15/2007
wwang : 9/12/2007
ckniffin : 9/10/2007
carol : 6/15/2007
alopez : 10/25/2006
carol : 8/29/2006
terry : 8/4/2006
wwang : 6/6/2006
terry : 6/2/2006
wwang : 5/22/2006
terry : 5/17/2006
wwang : 5/16/2006
wwang : 5/16/2006
terry : 5/12/2006
ckniffin : 4/18/2006
tkritzer : 3/30/2004
terry : 3/29/2004
carol : 3/8/2002
terry : 3/8/2002
terry : 3/16/1999
terry : 3/11/1999
alopez : 1/15/1999
alopez : 1/14/1999
joanna : 1/14/1999
carol : 12/28/1998
terry : 12/18/1998
carol : 4/30/1998
terry : 4/29/1998
terry : 8/13/1997
mark : 4/1/1996
mark : 2/14/1996
mark : 1/21/1996
terry : 1/17/1996
mark : 12/4/1995
terry : 9/13/1995
mark : 7/28/1995
carol : 2/25/1994
carol : 4/27/1993
carol : 4/26/1993
carol : 2/17/1993

* 126455

SOLUTE CARRIER FAMILY 6 (NEUROTRANSMITTER TRANSPORTER, DOPAMINE), MEMBER 3; SLC6A3


Alternative titles; symbols

DOPAMINE TRANSPORTER; DAT
DAT1


HGNC Approved Gene Symbol: SLC6A3

Cytogenetic location: 5p15.33     Genomic coordinates (GRCh38): 5:1,392,794-1,445,440 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
5p15.33 {Nicotine dependence, protection against} 188890 3
Parkinsonism-dystonia, infantile, 1 613135 Autosomal recessive 3

TEXT

Description

The dopamine transporter (DAT), which is encoded by the SLC6A3 gene, mediates the active reuptake of dopamine from the synapse and is a principal regulator of dopaminergic neurotransmission. The SLC6A3 gene has been implicated in human disorders such as parkinsonism, Tourette syndrome, and substance abuse (Vandenbergh et al., 1992).


Cloning and Expression

Vandenbergh et al. (1992) cloned cDNAs encoding the entire coding sequence and 3-prime untranslated sequence of the human DAT1 gene.

Kurian et al. (2009) noted that SLC6A3 contains 12 transmembrane domains with helically unwound regions in the first and sixth domains. Extracellular and intracellular loops include helical portions E2, E3, E4A, E4B, I1, and I5. Mature, glycosylated DAT has a molecular mass of 85 kD, whereas immature nonglycosylated DAT has a molecular mass of 55 kD and does not transport dopamine as efficiently as the mature protein.

Navaroli et al. (2011) reported that DAT has an endocytic signal near the C terminus that modulates both basal and protein kinase C (PKC; see 176960)-enhanced DAT internalization. Immunohistochemical analysis revealed that rat Dat localized to plasma membrane foci that corresponded to both lipid rafts and non-raft microdomains in PC12 pheochromocytoma cells.


Gene Function

By yeast 2-hybrid analysis of a human substantia nigra cDNA library, Navaroli et al. (2011) found that the small GTPase RIN (RIT2; 609592) interacted with the isolated C-terminal endocytic signal of human DAT. Coimmunoprecipitation and protein pull-down assays with PC12 cells that were transfected with human DAT revealed direct interaction between DAT and Rin. Rin did not interact with other members of the SLC6 transporter family. DAT colocalized with Rin at plasma membrane foci, predominantly at lipid rafts, in transfected PC12 cells. Phorbol ester-induced PKC activation enhanced DAT-Rin interaction, followed by their dissociation and DAT internalization into endocytic vesicles. These effects did not occur with a DAT mutant lacking the C-terminal endocytic signal or with a Rin mutant lacking GTPase activity. Knockdown of Rin via short hairpin RNA completely inhibited PKC-induced DAT internalization.


Gene Structure

The SLC6A3 gene contains 15 exons spanning approximately 60 kb (Vandenbergh et al., 1992).


Mapping

By in situ hybridization and by PCR amplification of rodent/human somatic cell hybrid DNAs, Vandenbergh et al. (1992) and Giros et al. (1992) mapped the SLC6A3 gene to chromosome 5p15.3.

Lossie et al. (1994) mapped the mouse Dat1 homolog to chromosome 15 by linkage analysis of an interspecific backcross.

Gelernter et al. (1995) demonstrated close linkage between the SLC6A3 gene and several markers previously mapped to distal chromosome 5p.


Biochemical Features

Crystal Structure

Zhou et al. (2007) determined the crystal structure at 2.9 angstroms of the bacterial leucine transporter (LeuT), a homolog of SERT (182138), NET (163970), and DAT, in complex with leucine and the antidepressant desipramine. Desipramine binds at the inner end of the extracellular cavity of the transporter and is held in place by a hairpin loop and by a salt bridge. This binding site is separated from the leucine-binding site by the extracellular gate of the transporter. By directly locking the gate, desipramine prevents conformational changes and blocks substrate transport. Mutagenesis experiments on human SERT and DAT indicate that both the desipramine-binding site and its inhibition mechanism are probably conserved in the human neurotransmitter transporters.


Molecular Genetics

Vandenbergh et al. (1992) identified a 40-bp variable-number tandem repeat (VNTR) polymorphism in the 3-prime untranslated region of the DAT1 gene with repeat copy numbers ranging from 3 to 11.

Byerley et al. (1993) demonstrated a VNTR polymorphism related to the DAT1 gene. Doucette-Stamm et al. (1995) established allele frequencies for the 40-bp VNTR polymorphism. Differences were found between black Americans and Caucasians or Hispanics, but there were no differences between Caucasians and Hispanics. A previously unreported allele was detected in all 3 populations.

Association with Attention-Deficit Hyperactivity Disorder

Attention-deficit hyperactivity disorder (ADHD; 143465) is presumably heritable in some cases. Some ADHD patients respond to medications that inhibit the dopamine transporter, including methylphenidate, amphetamine, pemoline, and bupropion. Cook et al. (1995) used the haplotype-based haplotype relative risk (HHRR) method to test for association between a VNTR polymorphism at the DAT1 locus and 49 cases of ADHD. Eight cases of undifferentiated attention-deficit disorder (UADD) were also studied. All cases were studied in trios composed of father, mother, and affected offspring. HHRR analysis revealed significant association between ADHD/UADD and the 480-bp DAT1 allele. When cases of UADD were dropped from the analysis, similar results were found. Cook et al. (1995) suggested that molecular analysis of DAT1 may identify mutations that increase susceptibility to this disorder and that biochemical analysis of such mutations may lead to development of more effective therapeutic interventions.

Gill et al. (1997) found a significant association between ADHD and the 480-bp DAT1 VNTR allele. Gill et al. (1997) reported concordance rates for ADHD in monozygotic and dizygotic twins of 81% and 29%, respectively.

Waldman et al. (1998) used 4 analytic strategies to examine the association and linkage of the DAT1 gene with ADHD. The study group comprised 122 children referred to psychiatric clinics for behavioral and learning problems that included but were not limited to ADHD, as well as their parents and sibs. Within-family analyses of linkage disequilibrium, using the transmission disequilibrium test (TDT), confirmed the 480-bp DAT1 allele as the high-risk allele. In between-family association analyses, levels of hyperactive-impulsive symptoms but not inattentive symptoms were related to the number of DAT1 high-risk alleles. Sibs discordant for the number of DAT1 high-risk alleles differed markedly in their levels of both hyperactive-impulsive and inattentive symptoms, such that the sibs with the higher number of high-risk alleles had much higher symptom levels. Within-family analyses of linkage disequilibrium, using the TDT, suggested association and linkage of ADHD with DAT1 and that this relationship was especially strong with the combined but not the inattentive subtype. The findings were believed to represent one of the first replicated relations of a candidate gene and a psychiatric disorder in children.

Kahn et al. (2003) found a significant association between homozygosity for the 480-bp DAT allele and hyperactivity-impulsivity and oppositional behaviors only in the presence of maternal prenatal smoking. They emphasized the importance of incorporating environmental cofactors in genetic studies of ADHD.

Langley et al. (2005) tested the DAT1 3-prime VNTR and 3 putative DAT1 promoter SNPs for association with ADHD in 263 parent-proband trios using family-based association methods. No evidence of association with any of the promoter region SNPs or the VNTR was seen. Haplotype analysis was also nonsignificant, and no association was found between the VNTR and response to stimulant medications. By case-control analysis of the VNTR in 263 cases and 287 controls, the 10-repeat allele showed no significant association compared to all other alleles combined.

Feng et al. (2005) investigated whether the DAT1 3-prime UTR contributed to ADHD by genotyping DNA variants around the VNTR region in a sample of 178 ADHD families. Variants included an MspI polymorphism (rs27072), a DraI T-C transition reported to influence DAT1 expression levels, and a BstUI polymorphism (rs3863145), in addition to the VNTR. They found an association between the G allele of the MspI SNP and ADHD (P = 0.009) but not with alleles of the VNTR polymorphism or the BstUI polymorphism, and they did not observe the DraI SNP. Feng et al. (2005) screened the VNTR region by direct sequencing to determine if there were additional variants within the repeats that could account for association with ADHD and found no variation in the VNTR region for either the 10- or 9-repeat alleles in the probands screened.

In both a Taiwanese ADHD sample and an English ADHD sample, Brookes et al. (2006) identified association with the DAT1 3-prime UTR VNTR and a novel DAT1 intron 8 repeat polymorphism. A risk haplotype composed of the 2 repeat polymorphisms was also associated with ADHD in both populations, and the authors found that the risk haplotype showed significant interactions with maternal use of alcohol during pregnancy in the English sample.

Hebebrand et al. (2006) found no evidence of association between ADHD and the DAT1 VNTR in 102 German families with 2 or more offspring who fulfilled diagnostic criteria for ADHD.

Cheuk et al. (2006) studied the association between the DAT1 VNTR and ADHD in Chinese children (64 ADHD cases, including 52 boys and 12 girls, their families, and 64 sex-matched normal controls) in Hong Kong. Patients were diagnosed with combined, hyperactive-impulsive, or inattentive subtypes of ADHD. The 10-repeat allele (92.6%) and the 10/10 (85.2%) repeat genotype were most prevalent. Both family-based and case-control analyses showed no association between the DAT1 polymorphisms and ADHD.

Animal studies suggest that the development of substance dependence (e.g., alcoholism) is associated with dopaminergic activity in striatum and the limbic system (Tiihonen et al., 1995). Several genetic studies had indicated that allele A1 at the DRD2 (126450) locus is associated with both density of dopamine receptor-2 and alcoholism. Tiihonen et al. (1995) studied striatal dopamine reuptake sites in 19 healthy controls, 19 habitually impulsive violent alcoholics, and 10 nonviolent alcoholics with single photon emission computed tomography (SPECT) using iodine-123-labeled beta-CIT as a tracer. Blind quantitative analysis revealed that the striatal dopamine transporter density was markedly lower in nonviolent alcoholics than in healthy controls (P less than 0.001), while violent alcoholics had slightly higher dopamine transporter densities than controls (P less than 0.10). Goldman (1995) suggested that the next step would be to look for inherited variants of the DAT1 gene that may influence vulnerability to alcoholism and other psychiatric diseases.

Association with Tourette Syndrome

In a large Canadian Mennonite pedigree segregating for Tourette syndrome (137580) (Kurlan et al., 1986), Gelernter et al. (1995) excluded linkage to SLC6A3. They cautiously pointed out, however, that these results did not exclude a role for the dopamine transporter in influencing risk for Tourette syndrome in combination with other loci.

Association with Cigarette Smoking

Dopaminergic genes are likely candidates for heritable influences on cigarette smoking (188890). Lerman et al. (1999) and Sabol et al. (1999) reported associations between allele 9 of a DAT1 polymorphism, SLC6A3*9 (126455.0001), and smoking status. While Lerman et al. (1999) reported association of the SLC6A3*9 allele with lack of smoking, late initiation of smoking, and length of quitting attempts, Sabol et al. (1999) reported that the significant association between SLC6A3*9 and smoking status was due to an effect on cessation rather than initiation. The SLC6A3*9 polymorphism was also associated with low scores for novelty seeking, which was the most significant personality correlate of smoking cessation.

Association with Bipolar Disorder

Greenwood et al. (2001) reported evidence for an association between DAT1 and bipolar disorder (MAFD1; 125480) in a sample of 50 parent-proband trios. Using the transmission disequilibrium test (TDT), they showed an association between a haplotype composed of 5 SNPs in the 3-prime region of the DAT1 gene, exon 9 through exon 15, and bipolar disorder (allele-wise TDT empirical P = 0.001; genotype-wise TDT empirical P = 0.0004). Greenwood et al. (2006) analyzed a total of 22 SNPs in the 50 previously studied parent-proband trios and an independent set of 70 parent-proband trios. Using TDT analysis, an intron 8 SNP and an intron 13 SNP were found to be moderately associated with bipolar disorder, each in 1 of the 2 independent samples. Analysis of haplotypes of all 22 SNPs in sliding windows of 5 adjacent SNPs revealed an association to the region near intron 7 and 8 in both samples (empirical P values of 0.002 and 0.001, respectively, for the same window). The haplotype block structure observed by Greenwood et al. (2001) was confirmed in this new sample with the greater resolution allowing for discrimination of a third haplotype block in the middle of the gene.

Infantile Parkinsonism-Dystonia 1

By linkage analysis followed by candidate gene sequencing of a consanguineous Pakistani family with infantile parkinsonism-dystonia (PKDYS1; 613135), Kurian et al. (2009) identified a homozygous mutation (L368Q; 126455.0002) in the SLC6A3 gene. A similarly affected individual from a second family had a different homozygous mutation (P395L; 126455.0003). In vitro functional expression studies showed that both mutant proteins had no dopamine uptake activity.

In 8 unrelated patients with dopamine transporter deficiency syndrome, Kurian et al. (2011) identified homozygous or compound heterozygous mutations in the SLC6A3 gene (see, e.g., 126455.0005-126455.0007). None of the patients shared a mutation, suggesting the absence of mutational hotspots. In vitro functional expression studies in HEK293 cells showed that the mutations caused a loss of transporter function and decreased expression of the normal protein. All patients presented in early infancy with a complex motor disorder involving both hypo- and hyperkinetic movements. There was no effective treatment, and several patients died in the teenage years.

By homozygosity mapping followed by exome sequencing of a Mennonite family in which 2 sisters had infantile parkinsonism-dystonia, Puffenberger et al. (2012) identified a homozygous splice site mutation in the SLC6A3 gene (126455.0004).


Animal Model

Giros et al. (1996) found that the disruption of the mouse Dat1 gene results in spontaneous hyperlocomotion despite major adaptive changes such as decreases in neurotransmitter and receptor levels. In homozygous-null mice, dopamine persisted at least 100 times longer in the extracellular space, providing a biochemical explanation of the hyperdopaminergic phenotype and demonstrating the critical role of DAT1 in regulating neurotransmission. The authors noted that the dopamine transporter is an obligatory target of cocaine and amphetamine, as demonstrated by the fact that these psychostimulants had no effect on locomotor activity or dopamine release and uptake in mice lacking the transporter. Giros et al. (1996) noted that there are similarities between the hyperdopaminergic phenotype of the knockout mice and some of the positive symptoms of patients with schizophrenia. The authors also postulated that specific blockade of DAT1 with high-affinity inhibitors may be beneficial in illnesses such as Parkinson disease (see 168600), where the effective levels of dopamine are markedly reduced.

Gainetdinov et al. (1999) continued the studies of Giros et al. (1992), which demonstrated mice lacking the gene encoding the plasma membrane dopamine transporter have elevated dopaminergic tone and are hyperactive. They found that this activity was exacerbated by exposure to a novel environment. Additionally, these mice were impaired in spatial cognitive function, and they showed a decrease in locomotion in response to psychostimulants. This paradoxical calming effect of psychostimulants depended on serotonergic neurotransmission, and emphasized the importance of a relative balance of the serotonin and dopamine systems for normal motor activity. The parallels between the DAT knockout mice and individuals with ADHD suggested that common mechanisms may underlie some of their behaviors and responses to psychostimulants.

Sotnikova et al. (2006) developed a novel acute mouse model of severe dopamine deficiency using Dat-null mice and pharmacologic inhibition of tyrosine hydroxylase. Dopamine-deficient Dat-null mice (DDD) demonstrated severe akinesia, rigidity, tremor, and ptosis, similar to behaviors observed in patients with Parkinson disease. Interestingly, DDD mice were able to swim in water, indicating that certain movements and conditions can occur independently of dopamine. Dopamine agonists such as L-DOPA temporarily restored locomotion in DDD mice, and amphetamine derivatives showed effectiveness in reducing motor abnormalities in DDD mice. Sotnikova et al. (2006) noted that the DDD mouse model provides a unique opportunity to screen potential therapeutic agents for the treatment of Parkinson disease.

Using an in silico search, followed by PCR, Hejjas et al. (2007) identified VNTR polymorphisms in genes of the dopaminergic system in 4 dog breeds and European gray wolves. Polymorphisms of the DRD4 (126452), DBH (609312), and DAT genes were associated with attention deficit, but not activity-impulsivity, in Belgian Tervuerens, a breed that had almost all genetic variants identified.


ALLELIC VARIANTS 7 Selected Examples):

.0001   NICOTINE DEPENDENCE, PROTECTION AGAINST

SLC6A3, 9-REPEAT VNTR
SNP: rs28363170, ClinVar: RCV000018247

Lerman et al. (1999) reported associations between allele 9 of the SLC6A3 gene (SLC6A3*9), a polymorphism containing 9 repeats of the 40-basepair 3-prime VNTR, and lack of smoking, late initiation of smoking, and length of quitting attempts (see 188890). The association with smoking risk was modified by the DRD2 (126450) genotype, resulting in a 50% reduction in smoking risk for individuals carrying both SLC6A3*9 and DRD2-A2. Sabol et al. (1999) extended this study by examining both smoking behavior and personality traits of 1,107 individuals in a diverse population of nonsmokers, current smokers, and former smokers. A significant association between SLC6A3*9 and smoking status was confirmed and was due to an effect on cessation rather than initiation. The SLC6A3*9 polymorphism was also associated with low scores for novelty seeking, which was the most significant personality correlate of smoking cessation. Sabol et al. (1999) hypothesized that individuals carrying the SLC6A3*9 polymorphism have altered dopamine transmission, which reduces their need for novelty and reward by external stimuli, including cigarettes. Sabol et al. (1999) found that individuals carrying the SLC6A3*9 allele were 1.5-fold more likely to have quit smoking than were individuals lacking this polymorphism. The results supported the finding of Lerman et al. (1999) of an association between SLC6A3 and length of previous cessation attempts in current smokers.


.0002   PARKINSONISM-DYSTONIA, INFANTILE, 1

SLC6A3, LEU368GLN
SNP: rs267607068, ClinVar: RCV000018249

In 2 affected members of a consanguineous Pakistani family with infantile parkinsonism-dystonia (PKDYS1; 613135), Kurian et al. (2009) identified a homozygous 1103T-A transversion in exon 8 of the SLC6A3 gene, resulting in a leu368-to-gln (L368Q) substitution in a highly conserved residue in transmembrane domain-7 toward the exterior of the protein surface. In vitro functional expression assays in HEK293 cells showed that L368Q-mutant DAT had no dopamine reuptake activity. Binding affinity for a cocaine analog was near normal, but the potency of dopamine in inhibiting cocaine analog binding was greatly reduced in the L368Q mutant, indicating reduced dopamine binding affinity. The mutant protein also resulted in a profound reduction in mature glycosylated DAT, which likely impaired transport function. The mutation was not identified in 544 alleles from Asian individuals or in 438 alleles from individuals of mixed European descent.


.0003   PARKINSONISM-DYSTONIA, INFANTILE, 1

SLC6A3, PRO395LEU
SNP: rs267607069, gnomAD: rs267607069, ClinVar: RCV000018250

In a patient of European descent, born of consanguineous parents, with infantile parkinsonism-dystonia (PKDYS1; 613135), Kurian et al. (2009) identified a homozygous 1184C-T transition in exon 9 of the SLC6A3 gene, resulting in a pro395-to-leu (P395L) substitution in a highly conserved residue of the E4B loop close to transmembrane domain-8. In vitro functional expression assays in HEK293 cells showed that the P395L-mutant protein had no dopamine reuptake activity. Binding affinity for a cocaine analog was near normal, and the potency of dopamine in inhibiting cocaine analog binding was near normal in the P395L mutant, suggesting no impact on dopamine binding affinity. The mutant protein did result in a profound reduction in mature glycosylated DAT, which likely impaired transport function. The mutation was not identified in 544 alleles from Asian individuals or in 438 alleles from individuals of mixed European descent.


.0004   PARKINSONISM-DYSTONIA, INFANTILE, 1

SLC6A3, IVS9DS, G-T, +1
SNP: rs431905504, gnomAD: rs431905504, ClinVar: RCV000022531, RCV000493130, RCV003593863

In 2 Mennonite sisters with infantile parkinsonism-dystonia (PKDYS1; 613135), Puffenberger et al. (2012) identified a homozygous G-to-T transversion in intron 9 of the SLC6A3 gene. The mutation was found by homozygosity mapping followed by exome sequencing of the candidate region. No carriers of this mutation were found among 201 Mennonite control samples. The proband developed irritability and feeding difficulties soon after birth, followed by generalized rigidity and dystonia during early infancy. She had impaired motor development and severe rigid parkinsonism by late childhood. She could not speak or use her hands to communicate, and it was difficult to assess cognitive function or thought content. Brain structure was normal. Cerebrospinal fluid showed increased homovanillic acid (HVA). A similarly affected sister had died.


.0005   PARKINSONISM-DYSTONIA, INFANTILE, 1

SLC6A3, IVS7DS, G-A, +1
SNP: rs431905514, ClinVar: RCV000083262, RCV002228329

In a patient, born of consanguineous Turkish parents, with infantile parkinsonism-dystonia (PKDYS1; 613135), Kurian et al. (2011) identified a homozygous G-to-A transition in intron 7 of the SLC6A3 gene (c.1031+1G-A), predicted to cause aberrant splicing.


.0006   PARKINSONISM-DYSTONIA, INFANTILE, 1

SLC6A3, LEU224PRO
SNP: rs431905515, ClinVar: RCV000083263

In a patient of mixed European descent, born of consanguineous parents, with PKDYS (PKDYS1; 613135), Kurian et al. (2011) identified a homozygous c.671T-C transition in exon 5 of the SLC6A3 gene, resulting in a leu224-to-pro (L224P) substitution at a highly conserved residue in mammalian species. In vitro functional expression studies of the mutation in HEK293 cells showed nonspecific uptake for the mutant transporter, and Western blot analysis indicated deficiency of the mature protein. These findings were consistent with a loss of function.


.0007   PARKINSONISM-DYSTONIA, INFANTILE, 1

SLC6A3, ARG521TRP
SNP: rs431905516, gnomAD: rs431905516, ClinVar: RCV000083264, RCV001854451

In a patient of mixed European descent, born of consanguineous parents, with PKDYS (PKDYS1; 613135), Kurian et al. (2011) identified a homozygous c.1561C-T transition in exon 12 of the SLC6A3 gene, resulting in an arg521-to-trp (R521W) substitution at a highly conserved residue. In vitro functional expression studies of the mutation in HEK293 cells showed decreased dopamine uptake (27% of wildtype), and Western blot analysis indicated deficiency of the mature protein. These findings were consistent with a loss of function.


REFERENCES

  1. Brookes, K.-J., Mill, J., Guindalini, C., Curran, S., Xu, X., Knight, J., Chen, C.-K., Huang, Y.-S., Sethna, V., Taylor, E., Chen, W., Breen, G., Asherson, P. A common haplotype of the dopamine transporter gene associated with attention-deficit/hyperactivity disorder and interacting with maternal use of alcohol during pregnancy. Arch. Gen. Psychiat. 63: 74-81, 2006. [PubMed: 16389200] [Full Text: https://doi.org/10.1001/archpsyc.63.1.74]

  2. Byerley, W., Hoff, M., Holik, J., Caron, M. G., Giros, B. VNTR polymorphism for the human dopamine transporter gene (DAT1). Hum. Molec. Genet. 2: 335, 1993. [PubMed: 8098980] [Full Text: https://doi.org/10.1093/hmg/2.3.335]

  3. Cheuk, D. K. L., Li, S. Y. H., Wong, V. No association between VNTR polymorphisms of dopamine transporter gene and attention deficit hyperactivity disorder in Chinese children. Am. J. Med. Genet. 141B: 123-125, 2006. [PubMed: 16402340] [Full Text: https://doi.org/10.1002/ajmg.b.30280]

  4. Cook, E. H., Jr., Stein, M. A., Krasowski, M. D., Cox, N. J., Olkon, D. M., Kieffer, J. E., Leventhal, B. L. Association of attention-deficit disorder and the dopamine transporter gene. Am. J. Hum. Genet. 56: 993-998, 1995. [PubMed: 7717410]

  5. Doucette-Stamm, L., Blakely, D. J., Tian, J., Mockus, S., Mao, J. Population genetic study of the human dopamine transporter gene (DAT1). Genet. Epidemiol. 12: 303-308, 1995. [PubMed: 7557351] [Full Text: https://doi.org/10.1002/gepi.1370120307]

  6. Feng, Y., Wigg, K. G., Makkar, R., Ickowicz, A., Pathare, T., Tannock, R., Roberts, W., Malone, M., Kennedy, J. L., Schachar, R., Barr, C. L. Sequence variation in the 3-prime-untranslated region of the dopamine transporter gene and attention-deficit hyperactivity disorder (ADHD). Am. J. Med. Genet. 139B: 1-6, 2005. [PubMed: 16082693] [Full Text: https://doi.org/10.1002/ajmg.b.30190]

  7. Gainetdinov, R. R., Wetsel, W. C., Jones, S. R., Levin, E. D., Jaber, M., Caron, M. G. Role of serotonin in the paradoxical calming effect of psychostimulants on hyperactivity. Science 283: 397-401, 1999. [PubMed: 9888856] [Full Text: https://doi.org/10.1126/science.283.5400.397]

  8. Gelernter, J., Vandenbergh, D., Kruger, S. D., Pauls, D. L., Kurlan, R., Pakstis, A. J., Kidd, K. K., Uhl, G. The dopamine transporter protein gene (SLC6A3): primary linkage mapping and linkage studies in Tourette syndrome. Genomics 30: 459-463, 1995. [PubMed: 8825631] [Full Text: https://doi.org/10.1006/geno.1995.1265]

  9. Gill, M., Daly, G., Heron, S., Hawi, Z., Fitgerald, M. Confirmation of association between attention deficit hyperactivity disorder and a dopamine transporter polymorphism. Molec. Psychiat. 2: 311-313, 1997. [PubMed: 9246671] [Full Text: https://doi.org/10.1038/sj.mp.4000290]

  10. Giros, B., El Mestikawy, S., Godinot, N., Zheng, K., Han, H., Yang-Feng, T., Caron, M. G. Cloning, pharmacological characterization, and chromosome assignment of the human dopamine transporter. Molec. Pharm. 42: 383-390, 1992. [PubMed: 1406597]

  11. Giros, B., Jaber, M., Jones, S. R., Wightman, R. M., Caron, M. G. Hyperlocomotion and indifference to cocaine and amphetamine in mice lacking the dopamine receptor. Nature 379: 606-612, 1996. [PubMed: 8628395] [Full Text: https://doi.org/10.1038/379606a0]

  12. Goldman, D. Dopamine transporter, alcoholism and other diseases. Nature Med. 1: 624-625, 1995. [PubMed: 7585136] [Full Text: https://doi.org/10.1038/nm0795-624]

  13. Greenwood, T. A., Alexander, M., Keck, P. E., McElroy, S., Sadovnick, A. D., Remick, R. A., Kelsoe, J. R. Evidence for linkage disequilibrium between the dopamine transporter and bipolar disorder. Am. J. Med. Genet. 105: 145-151, 2001. [PubMed: 11304827] [Full Text: https://doi.org/10.1002/1096-8628(2001)9999:9999<::aid-ajmg1161>3.0.co;2-8]

  14. Greenwood, T. A., Schork, N. J., Eskin, E., Kelsoe, J. R. Identification of additional variants within the human dopamine transporter gene provides further evidence for an association with bipolar disorder in two independent samples. Molec. Psychiat. 11: 125-133, 2006. [PubMed: 16261167] [Full Text: https://doi.org/10.1038/sj.mp.4001764]

  15. Hebebrand, J., Dempfle, A., Saar, K., Thiele, H., Herpertz-Dahlmann, B., Linder, M., Kiefl, H., Remschmidt, H., Hemminger, U., Warnke, A., Knolker, U., Heiser, P., Friedel, S., Hinney, A., Schafer, H., Nurnberg, P., Konrad, K. A genome-wide scan for attention-deficit/hyperactivity disorder in 155 German sib-pairs. Molec. Psychiat. 11: 196-205, 2006. [PubMed: 16222334] [Full Text: https://doi.org/10.1038/sj.mp.4001761]

  16. Hejjas, K., Vas, J., Kubinyi, E., Sasvari-Szekely, M., Miklosi, A., Ronai, Z. Novel repeat polymorphisms of the dopaminergic neurotransmitter genes among dogs and wolves. Mammalian Genome 18: 871-879, 2007. [PubMed: 18049838] [Full Text: https://doi.org/10.1007/s00335-007-9070-0]

  17. Kahn, R. S., Khoury, J., Nichols, W. C., Lanphear, B. P. Role of dopamine transporter genotype and maternal prenatal smoking in childhood hyperactive-impulsive, inattentive, and oppositional behaviors. J. Pediat. 143: 104-110, 2003. [PubMed: 12915833] [Full Text: https://doi.org/10.1016/S0022-3476(03)00208-7]

  18. Kurian, M. A., Li, Y., Zhen, J., Meyer, E., Hai, N., Christen, H.-J., Hoffmann, G. F., Jardine, P., von Moers, A., Mordekar, S. R., O'Callaghan, F., Wassmer, E., and 10 others. Clinical and molecular characterisation of hereditary dopamine transporter deficiency syndrome: an observational cohort and experimental study. Lancet Neurol. 10: 54-62, 2011. [PubMed: 21112253] [Full Text: https://doi.org/10.1016/S1474-4422(10)70269-6]

  19. Kurian, M. A., Zhen, J., Cheng, S.-Y., Li, Y., Mordekar, S. R., Jardine, P., Morgan, N. V., Meyer, E., Tee, L., Pasha, S., Wassmer, E., Heales, S. J. R., Gissen, P., Reith, M. E. A., Maher, E. R. Homozygous loss-of-function mutations in the gene encoding the dopamine transporter are associated with infantile parkinsonism-dystonia. J. Clin. Invest. 119: 1595-1603, 2009. [PubMed: 19478460] [Full Text: https://doi.org/10.1172/JCI39060]

  20. Kurlan, R., Behr, J., Medved, L., Shoulson, I., Pauls, D., Kidd, J. R., Kidd, K. K. Familial Tourette's syndrome: report of a large pedigree and potential for linkage analysis. Neurology 36: 772-776, 1986. [PubMed: 3458031] [Full Text: https://doi.org/10.1212/wnl.36.6.772]

  21. Langley, K., Turic, D., Peirce, T. R., Mills, S., Van Den Bree, M. B., Owen, M. J., O'Donovan, M. C., Thapar, A. No support for association between the dopamine transporter (DAT1) gene and ADHD. Am. J. Med. Genet. 139B: 7-10, 2005. [PubMed: 16082688] [Full Text: https://doi.org/10.1002/ajmg.b.30206]

  22. Lerman, C., Caporaso, N. E., Audrain, J., Main, D., Bowman, E. D., Lockshin, B., Boyd, N. R., Shields, P. G. Evidence suggesting the role of specific genetic factors in cigarette smoking. Health Psychol. 18: 14-20, 1999. [PubMed: 9925041] [Full Text: https://doi.org/10.1037//0278-6133.18.1.14]

  23. Lossie, A. C., Vandenbergh, D. J., Uhl, G. R., Camper, S. A. Localization of the dopamine transporter gene, Dat1, on mouse chromosome 13. Mammalian Genome 5: 117-118, 1994. [PubMed: 8180472] [Full Text: https://doi.org/10.1007/BF00292340]

  24. Navaroli, D. M., Stevens, Z. H., Uzelac, Z., Gabriel, L., King, M. J., Lifshitz, L. M., Sitte, H. H., Melikian, H. E. The plasma membrane-associated GTPase Rin interacts with the dopamine transporter and is required for protein kinase C-regulated dopamine transporter trafficking. J. Neurosci. 31: 13758-13770, 2011. [PubMed: 21957239] [Full Text: https://doi.org/10.1523/JNEUROSCI.2649-11.2011]

  25. Puffenberger, E. G., Jinks, R. N., Sougnez, C., Cibulskis, K., Willert, R. A., Achilly, N. P., Cassidy, R. P., Fiorentini, C. J., Heiken, K. F., Lawrence, J. J., Mahoney, M. H., Miller, C. J., and 13 others. Genetic mapping and exome sequencing identify variants associated with five novel diseases. PLoS One 7: e28936, 2012. Note: Electronic Article. [PubMed: 22279524] [Full Text: https://doi.org/10.1371/journal.pone.0028936]

  26. Sabol, S. Z., Nelson, M. L., Fisher, C., Gunzerath, L., Brody, C. L., Hu, S., Sirota, L. A., Marcus, S. E., Greenberg, B. D., Lucas, F. R., IV, Benjamin, J., Murphy, D. L., Hamer, D. H. A genetic association for cigarette smoking behavior. Health Psychol. 18: 7-13, 1999. [PubMed: 9925040] [Full Text: https://doi.org/10.1037//0278-6133.18.1.7]

  27. Sotnikova, T. D., Caron, M. G., Gainetdinov, R. R. DDD mice, a novel acute mouse model of Parkinson's disease. Neurology 67 (suppl. 2): S12-S17, 2006. [PubMed: 17030735] [Full Text: https://doi.org/10.1212/wnl.67.7_suppl_2.s12]

  28. Tiihonen, J., Kuikka, J., Bergstrom, K., Hakola, P., Karhu, J., Ryynanen, O.-P., Fohr, J. Altered striatal dopamine re-uptake site densities in habitually violent and non-violent alcoholics. Nature Med. 1: 654-657, 1995. [PubMed: 7585146] [Full Text: https://doi.org/10.1038/nm0795-654]

  29. Vandenbergh, D. J., Persico, A. M., Hawkins, A. L., Griffin, C. A., Li, X., Jabs, E. W., Uhl, G. R. Human dopamine transporter gene (DAT1) maps to chromosome 5p15.3 and displays a VNTR. Genomics 14: 1104-1106, 1992. [PubMed: 1478653] [Full Text: https://doi.org/10.1016/s0888-7543(05)80138-7]

  30. Vandenbergh, D. J., Persico, A. M., Uhl, G. R. A human dopamine transporter cDNA predicts reduced glycosylation, displays a novel repetitive element and provides racially-dimorphic TaqI RFLPs. Molec. Brain Res. 15: 161-166, 1992. [PubMed: 1359373] [Full Text: https://doi.org/10.1016/0169-328x(92)90165-8]

  31. Waldman, I. D., Rowe, D. C., Abramowitz, A., Kozel, S. T., Mohr, J. H., Sherman, S. L., Cleveland, H. H., Sanders, M. L., Gard, J. M. C., Stever, C. Association and linkage of the dopamine transporter gene and attention-deficit hyperactivity disorder in children: heterogeneity owing to diagnostic subtype and severity. Am. J. Hum. Genet. 63: 1767-1776, 1998. [PubMed: 9837830] [Full Text: https://doi.org/10.1086/302132]

  32. Zhou, Z., Zhen, J., Karpowich, N. K., Goetz, R. M., Law, C. J., Reith, M. E. A., Wang, D.-N. LeuT-desipramine structure reveals how antidepressants block neurotransmitter reuptake. Science 317: 1390-1393, 2007. [PubMed: 17690258] [Full Text: https://doi.org/10.1126/science.1147614]


Contributors:
Patricia A. Hartz - updated : 1/20/2015
Cassandra L. Kniffin - updated : 1/29/2014
Cassandra L. Kniffin - updated : 2/27/2012
Cassandra L. Kniffin - updated : 11/23/2009
Patricia A. Hartz - updated : 10/24/2008
Ada Hamosh - updated : 10/15/2007
Cassandra L. Kniffin - updated : 9/10/2007
John Logan Black, III - updated : 8/4/2006
John Logan Black, III - updated : 6/2/2006
John Logan Black, III - updated : 5/17/2006
John Logan Black, III - updated : 5/12/2006
Natalie E. Krasikov - updated : 3/29/2004
Victor A. McKusick - updated : 3/11/1999
Victor A. McKusick - updated : 1/14/1999
Victor A. McKusick - updated : 12/18/1998
Victor A. McKusick - updated : 4/29/1998
Victor A. McKusick - updated : 8/13/1997

Creation Date:
Victor A. McKusick : 1/8/1993

Edit History:
carol : 01/08/2020
carol : 07/09/2018
ckniffin : 07/05/2018
carol : 06/05/2017
carol : 06/02/2017
carol : 02/25/2015
mgross : 1/26/2015
mcolton : 1/20/2015
mcolton : 2/21/2014
mgross : 2/10/2014
ckniffin : 1/29/2014
carol : 3/5/2012
terry : 3/2/2012
ckniffin : 2/27/2012
terry : 12/16/2009
wwang : 12/11/2009
wwang : 12/11/2009
ckniffin : 11/23/2009
carol : 9/4/2009
wwang : 4/16/2009
mgross : 10/24/2008
mgross : 10/24/2008
carol : 7/25/2008
alopez : 10/26/2007
terry : 10/15/2007
wwang : 9/12/2007
ckniffin : 9/10/2007
carol : 6/15/2007
alopez : 10/25/2006
carol : 8/29/2006
terry : 8/4/2006
wwang : 6/6/2006
terry : 6/2/2006
wwang : 5/22/2006
terry : 5/17/2006
wwang : 5/16/2006
wwang : 5/16/2006
terry : 5/12/2006
ckniffin : 4/18/2006
tkritzer : 3/30/2004
terry : 3/29/2004
carol : 3/8/2002
terry : 3/8/2002
terry : 3/16/1999
terry : 3/11/1999
alopez : 1/15/1999
alopez : 1/14/1999
joanna : 1/14/1999
carol : 12/28/1998
terry : 12/18/1998
carol : 4/30/1998
terry : 4/29/1998
terry : 8/13/1997
mark : 4/1/1996
mark : 2/14/1996
mark : 1/21/1996
terry : 1/17/1996
mark : 12/4/1995
terry : 9/13/1995
mark : 7/28/1995
carol : 2/25/1994
carol : 4/27/1993
carol : 4/26/1993
carol : 2/17/1993