Sie sind auf Seite 1von 8

Neuropharmacology 47 (2004) 140–147

www.elsevier.com/locate/neuropharm

Molecular genetic underpinnings of human substance abuse


vulnerability: likely contributions to understanding addiction
as a mnemonic process
George R. Uhl 
Molecular Neurobiology Branch, NIDA-IRP, NIH, Box 5180 Baltimore, MD 21224, USA

Received 11 May 2004; received in revised form 20 July 2004; accepted 22 July 2004

Abstract

Classical genetic studies document strong complex genetic contributions to abuse of multiple addictive substances. These gen-
etic influences are more prominent in the later phases of individuals’ progressions toward substance dependence. Individual differ-
ences in human addiction vulnerability could thus derive, in part, from individual differences in mnemonic systems. These
variations could add to allelic variations that could produce effects on addiction vulnerability phenotypes by other routes that
could include (1) differences in drug metabolism or biodistribution, (2) differences in drug’s rewarding properties, (3) differences in
traits manifest by the addict, including personality differences and (4) differences in the addict’s psychiatric comorbidities. Data
from linkage and association genome scans now identify chromosomal regions that are likely to contain allelic gene variants that
contribute to human addiction vulnerability. Converging positive results are found in several different substance-abusing popula-
tions studied in several laboratories. This convergence supports the idea that common allelic variants contribute to individual dif-
ferences in vulnerability to substance dependence. Genomic markers that identify allelic variants that reproducibly alter addiction
vulnerability in several populations provide tools for research in addictions, tools to improve addiction treatments, tools to
improve addiction prevention, clues to the genetic bases of individual differences in mnemonic processes and clues to the genetic
bases of individual differences in the other traits and disorders that co-occur with substance dependencies.
Published by Elsevier Ltd.

Keywords: Drug abuse; Complex genetics; Association genome scanning

1. Classical genetic studies that delineate genetic diagnostic and statistical manual (DSM) diagnoses of
and environmental contributions to human addiction drug dependence and quantity/frequency of drug use.
vulnerabilities Studies of the ways in which addictions move
through families fail to support single-gene models for
We know about the dimensions of genetic contribu- inheritance of most addiction vulnerability. Addictions,
tions to drug abuse from family, adoption and twin like other human disorders that display both genetic
study data. Drug abuse vulnerability is familial and environmental contributions to vulnerability, are
(reviewed by Uhl et al., 1995). Twin data converge on a thus ‘‘complex disorders’’ that are likely to receive con-
common theme: drug abuse vulnerability has overall tributions from allelic variations in several genes.
genetic components that explain 40–60% of the overall Environmental features that contribute to addiction
vulnerability (Pickens et al., 1991; Tsuang et al., 1996, vulnerability have been identified in numerous epide-
1998, 1999; Kendler et al., 1997, 1999, 2000; Kendler miological studies. Nevertheless, some individuals
and Prescott, 1998; True et al., 1999a,b; Uhl, 1999; Uhl avoid addiction even in environments rich in addictive
et al., 2002). Phenotypes that are heritable include drugs. It seems likely that few addiction vulnerability
alleles will express themselves equally in all environ-

Tel.: +1-410-550-2843x146; fax: +1-410-550-1535. ments and that few environments will make addicts of
E-mail address: guhl@intra.nida.nih.gov (G.R. Uhl). all of the individuals who have highly protective genetic
0028-3908/$ - see front matter Published by Elsevier Ltd.
doi:10.1016/j.neuropharm.2004.07.029
G.R. Uhl / Neuropharmacology 47 Supplement No. 1 (2004) 140–147 141

backgrounds. Neither genetic nor environmental con- with these striking ADH/ALDH variants are pro-
tributions to addiction vulnerability can thus be tected against alcoholism in the Asian populations in
ignored. which these variants are common (Yamashita et al.,
Twin data support the idea that much of the genetic 1990). Interestingly, more subtle allelic variants in
vulnerability to abuse of different legal and illegal these same genes may produce the convergent
addictive substances is shared (Tsuang et al., 1998; association and linkage findings identified at the
Kendler et al., 1999). Although parts of addiction vul- chromosome four ADH/ALDH locus in European-
nerability are likely to be specific for specific sub- Americans and African Americans (see below).
stances, most genetic influences are common to Substance abusers have diagnoses of depression, anti-
different addictive substances. Genetically identical social personality disorder/conduct disorder and adult
cotwins of smokers are more likely to use cocaine than residua of attention deficit/hyperactivity disorder
fraternal cotwins of smokers who share only half of more frequently than control populations (Regier
their vulnerability alleles, for example. These studies’ et al., 1990; Rowland et al., 2002). Each of these
results buttress common clinical observations that most diagnoses is likely to display heritable components.
substance abusers abuse multiple addictive substances. Each also appears to be comorbid with substance
abuse more than with the other comorbid diagnoses.
Genetic contributions to addiction in individuals with
2. Working ideas about genetic architecture and antisocial personality disorder may thus differ from
commonly co-occurring diagnoses or traits those found in addicts with depression. Groups of
individuals with apparently similar addictions but
We are rapidly approaching a major goal of drug with different psychiatric comorbidities could provide
abuse molecular genetics: discovering which chromo- plausible samples for studies seeking evidence of mol-
somal regions, which genes, which haplotypes and ecular genetic heterogeneity of substance abuse.
which allelic variants provide the genetic influences on Personality types provide another perspective. There
drug abuse vulnerability that are documented in classi- are substantial heritabilities for several of the principal
cal genetic studies. Many of the initial results of mol- currently accepted personality domains (Costa and
ecular genetic studies have buttressed our initial Widiger, 1998; Cloninger et al., 1993). Individuals who
assumptions about the ‘‘genetic architecture’’ of these score at the extremes of several tests of heritable per-
disorders. sonality types display enhanced risk of addiction. High
One group of our assumptions about the genetic ‘‘neuroticism’’ scores are found in substance abusers
architecture of substance abuse comes from the high (Costa and Widiger, 1998; Carter et al., 2001), while
frequency of addictions in the population. The ‘‘com- this trait shows substantial heritability in twin studies
mon disease/common allele’’ hypothesis states that (Jang et al., 1996; Costa and Widiger, 1998). Some of
many of the same allelic variants could contribute to the same allelic variants that predispose to high neur-
vulnerability to drug abuse in many addicted indivi- oticism could thus also predispose to substance abuse.
duals in the population (Reich and Lander, 2001). Twin studies have addressed the impact of genetics
Since twin study observations suggest that a large on aspects of individuals’ progression from experimen-
amount of the genetic vulnerability to abuse of addic- tation with drugs to use to regular use to DSM diag-
tive substances of several chemical classes is shared noses of dependence. Few genetic influences impact
(Tsuang et al., 1998; Kendler et al., 1999), abusers of initiation of drug use (Tsuang et al., 1999; Kendler
multiple substances that are commonly found in clini- et al., 1999). Only a few influence progression from
cal and research volunteer populations appear to pro- initial use to regular use. Most genetic, heritable deter-
vide reasonable samples for molecular genetic studies. minants appear to influence progression from regular
However, there is also evidence for heterogeneity of the use to dependence. Some evidence does support genetic
genetics of substance vulnerability. Some heterogeneity mediation of individual differences in acute drug
of this genetic vulnerability fits with data from studies responses. Individuals who have elevated genetic risk
of abusers of different ethnic backgrounds, those with for developing alcoholism can display different respon-
different patterns of co-occurring psychiatric disease, ses to acute ethanol administration, for example,
those with different personality features, those with dif- Schuckit et al. (2000). Nevertheless, the major genetic
ferent cognitive features and those with different sub- influences on human addiction vulnerability are found
stances of choice. in the later stages of the progression toward drug
The allelic variations that have been most strongly dependence.
associated with differential vulnerability to addiction Many of the phenomena that accompany later stages
are the striking variants in the alcohol and acetalde- of progression toward drug dependence differ from
hyde dehydrogenase (ADH and ALDH) genes that those found at earlier stages of increasing drug use.
produce flushing when alcohol is consumed. Individuals Many of these differences appear likely to involve mnemonic
142 G.R. Uhl / Neuropharmacology 47 Supplement No. 1 (2004) 140–147

processes. Thus, increasing fractions of the drug use molecular genetic findings. Initial findings describing
manifest by more and more dependent individuals linkage-based genome scans for legal addictions were
appear to be driven to greater and greater extents by reported by Long et al. (1998) (172 sibpairs, 517 mar-
habitual, implicit mnemonic systems. Habit-driven epi- kers), Reich et al. (1998) (225 sibpairs, 291 markers),
sodes of drug administration may not be reliably Foroud et al. (2000) (266 additional sibpairs, 351
accompanied by drug craving or other manifestations markers), and Straub et al. (1999) (391 sibpairs; 451
of explicit drug memories. Nevertheless, habitual drug markers), using affected sibpairs from US and New
use takes more and more dominant roles in the beha- Zealand populations of several discrete ethnicities. We
vioral hierarchies of many of the individuals who are reported the first association-based scan for illegal
dependent. Habitual drug memory systems are thus addictions in two different samples of unrelated abusers
strong candidates for genetically mediated individual and controls (Uhl et al., 2001, 2002) and have com-
differences in substance abuse behaviors. Even in drug pleted a higher-density association genome scan of lar-
dependent individuals, however, some episodes of drug ger unrelated abuser and control samples (Q-R Liu,
use are likely to be triggered by other pathways and GRU et al., in preparation). The results of these stu-
thus to involve other mnemonic systems which can also dies can be compared to linkage studies of poly-
provide alternative sites for the influences of other gen- substance use in affected sibpairs in which the proband
etic variations. had conduct disorder (Stallings et al., 2003), smoking
Individual differences in performance on memory in pedigrees in which the proband was identified by
tests are substantially heritable. Heritability for work- anxiety or panic disorder phenotypes (Gelernter, 2002),
ing memory is reported to range from 33–64% (Wright and association results from COGA pedigrees (Beglei-
et al., 2001). The volumes of brain structures important ter, 2002). Each of these studies examined phenotypes
for mnemonic processes are also substantially heritable. whose heritability is supported by twin data: (a) depen-
Comparisons of MRI brain images of mono- and dizy- dence diagnoses made according to diagnostic and
gotic twin pairs suggest that ca. 60% of the individual statistical manual (DSM) criteria, and (b) quantity-
differences in the volume of frontal lobe regions are frequency estimates. Linkage-based studies examined
heritable, while hippocampal volumes display ca. 0.4 DNA from members of small pedigrees, studying the
heritabilities (Geschwind et al., 2002; Sullivan et al., ways in which allelic DNA markers and disease move
2001). Taken together, these lines of evidence support through pedigrees. Association approaches examined
substantial heritable contributions to the structures of the ways in which allelic DNA markers and disease
brain regions important for mnemonic tasks, perform- move through unrelated members of the population.
ance on memory tasks, and on the later aspects of We have approached these datasets by seeking evi-
addictions that are likely to be memory related. Sub- dence for converging findings. We have sought simi-
stantial inter-relationships between these three heritable larity of chromosomal locations of genomic markers
components appear likely. that have been linked to or associated with addiction
Substance abusers differ from nonabusers in tests of vulnerability in association and linkage studies. In our
several cognitive functions that can also be associated initial association genome scans using 1494 SNPs, we
with brain regions whose sizes are heritable (Fein et al., identified 41 markers with allelic variants that were
2002a, b). Substance abusers score lower on tests of associated with substance abuse vulnerability in both
‘‘executive function’’ (Sim et al., 2002; Simon et al., Caucasian and African American research volunteer
2002). Twin data suggest that executive function is samples (Uhl et al., 2001). When we examined the
heritable (Swan and Carmelli, 2002). Comparisons of chromosomal distribution of the 104 markers that
MRI brain images of twin pairs support heritability of included these 41 SNPs and the chromosomal markers
the volumes of prefrontal and frontal cortical regions linked to substance abuse vulnerability in at least one
(Carmelli et al., 2002). Substance abusers that differ other study, we found that 34 of these nominally posi-
from each other based on executive or other cognitive tive markers clustered into 15 relatively small chromo-
functions provide yet another way in which genetically somal regions (Uhl et al., 2001). Modeling studies
heterogeneous addict subgroups might be defined. suggested that this clustering was unlikely to all be due
to chance alone (Uhl et al., 2002). Additional evidence
for convergence comes from data from Stallings and
3. Molecular genetic genome scans and remarkable coworkers (Stallings et al., 2003), Gelernter (2002),
convergence of results association studies in a region of interest reported by
COGA investigators (Begleiter, 2002) and improving
We now have sufficient data from genome scans data concerning the chromosomal locations of nomin-
using linkage and association approaches to allow us ally positive markers. Table 1 lists the chromosomal
to test the fit between hypotheses concerning the gen- regions in which current genomic coordinates place at
etic architecture of addictions and currently available least two nearby (within 5 Mb) markers that are linked
G.R. Uhl / Neuropharmacology 47 Supplement No. 1 (2004) 140–147 143

Table 1
Candidate ‘‘rSA’’ replicated chromosomal areas likely to harbor substance abuse vulnerability alleles

SA# Ch MV34 WIAF/SSLP Reference Samples Substance


rSA16 2 233.6-234.15 na Gelertner et al. Four Tobacco
2 234667480 WIAF-1700 Uhl et al. Polysubstance
2 237522162 D2S338 Stallings et al. Polysubstance

rSA1 3 27450316 WIAF-1765 Uhl et al. Three Polysubstance


3 32137007 D3S2432 Long et al. Alcohol

rSA17 3 152346291 D3S1279 Stallings Two Polysubstance


3 153050650 D3S1746 Long et al. Alcohol

rSA2 3 168560613 D3S1763 Long et al. Four Alcohol


3 169531042 D3S1614 Stallings et al. Polysubstance
3 172361071 WIAF-847 Uhl et al. Polysubstance
3 172808611 D3S3053 Long et al. Alcohol

rSA3 4 35601324 D4S2632 Long et al. Four Alcohol


4 39947943 D4S2382 Long et al. Alcohol
4 40749467 D4S174 Long et al. Alcohol
4 44094355 D4S1627 Long et al. Alcohol
4 44622461 WIAF-452 Uhl et al. Polysubstance
4 46221275 rs279871 Beigleiter et al. Alcohol
4 46843212 D4S3242 Long et al. Alcohol

rSA4 4 61986356 D4S1645 Long et al. Two Alcohol


4 65494871 D4S244 Reich et al. Alcohol

rSA5 4 na D4S2456 Long et al. Two Alcohol


4 77952893 D4S2393 Reich et al. Alcohol

rSA6 4 95596000 WIAF-373 Uhl et al. (2001) Four Polysubstance


4 99893367 D4S1647 99.7 Long et al. Alcohol
4 101541576 WIAF-2765 Uhl et al. (2001) Polysubstance
4 103210000 ADH3 Reich et al. Alcohol

rSA18 4 107037800 D4S2457 Reich et al. Two Alcohol


4 108344788 D4S3256 Long et al. Alcohol
4 110253428 D4S3240 Long et al. Alcohol

SA7 4 125278307 WIAF-3821 Uhl et al. Two Polysubstance


4 125278333 WIAF-3818 Uhl et al. Polysubstance

rSA19 6 11210857 WIAF-1567 Uhl et al. Two Polysubstance


11267058 WIAF-967 Uhl et al. Polysubstance

rSA8 9 27316060 WIAF-1900 Uhl et al. Three Polysubstance


9 29549719 D9S319 Long et al. Alcohol

rSA9 10 102860940 D10S1239 Straub et al. Three Tobacco


10 106310791 D10S2469 Straub et al. Tobacco
10 106317898 WIAF-268 Uhl et al. Polysubstance

rSA10 11 1533898 D11S1984 Long et al. Four Alcohol


11 1533898 D11S1984 Uhl et al., un Polysubstance
11 1927951 D11S4046 Gelertner, un Tobacco

rSA19 11 17452815 D11S902 Stallings Polysubstance


11 19245375 D11S2368 Long et al. Alcohol

SA11 11 26648926 WIAF-2046 Uhl et al. Two Polysubstance


11 29083355 WIAF-1949 Uhl et al. Polysubstance

(continued on next page)


144 G.R. Uhl / Neuropharmacology 47 Supplement No. 1 (2004) 140–147

Table 1 (continued )

SA# Ch MV34 WIAF/SSLP Reference Samples Substance


SA12 12 122337633 WIAF-3624 Uhl et al. Two Polysubstance
12 122599343 WIAF-364 Uhl et al. Polysubstance

rSA13 13 90538821 WIAF-4520 Uhl et al. Three Polysubstance


13 nr 91,900,000 D13S762 Reich et al. Alcohol

rSA14 13 109100000 D13S895 Long et al. Three Alcohol


13 110743433 D13S285 Long et al. Alcohol
13 111559844 WIAF-2555 Uhl et al. Polysubstance

rSA20 17 69098093 D17S2059 Straub et al. Two Tobacco


17 73158564 D17S1535 Long et al. Alcohol

rSA15 X 150639232 DXS8061 Straub et al. Three Tobacco


X 151048287 WIAF-1743 Uhl et al. Polysubstance
SA# is the designation for each chromosomal area, as indicated in the text.
Chr is the chromosomal location.
WIAF/SSLP is the single nucleotide polymorphism (SNP) or simple sequence length polymorphism (SSLP).
MV34 chromosomal positions in bp for the markers based on coordinates calculated or inferred from the NCBI ‘‘Mapviewer’’program, version 34.3.
Reference is the reference for the study whose nominally positive results are displayed.
Samples refer to the number of independent samples for which positive data is displayed.
Substance indicates the primary substance reported to be abused by probands, although many subjects in all samples are polysubstance abusers.

or associated with human addiction vulnerability from It is also likely that there are many false-negative
at least two different populations. Twenty chromo- regions in current data No single study performed to
somal regions that now provide the ‘‘reproducible sub- date has power sufficient to detect all of the allelic var-
stance abuse vulnerability’’ regions (rSA1-20) that are iants that contribute to addiction, especially if there is
listed here. Identifying these regions is an ongoing task. a polygenic underlying genetic architecture and signifi-
It is nevertheless gratifying that more recent studies cant genetic heterogeneity. The rSA regions in Table 1
have provided confirmation of several previously nomi- are thus likely to represent an incomplete list of only
nated loci. It is also gratifying that several previously those allelic variants that provide enough influence on
identified nominally positive markers are now posi- abuse of so many different substances in so many
tioned closer to each other as the precision of the geno- populations that they can be jointly detected in each of
mic backbone on which they have been localized has several studies that have used different methodologies
improved. and/or different clinical samples.
Initial results from 11,500 SNP arrays appear to but- Data from candidate gene association studies also
tress and extend these results. SNPs that lie in several of provide evidence for likely false-negative results from
the previously defined rSA regions again display abuser- genome scanning studies. Our work and work from a
control differences anticipated if allelic variants number of other laboratories have identified allelic var-
that alter substance abuse vulnerability lie on blocks of iants of the catechol-O-methyl transferase (COMT)
restricted haplotype diversity that are marked by the gene that are present in different frequencies in some,
SNPs that we have employed in both initial and follow- though not all, abuser vs. control populations studied
up studies (Q.-R. Liu, GRU et al., in preparation). (Smith et al., 1992; Persico et al., 1996; Vandenbergh
et al., 1997, 2000). The COMT gene is located between
18.30 and 18.33 Mb on chromosome 22. However,
4. Caveats and limitations: there are no nominally positive association signals at
flanking chromosome SNP markers located at 17,356,651,
The low likelihood that all of the convergence 17,506,879, 19,773,582, 19,773,965, 20,306,803, 20,379,948
observed to date is based on chance does not exclude or 20,924,091 bp. Conceivably, the ca. 2 Mb gap in
the likelihood of some false-positive results (Romero the markers employed resulted in this apparent false-
et al., 2002; Glazier et al., 2002). Simulation studies negative result, although it does not explain the fail-
indicated that the chance that at least one, two, three ure of linkage-based genome scanning studies. Higher
or four of the 15 initially defined rSA regions were marker densities will help to improve the power of
identified by chance were 0.78, 0.42, 0.17, 0.06, and association-based genome scanning, and reduce false-
0.01, respectively (Uhl et al., 2002). negative results of this sort.
G.R. Uhl / Neuropharmacology 47 Supplement No. 1 (2004) 140–147 145

5. Example: candidate addiction vulnerability and substances, each allelic variant could provide a distinc-
memory-associated locus identified by positional tive pattern of influences. Each could manifest effects
cloning and as a drug-regulated gene on different aspects of addiction vulnerability and/or
on vulnerability to the other traits and behavioral dis-
As we identify the specific genetic haplotypes orders that often co-occur with substance disorders.
that confer human individual differences in addiction Alleles altering the metabolism of a drug, for example,
vulnerability and develop animal models of these varia- might produce a straightforward shift in the dose-effect
tions, we can also seek variations in other addiction- relationships for that drug without substantially alter-
associated phenotypes that might be conferred by some ing brain chemistries or vulnerabilities to comorbid
of these same allelic variants. We can seek effects of conditions. Additional studies will be able to better
modeling these allelic variations in animals which can identify the ways in which each allelic variant changes
provide better experimental control over both environ- specific features of addiction vulnerability. Such studies
mental and genetic background variables. can also help us better understand the ways in which
An example of this sort of progression comes from
genetic and environmental influences could interact and
our studies of the human gene locus for NrCAM. We
add to our understanding of addictions.
identified NrCAM and specific NrCAM 50 haplotypes
through positional cloning (Ishiguro, Uhl et al. in prep-
aration). We focused on this gene since we had pre-
viously discovered that its expression levels are 7. Potential impact
regulated by morphine administration. We also ident- We have recently described the large size of the com-
ified NrCAM expression in both dopaminergic neurons plex genetic contributions to addictions in US society
that modulate reward and memory-related brain areas (Uhl and Grow, 2004). This sizable impact continues to
including striatum and hippocampus.
motivate us to identify allelic variants that contribute
We have been able to test conditioned place pre-
to addiction vulnerability, even though we are increas-
ferences in NrCAM knockout mice that were originally
ingly certain that no single allelic variant is likely to
developed by Grumet, Sakarai and colleagues (Grumet,
contribute any large fraction of this vulnerability in
1997). Mouse conditioned place preference for drugs is
individuals from several different populations. Know-
almost invariably a good predictor of human abuse
ing which addiction-vulnerability allelic variants are
liability. Interestingly, NrCAM knockout mice failed to
present in vulnerable individuals should improve tar-
develop significant preferences for the place where they
geting of large addiction treatment and prevention
received morphine injections (Ishiguro, Uhl et al. in
expenditures. Individual and societal suffering could be
preparation). Also intriguingly, other studies of
substantially relieved by better understanding the com-
NrCAM knockouts have identified selective deficits in
plex human processes of human addiction through
several, but not all, memory tasks. Taken together,
careful application of complex human genetic approa-
these data suggest that we have identified NrCAM
ches.
haplotypes that are involved in human addiction vul-
nerability as well as effects of NrCAM deletion in both
mouse models of human addiction vulnerability and
mouse models of memory. Acknowledgments

I am grateful for the help with personal studies cited


here from Qing-Rong Liu, Daniel Naiman, Judith
6. Working hypotheses concerning the genetic Hess, Bruce O’Hara, Antonio Persico, Donna Walther,
architecture of substance abuse based on current, Fely Carillo, Brenda Campb Linda Kahler, Fred
convergent molecular genetic data Snyder, Carlo Contoreggi, Larry Rodriguez, Robert
Assembling the markers now linked to or associated Grow, David Gorelick, Zhicheng Lin, Andrew Shapiro
with vulnerability to substance abuse in at least one and Leslie Cope and financial support from NIDA-
published study now provides a brighter and sharper IRP.
picture of the molecular genetics of substance abuse.
The data support polygenic inheritance for substance
abuse vulnerability. They also support the idea that References
common allelic variants that are likely to contribute to
vulnerability to abuse of several substances exist in Begleiter, H., 2002. Neurophysiological endophenotypes in the search
for genes for alcoholism. ACNP.
humans from several racial/ethnic groups. Carmelli, D., Swan, G.E., et al., 2002. Quantitative genetic modeling
Although common allelic variants could thus play of regional brain volumes and cognitive performance in older
significant roles in vulnerabilities to abuse of multiple male twins. Biol. Psychol. 61 (1-2), 139–155.
146 G.R. Uhl / Neuropharmacology 47 Supplement No. 1 (2004) 140–147

Carter, J.A., Herbst, J.H., et al., 2001. Short-term stability of Romero, R., Kuivaniemi, H., et al., 2002. The design, execution, and
NEO-PI-R personality trait scores in opioid-dependent out- interpretation of genetic association studies to decipher complex
patients. Psychol. Addict. Behav. 15 (3), 255–260. diseases. Am. J. Obstet. Gynecol. 187 (5), 1299–1312.
Cloninger, C.R., Svrakic, D.M., Przybeck, T.R., 1993. A psychobio- Rowland, A.S., Lesesne, C.A., et al., 2002. The epidemiology of
logical model of temperament and character. Arch. Gen. attention-deficit/hyperactivity disorder (ADHD): a public health
Psychiatry 50 (12), 975–990. view. Ment. Retard Dev. Disabil. Res. Rev. 8 (3), 162–170.
Costa, P.T., Widiger, T.A., 1998. Personality disorders and the five- Schuckit, M.A., Smith, T.L., Kalmijn, J., Tsuang, J., Hesselbrock,
factor model of personality. American Psychological Association, V., Bucholz, K., 2000. Response to alcohol in daughters of alco-
Washington, DC. holics: a pilot study and a comparison with sons of alcoholics.
Fein, G., Di Sclafani, V., et al., 2002a. Cortical gray matter loss in Alcohol Alcohol 35 (3), 242–248.
treatment—naive alcohol dependent individuals. Alcohol Clin. Sim, T., Simon, S.L., et al., 2002. Cognitive deficits among metham-
Exp. Res. 26 (4), 558–564. phetamine users with attention deficit hyperactivity disorder
Fein, G., Di Sclafani, V., et al., 2002b. Prefrontal cortical volume symptomatology. J. Addict. Dis. 21 (1), 75–89.
reduction associated with frontal cortex function deficit in 6-week Simon, S.L., Domier, C.P., et al., 2002. Cognitive performance of
abstinent crack-cocaine dependent men. Drug Alcohol Depend. current methamphetamine and cocaine abusers. J. Addict. Dis. 21
68 (1), 87–93. (1), 61–74.
Foroud, T., Edenberg, H.J., et al., 2000. Alcoholism susceptibility Smith, S.S., O’Hara, B.F., et al., 1992. Genetic vulnerability to drug
loci: confirmation studies in a replicate sample and further map- abuse. The D2 dopamine receptor Taq I B1 restriction fragment
ping. Alcohol Clin. Exp. Res. 24 (7), 933–945. length polymorphism appears more frequently in polysubstance
Gelernter, 2002. A genome scan for nicotine addiction in panic dis- abusers. Arch. Gen. Psychiatry 49 (9), 723–727.
order clinic atendees. American Society for Human Genetics Stallings, M.C., Corley, R.P., et al., 2003. A genome-wide search for
satellite. QTLs influencing substance dependence vulnerabity in ado-
Geschwind, D.H., Miller, B.L., DeCarli, C., Carmelli, D., 2002. Heri- lescence. Drug Alcohol Depend 70, 295–307.
tability of lobar brain volumes in twins supports genetic models Straub, R.E., Sullivan, P.F., et al., 1999. Susceptibility genes for
of cerebral laterality and handedness. Proc. Natl. Acad. Sci. USA nicotine dependence: a genome scan and followup in an inde-
99, 3176–3181. pendent sample suggest that regions on chromosomes 2, 4, 10, 16,
Glazier, A.M., Nadeau, J.H., et al., 2002. Finding genes that underlie 17 and 18 merit further study. Mol. Psychiatry 4 (2), 129–144.
complex traits. Science 298 (5602), 2345–2349. Sullivan, E.V., Pfefferbaum, A., Swan, G.E., Carmelli, D., 2001.
Grumet, M., 1997. Nr-CAM: a cell adhesion molecule with ligand Heritability of hippocampal size in elderly twin men: equivalent
and receptor functions. Cell Tissue Res. 290 (2), 423–428, influence from genes and environment. Hippocampus 11 (6),
(November). 754–762.
Jang, K.L., Livesley, W.J., et al., 1996. Heritability of the big five Swan, G.E., Carmelli, D., 2002. Evidence for genetic mediation
personality dimensions and their facets: a twin study. J. Pers. 64 of executive control: a study of aging male twins. J. Gerontol. B
(3), 577–591. Psychol. Sci. Soc. Sci. 57 (2), 133–143.
Kendler, K.S., Prescott, C.A., 1998. Cocaine use, abuse and depen- True, W.R., Heath, A.C., et al., 1999a. Interrelationship of genetic
dence in a population-based sample of female twins. Br. J. and environmental influences on conduct disorder and alcohol
Psychiatry 173, 345–350. and marijuana dependence symptoms. Am. J. Med. Genet. 88 (4),
Kendler, K.S., Prescott, C.A., et al., 1997. Temperance board regis- 391–397.
tration for alcohol abuse in a national sample of Swedish male True, W.R., Xian, H., et al., 1999b. Common genetic vulnerability
twins, born 1902 to 1949. Arch. Gen. Psychiatry 54 (2), 178–184. for nicotine and alcohol dependence in men. Arch. Gen. Psy-
Kendler, K.S., Karkowski, L.M., et al., 1999. Genetic and environ- chiatry 56 (7), 655–661.
mental risk factors in the aetiology of illicit drug initiation and Tsuang, M.T., Lyons, M.J., et al., 1996. Genetic influences on DSM-
subsequent misuse in women. Br. J. Psychiatry 175, 351–356. III-R drug abuse and dependence: a study of 3,372 twin pairs.
Kendler, K.S., Thornton, L.M., et al., 2000. Tobacco consumption Am. J. Med. Genet. 67 (5), 473–477.
in Swedish twins reared apart and reared together. Arch. Gen. Tsuang, M.T., Lyons, M.J., et al., 1998. Co-occurrence of abuse of
Psychiatry 57 (9), 886–892. different drugs in men: the role of drug-specific and shared vulner-
Long, J.C., Knowler, W.C., et al., 1998. Evidence for genetic linkage abilities. Arch. Gen. Psychiatry 55 (11), 967–972.
to alcohol dependence on chromosomes 4 and 11 from an auto- Tsuang, M.T., Lyons, M.J., et al., 1999. Genetic and environmental
some-wide scan in an American Indian population. Am. J. Med. influences on transitions in drug use. Behav. Genet. 29 (6), 473–
Genet. 81 (3), 216–221. 479.
Persico, A.M., Bird, G., et al., 1996. D2 dopamine receptor gene Uhl, G.R., 1999. Molecular genetics of substance abuse vulnerability:
TaqI A1 and B1 restriction fragment length polymorphisms: a current approach. Neuropsychopharmacology 20 (1), 3–9.
enhanced frequencies in psychostimulant-preferring polysubstance Uhl, G.R., Grow, R.W., 2004. The burden of complex genetics in
abusers. Biol. Psychiatry 40 (8), 776–784. brain disorders. Arch. Gen. Psychiatry 61 (3), 223–229.
Pickens, R.W., Svikis, D.S., et al., 1991. Heterogeneity in the inherit- Uhl, G.R., Elmer, G.I., et al., 1995. Genetic influences in drug abuse.
ance of alcoholism. A study of male and female twins. Arch. Gen. In: Bloom, F.E., Kupfer, D.J. (Eds.), Psychopharmacology: the
Psychiatry 48 (1), 19–28. Fourth Generation. Raven Press, New York, pp. 1793–1806.
Reich, D.E., Lander, E.S., 2001. On the allelic spectrum of human Uhl, G.R., Liu, Q.R., et al., 2001. Polysubstance abuse-vulnerability
disease. Trends Genet. 17 (9), 502–510. genes: genome scans for association, using 1,004 subjects and
Regier, D.A., Farmer, M.E., et al., 1990. Comorbidity of mental 1,494 single-nucleotide polymorphisms. Am. J. Hum. Genet.
disorders with alcohol and other drug abuse. Results from the 69 (6), 1290–1300.
Epidemiologic Catchment Area (ECA) Study. JAMA 264 (19), Uhl, G.R., Liu, Q.R., et al., 2002. Substance abuse vulnerability loci:
2511–2518. converging genome scanning data. Trends Genet. 18 (8), 420–425.
Reich, T., Edenberg, H.J., et al., 1998. Genome-wide search for genes Vandenbergh, D.J., Rodriguez, L.A., et al., 1997. High-activity
affecting the risk for alcohol dependence. Am. J. Med. Genet. 81 catechol-O-methyltransferase allele is more prevalent in poly-
(3), 207–215. substance abusers. Am. J. Med. Genet. 74 (4), 439–442.
G.R. Uhl / Neuropharmacology 47 Supplement No. 1 (2004) 140–147 147

Vandenbergh, D.J., Rodriguez, L.A., et al., 2000. Long forms of the D., Geffen, G., Geffen, L., Kanba, S., Miyake, A., Martin, N.,
dopamine receptor (DRD4) gene VNTR are more prevalent in Boomsma, D., 2001. Genetics of cognition: outline of a collabora-
substance abusers: no interaction with functional alleles of the tive twin study. Twin Res. 4 (1), 48–56, (February).
catechol-o-methyltransferase (COMT) gene. Am. J. Med. Genet. Yamashita, I., Ohmori, T., et al., 1990. Biological study of alcohol
96 (5), 678–683. dependence syndrome with reference to ethnic difference: report
Wright, M., Degeus, E., Ando, J., Luciano, M., Posthuma, D., Ono, of a WHO Collaborative Study. Jpn. J. Psychiatry Neurol. 44 (1),
Y., Hansell, N., Van Baal, C., Hiraishi, K., Hasegawa, T., Smith, 79–84.

Das könnte Ihnen auch gefallen