5-ht1a gene variants and psychiatric disorders: a review of current

21
5-HT 1A gene variants and psychiatric disorders : a review of current literature and selection of SNPs for future studies Antonio Drago, Diana De Ronchi and Alessandro Serretti Institute of Psychiatry, University of Bologna, Italy Abstract 5-HT 1A receptors are key components of the serotonin system, acting both pre- and post- synaptically in different brain areas. There is a growing amount of evidence showing the importance of 5-HT 1A in different psychiatric disorders, from mood to anxiety disorders, moving through suicidal behaviour and psychotic disorders. Findings in the literature are not consistent with any definite 5-HT 1A influence in psychiatric disorders. 5-HT 1A gene variants have been reported to play some role in mood disorders, anxiety disorders and psychotic disorders. Again, the literature findings are not unequivocal. Concerning response to treatment, the C(-1019)G variant seems to be of primary interest in antidepressant response: C allele carriers generally show a better response to treatment, especially in Caucasian samples. Together with the C(-1019)G (rs6295) variant, the Ile28Val (rs1799921), Arg219Leu (rs1800044) and Gly22Ser (rs1799920) variants have been investigated in possible associations with psychiatric disorders, also with no definitive results. This lack of consistency can be also due to an incomplete gene investigation. To make progress on this point, a list of validated single nucleotide polymorphisms (SNPs) covering the whole gene is proposed for further investigations. Received 2 July 2007; Reviewed 20 September 2007; Revised 27 September 2007; Accepted 7 October 2007; First published online 30 November 2007 Key words : Depression, mood disorders, psychiatric genetics, serotonin, serotonin receptor 1A. Introduction Why this receptor is important : evidence Serotonin (5-hydroxytryptamine or 5-HT) is an in- trinsically fluorescent biogenic amine, which acts as a neurotransmitter by binding to a class of trans- membrane receptors termed serotonin receptors (Chattopadhyay et al., 1996). These receptors rep- resent one of the largest, evolutionarily ancient, and highly conserved families of G-protein-coupled re- ceptors (GPCR) (Peroutka and Howell, 1994). Through specific receptors, 5-HT mediates a large variety of cognitive and behavioural functions including sleep, mood, pain, addiction, locomotion, sexual activity, depression, anxiety, alcohol abuse, aggression, and learning (Artigas et al., 1996; Ramboz et al., 1998); since all those functions are receptor-mediated, gen- etic variations of the receptor coding sequences are of primary interest for psychiatric research. There is a number of known mammalian 5-HT re- ceptor genes (5-HT 1A/B/D/E/F , 5-HT 2A/B/C , 5-HT 3A/B and 5-HT 3C/D/E , 5-HT4, 5-HT 5A/B , 5-HT 6 , 5-HT 7 ) some of which encode additional receptor variants (Albert and Lemonde, 2004) ; within this group, 5-HT 1A is one of the most investigated ; more than 1750 research papers up to January 2007 are dedicated to this pro- tein. 5-HT 1A is a G protein-associated receptor, pre- ferentially coupled to Gi/o proteins to inhibit adenylyl cyclase. Many other second-messenger cascades have been associated with 5-HTR1A activation : MAPK pathways (which may be excitatory in post-synaptic cells or inhibitory in presynaptic neurons), c-Jun N-terminal kinase, GH/IGF-1 receptors and NMDA receptor channels (Adayev et al., 2003 ; Appert-Collin et al., 2005; Chen et al., 2002; Cowen et al., 1996, 2005; Millan et al., 2001; Stark et al., 2007; Sullivan et al., 2005; Turner et al., 2007; Yuen et al., 2005). Influence on inwardly rectified potassium channels (Andrade and Nicoll, 1987; Grunnet et al., 2004) and endo- crine hormones secretion (Serres et al., 2000) have also been reported. Interestingly, membrane proved to play a relevant functional role : Evans and colleagues Address for correspondence : A. Serretti, M.D., Institute of Psychiatry, University of Bologna, Viale Carlo Pepoli 5, 40123 Bologna, Italy. Tel. : +39 051 6584233 Fax : +39 051 521030 E-mail : [email protected] International Journal of Neuropsychopharmacology (2008), 11, 701–721. Copyright f 2007 CINP doi:10.1017/S1461145707008218 REVIEW ARTICLE CINP Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740 by guest on 01 February 2018

Upload: buithuan

Post on 30-Dec-2016

217 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: 5-HT1A gene variants and psychiatric disorders: a review of current

5-HT1A gene variants and psychiatric disorders :a review of current literature and selectionof SNPs for future studies

Antonio Drago, Diana De Ronchi and Alessandro Serretti

Institute of Psychiatry, University of Bologna, Italy

Abstract

5-HT1A receptors are key components of the serotonin system, acting both pre- and post- synaptically

in different brain areas. There is a growing amount of evidence showing the importance of 5-HT1A in

different psychiatric disorders, from mood to anxiety disorders, moving through suicidal behaviour and

psychotic disorders. Findings in the literature are not consistent with any definite 5-HT1A influence in

psychiatric disorders. 5-HT1A gene variants have been reported to play some role in mood disorders,

anxiety disorders and psychotic disorders. Again, the literature findings are not unequivocal. Concerning

response to treatment, the C(-1019)G variant seems to be of primary interest in antidepressant response:

C allele carriers generally show a better response to treatment, especially in Caucasian samples. Together

with the C(-1019)G (rs6295) variant, the Ile28Val (rs1799921), Arg219Leu (rs1800044) and Gly22Ser

(rs1799920) variants have been investigated in possible associations with psychiatric disorders, also with

no definitive results. This lack of consistency can be also due to an incomplete gene investigation. To make

progress on this point, a list of validated single nucleotide polymorphisms (SNPs) covering the whole

gene is proposed for further investigations.

Received 2 July 2007; Reviewed 20 September 2007; Revised 27 September 2007; Accepted 7 October 2007;

First published online 30 November 2007

Key words : Depression, mood disorders, psychiatric genetics, serotonin, serotonin receptor 1A.

Introduction

Why this receptor is important: evidence

Serotonin (5-hydroxytryptamine or 5-HT) is an in-

trinsically fluorescent biogenic amine, which acts

as a neurotransmitter by binding to a class of trans-

membrane receptors termed serotonin receptors

(Chattopadhyay et al., 1996). These receptors rep-

resent one of the largest, evolutionarily ancient, and

highly conserved families of G-protein-coupled re-

ceptors (GPCR) (Peroutka and Howell, 1994). Through

specific receptors, 5-HT mediates a large variety of

cognitive and behavioural functions including sleep,

mood, pain, addiction, locomotion, sexual activity,

depression, anxiety, alcohol abuse, aggression, and

learning (Artigas et al., 1996; Ramboz et al., 1998) ;

since all those functions are receptor-mediated, gen-

etic variations of the receptor coding sequences are of

primary interest for psychiatric research.

There is a number of known mammalian 5-HT re-

ceptor genes (5-HT1A/B/D/E/F, 5-HT2A/B/C, 5-HT3A/B

and 5-HT3C/D/E, 5-HT4, 5-HT5A/B, 5-HT6, 5-HT7) some

of which encode additional receptor variants (Albert

and Lemonde, 2004) ; within this group, 5-HT1A is

one of the most investigated; more than 1750 research

papers up to January 2007 are dedicated to this pro-

tein.

5-HT1A is a G protein-associated receptor, pre-

ferentially coupled to Gi/o proteins to inhibit adenylyl

cyclase. Many other second-messenger cascades have

been associated with 5-HTR1A activation: MAPK

pathways (which may be excitatory in post-synaptic

cells or inhibitory in presynaptic neurons), c-Jun

N-terminal kinase, GH/IGF-1 receptors and NMDA

receptor channels (Adayev et al., 2003; Appert-Collin

et al., 2005; Chen et al., 2002; Cowen et al., 1996, 2005;

Millan et al., 2001; Stark et al., 2007; Sullivan et al.,

2005; Turner et al., 2007; Yuen et al., 2005). Influence

on inwardly rectified potassium channels (Andrade

and Nicoll, 1987; Grunnet et al., 2004) and endo-

crine hormones secretion (Serres et al., 2000) have also

been reported. Interestingly, membrane proved to

play a relevant functional role : Evans and colleagues

Address for correspondence: A. Serretti, M.D., Institute of Psychiatry,

University of Bologna, Viale Carlo Pepoli 5, 40123 Bologna, Italy.

Tel. : +39 051 6584233 Fax : +39 051 521030

E-mail : [email protected]

International Journal of Neuropsychopharmacology (2008), 11, 701–721. Copyright f 2007 CINPdoi:10.1017/S1461145707008218

REVIEW ARTICLE

CINP

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 2: 5-HT1A gene variants and psychiatric disorders: a review of current

showed that the responsiveness of the 5-HT1A receptor

can be enhanced or depressed as a consequence of

activation of phospholipid-coupled receptor systems

(Evans et al., 2001), and Pucadyil and colleagues

showed that 5-HT1A activation is cholesterol-

dependent since there is evidence of 5-HT1A agonists

and antagonists inhibitory effects aftermembrane chol-

esterol oxidation (Pucadyil et al., 2005). This under-

lines the importance of the membrane environment

where receptor activation takes place, and gives fur-

ther details about the intracellular signal cascade,

highlighting the high complexity of cellular mech-

anisms. On this topic see also Kalipatnapu and Chat-

topadhyay (2005), for a review about the membrane

solubilization of 5-HT1A receptors. The aim of this

study is to review the literature evidence about asso-

ciations of 5-HT1A genetic variations and some of the

most important psychiatric disorders, clinical presen-

tation and treatment responses, attempting to high-

light the prerogatives and opportunities of this line of

research. This will follow a short review of receptor

distribution in the brain and a 5-HT1A coding sequence

description.

Where in the neurons?, where in the brain?

Brain 5-HT1A receptors are located both pre- and post-

synaptically. Presynaptic 5-HT1A autoreceptors are

located in the somatodendritic neuronal region, at the

site of the serotonergic cell bodies in the dorsal and

medial raphe nuclei, and they mediate a feedback

inhibition of the 5-HT system, the so called ‘short

feedback loop’, regulating the release of 5-HT by

modulating neurotransmitter synthesis, terminal re-

lease and cell discharge rate (Sprouse and Aghajanian,

1988). In greater detail, the short negative feedback

loop involves GABAergic cells which are stimulated

by serotonergic neurons via HTR2A/C, and in turn

stimulates the 5-HT1A receptors on the surface of the

serotonergic neurons which first stimulated them,

closing the auto-inhibition cycle. That is the reason

why blockade of the 5-HT autoreceptor 1A (and also

1B) can enhance the increase of extracellular 5-HT

levels achieved after selective serotonin reuptake

inhibitor (SSRI) treatment (see Hjorth et al., 2000

for a review). Moreover, the presynaptic 5-HT1A

down-regulation is believed to be associated with

therapeutic effects of serotonergic antidepressant

treatments and related to its time delay before onset

(de Montigny et al., 1984). Further, it is suggested that

multiple 5-HT1A subtypes govern different aspects of

5-HT function in the dorsal raphe nucleus (Stamford

et al., 2000), and the 5-HT system flexibility is also

enriched by action of opiate receptors and neurokinins

(substance P and neurokinin B) stimulating 5-HT

neurons via glutamatergic influences (see Guiard

et al., 2005 and Liu et al., 2002 for interesting papers on

this topic).

Mutual influences between deep serotonergic nuclei

and upper brain systems is the biological basis for

the ‘long feedback loop’ which is mainly mediated by

post-synaptic 5-HT1A receptors and involves cortico-

limbic areas probably influencing the activity of

GABAergic and glutamatergic neurons (Davis et al.,

2002). In fact, post-synaptic 5-HT1A receptors are

found at high density in limbic regions and in the

frontal, medial prefrontal and enthorhinal cortices

(Grunschlag et al., 1997; Pazos et al., 1985; Schmitz

et al., 1998), and the hippocampal regions and medial

prefrontal cortex (mPFC) seem to be of primary in-

terest for 5-HT1A investigation. In fact, the long feed-

back loop has been proposed to be of primary interest

in the antidepressant effect of drug treatments through

the down-regulation of post-synaptic 5-HT1A recep-

tors (Artigas et al., 1996). Focusing on the limbic sys-

tem, there is evidence that CA1 and CA3 hippocampal

pyramidal cells are mainly inhibited by 5-HT1A acti-

vation (Andrade and Nicoll, 1987; Okuhara and Beck,

1994; Segal et al., 1989; Zgombick et al., 1989). Indeed,

a neuronal activation can also be achieved after

5-HT1A stimulation, via 5-HT1A-mediated inhibition of

putative inhibitory interneurons (Schmitz et al., 1995;

Segal, 1990). This is quite interesting in view of the

recent great interest in hippocampal functions as-

sociated with psychiatric disorders, especially for the

possible influence of 5-HT tone on hippocampal

neurogenesis associated with depressive episodes or

disorders. This is consistent with the animal study by

Miquel and colleagues (1994), where 5-HT1A was

found to be highly expressed in the cortex and hippo-

campus of adult rats (for a review see also Elder et al.,

2006). Indeed, there is evidence that long-term anti-

depressant treatment (SSRI, tricyclics, ECT), is as-

sociated with increased 5-HT1A-mediated inhibitory

tone on CA3 hippocampal pyramidal cells (Haddjeri

et al., 1998), this being more evident after short-term

lithium augmentation (Haddjeri et al., 2000). There are

also conflicting findings: Knobelman and colleagues

suggested that 5-HT1A plays a larger role in regulating

5-HT release in the striatum and possibly other brain

regions innervated by the dorsal raphe nucleus (e.g.

cortex) than in the hippocampus, suggesting a major

role for 5-HT1B in that region (Knobelman et al., 2001).

Accordingly, Parsons and colleagues described a more

significant disinhibition of 5-HT release in the frontal

cortex than in the ventral hippocampus in 5-HT1A

702 A. Drago et al.

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 3: 5-HT1A gene variants and psychiatric disorders: a review of current

knockout rats after exposure to SSRIs (Parsons et al.,

2001). Conflicting with this, a recent interesting report

showed 5-HT1A reduced density in the ventral hippo-

campus, which is primarily implicated in emotional

processing in the offspring of stressed female rats

(Van den Hove et al., 2006).

An important role in brain functioning, reasonably

related to psychiatric disorders, is played by the

mPFC: pyramidal neurons in the PFC integrate corti-

cal, thalamic, hypothalamic, limbic inputs and project

to many widespread neuronal structures. In turn, the

mPFC receives a large amount of afferences from the

same neuronal net. On this basis, the PFC regulates a

large number of cognitive and associative functions

and is involved in the planning and execution of

complex tasks, which are commonly impaired in some

of the most important psychiatric disorders such as

schizophrenia or severe depression. There is evidence

that 5-HT1A is highly expressed in the mPFC (Read

et al., 1994) and 5-HT1A is localized at an axon hillock

in the pyramidal neurons providing key negative

regulation of these neurons. Particularly in this area,

highly complex interactions with 5-HT2A and 5-HT1A

occur at the level of individual neurons (Amargos-

Bosch et al., 2004), thus leading to a difficult in-

terpretation of findings. For example, it has recently

been reported that higher 5-HT1A density in the PFC in

rats is associated with increased aggressive behaviour

(Caramaschi et al., 2007), probably through a dimin-

ished 5-HT tone. This conflicts with a previous ima-

ging study in humans which found that a lower

density of 5-HT1A was associated with a history of

aggressive behaviours in psychiatric patients and

healthy controls (Parsey et al., 2002). More research

efforts in this direction are needed.

Finally, 5-HT1A is expressed in other brain struc-

tures such as the cerebellar Purkinje cells (Darrow

et al., 1990), the midbrain periaqueductal grey (PAG, a

region known to be involved in pain modulation and

fear responses), the amygdala (a brain structure highly

associated with anxiety) (Behbehani et al., 1993; Davis

et al., 2002), sensory neurons of dorsal root ganglia

(Scroggs and Anderson, 1990), nucleus prepositus

hypoglossi (Bobker and Williams, 1990, 1995), ven-

tromedial hypothalamus (Newberry, 1992), lateral

septum (Joels et al., 1987; Van den Hooff and Galvan,

1992) and laterodorsal tegmental nucleus (Thakkar

et al., 1998) where 5-HT1A interaction was found to be

associated to modified REM sleep patterns.

With regard to brain layers, 5-HT1A can be found on

both pyramidal and principal cells, throughout the

neocortex (Azmitia et al., 1996; Miquel et al., 1994),

and it may have a role in the control of motor neuronal

excitability. Moreover, diminished serotonergic ac-

tivity might increase the release of neurotoxic levels of

glutamate, or play a direct neurotrophic role (Turner

et al., 2005). Finally, 5-HT1A can also be found

in calbindin- and parvalbumin-containing neurons,

which generally define two different subtypes of in-

terneurons (Aznar et al., 2003).

Those data taken together suggest a complex and

highly integrated 5-HT1A action in the brain: it is con-

ceivable that this receptor influences key regulation

activities in different parts of the brain reasonably

associated with psychiatric disorders. Even if the

physiological fine mechanisms of these actions are still

far from being understood, there is enough genetic

association evidence to encourage an effort towards

an evidence-based individual genetic approach with

diagnostic and therapeutic benefits.

The gene and its variants

The 5-HT1A receptor is encoded by an intronless

gene located on human chromosome 5q11.2–q13 and

it spans about 1200 bp (http://www.ncbi.nlm.nih.

gov/). It was first cloned by Fargin and Albert (Albert

et al., 1990; Fargin et al., 1989), and it encodes a single

receptor isoform, a protein 422 aa long. Fifty HTR1A

single nucleotide polymorphisms (SNPs) are known

so far, and 23 have been validated (http://snpper.

chip.org/bio/snpper-enter).

The most important linkage study results were

found for association between the long arm on chro-

mosome 5 and schizophrenia (Bassett, 1992; Crowe

and Vieland, 1998, 1999; Gorwood et al., 1991; Owen

et al., 1990), even though there are many conflicting

reports of linkage analysis (Chavarria-Siles et al., 2007;

Choudhury et al., 2007; Christoforou et al., 2007;

DeLisi et al., 2002; Jang et al., 2007; Le Hellard et al.,

2007; Liu et al., 2006; Ni et al., 2007; Schosser et al.,

2007), but many other reports can be found in the

literature.

Regarding bipolar disorder, there is some evidence

coming from linkage association studies for an in-

volvement of chromosome 5 (Kerner et al., 2007), even

though it should be considered that the dopamine

transporter gene is within 5p (Homer et al., 1997), and

in the distal 5q there are the genes for the glucocorti-

coid receptor (GRL) and the b2 adrenergic receptor

(ADRB2), also putatively involved in bipolar disorder

(Mirow et al., 1994). Conflicting with this, other link-

age association studies did not mention chromosome 5

(Barden et al., 2006; Faraone et al., 2006; Schumacher

et al., 2005; Underwood et al., 2006). There is also

evidence of linkage study exclusion of association

HTR1A and psychiatric disorders 703

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 4: 5-HT1A gene variants and psychiatric disorders: a review of current

between bipolar disorder and the whole chromosome

5 (Detera-Wadleigh et al., 1992), although using a less

dense marker coverage. No article reporting linkage

association analysis and supporting involvement of

chromosome 5 with unipolar depressive disorders, or

with suicidal behaviour, was found. Generally, even

though SNP association analysis give some interest-

ing results, linkage analysis studies investigating

chromosome 5 in association with psychiatric dis-

orders, showed almost negative results.

The most investigated HTR1A variants are:

C(-1019)G (rs6295); Ile28Val (rs1799921) ; Arg219Leu

(rs1800044) and Gly22Ser (rs1799920). C(-1019)G can

be found in the promoter zone of the gene and this

variant was found to be quite prevalent in the

normal population (Wu and Comings, 1999). Ile28Val

is characterized by a base-pair substitution (ApG) at

the first position of codon 28 in the putative amino-

terminal domain of the receptor (Bruss et al., 1995) ; the

Arg219Leu variant can be found in the third intra-

cellular loop of the 5-HT1A receptor (Bruss et al., 2005)

while the Gly22Ser variant is located in the putative

amino-terminal domain of the receptor (Rotondo

et al., 1997). C(-1019)G (rs6295) has a balanced allele

frequency in Caucasians, this makes investigations

useful since there is a good chance of finding patients

with all the allelic re6295 variants. On the other hand,

Arg219Leu (rs1800044) and Gly22Ser (rs1799920) have

no frequency data in the general population, and the

Ile28Val (rs1799921) G allele has a small prevalence in

Caucasians (0.8%). No prevalence data is available

for the other populations in international databases

(for all prevalence distribution, see Table 1). A small

probability of finding a carrier of a mutant allele

impairs the clinical impact of association studies based

on cited variations. This issue will be further dis-

cussed.

Expression and functions

Table 2 gives a list of expression studies in association

with 5-HT1A genetic variants. The main results and

significance are reported.

The 5-HT1A G(-1019) allele in the 5-HT1A upstream

regulatory region fails to bind identified repressors

Deaf-1 and Hes5, abolishing Deaf-1 action and im-

pairing Hes5 action. This leads to the up-regulation

of receptor expression (Albert and Lemonde, 2004;

Lemonde et al., 2003). In fact, an increase in 5-HT1A

receptor expression in individuals with the G/G

genotype has been observed (Parsey et al., 2006b).

Moreover, Deaf-1 has been shown to have a different

function in presynaptic and post-synaptic cells or

non-serotonergic neurons: it does not repress but

actually enhances the 5-HT1A expression in post-

synaptic cells (Czesak et al., 2006). This might help

explain some of the conflicting findings reported in the

literature : observations of cortical region-specific

decreases in the 5-HT1A receptor in PET studies of

psychiatric patients could be mediated by the disrup-

tion of Deaf-1 signalling leading to down-regulation of

5-HT1A expression in post-synaptic neurons. Another

interesting evidence and proposed explanation, is

highlighted by a PET study by David et al. (2005):

following the evidence of a decreased expression in

5-HT1A receptors in 5-HT transporter knock-out rats,

especially in females (Li et al., 2000), David and col-

leagues studied the mutual relationship between two

different gene variations: the 5-HT transporter and

5-HT1A. These authors reported that a minor avail-

ability of 5-HT1A was detected only in subjects (heal-

thy volunteers) with 5-HTTLPR short (SS or SL)

genotypes with no influence of 5-HT1A variants. A

possible explanation proposed was that the lower

transcriptional efficiency associated with the S allele

of 5-HTTLPR may lead to decreased 5-HTT function,

which in turn could lead to a lifelong increase in 5-HT

tone, which may then desensitize and down-regulate

5-HT1A receptors. This is consistent with animal

experiments : SERT knockout rats show a decreased

5-HT1A density (Li et al., 2004). Moreover, the absence

of increased 5-HT1A density in normal subjects with

the G/G genotype might also suggest that healthy

subjects can better compensate for their unfavourable

genetic make up than depressed patients.

Interestingly, in the same year Arias and colleagues

found that a combined genetic effect of HTR1A and

5-HTTLPR variants, and not the 5-HTTLPR variant

alone, could influence the antidepressant response in a

sample of 130 depressed patients, being the S/S G/G

the at risk haplotype for failed remission achievement

(Arias et al., 2005). It is really difficult to discuss the

partially different findings in these papers: the first

one, by David et al., is based on healthy subjects

and the second one, by Arias et al., investigates the

antidepressant (citalopram) response in a sample

of depressed subjects. Moreover, sample sizes are

different : 35 in the study by David and colleagues,

130 in the study by Arias et al. Stratification factors

such as different sociodemographic variables, co-

morbidity, pre-treatment issues such as response,

and type and duration of previous therapies in the

depressed sample make it difficult to draw definitive

conclusions.

Finally, a third hypothesis was proposed in 2002 by

Gross and colleagues (Gross et al., 2002) and in 2004 by

704 A. Drago et al.

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 5: 5-HT1A gene variants and psychiatric disorders: a review of current

Table 1. rs allele prevalence in different populations

rs Position Caucasian Asians (Han Chinese) Asians ( Japanese) Sub-Saharan African

rs6295

(C(-1019)G)aPromoter C=0.675 No frequency data No frequency data No frequency data

G=0.325

rs6295 Promoter C=0.675 No frequency data No frequency data No frequency data

G=0.325

rs10042486 Promoter C=0.517 C=0.133 C=0.125 C=0.271

T=0.483 T=0.867 T=0.875 T=0.729

rs6878612 Promoter A=0.510 A=0.087 A=0.090 A=0.250

G=0.490 G=0.912 G=0.910 G=0.750

rs7448024 Promoter A=0.942 A=1 A=1 A=1

C=0.058 C=0.0 C=0.0 C=0.0

rs6449694 Promoter No frequency data No frequency data No frequency data No frequency data

rs6871276 Promoter No frequency data No frequency data No frequency data No frequency data

rs12653018 Promoter A=0.508 A=0.133 A=0.122 A=0.267

C=0.492 C=0.867 C=0.878 C=0.733

rs11960685 Promoter No frequency data No frequency data No frequency data No frequency data

rs1799921

(Ile28Val)aExon A=0.992 No frequency data No frequency data No frequency data

G=0.008

rs1800044

(Arg219Leu)abExon No frequency data No frequency data No frequency data No frequency data

rs1799920

(Gly22Ser)abExon No frequency data No frequency data No frequency data No frequency data

rs6294 Exon A=0.0 G=1 A=0.211c A=0.065 No frequency data No frequency data

G=0.789c G=0.935

rs10085024 3k-UTR C=0.517 C=0.144 C=0.125 C=0.181

G=0.483 G=0.856 G=0.875 G=0.819

rs4521432 3k-UTR C=0.492 C=0.856 C=0.878 C=0.817

T=0.508 T=0.144 T=0.122 T=0.183

rs1364043 3k-UTR G=0.217 G=0.622 G=0.700 G=0.183

T=0.783 T=0.378 T=0.300 T=0.817

rs970453 3k-UTR No frequency data No frequency data No frequency data No frequency data

rs1423691 3k-UTR C=0.429 C=0.850c C=0.800 C=0.875 C=0.817

T=0.571 T=0.150c T=0.200 T=0.125 T=0.183

rs7448318 3k-UTR No frequency data No frequency data No frequency data No frequency data

rs10062933 3k-UTR No frequency data No frequency data No frequency data No frequency data

rs749099 3k-UTR A=0.490 A=0.830 A=0.770 No frequency

G=0.510 G=0.170 G=0.230 data

rs749098 3k-UTR No frequency data No frequency data No frequency data No frequency data

rs878567 3k-UTR C=0.483 C=0.867 C=0.878 C=0.742

T=0.517 T=0.133 T=0.122 T=0.258

rs6449693 3k-UTR A=0.483 A=0.867 A=0.878 A=0.742

G=0.517 G=0.133 G=0.122 G=0.258

rs10046068 3k-UTR C=0.99 C=0.99 C=0.99 C=0.842

T=0.01 T=0.01 T=0.01 T=0.158

rs13436914 3k-UTR C=0.01 C=0.078 C=0.056 C=0.347

T=0.99 T=0.922 T=0.944 T=0.653

rs13361335 3k-UTR G=0.050 G=0.622 G=0.700 G=0.133

T=0.950 T=0.378 T=0.300 T=0.867

rs12515022 3k-UTR No frequency data No frequency data No frequency data No frequency data

aMost investigated; b not validated; c Afro Americans.

HTR1A and psychiatric disorders 705

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 6: 5-HT1A gene variants and psychiatric disorders: a review of current

Lesch and colleagues (Lesch and Gutknecht, 2004) : the

influence of the 5-HT1A coding sequence variations

should not be expected to reveal itself through ex-

pression on adult neuronal surfaces, but by the com-

plex neuronal mutual net influences involved in brain

formation. Thus, the genetically determined 5-HT1A

enrichment would influence the neuronal net forma-

tion eventually predisposing to psychiatric disorders.

Evidence shows that 5-HT1A knockout rats show an

anxiety-like behaviour (Groenink et al., 2003; Heisler

et al., 1998; Olivier et al., 2001; Parks et al., 1998;

Parsons et al., 2001; Ramboz et al., 1998; Toth, 2003):

future animal research should be able to selectively

shut down pre- or post-synaptic 5-HT1A receptors, and

this would provide unique evidence for understand-

ing the detection of the predictive utility of genetic

variations.

The Arg219Leu variant was found to be expressed

in virtually identical densities in wild and variant

transfected HEK293 cells (Bruss et al., 2005), suggest-

ing that this variant has no influence over the stability

or efficiency of transductional and translational sys-

tems of the 5-HT1A coding sequence, even though it

was found that the ability of G-coupling activity after

receptor activation, and the inhibition of forskolin-

stimulated accumulation, was decreased by 60–90% in

variant type. In view of that, this variant apparently

does not change the binding properties of 5-HT1A, but

it is associated with a drastic impairment of signal

transduction: it seems reasonable to expect that

patients carrying the variant will show different drug

treatment responses.

A reduced receptor activity together with a lower

density on cell surfaces was also reported for the

Table 2. Expression studies

Study rs Sample Result Significance

Lemonde

et al. (2003)

C(-1019)G 129 Caucasian major depressed

patients

G associated with depression

and suicide

p=0.0017 (allele –

depression)

134 Caucasian controls 102

Caucasian suicide completers

p=0.002 (allele –

suicide attempters)

116 normal controls

Parsey et al.

(2006a)

C(-1019)G 43 controls Different genotype distribution

between non-remitters and

controls

p=0.005

22 depressed (medication-free) A trend in non-remitters to be

G carriers

p=0.0058

Parsey et al.

(2006b)

C(-1019)G 43 controls GG genotype associated with

depression

p=0.059

28 depressed (medication-free)

Czesak et al.

(2006)

C(-1019)G RN46A, septal SN48,

neuroblastoma SKN-SH,

neuroblastoma/glioma

NG108-15 cells

The intrinsic activity of Deaf-1

at the 5-HT1A promoter is

opposite in presynaptic vs.

post-synaptic neuronal cells

and requires deacetylation

pf0.01

Li et al.

(2000)

C(-1019)G serotonin (5-HT) transporter

knock-out mice (5-HTT x/x)

Reduced 5-HT1A density in

raphe, more extensively in

females

pf0.05

Bruss et al.

(2005)

Arg219Leu HEK293 cells The variant allele is associated

with impaired signal

transduction

pf0.05

Rotondo

et al. (1997)

Ile28Val

Gly22Ser

BamHI linearized eukaryotic

expression vector

Ser22 variant is capable of

attenuating agonist-mediated

receptor down-regulation and

desensitization

pf0.05

Erdmann

et al. (1995)

Ile28Val 352 patients 210 controls No different prevalence of

mutant allele

Not significant

Bruss et al.

(1995)

Ile28Val Cells No different pharmacological

profile between alleles

Not significant

706 A. Drago et al.

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 7: 5-HT1A gene variants and psychiatric disorders: a review of current

Gly22Ser variant but only after 24 h treatment with a

5-HT1A agonist, whereas the binding affinity and

pharmacological profile were found to be similar in

wild an variant genotype-expressing cells (Gothert

et al., 1998; Rotondo et al., 1997). In the same study

the Ile28Val variant was found not to influence affin-

ity, pharmacologically promoted or agonist-promoted

down-regulation issues. Most part of those investiga-

tions are over-expression studies in non-neuronal

cells : further validation in neuronal models or in vivo

would be advisable. Given that, a genetic approach

to patients will offer the opportunity to highlight a

different and predictable, i.e. genetically determined,

receptor drug treatment determined down-regulation,

which is the supposed therapeutic mechanism of anti-

depressant drugs. Finally, the Ile28Val variant was

found to be equally expressed in psychiatric and

healthy subjects (Erdmann et al., 1995), and to share

the same pharmacological profile with the wild vari-

ant (Bruss et al., 1995).

Table 3. Major depressive disorder

Study rs Sample Result Significance

Parsey et al.

(2006a)

C(-1019)G 22 Caucasian depressed

(medication-free)

Different genotype distribution

between non-remitters and

controls

p=0.005

43 Caucasian controls A trend in non-remitters to be

G carriers

p=0.0058

Parsey et al.

(2006b)

C(-1019)G 28 Caucasian depressed

(medication-free)

GG genotype associated with

depression

p=0.059

43 Caucasian controls

Yu et al.

(2006)

C(-1019)G

Gly272Asp

222 Chinese major depressive

patients treated with 4-wk fluoxetine

G associated with worse

response in females

p=0.001

Hong et al.

(2006)

C-1019G 224 Chinese major depressive patients

treated with 4-wk fluoxetine

G associated with worse

response to treatment

p=0.009

Arias et al.

(2005)

C-1019G 130 Caucasian major depressive

patients treated with citalopram

G associated with worse

response to treatment when

associated with short HTTLPR

p=0.009

HTTLPR

Lemonde

et al. (2004)

C-1019G 118 Caucasian major depressive

patients treated with fluoxetine,

nefadozone, pindolol, flibanserin

G associated with worse

response to treatment

p=0.0497

Lemonde

et al. (2003)

C(-1019)G 129 Caucasian major depressed

patients

G associated with depression

and suicide

p=0.0017 (allele –

depression)

134 Caucasian controls 102 Caucasian

suicide completers

p=0.002 (allele –

suicide

attempters)

116 normal controls

Mandelli

et al. (2006)

C(-1019)G 686 Caucasian depressive or bipolar

patients

No significant association Not significant

Peters et al.

(2004)

Various

SNPs

96 Caucasian major depressive patients

treated with 12-week fluoxetine

No significant association Not significant

Serretti et al.

(2004)

C(-1019)G 151 Caucasian major depressed and

111 Caucasian bipolars treated with

6-wk fluoxetine

G associated with worse

treatment response in bipolars

p=0.036

Kraus et al.

(2007)

C(-1019)G 139 hepatitis C-infected outpatients

treated with interferon ax2b

G associated with depression as

interferon treatment unwanted

side-effect

p=0.011

Levin et al.

(2007)

C(-1019)G 130 Caucasian major depressive

patients treated with SSRIs

No significant association Not significant

Suzuki et al.

(2004)

Gly272Asp 65 Japanese depressed patients Gly associated with worse

treatment response

p=0.009

to p=0.031

HTR1A and psychiatric disorders 707

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 8: 5-HT1A gene variants and psychiatric disorders: a review of current

Depression

Table 3 gives a list of studies dealing with major de-

pressive disorder (MDD) and antidepressant response

in association with 5-HT1A genetic variants. The main

results and significance are reported.

The 5-HT system is implicated in major depression

and suicide and is negatively regulated by somato-

dendritic 5-HT1A autoreceptors (Albert and Lemonde,

2004) ; moreover, desensitization of 5-HT1A auto-

receptors is believed to be related to the therapeutic

effects of antidepressant drugs and it is probably im-

plicated in the 2- to 3-wk latency for antidepressant

treatments (Blier et al., 1998). A significant decrease in

5-HT1A mRNA was found in the dorsolateral PFC and

hippocampus in chronically stressed animal models

and in subjects with MDD (Lopez et al., 1998; Lopez-

Figueroa et al., 2004).

This evidence makes the 5-HT1A genetic variants

good candidates in the pathophysiology and treat-

ment of depression. The G allele of the C(-1019)G

polymorphism has been reported to be associated with

enriched expression of the gene and there is some

evidence suggesting that G allele carriers have higher

risks for a depressive episode and of being more re-

sistant to antidepressant therapy (Arias et al., 2005;

Hong et al., 2006; Lemonde et al., 2003, 2004; Parsey

et al., 2006b; Yu et al., 2006), even though conflicting

evidence can also be found (Mandelli et al., 2006;

Peters et al., 2004; Serretti et al., 2004). On this topic,

our group reported some results in 2004: the sample

under investigation was composed of 151 major

depressed and 111 bipolar patients, treated with

fluvoxamine for a period of 6 wk. In this sample, the

depressed patients responded to treatment indepen-

dently from 5-HT1A variants, whereas, in the bipolar

sample, HTR1A C/C genotype carriers showed a

better response to fluvoxamine.

In a interesting recent study, Kraus and colleagues

reported that interferon therapy associated with

depressive episode was found to be related to the

G-allele homozygosity of the C(-1019)G variant (Kraus

et al., 2007), whereas 5-HT promoter (SLC6A4 or

5-HTTLPR) or tryptophan hydroxylase 2 (TPH2) gene

variations were not associated with such an important

iatrogenic effect.

Finally, in 40 remitted mood disorders patients,

C/C genotype carriers had lower scores at TCI (tem-

perament and character inventory) subscales of total

transcendence, transpersonal identification and spiri-

tual acceptance (items, or item clusters) (Lorenzi et al.,

2005), this finding agrees with a converging imaging

study (Borg et al., 2003).

There are other gene variations that have been in-

vestigated as relevant for the antidepressant response:

in 2004 Suzuki and colleagues (Suzuki et al., 2004)

found that the Gly272Asp variant could influence flu-

voxamine response in a sample of 65 depressed

patients being Asp allele-associated with higher

Hamilton Depression Rating Scale (HAMD) score re-

ductions after 2, 6 and 12 wk of treatment. Conflicting

with this, lack of association between fluvoxamine re-

sponse and the Gly272Asp variant was found in a

sample of 222 Chinese major depressive patients (Yu

et al., 2006), even though the C(-1019)G variant, which

was reported as in strong linkage disequilibrium (LD)

with Gly272Asp, showed a significant association in

the female group; C carriers having better clinic re-

sults after fluvoxamine treatment. This observation

raises the issue of LD calculation, in fact in the paper

the authors define strong LD based on a highD’, while

the r2 low value (less sensitive to low frequencies and

small sample sizes) indicated a marginal LD. There-

fore it is not surprising to observe discrepant findings

between variants.

As far as Caucasian samples are considered, the

positive association studies by Parsey and colleagues

are based on small sample size (cases n=22 and n=28)

(Parsey et al., 2006a,b) : this is an important limitation.

Moreover, the significant result by Parsey et al. (2006a)

was found in genotype distribution within remitters

and the control group (p=0.005), while no significant

difference was found between remitters and non-

remitters (p=0.073). Analysis on allele distribution

reported non-significant results (p=0.058). Moreover,

the positive study by Arias and colleagues (Arias et al.,

2005) considered a sufficient sample size (cases

n=130) but revealed significant association between

5-HT1A genetic variations and antidepressant response

only after considering the genetic variation of the

HTR1A together with the functional polymorph-

ism in the promoter of the serotonin transporter

(HTTLPR), prompting the hypothesis that the statisti-

cal significance is mostly based on the latter. This is

consistent with the negative results our group re-

ported about Caucasian samples (Mandelli et al., 2006;

Serretti et al., 2004) together with other negative find-

ings obtained by other groups (Peters et al., 2004). The

positive findings reported in Asian samples (Hong

et al., 2006; Suzuki et al., 2004; Yu et al., 2006) could

be explained by the different prevalence of the in-

vestigated polymorphisms between different ethni-

cities. Another possible reason to explain conflicting

results is that complex phenotypes can not be ex-

plained by single gene mutations: considering haplo-

types involving different genes within the 5-HT

708 A. Drago et al.

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 9: 5-HT1A gene variants and psychiatric disorders: a review of current

system and beyond it, could be a good strategy. The

previously mentioned paper by Arias et al. (2005)

stressed this line of research. Finally, a more complete

investigation of the all the single gene variants is sug-

gested.

Suicide

Table 4 gives a list of studies dealing with suicidal

behaviour in association with 5-HT1A genetic variants.

The main results and significance are reported.

The C(-1019)G variant has been associated with

suicidal behaviour, even though there is also conflict-

ing evidence and generally this variant together with

the structural Gly272Asp and Pro16Leu variants

do not seem to be strongly associated with suicide

behaviour or attempt. In a recent study we performed

(Serretti et al., 2006), no significant association

was found between 5-HT1A variants and suicide.

Wassermann and colleagues (2006) had similar

results in a sample of 272 suicide attempter families :

the HTR1A promoter variant seemed to be not as-

sociated with suicide history except for a trend of

G allele over transmission in a subgroup of suicide

attempters characterized by high levels of traumatic or

stressful life events prior to the suicide attempt.

Interestingly, stressful life events were not found to

be associated with HTR1A variants and mood dis-

orders in a large Italian sample (n=686) recently in-

vestigated by our group (Mandelli et al., 2006). Those

lines of evidence taken together might suggest that

suicide could be associated with the HTR1A promoter

polymorphism and stressful life events independently

from mood disorders. A stronger boundary with

the impulsiveness cluster, might be a possible hy-

pothesis.

Other negative association results can be found in

some studies considering the promoter or the struc-

tural polymorphisms (Huang et al., 2004; Nishiguchi

et al., 2002; Ohtani et al., 2004; Videtic et al., 2006).

Positive association findings are also available

(Lemonde et al., 2003). Again, a possible explanation

of non unequivocal evidence could be addressed to

incomplete gene polymorphism analysis, to the un-

known and probably multifactorial pathogenesis of

psychiatric disorders, and to the small impact of a

single gene in such complex phenotypes (Kendler,

2005). According to the last point, an interesting study

performed by Hsiung and colleagues (2003) reported

that at least two independent second-messenger cas-

cades after 5-HT1A activation in the brain of suicide

victims were differently active when compared with

controls, this suggests that future studies should con-

sider complex haplotypes, choosing them on the basis

of evidence and rational study of internal cell mech-

anisms.

Anxiety disorders

Table 5 gives a list of studies dealing with anxiety

disorders in association with 5-HT1A genetic variants.

The main results and significance are reported. Several

lines of evidence show a significant relationship be-

tween 5-HT1A and anxiety symptoms.

Table 4. Suicide

Study rs Sample Result Significance

Serretti et al. (2006) C(-1019)G 411 Caucasian suicide attempters No significant association Not significant

rs1423691 443 controls

rs878567

Wasserman et al.

(2006)

C(-1019)G 272 Caucasian suicide attempter families G allele overtransmitted

in a subgroupa

p=0.0630

Videtic et al. (2006) C47T 226 Caucasian suicide victims 225

Caucasian healthy control subjects

No significant association Not significant

G815A

Huang et al. (2004) C(-1019)G 696 psychiatric subjects of different

ethnicities

No significant association Not significant

241 post-mortem brain cases

107 healthy volunteers

Ohtani et al. (2004) Pro16Leu 134 Japanese suicide victims No significant association Not significant

325 Japanese controls

Nishiguchi et al.

(2002)

Pro16Leu 163 Japanese suicide completers No significant association Not significant

Gly272Asp 163 Japanese controls

a High level of previous traumatic and/or stressful life events.

HTR1A and psychiatric disorders 709

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 10: 5-HT1A gene variants and psychiatric disorders: a review of current

Animal experiments show anxiety-like behaviour in

5-HT1A knockout rats (Groenink et al., 2003; Heisler

et al., 1998; Olivier et al., 2001; Parks et al., 1998;

Parsons et al., 2001; Ramboz et al., 1998; Toth, 2003),

probably because of a perturbation in the prefrontal

GABA/glutamate system (Bruening et al., 2006).

Moreover, severe acute stress as well as stress of 2 wk

duration has been found to be associated with down-

regulation of 5-HT1A mRNA synthesis in the rat hip-

pocampus (Lopez et al., 1998, 1999). Neumeister and

colleagues consistently reported lower 5-HT1A density

in the anterior cingulate, posterior cingulate and raphe

of patients suffering from panic disorders with or

without depressive comorbidity (Neumeister et al.,

2004). Anti-anxiety properties of 5-HT1A active drugs

are well known in everyday clinical practice. Con-

sistent with this, panic disorder was recently found

to be associated with HTR1A and COMT variants

(Freitag et al., 2006), and when HTR1A is considered

alone, a significant relationship is still found between

the promoter polymorphism and panic (Huang et al.,

2004; Rothe et al., 2004). Those findings seem to be

more significant when panic with agoraphobia is con-

sidered, prompting the idea that a more in-depth dis-

section of clinical phenotypic subtypes would be a

good strategy for future investigations. Post-traumatic

stress disorder (PTSD) does not seem to be associated

with 5-HT1A density variants (Bonne et al., 2005).

There is a lack of evidence regarding other anxiety

disorders.

Bipolar disorder

Imaging studies investigating the hypothesis of a

different density or affinity of 5-HT1A receptors in

hippocampal and cortex regions reported negative

findings (Dean et al., 2003; Gray et al., 2006), even

though an interesting dissociation between the num-

ber of receptors and the activity of G protein-

associated second-messenger cascade was reported in

the study performed by Gray and colleagues: this

could suggest a kind of segregation of 5-HT1A re-

ceptors leading to a normal number of receptors as-

sociated with a reduced activity in bipolar disorder. In

the same study a gender difference was reported, i.e.

5-HT1A receptors were expressed less in female brains

(Gray et al., 2006). A mRNA in-situ hybridization

study confirmed this report (Lopez-Figueroa et al.,

2004).

Conflicting with this, some animal studies in-

vestigated the possible role of 5-HT1A receptors in the

pharmacodynamics of mood stabilizers, reporting

positive association results. Lithium was found to en-

hance the sensitivity of post-synaptic 5-HT1A after

short-term administration (Blier et al., 1987). This

finding was replicated by Haddjeri and colleagues

(2000) : lithium augmentation of long-term anti-

depressant treatment in rats was found to be as-

sociated with an enhanced disinhibition of dorsal

hippocampal CA3 pyramidal neurons, an action

which is known to be mediated by post-synaptic

5-HT1A receptors. More recently, McQuade and col-

leagues reported an association with chronic lithium

treatment and reduced 5-HT1A mRNA in the hippo-

campus, whereas no different levels of 5-HT1A mRNA

could be found in the dorsal raphe nucleus where

5-HT1A receptors are present as autoreceptors

(McQuade et al., 2004). Post-synaptic 5-HT1A receptors

were found to be influenced also by lamotrigine,

another mood stabilizer (Bourin et al., 2005), whereas

Table 5. Anxiety

Study rs Sample Result Significance

Freitag et al.

(2006)

C(-1019)G 115 Caucasian patients

suffering from panic attacks

GG genotype associated with panic

when considered together with

COMT polymorphism

p=0.04

115 Caucasian controls

Rothe et al.

(2004)

C(-1019)G 134 Caucasian patients suffering

from panic attacks with (n=101)

or without agoraphobia

G allele associated with panic

disorder with agoraphobia

p=0.03

134 Caucasian controls

Huang et al.

(2004)

C(-1019)G 696 psychiatric subjects

of different ethnicities

G associated with schizophrenia,

substance use and panic attack

p=0.009

p=0.015

241 post-mortem brain cases

107 healthy volunteers

No significant association result in

post-mortem brain (mRNA levels

and genotype)

p=0.043

respectively

710 A. Drago et al.

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 11: 5-HT1A gene variants and psychiatric disorders: a review of current

negative findings were reported for valproate (Delva

et al., 2002; Khaitan et al., 1994). As mentioned

previously, in our 2004 study, we reported that

HTR1A C/C bipolar genotype carriers showed a bet-

ter response to fluvoxamine in regard to depressive

episodes (Serretti et al., 2004). Those lines of evidence

could suggest a possible pharmacological impact of

5-HT1A gene variants, but more studies and a larger

gene investigation are needed in this field of research.

Psychosis

Table 6 gives a list of studies dealing with psychotic

disorders in association with 5-HT1A genetic variants.

The main results and significance are reported.

5-HT1A seems to be highly involved in the patho-

logical neurotransmitter disequilibrium associated

with schizophrenia. Several post-mortem brain stud-

ies reported HTR1A altered expression in schizo-

phrenia patients (see Frankle et al., 2006 for a review),

and, consistent with this, two PET studies (Kasper

et al., 2002; Tauscher et al., 2002) reported increased

5-HT1A density in prefrontal and temporal cortices,

whereas a diminished density in amygdala or no dif-

ferent density compared to controls were found in

other PET studies (Frankle et al., 2006; Yasuno et al.,

2004). This diversity could be due to the different res-

olution of post- mortem and PET studies: only the

former can investigate the 5-HT1A density in different

layers of brain tissues.

Besides conflicting imaging studies, there is evi-

dence that 5-HT1A is involved in antipsychotic re-

sponse. Many atypical antipsychotics act on 5-HT1A

(clozapine, serizotan, bifeprunox, nomonapride, zi-

prasidone, aripiprazole, tiospirone) and even though

other atypicals such as olanzapine or risperidone are

not directly active on 5-HT1A, it has been suggested

that the improvement of depressive symptomatology,

which is characteristic of atypical antipsychotics,

could be associated with a dopamine-enhanced re-

lease in the mPFC, this being dependent also on

5-HT1A activity (Ichikawa et al., 2001; Newman-

Tancredi et al., 2005). The role of 5-HT1A receptors in

antipsychotic treatment, especially as far as negative

symptomatology is concerned, is suggested by

Richtand and colleagues who recently reported that

the strongest correlation with clinically effective drugs

and receptor activity profile is the HTR2C/D2 ratio for

the typical antipsychotics, and HTR2A/5-HT1A and

HTR2C/5-HT1A for the atypical antipsychotics : the

higher the 5-HT1A expression or function, the less ef-

fective the antipsychotic treatment (Richtand et al.,

Table 6. Psychosis

Study rs Sample Result Significance

Huang

et al. (2004)

C(-1019)G 696 psychiatric subjects of

different ethnicities

G associated with schizophrenia,

substance use and panic attack

p=0.009

p=0.015

241 post-mortem brain cases

107 healthy volunteers

No significant association result

in post-mortem brain (mRNA

levels and genotype)

p=0.043

respectively

Reynolds

et al. (2006)

C(-1019)G 63 drug naive Caucasian

psychotic patients treated

with antipsychotic for 3 months

G allele associated with worse

response to treatment (negative

symptoms)

p=0.001

Lane et al. (2006) Gly272Asp 123 Han Chinese psychotic

patients treated with

risperidone

No association with psychotic

symptoms

Not significant

Serretti et al.

(2000)

Ile28Val 72 Caucasian bipolar patients No association with psychotic

symptoms

Not significant

12 Caucasian depressed patients

Erdmann

et al. (1995)

Ile28Val 352 Caucasian schizophrenic No significant association Not significant

210 Caucasian controls

Kawanishi

et al. (1998)

Gly272Asp 61 Japanese schizophrenic No significant association Not significant

Pro16Leu 100 Japanese controls

C-51T

G-152C

C-321G

-480delA,

A-581C

HTR1A and psychiatric disorders 711

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 12: 5-HT1A gene variants and psychiatric disorders: a review of current

2007). Moreover, Cosi and colleagues reported that

atypical antipsychotics that are active on 5-HT1A, but

not haloperidol which is devoid of 5-HT1A, were as-

sociated with a reduced kainite-induced brain damage

in rats (Cosi et al., 2005). This is quite interesting be-

cause of the possible neurodegenerative aetiopatho-

genesis of schizophrenia, and the suggestion of a

possible use of 5-HT1A agonists as protectors against

excitotoxic injuries.

In regard of the benefit over negative symptoma-

tology, Sato and colleagues recently reported that ris-

peridone can increase acetylcholine release (which is

assumed to be associated with cognitive performance),

in the PFC of rats through a complex mechanism

enhanced by 5-HT1A activation: if its activation is

missing, the acetylcholine levels in prefrontal brain

structures are impaired after risperidone treatment,

whereas the simple 5-HT1A stimulation does not alter

acetylcholine levels in the brain (Sato et al., 2007). The

same result was obtained by Chung and colleagues in

2004, using clozapine (Chung et al., 2004). This is

consistent with other papers which reported the as-

sociation of 5-HT1A with cognitive functions

(Sumiyoshi et al., 2000; Sumiyoshi and Meltzer, 2004;

Tamminga, 2006). The benefit on antipsychotic-

induced motor side-effects could be also partially

associated with 5-HT1A function when atypical anti-

psychotics are used: it has been proposed that the

serotonergic modulation over dopaminergic system is

principally inhibitory, thus, the stimulation of pre-

synaptic 5-HT1A should lead to an inhibitory effect

over HTR2C which in dopaminergic neurons will lead

to a final effect of diminished inhibition and a benefit

for movement dyscontrol (Haleem, 2006).

Other evidence of 5-HT1A modified function in

schizophrenia comes from a study by Lee and Meltzer

(2001) reporting that 5-HT1A functionality is impaired

in schizophrenic females: the plasma cortisol concen-

tration increased after ipsapirone administration was

found to be blunted in schizophrenic female patients.

Other parameters such as hypotermina, nausea, diz-

ziness, irritability, and feeling less well did not differ

between schizophrenic and healthy subjects. It must

be underlined that the blunted plasma cortisol con-

centration stands for a hypofunction of 5-HT1A post-

synaptic receptors (Lee and Meltzer, 2001).

Even though all these lines of evidence should en-

courage investigations on HTR1A variants, there are

not many studies investigating 5-HT1A genetic vari-

ants and the study of clinical characteristics of

schizophrenia or schizophrenia-like disorders.

In 2004 Huang and colleagues (Huang et al., 2004)

reported that the C(-1019)G variant was associated

with schizophrenia together with substance use dis-

order and panic attacks, this result was replicated 2 yr

later by Reynold and colleagues (2006) who reported

that C/C carriers had better scores at PANSS (Positive

and Negative Syndrome Scale), but not for positive

symptoms, and at the Calgary Depression Scale than

G/G carriers after 3 months of a standard treatment

for acute psychosis. Interestingly, with the Calgary

Depression Scale G/G carriers showed worse con-

ditions at the end of treatment than at the beginning.

This is consistent with the literature findings dis-

cussed above about HTR1A variant and depression.

In 2006 Lane and colleagues investigated the hy-

pothesis that HTR1A variants, together with another

15 genetic variants across 10 candidate genes [5-HT1A,

5-HT2A, 5-HT2C, 5-HT6, D1, D2, D3, a1-adrenergic re-

ceptors, brain-derived neurotrophic factor (BDNF),

and cytochrome P450 2D6], could influence the ris-

peridone-associated weight gain side-effect. A sample

of 122 Han Chinese schizophrenia in-patients was

selected: the HTR1A Gly272Asp variant showed no

evidence of association (Lane et al., 2006). Finally, our

group investigated the Ile28Val polymorphism of the

5-HT1A coding sequence in a sample of 84 in-patients

affected by mood disorders (72 bipolar and 12 major

depressive disorder) verifying the hypothesis that

5-HT1A could be involved in psychotic symptoms of

mood disorders. No association result was found

(Serretti et al., 2000). The different ethnicity of the

samples investigated [Caucasians (Erdmann et al.,

1995; Huang et al., 2004; Reynolds et al., 2006; Serretti

et al., 2000) and Asians (Kawanishi et al., 1998; Lane

et al., 2006)], could explain the inconsistency of results.

As far as the study on the bipolar sample is concerned

(Serretti et al., 2000), the different psychiatric disorders

at the basis of the investigation, is probably the most

important cause of discrepancy.

The Ile28Val variant was also investigated in a large

case (n=352) control (n=210) study with no evidence

of association with schizophrenia. The sample also

included other diagnoses such as bipolar disorder and

Tourette’s syndrome (Erdmann et al., 1995). Lack of

association within HTR1A variants and schizophrenia

was also reported by Kawanishi and colleagues

(1998) : in this work a list of variants was considered

[Gly272Asp, Pro16Leu, A(294)G, T(549C), C(51T),

G(-152)C, C(-321)G, (-480)delA, and A(-581)C]. So

far, studies investigating the C(-1019)G variant tended

to find positive association results in Caucasian

samples. The other considered variants tended to give

negative association results. Study designs involving

all the known and validated genetic variants of 5-HT1A

are missing.

712 A. Drago et al.

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 13: 5-HT1A gene variants and psychiatric disorders: a review of current

Other findings

Anxiety, as measured by the NEO Personality

Inventory, showed a negative association with 5-HT1A

density in a PET study performed in a sample of 19

healthy volunteers (Tauscher et al., 2001). In another

PET study, the 5-HT1A density was found to be

negatively correlated with spiritual acceptance, a

sub-item of the TPQ (Tridimensional Personality

Questionnaire) in 15 healthy subjects (Borg et al.,

2003), and in 2005 our group investigated this field

from a genetic point of view, finding that C/C carriers

at the C(-1019)G variant were more likely to have sig-

nificantly lower scores at the total self-transcendence

and at the subscales of transpersonal identification

and spiritual acceptance (Lorenzi et al., 2005). Finally,

an association study considering personality traits and

the C(-1019)G HTR1A promoter variant in 284 healthy

students, reported an association with the G allele and

higher scores on the neuroticism scale on the NEO

Personality Inventory and more harm avoidance on

the TPQ (Strobel et al., 2003).

SNP suggestion

As previously seen, not all the validated SNPs of the

HTR1A sequence have been investigated over the re-

viewed studies. This is an important methodological

bias. A complete and commonly accepted investi-

gation of the gene is advised for the following reasons:

(1) to facilitate the interpretation of results throughout

different studies ;

(2) to cover important functional areas such as 5k-UTR,

3k-UTR, promoter, enhancers, silencers;

(3) to permit an inductive research approach to the

genetic sequence, identifying relevant mutations

still to be hypothesized;

(4) to permit the identification of mutations interfering

with important functional mechanisms such as:

DNA access, reading and transduction, mRNA

stability and translation, mRNA primary structure-

based functions. In all those mechanisms, intronic

sequences are as important as exonic ones.

Actual genome-wide techniques make it possible to

investigate several polymorphisms (up to 600 000) in a

single investigation scan, and the costs have started to

decrease substantially, not least because of compe-

tition between the leading commercial platforms,

Illumina and Affymetrix. This offers a unique oppor-

tunity to investigate haplotypes in a complete way. As

far as 5-HT1A is concerned, its possible involvement in

psychiatric disorders, following evidence of animal

and imaging studies, should be investigated at least

through all the validated variants. A list of validated

SNPs covering all of the gene is presented in Table 1;

data are collected from international databases

(http://www.ncbi.nlm.nih.gov/). To select the SNPs

two filters have been applied: complete coverage of the

gene and validation status. The first point has been

discussed previously. Validation status is advised

because it would be clinically useful and statistically

advisable, to concentrate research on genetic variants

that have a sufficient chance to be detected in the

general population. Table 1 gives the list of selected

variants with allele prevalence information.

Conclusions

Research suggesting that association studies may have

success in finding complex trait genes (De La Vega

et al., 2005; Risch and Merikangas, 1996) and the dis-

covery and validation of large numbers of SNPs in the

human genome (HapMap, 2003), have elicited a

growing interest in the performance of large-scale

genetic association studies using SNPs at the candi-

date-gene, whole-chromosome, and eventually whole-

genome level (De La Vega et al., 2005).

Genetic association studies require a large sample

size and strict investigation methodologies (Risch

andMerikangas, 1996) given that the reliably observed

gene variants odd ratios range around 1.2–1.5

(Kendler, 2005; Lasky-Su et al., 2005). The most part of

published work does not reach the theoretical meth-

odology threshold. It has recently been demonstrated

that going genome-wide makes the sample size a pre-

dominant determinant of the feasibility of the studies.

A number of 1.000 subjects per arm is advised for

common causal alleles with relatively large effect sizes

[genotype relative risk (GRR) 1.7] (Nannya et al.,

2007). Further, the definition of GRR depends also on

the disease model. If f0, f1, f2 are the probabilities of

being affected for individuals with 0, 1, or 2 copies of

the risk allele, then GRR can be: multiplicative

(GRR=f1/f0=f2/f1), additive (GRR=f1/f0), domi-

nant (GRR=f1/0=f2/f0), recessive (GRR=f2/f0=f2/

f1) (http://csg.sph.umich.edu/). Even though most

SNPs are believed to have a co-dominant, additive

effect, other models have been also proposed

(Greenberg et al., 1999; Hranilovic et al., 2004; Lesch,

2001).

However, as a practical example, using an additive

model and considering the prevalence of rs6295

(C(-1019)G), a sample size of 600 cases and 600 con-

trols is needed to obtain a power of 80% (one-stage

design) with a quite liberal p of 0.01 to detect and odds

HTR1A and psychiatric disorders 713

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 14: 5-HT1A gene variants and psychiatric disorders: a review of current

ratio of 1.5. For the rarest Ile28Val mutant allele, a

total sample size of over 50 000 subjects is needed!

(Cohen, 1988). This simulation is driven on the basis of

the investigation of a single gene, which is the most

common path in the reviewed papers. Consideration

of haplotypes further complicates the interpretation

of results given the risk of false positives and non-

independent observation (Purcell et al., 2003; Purcell

and Sham, 2001).

None of the studies reviewed in the present paper

reached the theorically requested sample size for the

investigated mutations. Moreover, many stratification

factors such as ethnicity, sociodemographic and clini-

cal issues, different treatment, different period of

treatment, different study design, multicentre vs.

monocentre investigations, different age of onset, pre-

vious treatments, comorbidity and other important

variables generally influence the validity of results.

A large part of this methodological issue has been

recently summarized and critically reviewed by our

group regarding pharmacogenetics (Serretti et al.,

in press) and may be responsible for inconclusive

findings even in very large sample sizes (Lewis et al.,

2003; Segurado et al., 2003; Wellcome Trust, 2007).

However, beyond clinical and endophenotipic dis-

sections, one important and easy-to-fix bias is the

complete gene variant coverage. In fact, 5-HT1A gene

variants have been reported to play some role in as-

sociation with psychiatric disorders such as mood

disorders, anxiety disorders and psychotic disorders,

but the literature findings are not unequivocal. The

C(-1019)G variant seems to be of primary interest in

antidepressant response, and preliminary studies

seem to indicate a role for 5-HT1A gene variants in

the psychotic spectrum but the evidence remains weak

and difficult to decipher. We suggest that genetic as-

sociation research would be reasonably more efficient

if all the validated SNP variants were included, this

point is easy to accomplish and technically feasible.

Acknowledgements

None.

Statement of Interest

None.

References

Adayev T, Ray I, Sondhi R, Sobocki T, Banerjee P (2003).

The G protein-coupled 5-HT1A receptor causes

suppression of caspase-3 through MAPK and protein

kinase C alpha. Biochimica et Biophysica Acta 1640, 85–96.

Albert PR, Lemonde S (2004). 5-HT1A receptors, gene

repression, and depression: guilt by association.

Neuroscientist 10, 575–593.

Albert PR, Zhou QY, Van Tol HH, Bunzow JR, Civelli O

(1990). Cloning, functional expression, and mRNA tissue

distribution of the rat 5-hydroxytryptamine1A receptor

gene. Journal of Biological Chemistry 265, 5825–5832.

Amargos-Bosch M, Bortolozzi A, Puig MV, Serrats J, Adell

A, Celada P, Toth M, Mengod G, Artigas F (2004). Co-

expression and in vivo interaction of serotonin1A and

serotonin2A receptors in pyramidal neurons of prefrontal

cortex. Cerebral Cortex 14, 281–299.

Andrade R, Nicoll RA (1987). Pharmacologically distinct

actions of serotonin on single pyramidal neurones of the

rat hippocampus recorded in vitro. Journal of Physiology

394, 99–124.

Appert-Collin A, Duong FH, Passilly Degrace P, Warter JM,

Poindron P, Gies JP (2005). MAPK activation via

5-hydroxytryptamine 1A receptor is involved in the

neuroprotective effects of xaliproden. International Journal

of Immunopathology and Pharmacology 18, 21–31.

Arias B, Catalan R, Gasto C, Gutierrez B, Fananas L

(2005). Evidence for a combined genetic effect of the

5-HT(1A) receptor and serotonin transporter genes in

the clinical outcome of major depressive patients

treated with citalopram. Journal of Psychopharmacology 19,

166–172.

Artigas F, Romero L, de Montigny C, Blier P (1996).

Acceleration of the effect of selected antidepressant drugs

in major depression by 5-HT1A antagonists. Trends in

Neuroscience 19, 378–383.

Azmitia EC, Gannon PJ, Kheck NM, Whitaker-Azmitia PM

(1996). Cellular localization of the 5-HT1A receptor in

primate brain neurons and glial cells.

Neuropsychopharmacology 14, 35–46.

Aznar S, Qian Z, Shah R, Rahbek B, Knudsen GM

(2003). The 5-HT1A serotonin receptor is located on

calbindin- and parvalbumin-containing neurons in the

rat brain. Brain Research 959, 58–67.

Barden N, Harvey M, Gagne B, Shink E, Tremblay M,

Raymond C, Labbe M, Villeneuve A, Rochette D,

Bordeleau L, et al. (2006). Analysis of single nucleotide

polymorphisms in genes in the chromosome 12Q24.31

region points to P2RX7 as a susceptibility gene to bipolar

affective disorder. American Journal of Medical Genetics. Part

B: Neuropsychiatric Genetics 141, 374–382.

Bassett AS (1992). Chromosomal aberrations and

schizophrenia. Autosomes. British Journal of Psychiatry 161,

323–334.

Behbehani MM, Liu H, Jiang M, Pun RY, Shipley MT

(1993). Activation of serotonin1A receptors inhibits

midbrain periaqueductal gray neurons of the rat. Brain

Research 612, 56–60.

Blier P, de Montigny C, Tardif D (1987). Short-term lithium

treatment enhances responsiveness of postsynaptic

5-HT1A receptors without altering 5-HT autoreceptor

sensitivity: an electrophysiological study in the rat brain.

Synapse 1, 225–232.

714 A. Drago et al.

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 15: 5-HT1A gene variants and psychiatric disorders: a review of current

Blier P, Pineyro G, el Mansari M, Bergeron R, de Montigny

C (1998). Role of somatodendritic 5-HT autoreceptors in

modulating 5-HT neurotransmission. Annals of the New

York Academy of Science 861, 204–216.

Bobker DH, Williams JT (1990). Serotonin-mediated

inhibitory postsynaptic potential in guinea-pig prepositus

hypoglossi and feedback inhibition by serotonin. Journal of

Physiology 422, 447–462.

Bobker DH, Williams JT (1995). The serotonergic inhibitory

postsynaptic potential in prepositus hypoglossi is

mediated by two potassium currents. Journal of

Neuroscience 15, 223–229.

Bonne O, Bain E, Neumeister A, Nugent AC, Vythilingam

M, Carson RE, Luckenbaugh DA, Eckelman W,

Herscovitch P, Drevets WC, Charney DS (2005). No

change in serotonin type 1A receptor binding in patients

with posttraumatic stress disorder. American Journal of

Psychiatry 162, 383–385.

Borg J, Andree B, Soderstrom H, Farde L (2003). The

serotonin system and spiritual experiences. American

Journal of Psychiatry 160, 1965–1969.

Bourin M, Masse F, Hascoet M (2005). Evidence for the

activity of lamotrigine at 5-HT(1A) receptors in the mouse

forced swimming test. Journal of Psychiatry and Neuroscience

30, 275–282.

Bruening S, Oh E, Hetzenauer A, Escobar-Alvarez S,

Westphalen RI, Hemmings Jr. HC, Singewald N,

Shippenberg T, Toth M (2006). The anxiety-like

phenotype of 5-HT receptor null mice is associated with

genetic background-specific perturbations in the prefrontal

cortex GABA-glutamate system. Journal of Neurochemistry

99, 892–899.

Bruss M, Buhlen M, Erdmann J, Gothert M, Bonisch H

(1995). Binding properties of the naturally occurring

human 5-HT1A receptor variant with the Ile28Val

substitution in the extracellular domain.

Naunyn-Schmiedeberg’s Archives of Pharmacology 352,

455–458.

Bruss M, Kostanian A, Bonisch H, Gothert M (2005). The

naturally occurring Arg219Leu variant of the human

5-HT1A receptor: impairment of signal transduction.

Pharmacogenetics and Genomics 15, 257–264.

Caramaschi D, de Boer SF, Koolhaas JM (2007).

Differential role of the 5-HT1A receptor in

aggressive and non-aggressive mice: an

across-strain comparison. Physiology & Behavior 90,

590–601.

Chattopadhyay A, Rukmini R, Mukherjee S (1996).

Photophysics of a neurotransmitter : ionization and

spectroscopic properties of serotonin. Biophysical Journal

71, 1952–1960.

Chavarria-Siles I, Walss-Bass C, Quezada P, Dassori A,

Contreras S, Medina R, Ramirez M, Armas R, Salazar R,

Leach RJ, et al. (2007). TGFB-induced factor (TGIF): a

candidate gene for psychosis on chromosome 18p.

Molecular Psychiatry 12, 1033–1041.

Chen J, Shen C, Meller E (2002). 5-HT1A receptor-mediated

regulation of mitogen-activated protein kinase

phosphorylation in rat brain. European Journal of

Pharmacology 452, 155–162.

Choudhury K, McQuillin A, Puri V, Pimm J, Datta S,

Thirumalai S, Krasucki R, Lawrence J, Bass NJ, Quested

D, et al. (2007). A genetic association study of chromosome

11q22–24 in two different samples implicates the FXYD6

gene, encoding phosphohippolin, in susceptibility to

schizophrenia. American Journal of Human Genetics 80,

664–672.

Christoforou A, Le Hellard S, Thomson PA, Morris SW,

Tenesa A, Pickard BS, Wray NR, Muir WJ, Blackwood

DH, Porteous DJ, Evans KL (2007). Association analysis of

the chromosome 4p15–p16 candidate region for bipolar

disorder and schizophrenia. Molecular Psychiatry 12,

1011–1025.

Chung YC, Li Z, Dai J, Meltzer HY, Ichikawa J (2004).

Clozapine increases both acetylcholine and dopamine

release in rat ventral hippocampus: role of 5-HT1A

receptor agonism. Brain Research 1023, 54–63.

Cohen J (1988). Statistical Power Analysis for the Behavioral

Sciences. Hillsdale, New Jersey: Lawrence Erlbaum

Associates.

Cosi C, Waget A, Rollet K, Tesori V, Newman-Tancredi A

(2005). Clozapine, ziprasidone and aripiprazole but not

haloperidol protect against kainic acid-induced lesion of

the striatum in mice, in vivo: role of 5-HT1A receptor

activation. Brain Research 1043, 32–41.

Cowen DS, Johnson-Farley NN, Travkina T (2005). 5-HT

receptors couple to activation of Akt, but not extracellular-

regulated kinase (ERK), in cultured hippocampal neurons.

Journal of Neurochemistry 93, 910–917.

Cowen DS, Sowers RS, Manning DR (1996). Activation of a

mitogen-activated protein kinase (ERK2) by the

5-hydroxytryptamine1A receptor is sensitive not only to

inhibitors of phosphatidylinositol 3-kinase, but to an

inhibitor of phosphatidylcholine hydrolysis. Journal of

Biological Chemistry 271, 22297–22300.

Crowe RR, Vieland V (1998). Chromosome 5 workshop.

Psychiatric Genetics 8, 7378.

Crowe RR, Vieland V (1999). Report of the Chromosome

5 Workshop of the Sixth World Congress on Psychiatric

Genetics. American Journal of Medical Genetics 88, 229–232.

Czesak M, Lemonde S, Peterson EA, Rogaeva A, Albert PR

(2006). Cell-specific repressor or enhancer activities of

Deaf-1 at a serotonin 1A receptor gene polymorphism.

Journal of Neuroscience 26, 1864–1871.

Darrow EJ, Strahlendorf HK, Strahlendorf JC (1990).

Response of cerebellar Purkinje cells to serotonin and the

5-HT1A agonists 8-OH-DPAT and ipsapirone in vitro.

European Journal of Pharmacology 175, 145–153.

David SP, Murthy NV, Rabiner EA, MunafoMR, Johnstone

EC, Jacob R, Walton RT, Grasby PM (2005). A functional

genetic variation of the serotonin (5-HT) transporter affects

5-HT1A receptor binding in humans. Journal of

Neuroscience 25, 2586–2590.

Davis KL, Charney D, Coyle JT, Nemeroff C (2002).

Neuropsychopharmacology: the Fifth Generation of Progress.

Philadelphia: Lippincott Williams & Wilkins.

HTR1A and psychiatric disorders 715

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 16: 5-HT1A gene variants and psychiatric disorders: a review of current

De La Vega FM, Gordon D, Su X, Scafe C, Isaac H, Gilbert

DA, Spier EG (2005). Power and sample size calculations

for genetic case/control studies using gene-centric

SNP maps: application to human chromosomes 6, 21,

and 22 in three populations. Human Heredity 60, 43–60.

de Montigny C, Blier P, Chaput Y (1984).

Electrophysiologically-identified serotonin receptors in

the rat CNS. Effect of antidepressant treatment.

Neuropharmacology 23, 1511–1520.

Dean B, Scarr E, Pavey G, Copolov D (2003). Studies on

serotonergic markers in the human hippocampus: changes

in subjects with bipolar disorder. Journal of Affective

Disorders 75, 65–69.

DeLisi LE, Shaw SH, Crow TJ, Shields G, Smith AB, Larach

VW, Wellman N, Loftus J, Nanthakumar B, Razi K, et al.

(2002). A genome-wide scan for linkage to chromosomal

regions in 382 sibling pairs with schizophrenia or

schizoaffective disorder. American Journal of Psychiatry 159,

803–812.

Delva NJ, Brooks DL, Franklin M, al-Said K, Hawken ER,

Merali Z, Lawson JS, Ravindran AV (2002). Effects of

short-term administration of valproate on serotonin-1A

and dopamine receptor function in healthy human

subjects. Journal of Psychiatry and Neuroscience 27, 429–437.

Detera-Wadleigh SD, Berrettini WH, Goldin LR, Martinez

M, Hsieh WT, Hoehe MR, Encio IJ, Coffman D, Rollins

DY, Muniec D, et al. (1992). A systematic search for a

bipolar predisposing locus on chromosome 5.

Neuropsychopharmacology 6, 219–229.

Elder GA, De Gasperi R, Gama Sosa MA (2006). Research

update: neurogenesis in adult brain and neuropsychiatric

disorders. Mt Sinai Journal of Medicine 73, 931–940.

Erdmann J, Shimron-Abarbanell D, Cichon S, Albus M,

Maier W, Lichtermann D, Minges J, Reuner U, Franzek E,

Ertl MA, et al. (1995). Systematic screening for mutations

in the promoter and the coding region of the 5-HT1A gene.

American Journal of Medical Genetics 60, 393–399.

Evans KL, Cropper JD, Berg KA, Clarke WP (2001).

Mechanisms of regulation of agonist efficacy at the

5-HT(1A) receptor by phospholipid-derived signaling

components. Journal of Pharmacology and Experimental

Therapeutics 297, 1025–1035.

Faraone SV, Lasky-Su J, Glatt SJ, Van Eerdewegh P, Tsuang

MT (2006). Early onset bipolar disorder: possible linkage

to chromosome 9q34. Bipolar Disorder 8, 144–151.

Fargin A, Raymond JR, Regan JW, Cotecchia S, Lefkowitz

RJ, Caron MG (1989). Effector coupling mechanisms of the

cloned 5-HT1A receptor. Journal of Biological Chemistry 264,

14848–14852.

Frankle WG, Lombardo I, Kegeles LS, Slifstein M, Martin

JH, Huang Y, Hwang DR, Reich E, Cangiano C, Gil R,

et al. (2006). Serotonin 1A receptor availability in

patients with schizophrenia and schizo-affective

disorder: a positron emission tomography imaging

study with [11C]WAY 100635. Psychopharmacology (Berlin)

189, 155–164.

Freitag CM, Domschke K, Rothe C, Lee YJ, Hohoff C,

Gutknecht L, Sand P, Fimmers R, Lesch KP, Deckert J

(2006). Interaction of serotonergic and noradrenergic gene

variants in panic disorder. Psychiatric Genetics 16, 59–65.

Gorwood P, Leboyer M, Jay M, Hillaire D, Carteault F,

Dugain AM, Berg S, Des Lauriers A, Feingold J (1991).

Familial forms of schizophrenia. Cytogenetic study.

Encephale 17, 525–529.

Gothert M, Propping P, Bonisch H, Bruss M, Nothen MM

(1998). Genetic variation in human 5-HT receptors:

potential pathogenetic and pharmacological role. Annals of

the New York Academy of Science 861, 26–30.

Gray L, Scarr E, Dean B (2006). Serotonin 1a receptor and

associated G-protein activation in schizophrenia and

bipolar disorder. Psychiatry Research 143, 111–120.

Greenberg BD, Tolliver TJ, Huang SJ, Li Q, Bengel D,

Murphy DL (1999). Genetic variation in the serotonin

transporter promoter region affects serotonin uptake in

human blood platelets. American Journal of Medical Genetics

88, 83–87.

Groenink L, Pattij T, De Jongh R, Van der Gugten J,

Oosting RS, Dirks A, Olivier B (2003). 5-HT1A receptor

knockout mice and mice overexpressing

corticotropin-releasing hormone in models of anxiety.

European Journal of Pharmacology 463, 185–197.

Gross C, Zhuang X, Stark K, Ramboz S, Oosting R, Kirby L,

Santarelli L, Beck S, Hen R (2002). Serotonin1A receptor

acts during development to establish normal anxiety-like

behaviour in the adult. Nature 416, 396–400.

Grunnet M, Jespersen T, Perrier JF (2004). 5-HT1A receptors

modulate small-conductance Ca2+-activated K+ channels.

Journal of Neuroscience Research 78, 845–854.

Grunschlag CR, Haas HL, Stevens DR (1997). 5-HT inhibits

lateral entorhinal cortical neurons of the rat in vitro by

activation of potassium channel-coupled 5-HT1A

receptors. Brain Research 770, 10–17.

Guiard BP, Froger N, Hamon M, Gardier AM, Lanfumey L

(2005). Sustained pharmacological blockade of NK1

substance P receptors causes functional desensitization of

dorsal raphe 5-HT1A autoreceptors in mice. Journal of

Neurochemistry 95, 1713–1723.

Haddjeri N, Blier P, de Montigny C (1998). Long-term

antidepressant treatments result in a tonic activation of

forebrain 5-HT1A receptors. Journal of Neuroscience 18,

10150–10156.

Haddjeri N, Szabo ST, de Montigny C, Blier P (2000).

Increased tonic activation of rat forebrain 5-HT(1A)

receptors by lithium addition to antidepressant treatments.

Neuropsychopharmacology 22, 346–356.

Haleem DJ (2006). Serotonergic modulation of dopamine

neurotransmission: a mechanism for enhancing

therapeutics in schizophrenia. Journal of the College of

Physicians and Surgeons of Pakistan 16, 556–562.

HapMap P (2003). The International HapMap Project. Nature

426, 789–796.

Heisler LK, Chu HM, Brennan TJ, Danao JA, Bajwa P,

Parsons LH, Tecott LH (1998). Elevated anxiety and

antidepressant-like responses in serotonin 5-HT1A

receptor mutant mice. Proceedings of the National Academy

of Sciences USA 95, 15049–15054.

716 A. Drago et al.

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 17: 5-HT1A gene variants and psychiatric disorders: a review of current

Hjorth S, Bengtsson HJ, Kullberg A, Carlzon D, Peilot H,

Auerbach SB (2000). Serotonin autoreceptor function and

antidepressant drug action. Journal of Psychopharmacology

14, 177–185.

Homer JP, Flodman PL, SpenceMA (1997). Bipolar disorder:

dominant or recessive on chromosome 5? Genetic

Epidemiology 14, 647–651.

Hong CJ, Chen TJ, Yu YW, Tsai SJ (2006). Response to

fluoxetine and serotonin 1A receptor (C-1019G)

polymorphism in Taiwan Chinese major depressive

disorder. Pharmacogenomics Journal 6, 27–33.

Hranilovic D, Stefulj J, Schwab S, Borrmann-Hassenbach

M, Albus M, Jernej B, Wildenauer D (2004). Serotonin

transporter promoter and intron 2 polymorphisms:

relationship between allelic variants and gene expression.

Biological Psychiatry 55, 1090–1094.

Hsiung SC, Adlersberg M, Arango V, Mann JJ, Tamir H,

Liu KP (2003). Attenuated 5-HT1A receptor signaling

in brains of suicide victims: involvement of adenylyl

cyclase, phosphatidylinositol 3-kinase, Akt and

mitogen-activated protein kinase. Journal of Neurochemistry

87, 182–194.

Huang YY, Battistuzzi C, Oquendo MA, Harkavy-Friedman

J, Greenhill L, Zalsman G, Brodsky B, Arango V, Brent

DA, Mann JJ (2004). Human 5-HT1A receptor C(-1019)G

polymorphism and psychopathology. International Journal

of Neuropsychopharmacology 7, 441–451.

Ichikawa J, Ishii H, Bonaccorso S, Fowler WL, O’Laughlin

IA, Meltzer HY (2001). 5-HT(2A) and D(2) receptor

blockade increases cortical DA release via 5-HT(1A)

receptor activation: a possible mechanism of atypical

antipsychotic-induced cortical dopamine release. Journal of

Neurochemistry 76, 1521–1531.

Jang YL, Kim JW, Lee YS, ParkDY, Cho EY, JeunHO, Lee D,

Hong KS (2007). Linkage of schizophrenia with

chromosome 1q32 in Korean multiplex families. American

Journal of Medical Genetics. Part B: Neuropsychiatric Genetics

144, 279–284.

Joels M, Shinnick-Gallagher P, Gallagher JP (1987). Effect of

serotonin and serotonin analogues on passive membrane

properties of lateral septal neurons in vitro. Brain Research

417, 99–107.

Kalipatnapu S, Chattopadhyay A (2005). Membrane protein

solubilization: recent advances and challenges in

solubilization of serotonin1A receptors. IUBMB Life 57,

505–512.

Kasper S, Tauscher J, Willeit M, Stamenkovic M,

Neumeister A, Kufferle B, Barnas C, Stastny J,

Praschak-Rieder N, Pezawas L, et al. (2002). Receptor and

transporter imaging studies in schizophrenia, depression,

bulimia and Tourette’s disorder–implications for

psychopharmacology. World Journal of Biological Psychiatry

3, 133–146.

Kawanishi Y, Harada S, Tachikawa H, Okubo T, Shiraishi

H (1998). Novel mutations in the promoter and coding

region of the human 5-HT1A receptor gene and association

analysis in schizophrenia. American Journal of Medical

Genetics 81, 434–439.

Kendler KS (2005). ‘A gene for …’: the nature of gene action

in psychiatric disorders. American Journal of Psychiatry

162, 1243–1252.

Kerner B, Brugman DL, Freimer NB (2007). Evidence of

linkage to psychosis on chromosome 5q33–34 in pedigrees

ascertained for bipolar disorder.American Journal of Medical

Genetics. Part B: Neuropsychiatric Genetics 144, 74–78.

Khaitan L, Calabrese JR, Stockmeier CA (1994). Effects of

chronic treatment with valproate on serotonin-1A receptor

binding and function. Psychopharmacology (Berlin) 113,

539–542.

Knobelman DA, Hen R, Lucki I (2001). Genetic regulation of

extracellular serotonin by 5-hydroxytryptamine(1A) and

5-hydroxytryptamine(1B) autoreceptors in different brain

regions of the mouse. Journal of Pharmacology and

Experimental Therapeutics 298, 1083–1091.

Kraus MR, Al-Taie O, Schafer A, Pfersdorff M, Lesch KP,

Scheurlen M (2007). Serotonin-1A receptor gene HTR1A

variation predicts interferon-induced depression in

chronic hepatitis C. Gastroenterology 132, 1279–1286.

Lane HY, Liu YC, Huang CL, Chang YC, Wu PL, Lu CT,

Chang WH (2006). Risperidone-related weight gain:

genetic and nongenetic predictors. Journal of Clinical

Psychopharmacology 26, 128–134.

Lasky-Su JA, Faraone SV, Glatt SJ, Tsuang MT (2005).

Meta-analysis of the association between two

polymorphisms in the serotonin transporter gene and

affective disorders. American Journal of Medical Genetics.

Part B: Neuropsychiatric Genetics 133, 110–115.

Le Hellard S, Lee AJ, Underwood S, Thomson PA, Morris

SW, Torrance HS, Anderson SM, Adams RR, Navarro P,

Christoforou A, et al. (2007). Haplotype analysis and a

novel allele-sharing method refines a chromosome 4p

locus linked to bipolar affective disorder. Biological

Psychiatry 61, 797–805.

Lee MA, Meltzer HY (2001). 5-HT(1A) receptor dysfunction

in female patients with schizophrenia. Biological Psychiatry

50, 758–766.

Lemonde S, Du L, Bakish D, Hrdina P, Albert PR (2004).

Association of the C(-1019)G 5-HT1A functional promoter

polymorphism with antidepressant response. International

Journal of Neuropsychopharmacology 7, 501–506.

Lemonde S, Turecki G, Bakish D, Du L, Hrdina PD, Bown

CD, Sequeira A, Kushwaha N, Morris SJ, Basak A, et al.

(2003). Impaired repression at a 5-hydroxytryptamine 1A

receptor gene polymorphism associated with major

depression and suicide. Journal of Neuroscience 23,

8788–8799.

Lesch KP (2001). Variation of serotonergic gene expression:

neurodevelopment and the complexity of response to

psychopharmacologic drugs. European

Neuropsychopharmacology 11, 457–474.

Lesch KP, Gutknecht L (2004). Focus on The 5-HT1A

receptor: emerging role of a gene regulatory variant in

psychopathology and pharmacogenetics. International

Journal of Neuropsychopharmacology 7, 381–385.

Levin GM, Bowles TM, Ehret MJ, Langaee T, Tan JY,

Johnson JA, Millard WJ (2007). Assessment of

HTR1A and psychiatric disorders 717

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 18: 5-HT1A gene variants and psychiatric disorders: a review of current

human serotonin 1A receptor polymorphisms and

SSRI responsiveness. Molecular Diagnosis & Therapy

11, 155–560.

Lewis CM, Levinson DF, Wise LH, DeLisi LE, Straub RE,

Hovatta I, Williams NM, Schwab SG, Pulver AE, Faraone

SV, et al. (2003). Genome scan meta-analysis of

schizophrenia and bipolar disorder, Part II : Schizophrenia.

American Journal of Human Genetics 73, 17–33.

Li Q, Holmes A, Ma L, Van de Kar LD, Garcia F, Murphy

DL (2004). Medial hypothalamic 5-hydroxytryptamine

(5-HT)1A receptors regulate neuroendocrine responses to

stress and exploratory locomotor activity: application of

recombinant adenovirus containing 5-HT1A sequences.

Journal of Neuroscience 24, 10868–10877.

Li Q, Wichems C, Heils A, Lesch KP, Murphy DL (2000).

Reduction in the density and expression, but not G-protein

coupling, of serotonin receptors (5-HT1A) in 5-HT

transporter knock-out mice: gender and brain region

differences. Journal of Neuroscience 20, 7888–7895.

Liu R, Ding Y, Aghajanian GK (2002). Neurokinins activate

local glutamatergic inputs to serotonergic neurons of the

dorsal raphe nucleus. Neuropsychopharmacology 27,

329–340.

Liu YL, Fann CS, Liu CM, Chen WJ, Wu JY, Hung SI, Chen

CH, Jou YS, Liu SK, Hwang TJ, et al. (2006). A single

nucleotide polymorphism fine mapping study of

chromosome 1q42.1 reveals the vulnerability genes for

schizophrenia, GNPAT and DISC1: association with

impairment of sustained attention. Biological Psychiatry 60,

554–652.

Lopez JF, Chalmers DT, Little KY, Watson SJ (1998).

Regulation of serotonin1A, glucocorticoid, and

mineralocorticoid receptor in rat and human

hippocampus: implications for the neurobiology of

depression. Biological Psychiatry 43, 547–573.

Lopez JF, Liberzon I, Vazquez DM, Young EA, Watson SJ

(1999). Serotonin 1A receptor messenger RNA regulation

in the hippocampus after acute stress. Biological Psychiatry

45, 934–937.

Lopez-Figueroa AL, Norton CS, Lopez-Figueroa MO,

Armellini-Dodel D, Burke S, Akil H, Lopez JF, Watson SJ

(2004). Serotonin 5-HT1A, 5-HT1B, and 5-HT2A receptor

mRNA expression in subjects with major depression,

bipolar disorder, and schizophrenia. Biological Psychiatry

55, 225–233.

Lorenzi C, Serretti A, Mandelli L, Tubazio V, Ploia C,

Smeraldi E (2005). 5-HT 1A polymorphism and

self-transcendence in mood disorders. American Journal of

Medical Genetics. Part B: Neuropsychiatric Genetics 137,

33–35.

Mandelli L, Serretti A, Marino E, Pirovano A, Calati R,

Colombo C (2006). Interaction between serotonin

transporter gene, catechol-O-methyltransferase gene and

stressful life events in mood disorders. International Journal

of Neuropsychopharmacology 10, 437–447.

McQuade R, Leitch MM, Gartside SE, Young AH (2004).

Effect of chronic lithium treatment on glucocorticoid and

5-HT1A receptor messenger RNA in hippocampal and

dorsal raphe nucleus regions of the rat brain. Journal of

Psychopharmacology 18, 496–501.

Millan MJ, Newman-Tancredi A, Duqueyroix D, Cussac D

(2001). Agonist properties of pindolol at h5-HT1A

receptors coupled to mitogen-activated protein kinase.

European Journal of Pharmacology 424, 13–17.

Miquel MC, Kia HK, Boni C, Doucet E, Daval G,

Matthiessen L, Hamon M, Verge D (1994).

Postnatal development and localization of

5-HT1A receptor mRNA in rat forebrain and

cerebellum. Brain Research. Developmental Brain Research

80, 149–157.

Mirow AL, Kristbjanarson H, Egeland JA, Shilling P,

Helgason T, Gillin JC, Hirsch S, Kelsoe JR (1994). A

linkage study of distal chromosome 5q and bipolar

disorder. Biological Psychiatry 36, 223–229.

Nannya Y, Taura K, Kurokawa M, Ogawa S (2007).

Evaluation of genome-wide power of genetic association

studies based on empirical data from the HapMap Project.

Human Molecular Genetics 16, 3494–3505.

Neumeister A, Bain E, Nugent AC, Carson RE, Bonne O,

Luckenbaugh DA, Eckelman W, Herscovitch P, Charney

DS, Drevets WC (2004). Reduced serotonin type 1A

receptor binding in panic disorder. Journal of Neuroscience

24, 589–591.

Newberry NR (1992). 5-HT1A receptors activate a potassium

conductance in rat ventromedial hypothalamic neurones.

European Journal of Pharmacology 210, 209–212.

Newman-Tancredi A, Assie MB, Leduc N, Ormiere AM,

Danty N, Cosi C (2005). Novel antipsychotics activate

recombinant human and native rat serotonin 5-HT1A

receptors: affinity, efficacy and potential implications for

treatment of schizophrenia. International Journal of

Neuropsychopharmacology 8, 341–356.

Ni X, Valente J, Azevedo M, Pato M, Pato C, Kennedy JL

(2007). Connexin50 gene on human chromosome 1q21 is

associated with schizophrenia in matched case-control and

family- based studies. Journal of Medical Genetics 44,

532–536.

Nishiguchi N, Shirakawa O, Ono H, Nishimura A, Nushida

H, Ueno Y, Maeda K (2002). Lack of an association

between 5-HT1A receptor gene structural polymorphisms

and suicide victims. American Journal of Medical Genetics

114, 423–425.

Ohtani M, Shindo S, Yoshioka N (2004). Polymorphisms of

the tryptophan hydroxylase gene and serotonin 1A

receptor gene in suicide victims among Japanese. Tohoku

Journal of Experimental Medicine 202, 123–133.

Okuhara DY, Beck SG (1994). 5-HT1A receptor linked

to inward-rectifying potassium current in hippocampal

CA3 pyramidal cells. Journal of Neurophysiology 71,

2161–2167.

Olivier B, Pattij T, Wood SJ, Oosting R, Sarnyai Z, Toth M

(2001). The 5-HT(1A) receptor knockout mouse and

anxiety. Behavioral Pharmacology 12, 439–450.

Owen M, Craufurd D, St Clair D (1990). Localisation of a

susceptibility locus for schizophrenia on chromosome 5.

British Journal of Psychiatry 157, 123–127.

718 A. Drago et al.

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 19: 5-HT1A gene variants and psychiatric disorders: a review of current

Parks CL, Robinson PS, Sibille E, Shenk T, Toth M (1998).

Increased anxiety of mice lacking the serotonin1A receptor.

Proceedings of the National Academy of Sciences USA 95,

10734–10739.

Parsey RV, Olvet DM, Oquendo MA, Huang YY, Ogden

RT, Mann JJ (2006a). Higher 5-HT1A receptor binding

potential during a major depressive episode predicts poor

treatment response: preliminary data from a naturalistic

study. Neuropsychopharmacology 31, 1745–1749.

Parsey RV, Oquendo MA, Ogden RT, Olvet DM, Simpson

N, Huang YY, Van Heertum RL, Arango V, Mann JJ

(2006b). Altered serotonin 1A binding in major depression:

a [carbonyl-C-11]WAY100635 positron emission

tomography study. Biological Psychiatry 59, 106–113.

Parsey RV, Oquendo MA, Simpson NR, Ogden RT, Van

Heertum R, Arango V, Mann JJ (2002). Effects of sex, age,

and aggressive traits in man on brain serotonin 5-HT1A

receptor binding potential measured by PET using

[C-11]WAY-100635. Brain Research 954, 173–182.

Parsons LH, Kerr TM, Tecott LH (2001). 5-HT(1A) receptor

mutant mice exhibit enhanced tonic, stress-induced and

fluoxetine-induced serotonergic neurotransmission.

Journal of Neurochemistry 77, 607–617.

Pazos A, Cortes R, Palacios JM (1985). Quantitative

autoradiographic mapping of serotonin receptors in the rat

brain. II. Serotonin-2 receptors. Brain Research 346, 231–249.

Peroutka SJ, Howell TA (1994). The molecular evolution of G

protein-coupled receptors: focus on 5-hydroxytryptamine

receptors. Neuropharmacology 33, 319–324.

Peters EJ, Slager SL, McGrath PJ, Knowles JA, Hamilton SP

(2004). Investigation of serotonin-related genes in

antidepressant response. Molecular Psychiatry 9, 879–889.

Pucadyil TJ, Shrivastava S, Chattopadhyay A (2005).

Membrane cholesterol oxidation inhibits ligand binding

function of hippocampal serotonin(1A) receptors.

Biochemical and Biophysical Research Communications 331,

422–427.

Purcell S, Cherny SS, Sham P (2003). Genetic Power

Calculator: design of linkage and association genetic

mapping studies of complex traits. Bioinformatics 19,

149–150.

Purcell S, Sham P (2001). Genetic Power Calculator. Social,

Genetic & Developmental Research Centre, London.

Ramboz S, Oosting R, Amara DA, Kung HF, Blier P,

Mendelsohn M, Mann JJ, Brunner D, Hen R (1998).

Serotonin receptor 1A knockout: an animal model of

anxiety-related disorder. Proceedings of the National

Academy of Sciences USA 95, 14476–14481.

Read HL, Beck SG, Dun NJ (1994). Serotonergic suppression

of interhemispheric cortical synaptic potentials. Brain

Research 643, 17–28.

Reynolds GP, Arranz B, Templeman LA, Fertuzinhos S, San

L (2006). Effect of 5-HT1A receptor gene polymorphism on

negative and depressive symptom response to

antipsychotic treatment of drug-naive psychotic patients.

American Journal of Psychiatry 163, 1826–1829.

Richtand NM, Welge JA, Logue AD, Keck Jr. PE,

Strakowski SM, McNamara RK (2007). Dopamine and

serotonin receptor binding and antipsychotic efficacy.

Neuropsychopharmacology 32, 1715–1726.

Risch N, Merikangas K (1996). The future of genetic studies

of complex human diseases. Science 273, 1516–1517.

Rothe C, Gutknecht L, Freitag C, Tauber R, Mossner R,

Franke P, Fritze J, Wagner G, Peikert G, Wenda B, et al.

(2004). Association of a functional 1019C>G 5-HT1A

receptor gene polymorphism with panic disorder with

agoraphobia. International Journal of

Neuropsychopharmacology 7, 189–192.

Rotondo A, Nielsen DA, Nakhai B, Hulihan-Giblin B,

Bolos A, Goldman D (1997). Agonist-promoted down-

regulation and functional desensitization in two naturally

occurring variants of the human serotonin1A receptor.

Neuropsychopharmacology 17, 18–26.

Sato M, Ago Y, Koda K, Nakamura S, Kawasaki T, Baba A,

Matsuda T (2007). Role of postsynaptic serotonin(1A)

receptors in risperidone-induced increase in acetylcholine

release in rat prefrontal cortex. European Journal of

Pharmacology 559, 155–160.

Schmitz D, Empson RM, Heinemann U (1995). Serotonin

reduces inhibition via 5-HT1A receptors in area CA1 of rat

hippocampal slices in vitro. Journal of Neuroscience 15,

7217–7225.

Schmitz D, Gloveli T, Empson RM, Heinemann U (1998).

Serotonin reduces polysynaptic inhibition via 5-HT1A

receptors in the superficial entorhinal cortex. Journal of

Neurophysiology 80, 1116–1121.

Schosser A, Fuchs K, Scharl T, Leisch F, Bailer U, Kasper S,

Sieghart W, Hornik K, Aschauer HN (2007). Additional

support for linkage of schizophrenia and bipolar disorder

to chromosome 3q29. European Neuropsychopharmacology

17, 501–505.

Schumacher J, Kaneva R, Jamra RA, Diaz GO, Ohlraun S,

Milanova V, Lee YA, Rivas F, Mayoral F, Fuerst R, et al.

(2005). Genomewide scan and fine-mapping linkage

studies in four European samples with bipolar affective

disorder suggest a new susceptibility locus on

chromosome 1p35-p36 and provides further evidence of

loci on chromosome 4q31 and 6q24. American Journal of

Human Genetics 77, 1102–1111.

Scroggs RS, Anderson EG (1990). 5-HT1 receptor

agonists reduce the Ca+ component of sensory neuron

action potentials. European Journal of Pharmacology 178,

229–232.

Segal M (1990). Serotonin attenuates a slow inhibitory

postsynaptic potential in rat hippocampal neurons.

Neuroscience 36, 631–641.

Segal M, Azmitia EC, Whitaker-Azmitia PM (1989).

Physiological effects of selective 5-HT1a and 5-HT1b

ligands in rat hippocampus: comparison to 5-HT. Brain

Research 502, 67–74.

Segurado R, Detera-Wadleigh SD, Levinson DF, Lewis CM,

Gill M, Nurnberger Jr. JI, Craddock N, DePaulo JR,

Baron M, Gershon ES, et al. (2003). Genome scan

Meta-analysis of schizophrenia and bipolar disorder. Part

III : Bipolar disorder. American Journal of Human Genetics 73,

34–48.

HTR1A and psychiatric disorders 719

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 20: 5-HT1A gene variants and psychiatric disorders: a review of current

Serres F, Li Q, Garcia F, Raap DK, Battaglia G, Muma NA,

Van de Kar LD (2000). Evidence that G(z)-proteins couple

to hypothalamic 5-HT(1A) receptors in vivo. Journal of

Neuroscience 20, 3095–3103.

Serretti A, Artioli P, Lorenzi C, Pirovano A, Tubazio V,

Zanardi R (2004). The C(-1019)G polymorphism of the

5-HT1A gene promoter and antidepressant response in

mood disorders: preliminary findings. International Journal

of Neuropsychopharmacology 7, 453–460.

Serretti A, Kato M, Kennedy JL (in press). Pharmacogenetic

studies in depression: a proposal for methodologic

guidelines. Pharmacogenomics Journal.

Serretti A, Lilli R, Lorenzi C, Lattuada E, Smeraldi E

(2000). Serotonin-2C and serotonin-1A receptor genes

are not associated with psychotic symptomatology of

mood disorders. American Journal of Medical Genetics 96,

161–166.

Serretti A, Mandelli L, Giegling I, Schneider B, Hartmann

AM, Schnabel A, Maurer K, Moller HJ, Rujescu D (2006).

HTR2C and HTR1A gene variants in German and Italian

suicide attempters and completers. American Journal of

Medical Genetics. Part B: Neuropsychiatric Genetics 144,

291–299.

Sprouse JS, Aghajanian GK (1988). Responses of

hippocampal pyramidal cells to putative serotonin 5-

HT1A and 5-HT1B agonists: a comparative study with

dorsal raphe neurons. Neuropharmacology 27, 707–715.

Stamford JA, Davidson C, McLaughlin DP, Hopwood SE

(2000). Control of dorsal raphe 5-HT function by multiple

5-HT(1) autoreceptors: parallel purposes or pointless

plurality? Trends in Neuroscience 23, 459–465.

Stark KL, Gross C, Richardson-Jones J, Zhuang X, Hen R

(2007). A novel conditional knockout strategy applied to

serotonin receptors. Handbook of Experimental Pharmacology

178, 347–363.

Strobel A, Gutknecht L, Rothe C, Reif A, Mossner R, Zeng

Y, Brocke B, Lesch KP (2003). Allelic variation in 5-HT1A

receptor expression is associated with anxiety- and

depression-related personality traits. Journal of Neural

Transmission 110, 1445–1453.

Sullivan NR, Crane JW, Damjanoska KJ, Carrasco GA,

D’Souza DN, Garcia F, Van de Kar LD (2005).

Tandospirone activates neuroendocrine and ERK (MAP

kinase) signaling pathways specifically through 5-HT1A

receptor mechanisms in vivo. Naunyn-Schmiedeberg’s

Archives of Pharmacology 371, 18–26.

Sumiyoshi T, Matsui M, Yamashita I, Nohara S, Uehara T,

Kurachi M, Meltzer HY (2000). Effect of adjunctive

treatment with serotonin-1A agonist tandospirone on

memory functions in schizophrenia. Journal of Clinical

Psychopharmacology 20, 386–388.

Sumiyoshi T, Meltzer HY (2004). Serotonin 1A receptors in

memory function. American Journal of Psychiatry 161, 1505.

(author reply 1505–1506).

Suzuki Y, Sawamura K, Someya T (2004). The effects of a

5-hydroxytryptamine 1A receptor gene polymorphism on

the clinical response to fluvoxamine in depressed patients.

Pharmacogenomics Journal 4, 283–286.

Tamminga CA (2006). The neurobiology of cognition in

schizophrenia. Journal of Clinical Psychiatry 67, e11.

Tauscher J, Bagby RM, Javanmard M, Christensen BK,

Kasper S, Kapur S (2001). Inverse relationship between

serotonin 5-HT(1A) receptor binding and anxiety: a

[(11)C]WAY-100635 PET investigation in healthy

volunteers. American Journal of Psychiatry 158, 1326–1328.

Tauscher J, Kapur S, Verhoeff NP, Hussey DF, Daskalakis

ZJ, Tauscher-Wisniewski S, Wilson AA, Houle S, Kasper

S, Zipursky RB (2002). Brain serotonin 5-HT(1A) receptor

binding in schizophrenia measured by positron emission

tomography and [11C]WAY-100635. Archives of General

Psychiatry 59, 514–520.

Thakkar MM, Strecker RE, McCarley RW (1998). Behavioral

state control through differential serotonergic inhibition in

the mesopontine cholinergic nuclei : a simultaneous unit

recording and microdialysis study. Journal of Neuroscience

18, 5490–5497.

Toth M (2003). 5-HT1A receptor knockout mouse as a genetic

model of anxiety. European Journal of Pharmacology 463,

177–184.

Turner JH, Garnovskaya MN, Coaxum SD, Vlasova TM,

Yakutovich M, Lefler DM, Raymond JR (2007). Ca2+-

calmodulin and janus kinase 2 are required for activation

of sodium-proton exchange by the Gi-coupled

5-hydroxytryptamine 1a receptor. Journal of Pharmacology

and Experimental Therapeutics 320, 314–322.

TurnerMR, Rabiner EA, Hammers A, Al-Chalabi A, Grasby

PM, Shaw CE, Brooks DJ, Leigh PN (2005). [11C]-

WAY100635 PET demonstrates marked 5-HT1A receptor

changes in sporadic ALS. Brain 128, 896–905.

Underwood SL, Christoforou A, Thomson PA, Wray NR,

Tenesa A, Whittaker J, Adams RA, Le Hellard S,

Morris SW, Blackwood DH, et al. (2006). Association

analysis of the chromosome 4p-located G

protein-coupled receptor 78 (GPR78) gene in bipolar

affective disorder and schizophrenia. Molecular Psychiatry

11, 384–394.

Van den Hooff P, Galvan M (1992). Actions of 5-

hydroxytryptamine and 5-HT1A receptor ligands on rat

dorso-lateral septal neurones in vitro. British Journal of

Pharmacology 106, 893–899.

Van den Hove DL, Lauder JM, Scheepens A, Prickaerts J,

Blanco CE, Steinbusch HW (2006). Prenatal stress in the

rat alters 5-HT1A receptor binding in the ventral

hippocampus. Brain Research 1090, 29–34.

Videtic A, Pungercic G, Pajnic IZ, Zupanc T, Balazic J,

Tomori M, Komel R (2006). Association study of seven

polymorphisms in four serotonin receptor genes on suicide

victims. American Journal of Medical Genetics. Part B:

Neuropsychiatric Genetics 141, 669–672.

Wasserman D, Geijer T, Sokolowski M, Rozanov V,

Wasserman J (2006). The serotonin 1A receptor C(-1019)G

polymorphism in relation to suicide attempt. Behavioral and

Brain Functions 2, 14.

Wellcome Trust CCC (2007). Genome-wide association study

of 14,000 cases of seven common diseases and 3,000 shared

controls. Nature 447, 661–678.

720 A. Drago et al.

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018

Page 21: 5-HT1A gene variants and psychiatric disorders: a review of current

Wu S, Comings DE (1999). A common C-1018G

polymorphism in the human 5-HT1A receptor gene.

Psychiatric Genetics 9, 105–106.

YasunoF, SuharaT, IchimiyaT, TakanoA,AndoT,OkuboY

(2004). Decreased 5-HT1A receptor binding in amygdala of

schizophrenia. Biological Psychiatry 55, 439–444.

Yu YW, Tsai SJ, Liou YJ, Hong CJ, Chen TJ (2006).

Association study of two serotonin 1A receptor gene

polymorphisms and fluoxetine treatment response in

Chinese major depressive disorders. European

Neuropsychopharmacology 16, 498–503.

Yuen EY, Jiang Q, Chen P, Gu Z, Feng J, Yan Z (2005).

Serotonin 5-HT1A receptors regulate NMDA receptor

channels through a microtubule-dependent mechanism.

Journal of Neuroscience 25, 5488–5501.

Zgombick JM, Beck SG, Mahle CD, Craddock-Royal B,

Maayani S (1989). Pertussis toxin-sensitive guanine

nucleotide-binding protein(S) couple adenosine A1 and

5-hydroxytryptamine1A receptors to the same effector

systems in rat hippocampus: biochemical and

electrophysiological studies. Molecular Pharmacology 35,

484–494.

HTR1A and psychiatric disorders 721

Downloaded from https://academic.oup.com/ijnp/article-abstract/11/5/701/968740by gueston 01 February 2018