5-HT1A receptor function in major depressive disorder

https://doi.org/10.1016/j.pneurobio.2009.01.009Get rights and content

Abstract

Dysfunction of the serotonin 1A receptor (5-HT1A) may play a role in the genesis of major depressive disorder (MDD). Here we review the pharmacological, post-mortem, positron emission tomography (PET), and genetic evidence in support of this statement. We also touch briefly on two MDD-associated phenotypes, cognitive impairment and somatic pain. The results of pharmacological challenge studies with 5-HT1A receptor agonists are indicative of blunted endocrine responses in depressed patients. Lithium, valproate, selective serotonin reuptake inhibitors (SSRIs), tricyclic antidepressants (TCAs), and other treatment, such as electroconvulsive shock therapy (ECT), all increase post-synaptic 5-HT1A receptor signaling through either direct or indirect effects. Reduced somatodendritic and postsynaptic 5-HT1A receptor numbers or affinity have been reported in some post-mortem studies of suicide victims, a result consistent with well-replicated PET analyses demonstrating reduced 5-HT1A receptor binding potential in diverse regions such as the dorsal raphe, medial prefrontal cortex (mPFC), amygdala and hippocampus. 5-HT1A receptor knockout (KO) mice display increased anxiety-related behavior, which, unlike in their wild-type counterparts, cannot be rescued with antidepressant drug (AD) treatment. In humans, the G allele of a single nucleotide polymorphism (SNP) in the 5-HT1A receptor gene (HTR1A; rs6295), which abrogates a transcription factor binding site for deformed epidermal autoregulatory factor-1 (Deaf-1) and Hes5, has been reported to be over-represented in MDD cases. Conversely, the C allele has been associated with better response to AD drugs. We raise the possibility that 5-HT1A receptor dysfunction represents one potential mechanism underpinning MDD and other stress-related disorders.

Introduction

The decreased levels of serotonin metabolites in cerebrospinal fluid (CSF), coupled with the mood-lowering effects of tryptophan depletion and the efficacy of serotonin-modulating antidepressants, have lent support to the notion that a dysfunctional serotonergic system is a vulnerability factor for major depressive disorder (MDD; or “unipolar depression”) and other forms of affective illness (Jans et al., 2006).

At least 14 different serotonin receptors have been identified (Hoyer et al., 2002). These receptors can be divided into distinct families – denoted 1, 2, 3, 4, 5, 6, and 7, with subtypes in each family denoted by letters such as a, b, and c. A number of these receptors may play a role in the genesis of psychiatric illness. For example, polymorphisms of the HTR2A gene [Entrez ID 3356], which codes for the 5-HT2A receptor, have been associated with major depressive illness (MDD) (Christiansen et al., 2007) and the efficacy of antidepressant drug (AD) treatment (McMahon et al., 2006). In addition, variants of the HTR3A gene [Entrez ID 3359], which codes for the A subunit of the 5-HT3 receptor, have been associated with bipolar disorder and schizophrenia (Niesler et al., 2008). This study will, however, focus on one of the best characterized of these receptors, the serotonin 1A receptor (5-HT1A), which is a key mediator of serotonergic signaling in the central nervous system.

Central serotonergic neurons are located in the raphe nuclei in the brain stem (Jans et al., 2006). The 5-HT1A receptor is a G-protein-coupled receptor widely distributed in regions that receive serotonergic input from the raphe nuclei: the frontal cortex, septum, amygdala, hippocampus, and hypothalamus (Lesch and Gutknecht, 2004, Sharp et al., 2007). In these cortico-limbic regions 5-HT1A is distributed post-synaptically (Celada et al., 2004, Sharp et al., 2007). The 5-HT1A receptor also serves as the predominant (somatodendritic) autoreceptor of the raphe nuclei, reducing the firing rate of these neurons, the amount of serotonin released per action potential, and the synthesis of the neurotransmitter; and thus by implication, the serotonergic activity of its projection areas (Wang and Aghajanian, 1977, Verge et al., 1985, Sprouse and Aghajanian, 1986, Blier and de Montigny, 1987, Hutson et al., 1989, Meller et al., 1990, Hjorth and Sharp, 1991, Kreiss and Lucki, 1994). Activation of the somatodendritic (i.e., presynaptic) 5-HT1A receptors may also indirectly reduce serotonergic transmission through the inhibition of tyrosine hydroxylase synthesis (reviewed in Drevets et al. (2007)), as well as an excitatory, glutamatergic pathway that originates in the medial prefrontal cortex (mPFC) and projects to the raphe nuclei (Sharp et al., 2007).

The 5-HT1A receptor not only contributes not only to the dynamic modulation of serotonergic activity impacting diverse functions such as cognition and emotion, but is thought to play a crucial role in the neuronal migration, neurite outgrowth and synapse formation inherent to the neurodevelopmental process (Whitaker-Azmitia et al., 1996). The 5-HT1A receptor is therefore a natural candidate for impacting an array of phenotypes, among them affective disorders.

There are four strands of evidence implicating the 5-HT1A receptor in affective illness, especially MDD: pharmacological challenge studies; post-mortem studies; neuroreceptor imaging, and genetic analyses. We will discuss each of these in turn.

Section snippets

Pharmacological challenge studies

The serotonin system has a facilitatory influence on cortisol, adrenocorticotrophic hormone (ACTH) and prolactin release. It also plays a role in the regulation of body temperature, and therefore a number of studies have evaluated 5-HT1A receptor function in depression through the administration of 5-HT1A agonists which elicit these endocrine and hypothermic responses. The data are difficult to interpret because known 5-HT1A agonists and antagonists bind to both somatodendritic and

Post-mortem analyses

An early post-mortem study reported reduced 5-HT1A receptor numbers and lowered receptor affinity in the hippocampi and amygdalae of depressed suicide victims, respectively [n = 19; six medicated with antidepressants (AD; Cheetham et al., 1990)]. Other post-mortem studies have also produced evidence for reduced 5-HT1A receptor gene expression, binding and/or numbers in the ventrolateral prefrontal cortex and the temporal polar cortex [all subjects received AD, tranquilizers or electroconvulsive

Positron emission tomography (PET) studies of receptor binding potential

In a [11C]WAY-100635 5-HT1A receptor antagonist positron emission tomography (PET) study, Drevets et al., 1999, Drevets et al., 2000 showed that unipolar and bipolar depressives with a familial form of illness exhibited reduced binding potential (BP) in the medial temporal cortex and hippocampus (25–33%), as well as the midbrain raphe (42%), compared with healthy controls. This result has been independently replicated: Sargent et al. (2000) reported a wide-spread reduction (frontal, temporal

Genetic studies

Serotonin transporter (SERT) KO mice show behavioral effects akin to depression in humans (Holmes et al., 2003, Lira et al., 2003, Wellman et al., 2007). Congruent with the PET imaging data, these SERT KO mice have also been reported to display a reduced density of 5-HT1A receptors in the hypothalamus, amygdala and dorsal raphe nucleus (Li et al., 2004). Similarly, an earlier study reported a desensitization of somatodendritic 5-HT1A receptors in the dorsal raphe nucleus, but not post-synaptic

MDD-associated phenotypes

The 5-HT1A receptor has been implicated in cognition and the regulation of pain perception. Since cognitive dysfunction and somatic symptoms are associated with mood disorders (Savitz et al., 2005, Vaccarino et al., 2008), understanding the impact of the 5-HT1A receptor on these two phenotypes may have relevance for MDD.

Long-term potentiation (LTP) is believed to be the neurobiological mechanism underlying learning and memory. At least one type of LTP is dependent on glutamatergic

Cause or effect?

It remains unclear whether the depression-related changes in 5-HT1A receptor binding discussed above are developmentally or genetically driven, or whether they are simply an adaptation to increased or decreased serotonergic neurotransmission. At least in the case of the dorsal raphe, receptor density is modified by pharmacological interventions. Acute administration of fluoxetine to rats may cause approximately one third of 5-HT1A receptors in the raphe to become internalized (Riad et al., 2001

Conclusion

The preclinical literature suggests that acute stress or administration of corticosteroids down-regulates or desensitizes both somatodendritic and postsynaptic 5-HT1A receptors (Fig. 1). This phenomenon may explain why MDD and other psychiatric conditions that have been associated with elevated secretion of cortisol show blunted endocrine or hormonal responses to 5-HT1A receptor agonists, reduced somatodendritic and postsynaptic 5-HT1A receptor numbers or binding at post-mortem, and reduced 5-HT

References (283)

  • A. Berney et al.

    An index of 5-HT synthesis changes during early antidepressant treatment: alpha-[(11)C]methyl-l-tryptophan PET study

    Neurochem. Int.

    (2008)
  • P. Blier et al.

    Serotonin 1A receptor activation and hypothermia in humans: lack of evidence for a presynaptic mediation

    Neuropsychopharmacology

    (2002)
  • P. Blier et al.

    Is there a role for 5-HT1A agonists in the treatment of depression?

    Biol. Psychiatry

    (2003)
  • M. Boldrini et al.

    Serotonin-1A autoreceptor binding in the dorsal raphe nucleus of depressed suicides

    J. Psychiatr. Res.

    (2008)
  • L. Christiansen et al.

    Candidate gene polymorphisms in the serotonergic pathway: influence on depression symptomatology in an elderly population

    Biol. Psychiatry

    (2007)
  • A.J. Cleare et al.

    Brain 5-HT1A receptor binding in chronic fatigue syndrome measured using positron emission tomography and [11C]WAY-100635

    Biol. Psychiatry

    (2005)
  • M.A. Cooper et al.

    Activation of 5-HT1A autoreceptors in the dorsal raphe nucleus reduces the behavioral consequences of social defeat

    Psychoneuroendocrinology

    (2008)
  • R.F. Cowburn et al.

    Adenylyl cyclase activity and G-protein subunit levels in postmortem frontal cortex of suicide victims

    Brain Res.

    (1994)
  • P.J. Cowen

    Psychopharmacology of 5-HT(1A) receptors(1)

    Nucl. Med. Biol.

    (2000)
  • G. Daval et al.

    Transient expression of 5-HT1A receptor binding sites in some areas of the rat CNS during postnatal development

    Int. J. Dev. Neurosci.

    (1987)
  • E. del Olmo et al.

    Aminergic receptors during the development of the human brain: the contribution of in vitro imaging techniques

    J. Chem. Neuroanat.

    (2001)
  • W.C. Drevets et al.

    Serotonin type-1A receptor imaging in depression

    Nucl. Med. Biol.

    (2000)
  • W.C. Drevets et al.

    PET imaging of serotonin 1A receptor binding in depression

    Biol. Psychiatry

    (1999)
  • W.C. Drevets et al.

    Serotonin-1A receptor imaging in recurrent depression: replication and literature review

    Nucl. Med. Biol.

    (2007)
  • R.S. Duman et al.

    A neurotrophic model for stress-related mood disorders

    Biol. Psychiatry

    (2006)
  • Y. Dwivedi et al.

    Protein kinase A in postmortem brain of depressed suicide victims: altered expression of specific regulatory and catalytic subunits

    Biol. Psychiatry

    (2004)
  • Y. Edagawa et al.

    5-HT1A receptor-mediated inhibition of long-term potentiation in rat visual cortex

    Eur. J. Pharmacol.

    (1998)
  • M. Elena Castro et al.

    Chronic fluoxetine induces opposite changes in G protein coupling at pre and postsynaptic 5-HT1A receptors in rat brain

    Neuropharmacology

    (2003)
  • G. Fairchild et al.

    Acute and chronic effects of corticosterone on 5-HT1A receptor-mediated autoinhibition in the rat dorsal raphe nucleus

    Neuropharmacology

    (2003)
  • S.E. File et al.

    Anxiolytic effects in the plus-maze of 5-HT1A-receptor ligands in dorsal raphe and ventral hippocampus

    Pharmacol. Biochem. Behav.

    (1996)
  • A.D. Fricker et al.

    Serotonin receptor activation leads to neurite outgrowth and neuronal survival

    Brain Res. Mol. Brain Res.

    (2005)
  • N. Galeotti et al.

    5-HT1A agonists induce central cholinergic antinociception

    Pharmacol. Biochem. Behav.

    (1997)
  • O. Gandolfi et al.

    The activation of serotonin receptors prevents glutamate-induced neurotoxicity and NMDA-stimulated cGMP accumulation in primary cortical cell cultures

    Pharmacol. Res.

    (2002)
  • C. Geretsegger et al.

    Paroxetine with pindolol augmentation: a double-blind, randomized, placebo-controlled study in depressed in-patients

    Eur. Neuropsychopharmacol.

    (2008)
  • J. Giordano et al.

    Putative mechanisms of buspirone-induced antinociception in the rat

    Pain

    (1992)
  • G. Giovacchini et al.

    Differential effects of paroxetine on raphe and cortical 5-HT1A binding: a PET study in monkeys

    Neuroimage

    (2005)
  • L. Gray et al.

    Serotonin 1a receptor and associated G-protein activation in schizophrenia and bipolar disorder

    Psychiatry Res.

    (2006)
  • W.C. Griffin et al.

    Prenatal stress influences 8-OH-DPAT modulated startle responding and [3H]-8-OH-DPAT binding in rats

    Pharmacol. Biochem. Behav.

    (2005)
  • C. Gross et al.

    Altered fear circuits in 5-HT(1A) receptor KO mice

    Biol. Psychiatry

    (2000)
  • O. Gureje et al.

    The relation between multiple pains and mental disorders: results from the World Mental Health Surveys

    Pain

    (2008)
  • T. Harkany et al.

    Oral post-lesion administration of 5-HT(1A) receptor agonist repinotan hydrochloride (BAY × 3702) attenuates NMDA-induced delayed neuronal death in rat magnocellular nucleus basalis

    Neuroscience

    (2001)
  • A. Harkin et al.

    Activity and onset of action of reboxetine and effect of combination with sertraline in an animal model of depression

    Eur. J. Pharmacol.

    (1999)
  • J.G. Hensler

    Differential regulation of 5-HT1A receptor-G protein interactions in brain following chronic antidepressant administration

    Neuropsychopharmacology

    (2002)
  • P. Albert et al.

    Increased DNA methylation of the 5-HT1A receptor promoter in suicide brain

    Int. J. Neuropsychopharmacol.

    (2008)
  • P.R. Albert et al.

    5-HT1A receptors, gene repression, and depression: guilt by association

    Neuroscientist

    (2004)
  • C. Alexandre et al.

    Early life blockade of 5-hydroxytryptamine 1A receptors normalizes sleep and depression-like behavior in adult knock-out mice lacking the serotonin transporter

    J. Neurosci.

    (2006)
  • H. Anisman et al.

    Serotonin receptor subtype and p11 mRNA expression in stress-relevant brain regions of suicide and control subjects

    J. Psychiatry Neurosci.

    (2008)
  • S. Anttila et al.

    Interaction between 5-HT1A and BDNF genotypes increases the risk of treatment-resistant depression

    J. Neural Transm.

    (2007)
  • B. Arias et al.

    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

    J. Psychopharmacol.

    (2005)
  • F. Artigas et al.

    Mechanism of action of antidepressants

    Psychopharmacol. Bull.

    (2002)
  • Cited by (481)

    • The bidirectional relationship of depression and disturbances in B cell homeostasis: Double trouble

      2024, Progress in Neuro-Psychopharmacology and Biological Psychiatry
    View all citing articles on Scopus
    View full text