Elsevier

Neuropharmacology

Volume 65, February 2013, Pages 213-222
Neuropharmacology

Adolescent male rats are less sensitive than adults to the anxiogenic and serotonin-releasing effects of fenfluramine

https://doi.org/10.1016/j.neuropharm.2012.10.010Get rights and content

Abstract

Risk taking behavior increases during adolescence, which is also a critical period for the onset of drug abuse. The central serotonergic system matures during the adolescent period, and its immaturity during early adolescence may contribute to adolescent risk taking, as deficits in central serotonergic function have been associated with impulsivity, aggression, and risk taking. We investigated serotonergic modulation of behavior and presynaptic serotonergic function in adult (67–74 days old) and adolescent (28–34 days old) male rats. Fenfluramine (2 mg/kg, i.p.) produced greater anxiogenic effects in adult rats in both the light/dark and elevated plus maze tests for anxiety-like behavior, and stimulated greater increases in extracellular serotonin in the adult medial prefrontal cortex (mPFC) (1, 2.5, and 10 mg/kg, i.p.). Local infusion of 100 mM potassium chloride into the mPFC also stimulated greater serotonin efflux in adult rats. Adult rats had higher tissue serotonin content than adolescents in the prefrontal cortex, amygdala, and hippocampus, but the rate of serotonin synthesis was similar between age groups. Serotonin transporter (SERT) immunoreactivity and SERT radioligand binding were comparable between age groups in all three brain regions. These data suggest that lower tissue serotonin stores in adolescents limit fenfluramine-stimulated serotonin release and so contribute to the lesser anxiogenic effects of fenfluramine.

Highlights

► Fenfluramine was more anxiogenic to adult male rats than adolescents. ► Adolescents had lower fenfluramine-stimulated cortical serotonin release. ► Lower tissue serotonin stores may limit the effects of fenfluramine in adolescents. ► Serotonin-releasing drugs of abuse could also be less anxiogenic to adolescents.

Introduction

Adolescence is the period of transition from childhood to adulthood (Spear, 2000). This transitional period includes the time from around 12 to 18 years of age in humans (reviewed in Spear, 2000). In rodents, adolescence encompasses postnatal days 28–42 (PN28–42), though adulthood is not considered to begin until around PN60 (reviewed in McCutcheon and Marinelli, 2009; Spear, 2000). Behavior changes during adolescence, with risk taking, novelty seeking, and social behavior expressed at higher levels than in childhood or adulthood (Stansfield and Kirstein, 2006; Steinberg et al., 2008, 2009; reviewed in Spear, 2000). Impulsive, risk taking behavior is part of normal development, but also contributes to major causes of adolescent injury and mortality such as reckless driving, suicide, unsafe sexual behavior, and experimentation with drugs (Chen and Kandel, 1995; Eaton et al., 2010; SAMHSA, 2011; Steinberg, 2008; reviewed in Spear, 2000).

Immature function of the neural circuits that mediate goal directed behavior contributes to adolescent risk taking. The balance between systems mediating approach to rewarding stimuli and avoidance of aversive stimuli may be biased toward approach during adolescence (reviewed in Ernst and Fudge, 2009; Ernst et al., 2006). Prefrontal cortical regulation of limbic brain regions is immature, limiting the regulation of these approach and avoidance drives (reviewed in Casey et al., 2011; Ernst and Fudge, 2009; Ernst et al., 2006; Steinberg, 2010; Sturman and Moghaddam, 2011). Immaturity of dopaminergic and serotonergic function in the forebrain may also contribute to this approach/avoidance imbalance in adolescents (reviewed in Chambers et al., 2003; Crews et al., 2007; Ernst et al., 2006). Serotonin is an important mediator of behavioral inhibition in response to aversive situations, and low central serotonergic function has been associated with risk taking, impulsivity, and aggression (Brown et al., 1979; Crockett et al., 2009; Higley and Linnoila, 1997; Higley et al., 1996; Mehlman et al., 1994; Soubrie, 1986; Virkkunen et al., 1995). Serotonin also contributes to the aversive effects of some drugs of abuse, and adolescents are less sensitive to aversive effects of drugs in animal models (Ettenberg and Bernardi, 2006, 2007; Ettenberg et al., 2011; Infurna and Spear, 1979; Jones et al., 2009, 2010; Rocha et al., 2002; Schramm-Sapyta et al., 2006; Serafine and Riley, 2010). Lower serotonergic function in adolescents could therefore contribute to increased risk taking behavior and reduce the aversive effects of drugs of abuse. These effects could factor into the increased experimentation with drugs seen during adolescence (Chen and Kandel, 1995; SAMHSA, 2011).

Animal studies suggest that forebrain serotonergic function during early adolescence may be lower than in adults, especially in the cortex. While serotonin receptor expression, dorsal raphe firing rates, and the anatomic pattern of serotonergic innervation appear to be mature by adolescence, neurochemical markers of presynaptic serotonergic function increase between adolescence and adulthood (Beique et al., 2004; Daval et al., 1987; Garcia-Alcocer et al., 2006; Lanfumey and Jacobs, 1982; Lidov and Molliver, 1982; Miquel et al., 1994; Pranzatelli and Galvan, 1994; Vizuete et al., 1997; Waeber et al., 1996, 1994). Serotonin transporter (SERT) binding is lower in the cortex of early adolescent rats (PN28–35), and some studies show lower SERT binding in subcortical regions such as the amygdala and striatum (Dao et al., 2011; Galineau et al., 2004; Moll et al., 2000; Tarazi et al., 1998). Serotonin tissue content and synaptosomal uptake are also lower in the cortex and striatum of early adolescent rats compared to adults (Kirksey and Slotkin, 1979; Loizou, 1972; Loizou and Salt, 1970; Mercugliano et al., 1996). Studies during later adolescence have reported more adult-like serotonin function. Older adolescent rats (PN45–50) and adults have similar baseline and methamphetamine-stimulated extracellular serotonin in medial prefrontal cortex (mPFC) (Staiti et al., 2011). SERT binding and serotonin uptake are also mature by this age (Kirksey and Slotkin, 1979; Tarazi et al., 1998). These studies show that neurochemical markers of presynaptic serotonergic function reach adult levels during the adolescent period, and that these markers are lower than in adults during early adolescence from PN28 to PN35.

While the ontogeny of forebrain serotonergic innervation has been described in young adolescents, serotonin release in response to pharmacologic or behavioral stimuli has not been evaluated over this critical developmental window. Additionally, the ability of serotonergic drugs to influence some behaviors has been studied in adolescents of several species, but a direct comparison of serotonergic regulation of behavioral inhibition in adolescents and adults has not been reported (Higley et al., 1996; LeMarquand et al., 1998; Mehlman et al., 1994; Zepf et al., 2008). The purpose of this study is to address this gap in our understanding of how serotonin regulates behavior during adolescence by assessing behavioral and neurochemical responses to pharmacologic challenge of the serotonergic system in adults (PN67–74) and early adolescents (PN28–34). We used the serotonin-releasing drug fenfluramine to compare the ability to mobilize serotonin stores in adults and early adolescents. Fenfluramine mobilizes serotonin stores by a similar mechanism as drugs of abuse such as methamphetamine and methylenedioxymethamphetamine (MDMA), but is more selective for the serotonergic system than these drugs (Rothman et al., 2001; reviewed in Sulzer et al., 2005). The behavioral effects of fenfluramine treatment were evaluated in the light/dark (LD) and the elevated plus maze (EPM) tests for anxiety-like behavior. Unconditioned tests for anxiety-like behavior such as the LD and EPM are thought be an ethologically relevant model of risk taking behavior because they measure inhibition of species-typical behaviors in novel, aversive, and potentially risky environments (Harro, 2002; Macri et al., 2002; Olausson et al., 1999). We then used microdialysis to assess the effect of fenfluramine and potassium on extracellular serotonin in the mPFC, and further investigated presynaptic serotonin function by measuring serotonin content, synthesis, innervation density, and SERT levels in the prefrontal cortex, amygdala, and hippocampus.

Section snippets

Animals

Young adult (PN60–63) and juvenile (PN21) male Sprague–Dawley (CD) rats were purchased from Charles River Laboratories (Raleigh, NC). The rats were housed in ventilated plastic cages (Techniplast USA, Exton, PA) or standard rat cages (Allentown Caging, Allentown, NJ) with corn cob bedding on a 12:12 h light/dark cycle with lights on at 06:00 and lights off at 18:00. All rats were allowed to acclimate to our AALAC accredited facility for at least 7 days before behavior testing or collection of

Light/dark test

The time spent in the light compartment and the latency to emerge into the light were used as measures of anxiety-like behavior, and the total distance traveled was used to assess locomotion (Morley et al., 2005; Schramm-Sapyta et al., 2007). Fenfluramine increased anxiety-like behavior in both ages (n = 12 per experimental group) as shown by a reduction in time spent in the light compartment (Fig. 2A) [main effect of Treatment, F(1,39) = 77.92, p < 0.001]. This effect was significantly greater

Discussion

This study shows that adolescent male rats are less sensitive than adult males to the anxiogenic effects of the serotonin-releasing drug fenfluramine, and produces neurochemical data which suggest this behavioral effect may result from lower fenfluramine-stimulated serotonin release in adolescents. Fenfluramine did not induce anxiety-like behavior as effectively in adolescent rats as in adults in either the LD test or EPM. The anxiogenic effects of fenfluramine in adult rats are consistent with

Acknowledgment

This work was supported by National Institute on Drug Abuse grants DA019114 and 1F31DA032532. The authors wish to thank Sam Johnson and the Duke Light Microscopy Core Facility for technical help with imaging and Jacob Jacobsen for technical help with microdialysis.

References (119)

  • 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)
  • D. Drapier et al.

    Effects of acute fluoxetine, paroxetine and desipramine on rats tested on the elevated plus-maze

    Behav. Brain Res.

    (2007)
  • M. Ernst et al.

    A developmental neurobiological model of motivated behavior: anatomy, connectivity and ontogeny of the triadic nodes

    Neurosci. Biobehav. Rev.

    (2009)
  • A. Ettenberg et al.

    Anxiolytic-like actions of buspirone in a runway model of intravenous cocaine self-administration

    Pharmacol. Biochem. Behav.

    (2006)
  • A. Ettenberg et al.

    Effects of buspirone on the immediate positive and delayed negative properties of intravenous cocaine as measured in the conditioned place preference test

    Pharmacol. Biochem. Behav.

    (2007)
  • A. Ettenberg et al.

    Inactivation of the dorsal raphe nucleus reduces the anxiogenic response of rats running an alley for intravenous cocaine

    Pharmacol. Biochem. Behav.

    (2011)
  • L. Galineau et al.

    Ontogeny of the dopamine and serotonin transporters in the rat brain: an autoradiographic study

    Neurosci. Lett.

    (2004)
  • K.S. Gomes et al.

    Contrasting effects of acute and chronic treatment with imipramine and fluoxetine on inhibitory avoidance and escape responses in mice exposed to the elevated T-maze

    Brain Res. Bull.

    (2009)
  • F.G. Graeff et al.

    Role of 5-HT in stress, anxiety, and depression

    Pharmacol. Biochem. Behav.

    (1996)
  • G. Griebel

    5-Hydroxytryptamine-interacting drugs in animal models of anxiety disorders: more than 30 years of research

    Pharmacol. Ther.

    (1995)
  • T.G. Heffner et al.

    A rapid method for the regional dissection of the rat brain

    Pharmacol. Biochem. Behav.

    (1980)
  • R.N. Infurna et al.

    Developmental changes in amphetamine-induced taste aversions

    Pharmacol. Biochem. Behav.

    (1979)
  • J.D. Jones et al.

    Differential involvement of the norepinephrine, serotonin and dopamine reuptake transporter proteins in cocaine-induced taste aversion

    Pharmacol. Biochem. Behav.

    (2009)
  • J.D. Jones et al.

    Dopamine, norepinephrine and serotonin transporter gene deletions differentially alter cocaine-induced taste aversion

    Pharmacol. Biochem. Behav.

    (2010)
  • L. Lanfumey et al.

    Developmental analysis of raphe dorsalis unit activity in the rat

    Brain Res.

    (1982)
  • D.G. LeMarquand et al.

    Tryptophan depletion, executive functions, and disinhibition in aggressive, adolescent males

    Neuropsychopharmacology

    (1998)
  • H.G. Lidov et al.

    Immunohistochemical study of the development of serotonergic neurons in the rat CNS

    Brain Res. Bull.

    (1982)
  • L.A. Loizou

    The postnatal ontogeny of monoamine-containing neurones in the central nervous system of the albino rat

    Brain Res.

    (1972)
  • L.A. Loizou et al.

    Regional changes in monoamines of the rat brain during postnatal development

    Brain Res.

    (1970)
  • W.H. Lyness et al.

    Destruction of dopaminergic nerve terminals in nucleus accumbens: effect on d-amphetamine self-administration

    Pharmacol. Biochem. Behav.

    (1979)
  • W.H. Lyness et al.

    Increased self-administration of d-amphetamine after destruction of 5-hydroxytryptaminergic neurons

    Pharmacol. Biochem. Behav.

    (1980)
  • P.D. Mabry et al.

    Serotonergic inhibition of catecholamine-induced behavioral arousal

    Brain Res.

    (1973)
  • S. Macri et al.

    Risk taking during exploration of a plus-maze is greater in adolescent than in juvenile or adult mice

    Anim. Behav.

    (2002)
  • B.A. McMillen

    CNS stimulants: two distinct mechanisms of action for amphetamine-like drugs

    Trends Pharmacol. Sci.

    (1983)
  • M. Mercugliano et al.

    Developmental alterations in 5-hydroxytryptamine concentration and turnover after treatment of neonatal rats with 5,7-dihydroxytryptamine

    Neurobiol. Learn. Mem.

    (1996)
  • M.C. Miquel et al.

    Postnatal development and localization of 5-HT1A receptor mRNA in rat forebrain and cerebellum

    Brain Res. Dev. Brain Res.

    (1994)
  • G.H. Moll et al.

    Age-associated changes in the densities of presynaptic monoamine transporters in different regions of the rat brain from early juvenile life to late adulthood

    Brain Res. Dev. Brain Res.

    (2000)
  • K.C. Morley et al.

    Serotonin (1A) receptor involvement in acute 3,4-methylenedioxymethamphetamine (MDMA) facilitation of social interaction in the rat

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2005)
  • D.H. Park et al.

    Postnatal developmental changes of tryptophan hydroxylase activity in serotonergic cell bodies and terminals of rat brain

    Brain Res.

    (1986)
  • S. Pellow et al.

    Anxiolytic and anxiogenic drug effects on exploratory activity in an elevated plus-maze: a novel test of anxiety in the rat

    Pharmacol. Biochem. Behav.

    (1986)
  • M.R. Pranzatelli et al.

    Ontogeny of [125I]iodocyanopindolol-labelled 5-hydroxytryptamine1B-binding sites in the rat CNS

    Neurosci. Lett.

    (1994)
  • D.C. Roberts et al.

    On the role of ascending catecholaminergic systems in intravenous self-administration of cocaine

    Pharmacol. Biochem. Behav.

    (1977)
  • R. Rogerio et al.

    Anxiogenic properties of cocaine in the rat evaluated with the elevated plus-maze

    Pharmacol. Biochem. Behav.

    (1992)
  • N.L. Schramm-Sapyta et al.

    Adolescent rats are protected from the conditioned aversive properties of cocaine and lithium chloride

    Pharmacol. Biochem. Behav.

    (2006)
  • K.M. Serafine et al.

    Preexposure to cocaine attenuates aversions induced by both cocaine and fluoxetine: Implications for the basis of cocaine-induced conditioned taste aversions

    Pharmacol. Biochem. Behav.

    (2010)
  • L.P. Spear

    The adolescent brain and age-related behavioral manifestations

    Neurosci. Biobehav Rev.

    (2000)
  • A.M. Staiti et al.

    A microdialysis study of the medial prefrontal cortex of adolescent and adult rats

    Neuropharmacology

    (2011)
  • L. Steinberg

    A social neuroscience perspective on adolescent risk-taking

    Dev. Rev.

    (2008)
  • W. Adriani et al.

    Elevated levels of impulsivity and reduced place conditioning with d-amphetamine: two behavioral features of adolescence in mice

    Behav. Neurosci.

    (2003)
  • J.C. Beique et al.

    Serotonergic regulation of membrane potential in developing rat prefrontal cortex: coordinated expression of 5-hydroxytryptamine (5-HT)1A, 5-HT2A, and 5-HT7 receptors

    J. Neurosci.

    (2004)
  • Cited by (28)

    • Stressor during Early Adolescence in Hyperreactive Female Wistar Kyoto Rats Induces a ‘Double Hit’ Manifested by Variation in Neurobehaviors and Brain Monoamines

      2019, Neuroscience
      Citation Excerpt :

      The 5-HT system has been shown to respond variably to different intensities and durations of stressors (De La Garza and Mahoney, 2004). Moreover, the 5-HT system is still developing (Arrant et al., 2013) during early adolescence, so along with the flux in serotonergic activity, stress may have strengthened the metabolic variations observed here, particularly in mPFC, St and NAc regions. Since 5-HT levels correlate positively with immobility, the possibility of it being the substrate for behavioral despair and underlying restraint stress-induced neurobehaviors cannot be overlooked.

    • Early onset cigarette smokers exhibit greater P300 reactivity to smoking-related stimuli and report greater craving

      2018, Brain Research
      Citation Excerpt :

      To this end, preclinical findings have shown distinct differences in nicotine self-administration behaviors between adult rats treated with nicotine during either periadolescence or postadolescence (Adriani et al., 2003). Periadolescent rats treated with nicotine between postnatal days 34–43 (an age range comparable to ‘early onset’ or early adolescence (Arrant et al. 2013)), compared to postadolescent rats treated with nicotine between postnatal days 60–69 (an age range comparable to ‘late onset’ or late adolescence/early adulthood), exhibited greater motivation and higher rates of responding to self-administer nicotine when tested as adults (approximately postnatal day 105) (Adriani et al., 2003). Thus, periadolescent (e.g. ‘early onset’) exposure to nicotine can produce distinct sensitized responses to the salient appetitive properties of the drug that correspond to greater self-administration behavior and facilitate the transition to long-term drug dependence in adulthood (Morgan et al., 2006).

    View all citing articles on Scopus
    View full text