Abstract
Studies using animal models of adolescent exposure to alcohol, nicotine, cannabinoids, and the stimulants cocaine, 3,4-Methylenedioxymethampethamine and methamphetamine have revealed a variety of persisting neural and behavioral consequences. Affected brain regions often include mesolimbic and prefrontal regions undergoing notable ontogenetic change during adolescence, although it is unclear whether this represents areas of specific vulnerability or particular scrutiny to date. Persisting alterations in forebrain systems critical for modulating reward, socioemotional processing and cognition have emerged, including apparent induction of a hyper-dopaminergic state with some drugs and/or attenuations in neurons expressing cholinergic markers. Disruptions in cognitive functions such as working memory, alterations in affect including increases in social anxiety, and mixed evidence for increases in later drug self-administration have also been reported. When consequences of adolescent and adult exposure were compared, adolescents were generally found to be more vulnerable to alcohol, nicotine, and cannabinoids, but generally not to stimulants. More work is needed to determine how adolescent drug exposure influences sculpting of the adolescent brain, and provide approaches to prevent/reverse these effects.
Keywords: Adolescent, Affect, Alcohol, Animal models, Brain, Cannabinoids, Cocaine, Cognition, Ethanol, MDMA, Methamphetamine, Nicotine, Rat
1. Introduction
Drug use is frequently initiated during adolescence. The latest Monitoring the Future survey reported that 13, 20 and 31% of 8th, 10th and 12th graders endorse having tried cigarettes, with over 10% of 12th graders affirming that they have smoked cigarettes within the past month (Johnston et al, 2016). Annual prevalence rates for use of alcohol and marijuana/hashish across these age ranges were 21, 42 and 58% and 12, 25 and 35%, respectively. Some of this use reaches high levels with, for instance, >10% of 12th grade students reporting consumption of 10 or more alcohol drinks in a row in the past 2 weeks, and >5% reporting consumption of >15 drinks per occasion over the same 2 week interval (Patrick et al, 2013). Illicit drugs other than marijuana used by adolescents include psychostimulants such as cocaine and the “club drugs” 3.4-methylenedioxymethamphetamine (MDMA, commonly called “ecstasy”) and methamphetamine (METH), with 2.5% of high school seniors in the United States reporting annual use of cocaine, 3.6% reporting MDMA use, and 0.6% endorsing use of METH (Johnston et al, 2016).
Among the likely contributors to the elevation of alcohol and drug use during adolescence are maturational changes occurring in the brain at this time – many of which are described in detail in other reviews in this special issue. Although notably oversimplified for the purposes here, prominent among these brain changes is a developmental dissociation between earlier maturation or even enhanced reactivity to rewarding and motivationally relevant stimuli in subcortical regions during adolescence that contrasts with relatively delayed maturation throughout adolescence of frontal brain regions that are critical for control of these subcortical regions (e.g., see Casey et al, 2011; Doremus-Fitzwater et al, 2010). That is, on the one hand, reactivity and cross-reactivity appears to be enhanced during adolescence in mesolimbic regions such as the nucleus accumbens (nAc) and amygdala (AMYG) that are critical for processing and responding to rewarding, aversive, and emotionally arousing stimuli, including social stimuli (e.g., Ernst & Fudge, 2009; Spear, 2011). This enhanced reactivity contrasts with delayed development of prefrontal cortex (PFC) and other frontal regions that mature only slowly during adolescence and are critical for cognitive control (Casey et al, 2011). These and other developmental changes occurring in the adolescent brain, such as developmental declines in cortical gray matter, and increases in myelination and in the portion of the brain partitioning as white matter, appear to have been highly conserved evolutionarily. That is, similar developmental alterations are evident during this ontogenetic transition across a variety of mammalian species, despite notable species differences in overall brain complexity and the relative length of the adolescent period (see Spear, 2000, 2016a, for review). For instance, in rats, the two week period between postnatal days (P) 28–42 is thought to roughly subsume the early-mid adolescent period (~ 12–17 years in humans), with the interval from ~ P43–55 more comparable to the late adolescence/emerging adulthood period in humans (~18–25 years) (see Spear, 2015). The conserved nature of the brain transformations of adolescence support the use of animal models to explore some of the critical questions regarding contributors to drug use and consequences of that use that may be challenging to study empirically and ethically in underage humans (see Spear, 2016a).
Alcohol and other drugs exert many of their effects through action in mesolimbic and frontal cortical regions undergoing particularly marked remodeling during adolescence. Hence, the maturational state of these regions could potentially influence the propensity for experimentation and continued drug use during adolescence. To the extent that initiation/escalation of alcohol/drug use during adolescence has a biological basis, and because the developmental timing is similar across species (see Spear, 2000; Spear, 2016a, for review), one would predict that enhanced propensities for drug initiation and elevated use seen in human adolescents would also be evident in adolescents of other species. Indeed, studies in laboratory animals have shown that voluntary self-administration of and sensitivity to the effects of alcohol and other drugs differs during adolescence from that seen in adulthood. Using alcohol as an example, adolescent rats (like their human counterparts) (SAMHSA, 2013) often drink 2–3 times more per drinking occasion than do adults (e.g., Doremus et al, 2005; Vetter et al, 2007; but see also Bell et al, 2006). Along with this elevation in intake, studies with adolescent rats have shown them to be less sensitive to many of the effects of alcohol (ethanol [EtOH]) that presumably serve as cues to limit intake (such as EtOH-induced motor impairing, sedative, aversive, and socially impairing effects), along with an enhanced sensitivity to desired effects of EtOH, including its rewarding and social facilitating effects (e.g. Spear et al, 2016b). Adolescents show an enhanced sensitivity to the rewarding effects not only of alcohol, but also nicotine and cocaine, with aversive effects attenuated during adolescence not only to alcohol, but also to nicotine, cocaine, amphetamine and delta-9-tetrahydrocannabinol ([THC] – one of the major cannabinoids in marijuana) (see Doremus-Fitzwater et al, 2010, for review). Collectively, these age-related propensities for accentuated appetitive and attenuated aversive properties of drugs could contribute to the enhanced susceptibility for initiation and escalation of drug use during adolescence.
That alcohol and drug use is often initiated (and sometimes escalates into high levels of use) during the time of the notable brain remodeling during adolescence leads to the question of whether drug exposure during this time may influence that development and exert persisting neural and behavioral alterations. For an extensive review of human imaging studies of adolescent alcohol and drug users, see Silveri et al in this special issue. Studies using animal models have also proven useful in examining this question empirically and with a rapidity of data accumulation that is often not possible in cross-sectional or time-consuming longitudinal studies with human adolescents (see Spear, 2016a, for review). From studies in laboratory animals, the evidence is mounting that indeed adolescent exposure to a variety of drugs influences neural, behavioral, affective and cognitive function long after adolescence. The present review will focus on studies in rodents and consequences of the three primary drugs used by adolescents – alcohol, nicotine, and marijuana/cannabinoids (CBs) – as well as cocaine and other stimulants, particularly MDMA and METH. Although not a focus of the current review, it should be noted that, to the extent that roughly comparable data are available in human research and studies with rodents, signs of across-species consiliences have emerged in the consequences of adolescent exposures to drugs such as alcohol, nicotine, and CBs (for reviews, see Spear, 2016a; Counotte et al, 2011a; Jager & Ramsey, 2008, respectively). These across-species consiliences are evident despite a major difference in exposure-to-test timing between investigations in humans and laboratory animals. In studies of drug use in human adolescents, drug-using individuals are typically assessed during adolescence or shortly thereafter, either while still using the drug or after a relatively short abstinence period. In contrast, lasting consequences of repeated drug exposure during adolescence in animal studies are typically examined in adulthood – well after exposure termination – rather than during or immediately following the exposure period.
In some, but not a majority, of the studies reviewed here, consequences of adolescent exposure were compared with equivalent exposure at some other point in life, most typically, adulthood, to assess whether adolescence is a particularly vulnerable period. As outlined in the sections to follow, for 3 of the 4 drug classes reviewed, when consequences of adolescent exposure were compared with exposure in adulthood (stimulants being the exception), effects were generally found to be more pronounced after exposures during adolescence. When considering differences in exposure age, it is important to note that there may be ontogenetic differences in pharmacokinetics that could influence differential drug vulnerabilities across age. Overall metabolic activity is typically greater in adolescence than in adulthood, and may extend to drugs of abuse as well (see Spear, 2000, for discussion and references). Thus, for those drugs where consequences of adolescent exposure are typically more pronounced than in adulthood, greater adverse consequences may be evident despite perhaps the tendency for these drugs to be metabolized more quickly during adolescence than in adulthood. In the case of stimulants where consequences of adolescent exposures may be attenuated relative to exposure in adulthood, potential age differences in metabolism are of particular concern, an issue that will be revisited later.
Note in the sections to follow that in some cases, studies examining neurobehavioral alterations after adolescent exposures have been complemented by pharmacological studies to assess the efficacy of manipulations of the altered neural system(s) for reversing observed deficits. These studies are not only crucial for testing hypotheses regarding neural contributors to observed consequences of adolescent exposure, but also may provide data of potential translational use for future prevention/intervention efforts.
Finally, although not a focus of this review, occasional examples of sex differences in the persisting effects of adolescent drug exposure are included. These sex-specific findings are consistent with evidence for notable sex differences in maturation of the brain during adolescence (see Viveros et al, 2011) as well as data showing “significant differences in the phenotypes of females and males in the domain of addiction” (Becker & Koob, 2016, p.243). It should be noted, however, that at this point the data are not plentiful enough for systematic characterization of sex differences in vulnerability to lasting effects of adolescent drug exposure, let alone for assessment of the neural substrates underlying these differential sex vulnerabilities (although progress in this area is beginning to emerge – e.g., see Viveros et al, 2011
2. Alcohol
There has been a rapid escalation of research using laboratory animals to investigate effects of repeated exposure to EtOH during adolescence. Evidence is building for a variety of lasting behavioral, cognitive, affective, and neural alterations after such exposure. These consequences have typically (although not always – e.g., see Schindler et al, 2014) been explored after adolescent exposures producing blood EtOH concentrations [BECs] above the threshold defining “binge” use (BECs > 80 mg%). In such studies, BECs often average around 150–200 mg% -- well within the range observed in field studies of late adolescents (e.g., Day et al, 2013), with little if any effort to date focused on determination of potential dose-dependency of effects. When studied, adolescent exposures to EtOH appear to induce more pronounced effects than equivalent EtOH exposures in adulthood, with some evidence to suggest that early-mid adolescence may be a time of particular vulnerability to lasting EtOH exposure effects (Spear, 2015).
2.1. Cognition, Behavior and Affect
Later effects on cognition after adolescent EtOH exposure are highly specific, with learning of a variety of less cognitive-challenging tasks largely unaffected (e.g., Popović et al, 2004; Risher et al, 2013). Reports of deficits on hippocampal-dependent tasks are mixed, with reports of performance decrements on trace conditioning (Yttri et al, 2004), spatial memory (Sircar & Sircar, 2005) and spatial-temporal object recognition (Swartzwelder et al, 2015) tasks contrasting with other findings of no alterations in spatial memory performance (Silvers et al, 2006; Acheson et al, 2013). Deficits have been suggested to be more likely to emerge on tasks tapping various “executive functions” (see Crews et al, 2007) such as conditional discrimination and object recognition tasks (Pascual et al, 2007). Task circumstances that demand some degree of cognitive and behavioral flexibility – i.e., the ability to modify behavior with changing environmental demands (see Hamilton & Brigman, 2015) – have been found to be particularly vulnerable to performance decrements after adolescent EtOH exposure, including reversal acquisition (Coleman et al, 2011), extinction, and set-shifting (Gass et al, 2014). Greater risk preferences in a probability discounting task have also been observed (Nasrallah et al, 2009; Boutros et al, 2014), along with signs interpreted to reflect increases in impulsivity and/or greater disinhibition (indexed, for example, via increased center time on an open field conflict test [Ehlers et al, 2011], more rears in the light side of a light-dark box [Desikan et al, 2014] and increased open arm time in an elevated plus maze under low light conditions [Gass et al, 2014]).
Alterations in affective measures have also been observed in adulthood after adolescent EtOH exposure. Depressive-like signs have emerged when indexed via decreases in sucrose consumption (used as a measure of anhedonia) or increased immobility in the Porsolt swim test (Slawecki et al, 2004; Ehlers et al, 2011; Briones & Woods, 2013). Reliable increases in social anxiety have been observed in adult males after EtOH exposure during early/mid adolescence, an effect that is sex-specific and not evident in females (Varlinskaya et al, 2014; Varlinskaya & Spear, 2015). Increases in general anxiety indexed via decreased open arm time in the elevated plus maze [EPM] (Slawecki et al, 2004; Montesinos et al, 2015a; Pandey et al, 2015) or increased defecation when in a novel environment (Popović et al, 2004) have also been reported in male rats, although these effects are not ubiquitous (Ehlers et al, 2011; Gilpin et al, 2012; Quoilin et al, 2012; White et al, 2000). Part of the challenge in interpreting data from EPM and similar tests of anxiety is that increased time in more “risky” environments (such as the open arms) could potentially reflect elevated “disinhibition” rather than an anxiolytic profile (see Desikan et al, 2014; Gass et al, 2014, for discussion). To the extent that adolescent EtOH exposure may be characterized by profiles both of increased anxiety as well as disinhibition, competition between these propensities might well lead to varied outcomes, depending on variables such as perceived stressfulness of the situation, prior handling or other test experiences.
2.2. Retention of adolescent phenotypes into adulthood
In a number of studies, adolescent EtOH exposure resulted in the retention of adolescent-typical phenotypes into adulthood (e.g., Fleming et al, 2012). Although these effects were sometimes evident in baseline behavior, cognition and electrophysiology, they appear particular pronounced in terms of the retention of adolescent-typical EtOH sensitivities into adulthood (see Spear & Swartzwelder, 2014, for review). For instance, adolescent EtOH exposure has been shown to disrupt normal developmental declines in sensitivity to EtOH-induced deficits in spatial working memory that typically occur between adolescence and adulthood; thus, adults exposed as adolescents to EtOH remain “adolescent-like” in being more vulnerable to EtOH-induced memory impairments than adults without a history of adolescent EtOH exposure (White et al, 2000). Adolescent-typical enhanced sensitivity to EtOH’s locomotor stimulant and rewarding effects were also retained into adulthood after adolescent EtOH exposure (Maldonado-Devincci et al, 2010; Quoilin et al, 2012; Toalston et al, 2014). Enhanced sensitivity to these desired effects of EtOH could serve to promote EtOH consumption in adulthood. Similar retentions of adolescent-like responses to EtOH after adolescent EtOH exposure are also evident in terms of adolescent-typical insensitivities to sedative, motor impairing, and aversive effects of EtOH (White et al, 2002; Diaz-Granados & Graham, 2007; Matthews et al, 2008; Quiolin et al, 2012; Saalfield & Spear, 2015). To the extent that these measures serve in part as feedback cues to moderate intake, retention of adolescent-typical insensitivities to these cues could serve to increase capacity for the maintenance of elevated drinking in adulthood after adolescent EtOH exposure. Later expression of adolescent-like phenotypes was specific to EtOH exposure during adolescence (and was not evident following comparable exposure in adulthood) (e.g., White et al, 2002; Diaz-Granados & Graham, 2007) and was more pronounced after episodic than continuous EtOH exposure (e.g., Diaz-Granados & Graham, 2007). The production of multiple “withdrawal” episodes of EtOH exposure and clearance associated with episodic exposure may be of particular importance, given that adolescents have been shown to be more sensitive than adults to escalation in withdrawal signs emerging over repeated withdrawals (Wills et al, 2008, 2009).
2.3. Later alcohol consumption
The data are mixed as to whether adolescent EtOH exposure increases later EtOH consumption in adulthood. Alcohol-preferring (P) rats given voluntary access to EtOH in their home cages throughout adolescence later acquired operant self-administration of ETOH more quickly than animals not given prior EtOH access; these animals also showed greater resistance to extinction of self-administration, more spontaneous recovery and more rapid reacquisition of the operant task than animals without a history of prior EtOH intake in adolescence (Rodd-Henricks et al, 2002a). None of these effects were evident when home cage EtOH access was not provided until adulthood (Rodd-Henricks et al, 2002b). Similarly, mice given voluntary access to EtOH in their home cages from weaning through adolescence and into adulthood consumed more EtOH than mice whose home cage access was delayed until adulthood (Ho et al, 1989). Intermittent intraperitoneal administration of EtOH during early-mid adolescence has been reported by two groups to increase later EtOH consumption in 2-bottle choice tests (Alaux-Cantin et al, 2013; Pandey et al, 2015), an effect that was not evident with comparable exposure late in adolescence (Alaux-Cantin et al, 2013).
Some studies, however, found no subsequent increases in consumption among rats exposed as adolescents to EtOH vapor (Slawecki & Bentacourt, 2002) or in mice or rats given voluntary access to EtOH in the home cage (Tambour et al, 2008; Vetter et al, 2007). In some cases, although adolescent-exposed animals did not differ from controls in their operant responding for EtOH in adulthood, they nevertheless demonstrated exacerbated dependence-induced increases in operant intake (Gilpin et al, 2012) or increased resistance to extinction of operant responding for EtOH (Gass et al, 2014). Critical variables that may influence whether later consumption is increased include sex, genetic background, amount and mode of adolescent EtOH exposure, and how intake is examined in adulthood (e.g., Blizard et al, 2004; Siciliano & Smith, 2001; Strong et al, 2010; Walker & Ehlers, 2009; Pascual et al, 2009). When rats were given voluntary access to sweetened EtOH or the sweetener alone during adolescence, animals increased their later intake of only the solution that they received as adolescents, suggesting that in some cases increases in later intake may be related to familiarity and enhanced solution acceptability rather than an effect specific to EtOH (Broadwater et al, 2013); such controls are rarely included. More remains to be learned regarding the constraints and circumstances that influence EtOH consumption in adulthood after adolescent exposure to EtOH.
2.4. Neural alterations
A variety of replicable and persisting alterations in brain have been observed after repeated EtOH exposure during adolescence in brain regions such as the PFC, hippocampus (HPC), AMYG and nAc that undergo notable developmental transformations during adolescence. Among the consistent findings reported after adolescent EtOH exposure are decreases in neurogenesis along with the induction of regionally-specific brain damage and cell death in regions such as the frontal cortex, HPC and cerebellum (Crews et al, 2000; Ehlers et al, 2013; Broadwater et al, 2014; Vetreno & Crews, 2015). Although binge-like levels of EtOH exposure can also induce brain damage in adult rats, the regions affected are different, with generally more pronounced effects evident after exposure during adolescence (Crews et al, 2000). A similar enhanced vulnerability of adolescents was also reported by Broadwater et al (2014) who observed lasting disruptions in neurogenesis and increases in cell death in adult rats after EtOH exposure during adolescence but not following equivalent exposure in adulthood. At the synaptic level, there is recent evidence that adolescent exposure to EtOH results in expression of a greater proportion of immature, more “plastic” synaptic spines in the adult HPC (Risher et al, 2015). Alterations in brain activity in this region were evident in terms of long-term potentiation (LTP), with adults exposed to EtOH during adolescence exhibiting more robust LTP than non-exposed animals, suggesting a surprising enhanced state of synaptic plasticity (Risher et al, 2015). Electrophysiological studies have revealed long-lasting decreases in slow-wave sleep, as well as attenuations in the P3 component of evoked potentials (Ehlers & Criado, 2010).
Adolescent EtOH exposure targets functioning of a number of neurotransmitter systems. Replicable and long-lasting decreases have been observed in the number of neurons in the basal forebrain displaying choline acetyltransferase (ChAcT) activity, a marker of cholinergic neurons (Coleman et al, 2011; Ehlers et al, 2011; Boutros et al, 2014; Vetreno et al, 2014; Swartzwelder et al, 2015). This effect is suggestive of a likely disruption in acetylcholine (ACh) function that is not evident after equivalent exposure in adulthood (Vetreno et al, 2014). In contrast to apparent ACh hypofunction, apparent elevations in dopamine (DA) function have often been reported in DA projections to reward-critical regions undergoing ontogenetic change during adolescence such as the nAc. These indices of elevated DA function include elevated basal levels of DA neurotransmission (Sahr et al, 2004) and extracellular DA levels (Badanich et al, 2007; Pascual et al, 2009), along with greater DA responses to EtOH challenge (Sahr et al, 2004; Pascual et al, 2009; Philpot et al, 2009). Alterations in the major excitatory (glutamate) and inhibitory (gamma aminobutyric acid [GABA]) neurotransmitter systems in brain have also been reported after adolescent EtOH exposure. Included are alterations in HPC GABAA receptor expression and increases in their EtOH sensitivity (Fleming et al, 2012), as well as increases in glutamate N-methyl-D-aspartate (NMDA) receptors (GluN) along with increased expression and alterations in phosphorylation of the GluN2B subtype of this receptor (Pascual et al, 2009; Sircar & Sircar, 2006). Administration of a positive allosteric modulator of the glutamate receptor mGlu5 (CDPPB) was found to reverse cognitive deficits induced by adolescent EtOH exposure (Gass et al, 2014).
EtOH exposure during adolescence has also been shown to influence epigenetic regulation through alterations in histone acetylation, thereby influencing DNA methylation and hence regulating propensity for DNA transcription at specific portions of the genome (Pascual et al, 2009, 2012; Pandey et al, 2015). For instance, persisting increases in histone deacetylase (HDAC) were seen in the AMYG of adolescent-exposed rats, along with decreases in levels of histone acetylation (see Pandey et al, 2015). Blockade of histone deacetylation with a HDAC inhibitor, trichostatin A (TSA), normalized regional deficits in histone acetylation and restored transcription of genes such as brain-derived neurotrophic factor (BDNF), while also attenuating the anxiety-like behavior and elevated EtOH intake seen in adulthood after adolescent EtOH exposure while the drug was “on board” (Pandey et al, 2015).
One other notable target of EtOH is neuroinflammation. Research in adult animals and humans has shown that neuroimmune signaling can influence ethanol intake and ethanol actions, with ethanol exposure conversely inducing proinflammatory signaling in brain (Robinson et al, 2014). Hence, it is not surprising that alcohol exposure during adolescence induces lasting increases in expression of neuroimmune genes, including a variety of proinflammatory signaling molecules, along with activation of the innate immune system receptor, Toll-like receptor 4 (TLR4) (see Crews & Vetreno, 2014, for review). Activation of TLR4 receptors by EtOH triggers transcription factors such as NFκB and AP-1 to induce cytokine production and mediators of inflammation that are thought to lead to the brain damage produced by EtOH (see Montesinos et al, 2015b). Evidence supports the suggestion that immune activation via TLR4 may be critical for the production of certain neural and behavioral consequences of EtOH exposure during adolescence. Induction of TRL4 activation with an agonist (lipopolysaccharide) was found to induce lasting reductions in neurogenesis similar to that observed after adolescent EtOH administration (Vetreno & Crews, 2015). Conversely, TLR-4 deficient (TLR4-KO) mice exposed to EtOH during adolescence did not show the inflammatory response nor cognitive and behavioral alterations characteristic of adolescent EtOH exposure (Montesinos et al, 2015a,b). Likewise, administration of an anti-inflammatory agent (indomethacin) was found to block the pattern of cell death and behavioral deficits associated with EtOH exposure during adolescence (Pascual et al, 2007). Thus, antiinflammatory agents join other possible pharmacotherapeutic agents, including positive allosteric modulators of the glutamate receptor mGlu5 and HDAC inhibitors, as possible strategies for preventing or reversing some of the lasting consequences of EtOH exposure during adolescence.
3. Nicotine
Substantial evidence has been obtained in rodent studies that adolescent nicotine exposure induces persisting neural consequences. Although less studied, cognitive and behavioral effects have also been reported, including increases in later self-administration of nicotine and other drugs.
3.1. Cognition, Behavior and Affect
Although not extensively explored, several persisting alterations in cognitive functioning, attention, impulsivity, and affective behavior have been reported after adolescent nicotine exposure (see Counotte et al, 2011a). Cognitive alterations reported in adulthood after adolescent nicotine exposure include alterations in serial pattern learning (Fountain et al, 2008) and attentional disruptions (Counotte et al, 2009, 2011b). Adult rats exposed to nicotine during adolescence (but not in adulthood) also exhibited increases in impulsive action (indexed by responding prematurely in a serial reaction time test) but not impulsive choice (indexed via delay discounting – i.e., the willingness to wait longer to receive a larger reward) (Counotte et al, 2009). These findings are consistent with other evidence that impulsivity indexed via action versus choice behavior are partially separable in their neural substrates, and hence sometimes differentially vulnerable to pharmacological manipulations (see Counotte et al, 2009, for discussion) , Increases in anxiety-like behavior have been reported in the open field and EPM (indexed via decreased time in the center of the field and the open arms, respectively) (Slawecki et al, 2003, 2005). Likewise, after adolescent nicotine exposure, adult rats acquired a conditioned fear response more rapidly, and extinguished this fear more slowly than control animals (Smith et al, 2006), findings also consistent with an anxiogenic profile.
3.2. Later Drug Self-administration
Adolescent nicotine exposure has also been shown to influence later responses to, and self-administration of, nicotine and other drugs of abuse. Indeed, nicotine has been suggested to be a gateway drug, often leading to the use of other drugs such as marijuana and cocaine (e.g., see Kandel & Kandel, 2014). For instance, exposure to nicotine during adolescence has been reported to increase locomotor sensitization to later amphetamine, cocaine, and amphetamine (Collins et al, 2004; Kelley & Rowan, 2004; Nolley & Kelley, 2007), effects that, where studied, were sex specific (evident only in adolescent males) and not apparent in animals who received equivalent nicotine exposure in adulthood (e.g., Collins et al, 2004). In contrast, adolescent nicotine exposure has been reported to decrease the rewarding properties of cocaine and methylphenidate in adulthood when indexed via conditioned place preferences (i.e., development of a preference for a location paired with drug administration)(Nolley & Kelley, 2007). Given that the nAc is thought to modulate both locomotor activity and the rewarding value of stimuli, with particular involvement of the core and shell regions, respectively, Kelley and Rowan (2004) have suggested that nicotine exposure during adolescence may differentially influence these regions to accentuate motor activation while conversely attenuating rewarding effects. Generally speaking, decreases in drug reward sensitivity are often associated with compensatory increases in drug self-administration under relatively low levels of response effort (see Koob & LeMoal, 2006). Indeed, age-specific enhancement of self-administration of nicotine (Adriani et al, 2003, 2006) and cocaine (McQuown et al, 2007; Dao et al, 2011) has sometimes (albeit not always – see Weaver et al, 2012; Levin et al, 2011) been reported after adolescent exposure to nicotine, with these effects generally restricted to exposures during early-mid adolescence, and not late adolescence or adulthood. This enhanced self-administration does not extend to all types of rewards given that similar effects were not evident when animals were given the opportunity to self-administer sucrose pellets (McQuown et al, 2007).
3.3. Age-dependent vulnerabilities
Where studied, persisting effects of adolescent exposure are generally more pronounced than observed after comparable nicotine exposure in adulthood. As one example, genome-wide microarray analyses were used to compare effects of repeated nicotine exposure during adolescence versus adulthood on gene expression in the VTA both immediately following the end of the 2 week exposure period as well as 1 month later (Doura et al, 2010). In this study, most nicotine-induced changes in gene expression in adults were observed immediately following the exposure period and had returned to control levels 1 month later, whereas persisting expression changes were most commonly seen in adolescents, with these genes being over-represented in networks involved in developmental processes, brain plasticity and various other neural functions. Effects of adolescent nicotine exposure have been shown to vary not only from adult exposures, but also from nicotine exposure earlier in life– i.e., during the prenatal or early postnatal period (see Slotkin, 2002; Dwyer et al, 2009). The timing specificity of developmental nicotine exposures has been suggested to be dependent on maturational changes in expression of nicotinic ACh receptors (nAchRs) and their subunit composition, as well as the structures undergoing nicotine-regulated development at that time. For instance, nAChRs are thought to be critical for regulating catecholamine and autonomic development during the prenatal period but to influence limbic and later stages of catecholamine development during the adolescent period (Dwyer et al, 2009). Indeed, alterations in limbic regions and other forebrain targets of the catecholamine, DA, are among the regions shown to be vulnerable to nicotine exposure during adolescence, as discussed in the following section.
3.4. Neural consequences
Cell losses in the brain (indexed via decreases in DNA) along with enlargement of surviving cells (indexed via increases in protein/ DNA ratios) have been reported after adolescent nicotine exposure, effects that were particularly pronounced in the cortex (Abreu-Villaça et al, 2003). Reported structural changes after adolescent nicotine exposure include increases in the number of spines on medium spiny neurons in the nAc and greater dendritic branching in regions such as the medial PFC, nAc shell, basolateral nucleus of the AMYG, bed nucleus of the stria terminalis, and HPC dentate gyrus (see Smith et al, 2015, for review). These consequences were found to persist into adulthood and were largely specific to adolescence exposure, with different regions affected and higher doses generally required to produce effects with adult exposure. Such dendritic remodeling was blocked by coadministration of a DA D1 receptor antagonist during the adolescent exposure period, suggesting that these dendritic modifications are D1 receptor dependent (Ehlinger et al, 2016). Indeed, a variety of neurotransmitter systems, including the DA system, show persisting alterations after exposure to nicotine during adolescence.
Nicotine acts in the brain through activation of nAChR, and hence it is not surprising that the cholinergic system upon which nicotine exerts direct effects is affected by adolescent nicotine exposure. Transient age- and subunit- dependent up- and down-regulation of nAChRs have been reported at the end of the nicotine exposure periods in adolescence as well as in adulthood (Doura et al, 2008), although these effects largely resolve with time post-exposure (e.g., see Slotkin et al, 2004; Counotte et al, 2009). In contrast, longer-term decreases in cholinergic markers (ChAcT and binding to the ACh transporter) have been reported in the forebrain after adolescent nicotine exposure (Slotkin et al, 2007), a pattern reminiscent of that seen after adolescent EtOH exposure as well. Increases in these cholinergic markers were observed, however, in hippocampus and midbrain, suggesting regional specificity in observed effects (Slotkin et al, 2007). Other persisting alterations reported after adolescent nicotine exposure include an increase in electrically stimulated DA efflux from PFC slices (Counotte et al, 2009), elevated cortical levels of the serotonin (5HT) transporter, as well as enhanced binding to 5HT1A and 5HT2 receptors (Slotkin et al, 2007). Lasting glutaminergic alterations in PFC have also been reported, characterized by a downregulation of mGlu2 glutamate receptors along with an associated reduction in mGlu2-mediated presynaptic inhibition (Counotte et al, 2011b). These disruptions in mGlu2 were not evident after comparable nicotine exposure in adulthood and appear critical for the deficits in attention observed after adolescent nicotine exposure, given that restoration of mGlu2 activity via local infusion of a mGlu2 agonist reversed the attentional deficit (Counotte et al, 2011b).
4. Cannabinoids
There has been a recent onslaught of research using animal models to assess potential neurobehavioral consequences of adolescent exposure to CBs. Findings of long-lasting neural and behavioral consequences of adolescent exposure to THC and a variety of synthetic CB agonists have emerged with a variety of measures; these findings have been reviewed recently in several excellent reviews to which the reader can refer for additional information, critique and references (Trezza et al, 2008; Renard et al, 2014; Higuera-Matas et al, 2015; Lubman et al, 2015). Although not commonly assessed, where examined, observed effects of adolescent CB exposure have sometimes been found to be sex-specific (e.g., Lee et al, 2014; Higuera-Matas et al, 2015; see Viveros et al, 2011, for review) and generally not evident after comparable exposures in adulthood (e.g., Viveros et al, 2012). Periods of particular vulnerability during adolescence have been little investigated to date, with work suggesting that early/mid adolescence (e.g., P35–40 and P40–45) is a time of greater vulnerability than during late adolescence (P50–55 or later) (Cass et al, 2014) contrasting with other findings that effects are more pronounced with exposures during late adolescence/emerging adulthood (P40–65) than during the juvenile-to-mid-adolescent period (P15–40) or in adulthood (>P70) (see Schneider, 2008).
4.1. Cognition, Behavior and Affect
Particularly well studied have been tests of working memory, including object recognition tests along with some work examining object location and social recognition; such tests expose animals briefly to a particular social or non-social stimulus (or object location) and then, shortly thereafter, give animals access to both the familiar and an unfamiliar stimulus (or location) and assess the extent to which animals direct their attention to the novel stimulus, thereby displaying evidence of remembering the initially presented stimulus. The most typical findings are long-lasting disruptions in these tests of working memory after repeated adolescent CB exposure (see Renard et al, 2014, for review). Effects were often, although not always (O’Tuathaugh et al, 2010) found to be specific to adolescent exposures. Cognitive effects of adolescent CB exposure appear highly specific, with working and spatial memory performance on a water maze task (location of a hidden platform) reported to be unaffected (e.g., see Higuera-Matas et al, 2009).
Alterations in social and affective measures have also been observed after adolescent CB exposure. Several studies have observed reductions in social behavior (O’Shea et al, 2004, 2006; Schneider, 2008; Schneider et al, 2008; Realini et al, 2011), perhaps associated in part with decreases in social motivation (Renaud et al, 2016) and increases in social anxiety (O’Shea et al, 2004, 2006). Social anxiety has occasionally (O’Shea et al, 2006; Quinn et al, 2008) but not typically (O’Shea et al, 2004; Schneider, 2008; Schneider et al, 2008) been reported after equivalent CB exposure in adulthood. Observed alterations in social behavior do not appear to be solely related to increases in social anxiety given that disruptions in the microstructure of social behavior are evident after adolescent CB exposure both when animals interact with an unfamiliar (and hence potentially stressful) social partner as well as when they interact with a familiar cage-mate (Schneider et al, 2008). Studies using non-social measures of anxiety have reported mixed results. Some studies, for instance, have reported increased anxiety after adolescent CB exposure in the light-dark box (indexed via fewer entries and longer latencies to enter the light side - Renaud et al, 2016) and in the EPM (Stopponi et al, 2014). Others, in contrast, have reported no notable alterations in anxiety (e.g., Higuera-Matas et al, 2009; O’Tuathaigh et al, 2010; Keeley et al, 2015) or in some cases even decreases in anxiety (e.g., Biscaia et al, 2003; also, see O’Tuathaigh et al, 2010; Keeley et al, 2015) in EPM tests – a diversity that, as discussed earlier, could potentially reflect the same tests reflecting anxiety versus “disinhibitory” profiles, depending on the specific pre-test or test conditions. Induction of a depressive-like phenotype (indexed via increased immobility in the forced swim test) has also been reported after adolescent CB exposure, along with signs of anhedonia (as indexed via a decreased preference for sucrose or palatable foods - e.g., Rubino et al, 2008; Bambico et al, 2010; Realini et al, 2011). Females may be somewhat more vulnerable to these depressive-like effects, with effects sometimes (Rubino et al, 2008; Realini et al, 2011), although not always (e.g., Rubino et al, 2008; Bambico et al, 2010) restricted to females.
4.2. Endophenotypes of schizophrenia
Given evidence from human studies that adolescent use of marijuana may be one factor that increases risk for later emergence of schizophrenia among otherwise vulnerable individuals (e.g., Semple et al, 2005), one focus of studies using animal models of adolescent CB exposure has been on assessment measures thought to reflect endophenotypes of schizophrenia (see Malone et al, 2010). These include the deficits in working memory, disruptions in social behavior, anxiety, and anhedonia discussed above, although it should be noted that these endophenotypes are not specific to schizophrenia and are evident with other conditions as well. One alteration particularly characteristic of schizophrenia is a disruption in sensory-motor gating. This type of cognitive filtering is often indexed in rodent studies via assessing the reduction in startle response to a loud tone when that stimulus is immediately preceded by a weaker, non-startle-inducing tone – a phenomenon known as pre-pulse inhibition (PPI). Long-term deficits in PPI have been frequently observed in rodent studies after CB exposure during adolescence, but not when animals received equivalent exposure in adulthood (e.g., Renaud et al, 2016), supporting the association of CB action on the adolescent brain with the later induction of schizophrenia-like alterations.
Although little studied to date, some studies have found that emergence of schizophrenia-relevant endophenotypes after adolescent CB exposure are even more pronounced among genetically vulnerable organisms or after early brain damage. For instance, adolescent CB exposure-induced anxiolytic effects were not evident in wild-type male mice but were evident in male mice with a knock-out of the catechol-O-methyl-transferase (COMT) gene (responsible for the breakdown of DA and norepinephrine); a similar synergism was not evident in males exposed to CB in adulthood, suggesting a critical genotype X exposure X age interaction in establishing vulnerabilities to lasting consequences of CB exposure. Likewise, in animals with early lesions of the medial PFC, disruptions in working memory and social behavior induced by adolescent CB exposure were exacerbated relative to animals that were not subjected to this early neural perturbation (Schneider & Koch, 2007). These findings are reminiscent of human data suggesting that separate vulnerabilities may interact or synergize to exacerbate risk for later emergence of schizophrenia and other neurodevelopmental disorders.
It should be noted, though, that in contrast to the idea that CB exposure may increase risk of schizophrenia in individuals with pre-existing vulnerabilities, there are reports that under certain circumstances adolescent exposure to cannabinoids may reverse adverse effects of exposure to other perturbations that include drugs (e.g., Lopez-Rodriguez et al, 2014), stressors (Abush & Akirav, 2013), or early maternal deprivation (López-Gallardo et al, 2012), leading some researchers to suggest that “cannabinoid exposure during adolescence may… have a protective role when individuals have faced severe life challenges, such as intense stress or maternal deprivation” (Higuerra-Matas et al, 2015, p.140). For example, administration of a cannabinoid agonist 1 hr following daily restraint stress during late adolescence (P45–60) reversed the stress-associated disruptions in LTP and short-term memory in a spatial location task seen one month later (Abush & Akirav, 2013). However, CB reversal effects are often highly measure-specific (e.g., see Abush & Akirav, 2013), with the CB agonist sometimes producing alterations alone or exacerbating effects of the other manipulation (e.g.,Schneider & Koch, 2005, 2007; Zamberletti et al, 2012). These reversal effects also do not appear to be adolescent-specific; similar effects have been reported in a number of cases following CB manipulations in adulthood (e.g., Schulz et al, 2013; Segev et al, 2014), with adult CB activation even found to reverse some effects of adolescent CB exposure (Realini et al, 2011). Certainly, although consequences of CB exposure may interact with other perturbations, potential beneficial vs. detrimental effects of these manipulations need careful further study before contemplating their potential translational relevance.
4.3. Later drug self-administration
A number of studies have used animal models to explore the notion of marijuana as a “gateway” drug – i.e. the propensity for adolescent CB exposure to increase the later probability of use of other illicit drugs. These studies have generally found evidence for increases in the later self-administration of cocaine, heroin and other opiates after repeated CB exposure during adolescence (e.g., Biscaia et al, 2008; Ellgren et al , 2008; Higuera-Matas et al, 2008), although observed effects were sometimes evident only in one sex or emerged only with stress-induced relapse (Stopponi et al, 2014). The nature of the sex-specificity varied across studies, with for instance adolescent CB exposure reported to increase self-administration of morphine in males but not females (Biscaia et al, 2008), whereas females (but not males) showed elevations in cocaine self-administration (Higuera-Matas et al, 2008). Later increases in self-administration after adolescent CB exposure may not reflect alterations in the processing of all rewards given that under the same circumstances where exposure-induced increases in cocaine self-administration were observed, no increases were evident when food was used as the reinforcer (Higuera-Matas et al, 2008). Whether elevations in later drug self-administration would also be evident when initial CB exposure is delayed until adulthood have yet to be explored systematically.
4.4. Neural effects
Neural consequences of adolescent CB exposure have been observed in a variety of brain regions critical for social/emotional behavior, working memory, cognitive filtering, and drug-self-administration, including regions such as the PFC, AMGY, nAc, HPC and other forebrain regions. A variety of neurotransmitter systems and signaling systems have been studied, although effects observed often vary across studies. Even when looking at effects of adolescent CB exposure on later functioning of the endogenous CB system, observed effects vary widely from no alterations in binding or functional activation of CB receptors to CB receptor 1 (CBR1) up-regulation or down-regulation (see Higuera-Matas et al, 2015, for review). As pointed out in that review, some of the variation may depend in part on whether adolescents were repeatedly exposed to THC or synthetic CB agonists, with these substances often producing opposite effects on CBR1 density and function (decreases with THC and increases with synthetic agonists).
Alterations in other neurotransmitter systems have also be observed. Diverse alterations in activity of the endogenous opioid system after adolescent CB exposure include complex patterns of sometimes opposing sex differences (Biscaia et al, 2008). Glutamate and GABA activity in a variety of brain regions is also altered after adolescence CB exposure (see Higuera-Matas et al, 2015). For instance, decreases in HPC glutamate tone (along with decreases in number of HPC dendritic spines receiving glutamate input – Rubino et al, 2009b) have been suggested to be related to the cognitive impairments evident after adolescent CB exposure (Higuera-Matas et al, 2015). Downregulated GABA transmission in PFC has also been observed after adolescent CB exposure. This GABA downregulation was linked to enduring PFC disinhibition in adulthood, a pattern of PFC disinhibition reminiscent of that seen normally early in adolescence (Cass et al, 2014). PFC dysregulation consistent with cortical disinhibition has likewise been reported by Renaud and colleagues (2016) after CB exposure during adolescence (but not in adulthood). In this case, however, they linked the cortical disinhibition to a hyper-DA state reflected by notable increases in basal firing and burst firing of DA neurons in the ventral tegmental area that project to the PFC.
Studies at the molecular level have proved to be a sensitive approach for assessing persisting consequences of adolescent CB exposure. Molecular signatures of compromised synaptic efficacy have been reported in the PFC (Rubino et al, 2009a), along with abnormalities in Akt, Wnt, and mTOR signaling cascades that “bear… remarkable similarity to adaptations observed in neuropsychiatric populations” (Renaud et al, 2016, p. 2). Adolescent CB exposure has also been reported to disrupt regulation of transcription factors induced by DA manipulations in the AMYG, and to alter receptor expression, transcription factors, opiate- and CB-sensitive signaling cascades and immediate early gene expression in forebrain DA terminal regions such as the nAc (Ellgren et al, 2008; Rubino et al, 2008; Wegener & Koch 2009). Although many of the molecular studies to date have focused only on consequences of adolescent exposures, in recent studies where exposure age has been varied, the adolescent brain has been shown to be either particularly vulnerable or to reveal different persisting alterations following repeated CB exposure than that observed in the adult brain (e.g., see Renaud et al, 2016).
5. Cocaine
After marijuana, the next most frequently used illicit drug globally is cocaine (Karila et al, 2014), although as discussed earlier the incidence of its use among adolescents is far lower than that of marijuana. It is a psychostimulant that, like other psychostimulants to be discussed later, inhibits reuptake of dopamine, norepinephrine, and serotonin, enhancing availability of these monoamine neurotransmitters in the synaptic region where receptors for these neurotransmitters are located (e.g., Koob & LeMoal, 2006). Reminiscent of the rodent data with alcohol, although the literature is somewhat mixed, systematic investigation of cocaine intravenous self-administration in adolescent and adult rats have shown that adolescent rats are more prone to cocaine self-administration than are adults when indexed via levels of intake at low doses of cocaine, escalation of cocaine intake and resistance to increases in “price” of the cocaine (Wong et al, 2013). Intriguingly, this effect was evident in mid-adolescent (~P42–49) but not early adolescent (~P35–42) rats, reminiscent of the increase in cocaine use seen across this age span in human adolescents (Johnston et al, 2016). As outlined below, various lasting consequences of repeated cocaine exposure during adolescence have been reported in rats and mice, although in most cases comparable adult exposure groups have not been studied, and hence little is known as to whether these effects are more or less pronounced than comparable exposures in adulthood.
5.1. Cognition, Behavior and Affect
Although studies are limited, the work available suggests that adolescent cocaine exposure induces a number of modest and sometimes unexpected behavioral and affective alterations in adulthood consistent with a decrease in threat evaluation and attenuated cautiousness. Evidence for alterations in spontaneous activity in adulthood are mixed, with observations of increases in spontaneous locomotor activity (Mateos-García et al, 2015) contrasting with other findings of decreased activity in a novel environment (Black et al, 2006). No effect of cocaine during adolescence on adult social interactions was observed (Mateos-García et al, 2015). Likewise, no increases in anxiety were observed in either the EPM or resident-intruder testing in adult rats after adolescent ethanol exposure (Alves et al, 2014). However, these animals exhibited several atypical behavioral alterations interpreted as suggesting possible impairment in the recognition and evaluation of potentially threatening stimuli. In the EPM, this was evinced via decreased time in the center of the maze (the relatively safe area between the open and closed arms) and in the resident-intruder test, as a longer latency to retreat from the aggressive “resident” (Alves et al, 2014). Intriguingly similar evidence for decreases in “cautious” behavior in adulthood after adolescent cocaine exposure were reported by Sillivan et al (2011) as indexed in their work via increased open arm time in the EPM, along with attenuated fear conditioning, increased center time in the open field and hole board tests, and increased hole poke behavior when no food was present in the holes (used as an index of novelty seeking).
Cognitive functions have been little investigated, with few notable differences reported to date. No evidence of adolescent exposure-associated alterations in working or reference memory was seen during acquisition or reversal of a hole board food search task and Morris water maze (Sillivan et al, 2011). Performance on an attentional set-shifting task revealed no performance deficits after adolescent cocaine exposure, with adolescent exposed rats conversely exhibiting more rapid shifts in performance on the set-shifting task than controls, perhaps due to alterations in attentional mechanisms; this effect was evident when exposed animals were tested in adulthood (P70) but not late adolescence (P56) (Black et al, 2006). These findings differ somewhat from those of Kantak et al, (2014) where both adolescent (P37–55) and adult (P77–95) rats were allowed to either self-administer cocaine or were given cocaine non-contingently, and tested >2 weeks later on a strategy set-shifting task and reversal. Adolescent cocaine exposure did not disrupt performance on the set-shifting task, although non-contingent (but not self-administered) cocaine during adolescence impaired reversal acquisition. In contrast, animals exposed to cocaine in adulthood exhibited faster acquisition on the set-shifting task and its reversal than controls, regardless of whether cocaine was delivered contingently or non-contingently. This lack of notable effects of adolescent cocaine exposure on “executive function”-type tasks that rely in part on circuitry involving the PFC contrast with compelling evidence for neural alterations in these regions, as discussed below. Indeed, Kantak and colleagues (2014) suggested that the immaturity of the PFC during adolescence may exert a protective effect against cognitive alterations from repeated cocaine exposure. Clearly, more studies in this area are warranted.
5.2. Later drug-self-administration
Repeated adolescent exposure to cocaine has been reported to induce sensitization to the locomotor stimulant effects of cocaine in rats that is evident when the animals are tested in adulthood (P70) but not during late adolescence (P56) (Black et al, 2006). In one of the few studies comparing repeated exposure to cocaine during adolescence with analogous exposure in adulthood, latter locomotor sensitization to cocaine was found to be more marked after cocaine exposure during adolescence (Wheeler et al, 2013). Cross-sensitization to METH has also been reported in both male and female adult mice exposed to cocaine as adolescents (Shanks et al, 2015). In the latter study, similar cross-sensitization to METH was also seen when the adolescents were exposed repeatedly to amphetamine or methylphenidate, although the former effect was only evident in males and the latter was specific to females (Shanks et al, 2015). Evidence for enhanced later sensitivity to cocaine after adolescent cocaine exposure has also been reported for the rewarding effects of cocaine, with high (although not low) novelty seeking adolescents repeatedly exposed to cocaine showing greater CPP to both cocaine and MDMA in adulthood relative to their non-exposed counterparts (Mateos-García et al, 2015).
5.3. Neural effects
Using magnetic resonance imaging in mice, Wheeler and colleagues (2013) observed persistent structural alterations in a network of regions innervated by DA, including decreases in gray matter volume of the nAC, anterior cingulate cortex, amygdala and insula, increases in volume of the substantia nigra and the orbital frontal cortex, but no changes observed in the volume of PFC. This study was one of the few to include animals that were equivalently exposed to cocaine in adulthood, and observed that the effects in these dopaminergic-innervated regions were more pronounced after adolescent than adult exposure. Direct evidence for functional alterations in dopaminergic projections have also been reported. For instance, Alves et al (2014) observed greater levels of DA turnover (indexed via the ratio of the DA metabolite DOPAC to DA) in the amygdala following EPM testing (in the absence of any alterations in anxiety in this test, as discussed above) (Alves et al, 2012). In this same study, adults exposed to cocaine as adolescents exhibited less notable increases in DA in the hippocampus following resident-intruder testing in adult rats following adolescent cocaine exposure, although the adolescent exposed animals showed greater corticosterone elevation post-test than did controls, findings they interpreted as evidence for greater stress responsivity in these animals (Alves et al, 2014). Greater stress sensitivity has also been reported in the PFC glutamatergic system, as discussed further below (Caffino et al, 2013).
Although the PFC was reported to be resistant to adolescent cocaine exposure in terms of gross structural alterations (Wheeler et al, 2013), such exposure has been reported to induce long-lasting increases in metabolic activity in the PFC, a “hypermetabolic PFC” than contrasts with the hypometabolic state reported after repeated cocaine exposure in adulthood (Cass et al, 2013). The consequence of both alterations, however, has been postulated to reflect decreased PFC functional capacity, consistent with an inverted U-shaped relationship between levels of PFC activity and efficacy of PFC functions (Cass et al, 2013).The increased PFC excitability after adolescent cocaine exposure is thought to be related in part to attenuated GABA activity in PFC (Caffino et al, 2013) associated with reduced GABAergic inhibition from the HPC to PFC as well as attenuated in GABA interneuron activity in the PFC (Cass et al, 2013). Such PFC hyperexcitability has also been to greater glutamatergic activity in the PFC, particularly in response to stressors. That is, adolescent cocaine exposure was found to sensitize mPFC glutaminergic synapses to a stress challenge three days later when indexed via increases in glutamate release, GluN1 responsivity, and spine hyperresponsiveness, along with decreases in glutamate transporter activity (involved in the clearance of glutamate) (Caffino et al, 2013).
Neural alterations induced by cocaine exposure in adolescence may be associated with transient drug-induced disruptions in gene expression. Alterations include increases in expression of genes encoding for cell adhesion molecules and declines in those encoding transcription factors observed in PFC after binge-like exposure to cocaine during adolescence, expression changes that were generally transient and declined with time following exposure termination (Black et al, 2006). Transient expression changes associated with adolescent cocaine exposure have also been reported in the amygdala in genes critical for synaptic function, axonal guidance and Wnt signaling (Sillivan et al, 2011). Decreases in histone methylation in PFC were also observed (Black et al, 2006). How these drug-induced changes in gene expression and epigenetic regulation relate to lasting neurobehavioral changes after adolescent cocaine exposure remain to be determined.
6. Other Psychostimulants
All drugs of the psychostimulant class, amphetamine (AMPHET), MDMA and METH, are indirect monoamine (MA) agonists, with AMPHET and METH having greater acute actions on DA, and MDMA having the most pronounced 5HT effects. The focus here will be on the “club” drugs METH and MDMA – stimulants used for recreational purposes, although it should be noted that the use of prescription stimulants such as methylphenidate as “cognitive enhancers” is increasing among some populations of youth (Clemow & Walker, 2014). Studies exploring consequences of adolescent exposure to METH and MDMA have been shown to produce a variety of persisting neural and behavioral alterations (see Teixeira-Gomes et al, 2015, for review and references) although, intriguingly, as reviewed below the adolescent period may not be a particularly vulnerable period for these effects, and in some cases may even be protective.
6.1. Cognition, Behavior and Affect
A number of persisting functional alterations have been observed after adolescent stimulant exposure. Exposure to MDMA has been observed to impair working memory (e.g., Piper & Meyer, 2004) and decrease social interactions (e.g., Bull et al, 2004). Increases in social anxiety-like behavior in the social interaction test contrast, however, with reports of no alterations in anxiety (Bull et al, 2004) or even attenuated anxiety (e.g., Piper & Meyer, 2004) when indexed via open arm time in the EPM. Stimulant exposure during adolescence has also been reported to sensitize animals to later stimulant challenge (McPherson & Lawrence, 2006; Klomp et al, 2012; although see also Piper et al, 2006). Measure-specific age differences have been reported, with for instance repeated AMPHET exposure during adolescence inducing less persisting stereotypy to later AMPHET challenge than after equivalent exposure beginning in adulthood, although the adolescent-exposed animals were reported to exhibit greater persisting cognitive deficits than those exposed as adults in a working memory task and during acquisition of a delayed non-matching to sample task (Sherill et al, 2013).
6.2. Neurotoxicity
Neurotoxicity observed after adolescent stimulant exposure includes decreases in MA levels or levels of their transporters, damage to MA nerve terminals, apoptotic neuronal death and reactive gliosis indexed via increases in glial fibrillary acidic protein (see Teixeira-Gomes et al, 2015). Although there are scattered reports that adolescent exposure to these stimulants alters functioning of a variety of neurotransmitters, including ACh (Siegel et al, 2011) and glutamate (Kindlundh-Högberg et al, 2008), the DA and 5HT systems appear to be particularly targeted. For instance, persisting decreases in striatal levels of DA and 5HT (Cappon et al, 1997), along with occasional reports of decreases in 5HT levels in HPC, frontal regions of cortex (Bull et al, 2004) and AMYG (Faria et al, 2006) have been reported following adolescent exposure to METH or MDMA. Studies comparing consequences of adolescent versus adult stimulant exposure, however, have generally found neurotoxicity of adolescent stimulant exposure to be less severe than after exposure in adulthood, at least with some response measures. For instance, after METH exposure in adulthood, decreases in DA uptake and binding to the DA transporter were seen in striatum 1 week later, whereas neither DA alteration was seen in animals receiving METH exposure as adolescents (Riddle et al, 2002). In a study examining both short-term (1 day) and persisting (1 week) consequences of METH exposure, similar short-term DA alterations were seen following METH exposure in both adolescents and adults, whereas only adults exhibited persisting disruptions in the DA system, indexed via decreases in DA synthesis, uptake and transporter activity (Kokoshka et al, 2000). In this same study, however, no age differences in 5HT synthesis were observed, with short-term and persisting decreases evident after exposure at either age (Kokoshka et al, 2000). Although Klomp and colleagues (2012) also reported persistent decreases in the serotonin transporter after MDMA exposure at both ages, the extent of this loss was less pronounced in frontal brain regions after exposure during adolescence than in adulthood.
6.3. Age-dependent vulnerabilities continued
The relative resistance of adolescents to persisting MA neurotoxicity may represent the later ontogenetic stages of an extended developmental delay in emergence of stimulant neurotoxicity. This conclusion was reached from rare and valuable research examining the ontogeny of stimulant neurotoxicity across a wide age span ranging from prenatal, pre-weanling, juvenile, adolescent and adult exposures. In this work, minimal MDMA neurotoxicity was observed after exposure at younger ages, with evidence of neural alterations (indexed via decreases in serotonin transporter binding sites) beginning to emerge after exposure during the late juvenile/early adolescent period (i.e., ~ P25–30), although toxicity at those ages was still attenuated relative to that seen after equivalent exposure in adulthood (Kelly et al, 2002). A similar ontogeny was revealed in a study of MDMA-induced decreases in serotonin transporter density and 5HT concentrations in frontal cortex 1 week following acute MDMA exposure where decreases were evident after exposure at P35, but not at P14, 21 or 28 (Aguirre et al, 1998). Similar ontogenetic delays in the emergence of neurotoxicity were reported with METH, with METH decreasing striatal DA, increasing glial fibrillary acidic protein and inducing hyperthermia only after administration at P40 and P60, but not P20 (Cappon et al, 1997). Monophasic ontogenetic increases in stimulant neurobehavioral toxicity have not always been observed, however, with for instance an inverted U-shaped curve peaking later in adolescence observed in a study where rats exhibited greater learning deficits after METH exposure at P41–50 than at P21–30, P31–40 or P51–60 (Vorhees et al, 2005).
A number of potential contributors to the attenuated toxicity of adolescents relative to adults to these stimulants has been suggested. One primary suspect has been the induction of drug-induced hyperthermia, given that METH-induced neurotoxicity has been reported to be exacerbated by hyperthermia and this hyperthermia emerges only gradually during ontogeny (e.g. Riddle et al, 2002). Yet, while METH-induced hyperthermia could be environmentally facilitated at both P20 and P40 (by placement in a context with a temperature above room temperature), this hyperthermia precipitated METH-induced neurotoxicity only at P40 and not P20 (Cappon et al, 1997), suggesting that hyperthermia alone is insufficient for inducing neurotoxic effects, at least in very young animals. Pharmacokinetic factors may also contribute to the stimulant insensitivity of young animals, with higher brain levels of stimulants typically produced in adulthood after administration of the same doses to adults and younger animals (e.g., see Kokoshka et al, 2000). Yet, even when brain METH concentrations were equated by administering a higher dose to adolescents than adults, greater and longer-lasting METH neurotoxicity was evident in adults (Truong et al, 2005; Rau et al, 2006), suggesting that, as with other drugs of abuse, age differences in drug effects are unlikely to be due merely to across-age pharmacokinetic differences. Maturational stage of MA systems has also been suggested to contribute to the resistance of adolescents to MDMA (e.g., Klomp et al, 2012) and METH (Truong et al, 2005; Volz et al, 2009) neurotoxicity. For instance, adolescent animals exhibit both enhanced DA efflux as well as greater kinetic activity of MA transporters which could result in greater capacity to sequester DA in synaptic vesicles. Together these factors could protect against METH-induced elevations of cytoplasmic DA levels and subsequent neurotoxicity induced via production of DA-associated reactive oxygen species (see Truong et al, 2005; Volz et al, 2009). Thus, a variety of factors may contribute to the relative resistance of younger animals to stimulant neurotoxicity.
Intriguingly, a number of studies have reported that repeated stimulant exposure during adolescence may protect against later deficits induced by subsequent METH or MDMA exposure in adulthood (e.g., Piper et al, 2006, McFadden et al, 2011). For instance, periodic MDMA exposure from P35–60 attenuated the locomotor hypoactivity and decreases in serotonin transporters typically seen following MDMA exposure in adulthood (Piper et al, 2006). Similar attenuations in subsequent neurotoxicity, however, have been reported when the initial stimulant exposure was delayed into adulthood. Consequently, these effects are not adolescent-specific and may be associated with the emergence of tolerance to stimulant-induced hyperthermia (McFadden et al, 2012) given that, as discussed previously, hyperthermia has been shown to exacerbate neurotoxicity.
Collectively, these data provide compelling evidence that stimulants show a markedly different pattern of ontogenetic sensitivity to drug neurotoxicity than the other drugs reviewed here, with young animals through adolescence being relatively resistant to the neurotoxic effects of METH and MDMA, perhaps due in part to the maturational state of DA systems that may confer partial protection against neurotoxicity. Despite this relative resistance, however, lasting neural, behavioral and cognitive consequences of adolescent exposure to these substances have nevertheless been observed, suggesting that adolescent exposures are not innocuous. For more detailed discussion of studies in laboratory animals examining the effects of adolescence stimulant exposure, see recent excellent reviews by Buck & Siegel (2015) and Teixeira-Gomes et al (2015).
7. Summary, conclusions and future directions
A summary of the major findings discussed in this review are provided in Table 1. Although these findings are not exhaustive (an unfeasibility given the rapid escalation of work in much of this area), the data reviewed provide convincing evidence that adolescent exposure to these drugs induces drug-specific patterns of sustained neurobehavioral alterations. Particularly common consequences evident across studies are disruptions in cognitive functions such as working memory, alterations in affect including increases in social anxiety, as well as increased cell death. Long-lasting alterations in forebrain neurotransmitter systems critical for modulating reward, socioemotional processing and cognition have also emerged, including some evidence for induction of a hyper-DA state after EtOH, CBs and nicotine (though not stimulants), and for attenuations in neurons expressing cholinergic markers after EtOH and nicotine. Alterations in later self-administration of the same or other drugs have also been reported under some circumstances, although null findings are also common.
Table 1.
EtOH | NIC | CB | COC | STIM | ||||||
---|---|---|---|---|---|---|---|---|---|---|
General Age Vulnerability | Adol>Adult | Adol>Adult | Adol>Adult | ? | Adol<Adult | |||||
Cognitive/behav. | ||||||||||
Spatial memory | ↓= | = | ||||||||
Conditional discrimin./pattern learning |
↓ | ↓ | ||||||||
Attention | ↓ | (↓) | ||||||||
Obj. recognit./working memory |
↓ | ↓ | = | ↓ | ||||||
Pre-pulse inhibition |
↓ | |||||||||
Congitive flexibility |
↓ | |||||||||
Risk preference | ↑ | |||||||||
Impulsivity/disinhibition | ↑ | ↑ | ||||||||
Retent. of adoles-typical phenotypes | ↑ | |||||||||
Affective/Social behavior | ||||||||||
Depression-like behaviors | ↑ | ↑ | ||||||||
Social interactions | ↓ | ↓ | = | ↓ | ||||||
Social anxiety-like behaviors |
↑ | ↑ | ↓ | ↑ | ||||||
Other anxiety-like behaviors |
↑, ↓ | ↑ | ↑, ↓, = | =, ↓ | ||||||
Later self-admint. (same/different drugs) |
↑, = | ↑, = | ↑, = | |||||||
Neural | ||||||||||
Neurogenesis | ↓ | |||||||||
Cell death |
↑ | ↑ | ↑ | |||||||
Spines/dendritic branching | ↑ | |||||||||
Electrophysiol. Alterations | Y | Y | ||||||||
Neuroimmune activation | Y | |||||||||
Histone acetylation/epigentic regulat. |
Y | Y | ||||||||
Alterations in: | ||||||||||
Ach | Y | Y | Y | |||||||
Glutmate/GABA | Y | Y | Y | Y | Y | |||||
DA | Y | Y | Y | Y | Y | |||||
5HT | Y | Y | Y | |||||||
CB | Y | |||||||||
Affected brain regions: | ||||||||||
PFC | Y | Y | Y | Y | Y | |||||
HPC | Y | Y | Y | Y | Y | |||||
nAc | Y | Y | Y | Y | ||||||
AMYG | Y | Y | Y | Y | Y |
impaired/attenuated; ↑ enhanced; = no notable exposure effects; Y alterations reported (often complex)
Empty cells in the Table could represent dependent measures that have yet to be systematically explored, or negative findings. As examples of potentially promising areas for further inquiry, note that although neuroimmune activation, alterations in neurogenesis, histone regulation, and sleep and other electrophysiological indices have been recently reported after adolescent exposure to EtOH, these sensitive measures have seemingly received little attention in the literature after adolescent exposure to other drugs. Likewise, alterations in cognitive flexibility have been reported in a number of instances after adolescent exposure to EtOH, but have seemingly been little explored after exposure to the other drugs. As another example, despite frequent reports of DA alterations after adolescent alcohol/drug exposure, and evidence that endogenous CBs play critical roles in regulation of DA neurons (e.g. Wang & Lupica, 2014), there has yet been little apparent investigation of alterations in endogenous CBs and their receptors after adolescent exposure to any of the drugs other than THC and other CBs. Thus, despite rapid progress to date, more work remains to determine whether apparent drug-selective findings reflect meaningful differences across drugs or merely areas of research focus to date, and to assess whether comparable findings observed across drugs reflect similarly altered neural substrates.
Another understudied area at present is that of sex differences. When both sexes have been investigated, sex differences have often emerged. Inclusion of both sexes in studies of lasting consequences of adolescent drug exposure has been sufficiently rare, though, to make it difficult to characterize the extent and nature of such sex differences. Even when sex-specific vulnerabilities in adolescent exposure effects have emerged, there often has been little exploration of the critical mechanisms that underlie these differential vulnerabilities. Assessment of sex differences in the functional and neural consequences of adolescent drug effects will undoubtedly be aided in the future by the rapid progress being made in the area of puberty and the emergence of sex differences in neurobehavioral function (see reviews by Gur & Gur and by Shultz & Sisk in this Special Issue for examples of recent work in this area).
One type of measure where notable variations in findings has been observed is in the assessment of general anxiety-like behaviors. In contrast to generally consistent findings of persisting decreases in social behavior and increases in social-like anxiety after adolescent drug exposure, findings have been more variable when using tests such as the EPM, light/dark box and open field. As discussed earlier, increased time spent in the open could varyingly reflect low anxiety levels or greater “disinhibition”. And the extent to which possible drug-induced increases in anxiety versus greater disinhibition would predominate in a particular test situation might well vary depending on prior experiences and the particular test circumstances. Development of test procedures to disentangle detection of these two constructs would be useful.
Only a modest amount of work has been conducted to assess whether adolescence is a time of particular vulnerability for the production of lasting drug effects. When other aged exposure groups have been studied, consequences typically were found to be more pronounced after exposures during adolescence than in adulthood for three of the four drugs/drug classes examined. The stimulants METH and MDMA were the exception, with neurotoxicity very low in young animals, more marked by adolescence, and continuing to increase into adulthood. Given this precedent for greater toxicity after adult than adolescent exposure for at least some drugs, it would appear premature to assume greater adolescent vulnerability and dismiss the importance of periodically including adult exposure groups to assess relative across-age vulnerabilities in programmatic research in this area.
Thus, there is compelling evidence that adolescent drug exposure can induce persisting neural, cognitive, affective and behavioral consequences. To date, brain regions undergoing notable development during adolescence appear to be particularly sensitive to lasting perturbations after adolescent drug exposure. Yet, it is not clear whether these regions are truly more vulnerable or if they have merely received particular scrutiny due to their developmental “volatility” during adolescence. Work remains to determine the breadth of findings within drugs, consistencies across drugs, exposure-age specificity, underlying mechanisms, and consilience of findings with human drug-abusing adolescents, with a goal of identifying promising translational approaches for the development of effective prevention/intervention efforts.
Highlights.
Rodent studies reveal persisting neurobehavioral effects of adolescent drug exposure
Ethanol, nicotine and cannabinoid vulnerability is greater in adolescents than adults
MDMA and methamphetamine show a converse age-related vulnerability
Affected brain regions include those undergoing developmental change at this time
Alterations include disruptions in cognition, affect, and drug self-administration
Acknowledgments
Preparation of this review was supported by the National Institute on Alcohol Abuse and Alcoholism of the National Institutes of Health under award numbers U01 AA019972 and P50 AA017823. The content is solely the responsibility of the author and does not necessarily represent the official views of the National Institutes of Health.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFERENCES
- Abreu-Villaça Y, Seidler FJ, Tate CA, Slotkin TA. Nicotine is a neurotoxin in the adolescent brain: critical periods, patterns of exposure, regional selectivity, and dose thresholds for macromolecular alterations. Brain Res. 2003;979(1–2):114–128. doi: 10.1016/s0006-8993(03)02885-3. [DOI] [PubMed] [Google Scholar]
- Abush H, Akirav I. Cannabinoids ameliorate impairments induced by chronic stress to synaptic plasticity and short-term memory. Neuropsychopharmacology. 2013;38(8):1521–1534. doi: 10.1038/npp.2013.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Acheson SK, Bearison C, Risher ML, Abdelwahab SH, Wilson WA, Swartzwelder HS. Effects of acute or chronic ethanol exposure during adolescence on behavioral inhibition and efficiency in a modified water maze task. PLoS One. 2013;8(10):e77768. doi: 10.1371/journal.pone.0077768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adriani W, Spijker S, Deroche-Gamonet V, Laviola G, LeMoal M, Smit AB, Piazza PV. Evidence for enhanced neurobehavioral vulnerability to nicotine during periadolescence in rats. J Neurosci. 2003;23(11):4712–4718. doi: 10.1523/JNEUROSCI.23-11-04712.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adriani W, Deroche-Gamonet V, Le Moal M, Laviola G, Piazza PV. Preexposure during or following adolescence differentially affects nicotine-rewarding properties in adult rats. Psychopharmacology. 2006;184(3–4):382–390. doi: 10.1007/s00213-005-0125-1. [DOI] [PubMed] [Google Scholar]
- Aguirre N, Barrionuevo M, Lasheras B, Del Rio J. The role of dopaminergic systems in the perinatal sensitivity to 3,4-methyloxymethamphetamine-induced neurotoxicity in rats. J Pharmacol Exp Ther. 1998;286(3):1159–1165. [PubMed] [Google Scholar]
- Alaux-Cantin S, Warnault V, Legastelois R, Botia B, Pierrefiche O, Vilpoux C, Naassila M. Alcohol intoxications during adolescence increase motivation for alcohol in adult rats and induce neuroadaptations in the nucleus accumbens. Neuropharmacology. 2013;67:521–531. doi: 10.1016/j.neuropharm.2012.12.007. [DOI] [PubMed] [Google Scholar]
- Alves CJ, Magalhaes A, Melo P, deSousa L, Tavares MA, Monteiro PR, Summavielle T. Long-term effects of chronic cocaine exposure throughout adolescence on anxiety and stress responsivity in a Wistar rat model. Neuroscience. 2014;277:343–355. doi: 10.1016/j.neuroscience.2014.07.008. [DOI] [PubMed] [Google Scholar]
- Badanich KA, Maldonado AM, Kirstein CL. Chronic ethanol exposure during adolescence increases basal dopamine in the nucleus accumbens septi during adulthood. Alcohol Clin Exp Res. 2007;31(5):895–900. doi: 10.1111/j.1530-0277.2007.00370.x. [DOI] [PubMed] [Google Scholar]
- Bambico FR, Nguyen NT, Katz N, Gobbi G. Chronic exposure to cannabinoids during adolescence but not during adulthood impairs emotional behaviour and monaminergic neurotransmission. Neurobiol Dis. 2010;37(3):641–655. doi: 10.1016/j.nbd.2009.11.020. [DOI] [PubMed] [Google Scholar]
- Becker JB, Koob GF. Sex differences in animal models: Focus on addiction. Pharmacol Rev. 2016;68:242–263. doi: 10.1124/pr.115.011163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bell RL, Rodd ZA, Sable HJ, Schultz JA, Shu CC, Lumeng L, Murphy JM, McBride WJ. Daily patterns of ethanol drinking in peri-adolescent and adult alcohol-preferring (P) rats. Pharmacol Biochem Behav. 2006;83(1):35–46. doi: 10.1016/j.pbb.2005.12.004. [DOI] [PubMed] [Google Scholar]
- Biscaia M, Marin S, Fernandez B, Marco EM, Rubio M, Gauza C, Ambrosio E, Viveros MP. Chronic treatment with CP 55,940 during the peri-adolescent period differentially affects the behavioural responses of male and female rats in adulthood. Psychopharmacology. 2003;170(3):301–308. doi: 10.1007/s00213-003-1550-7. [DOI] [PubMed] [Google Scholar]
- Biscaia M, Fernandez B, Higuera-Matas A, Miguens M, Viveros MP, Garcia-Lecumberri C, Ambrosio E. Sex-dependent effects of periadolescent exposure to the cannabinoid agonist CP-55,940 on morphine self-administration behaviour and the endogenous opioid system. Neuropsychopharmacology. 2008;54(5):863–873. doi: 10.1016/j.neuropharm.2008.01.006. [DOI] [PubMed] [Google Scholar]
- Blizard D, Vandenbergh D, Jefferson A, Chatlos C, Vogler G, McClearn G. Effects of periadolescent ethanol exposure on alcohol preference in two BALB substrains. Alcohol. 2004;34(2–3):177–185. doi: 10.1016/j.alcohol.2004.08.007. [DOI] [PubMed] [Google Scholar]
- Black YD, Maclaren FR, Naydenov AV, Carlezon WA, Jr, Baxter MG, Konradi C. Altered attention and prefrontal cortex gene expression in rats after binge-like exposure to cocaine during adolescence. J Neurosci. 2006;26(38):9656–9665. doi: 10.1523/JNEUROSCI.2391-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boutros N, Semenova S, Liu W, Crews FT, Markou A. Adolescent intermittent ethanol exposure is associated with increased risky choice and decreased dopaminergic and cholinergic neuron makers in adult rats. Int J Neuropsychopharmacol. 2014;18(2) doi: 10.1093/ijnp/pyu003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Briones TL, Woods J. Chronic binge-like alcohol consumption in adolescence causes depression-like symptoms possibly mediated by the effects of BDNF on neurogenesis. Neuroscience. 2013;254:324–334. doi: 10.1016/j.neuroscience.2013.09.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Broadwater MA, Varlinskaya EI, Spear LP. Effects of voluntary access to sweetened ethanol during adolescence on intake in adulthood. Alcohol Clin Exp Res. 2013;37(6):1048–1055. doi: 10.1111/acer.12049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Broadwater MA, Liu W, Crews FT, Spear LP. Persistent loss of hippocampal neurogenesis and increased cell death following adolescent, but not adult, chronic ethanol exposure. Dev Neurosci. 2014;36(3–4):297–305. doi: 10.1159/000362874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buck JM, Siegel JA. The effects of adolescent methamphetamine exposure. Front Neurosci. 2015;9:151. doi: 10.3389/fnins.2015.00151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bull EJ, Hutson PH, Fone KC. Decreased social behavior following 3,4-methylenedioxymethamphetamine (MDMA) is accompanied by changes in 5-HT2A receptor responsivity. Neuropharmacology. 2004;46(2):202–210. doi: 10.1016/j.neuropharm.2003.08.004. [DOI] [PubMed] [Google Scholar]
- Caffino L, Calabrese F, Giannotti G, Barbon A, Verheij MM, Racagni G, Fumagalli F. Stress rapidly dysregulates the glutamatergic synapse in the prefrontal cortex of cocaine-withdrawn adolescent rats. Addict Biol. 2015;20(1):158–169. doi: 10.1111/adb.12089. [DOI] [PubMed] [Google Scholar]
- Cappon GD, Morford LL, Voorhees CV. Ontogeny of methamphetamine-induced neurotoxicity and associated hyperthermic response. Brain Res Dev Brain Res. 1997;103(2):155–182. doi: 10.1016/s0165-3806(97)81791-9. [DOI] [PubMed] [Google Scholar]
- Casey BJ, Jones RM, Somerville LH. Braking and accelerating of the adolescent brain. J Res Adolesc. 2011;21(1):21–33. doi: 10.1111/j.1532-7795.2010.00712.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cass DK, Thomases DR, Caballero A, Tseng KY. Developmental disruption of gamma-aminobutyric acid function in the medial prefrontal cortex by noncontingent cocaine exposure during early adolescence. Biol Psychiatry. 2013;74(7):490–501. doi: 10.1016/j.biopsych.2013.02.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cass DK, Flores-Barrera E, Thomases DR, Vital WF, Caballero A, Tseng KY. CB1 cannabinoid receptor stimulation during adolescence impairs the maturation of GABA function in the adult rat prefrontal cortex. Mol Psychiatry. 2014;19(5):536–543. doi: 10.1038/mp.2014.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clemow DB, Walker DJ. The potential for misuse and abuse of medications in ADHD: a review. Postgrad Med. 2014;126(5):64–81. doi: 10.3810/pgm.2014.09.2801. [DOI] [PubMed] [Google Scholar]
- Coleman LG, Jr, He J, Lee J, Styner M, Crews F. Adolescent binge drinking alters adult brain neurotransmitter gene expression, behavior, brain regional volumes, and neurochemistry in mice. Alcohol Clin Exp Res. 2011;35(4):671–688. doi: 10.1111/j.1530-0277.2010.01385.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Collins SL, Montano R, Izenwasser S. Nicotine treatment produces persistentincreases in amphetamine-stimulated locomotor activity in periadolescent male but not female or adult male rats. Brain Res Dev Brain Res. 2004;153(2):175–187. doi: 10.1016/j.devbrainres.2004.08.004. [DOI] [PubMed] [Google Scholar]
- Counotte DS, Spijker S, Van de Burgwal LH, Hogenboom F, Schoffelmeer AN, DeVries TJ, Smit AB, Pattij T. Long-lasting cognitive deficits resulting from adolescent nicotine exposure in rats. Neuropsychopharmacology. 2009;34(2):299–306. doi: 10.1038/npp.2008.96. [DOI] [PubMed] [Google Scholar]
- Counotte DS, Smit AB, Pattij T, Spijker S. Development of the motivational system during adolescence, and its sensitivity to disruption by nicotine. Dev Cogn Neurosci. 2011a;1(4):430–443. doi: 10.1016/j.dcn.2011.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Counotte DS, Goriounova NA, Li KW, Loos M, van der Schors RC, Schetters D, Schoffelmeier AN, Smit AB, Mansvelder HD, Pattij T, Spijker S. Lasting synaptic changes underlie attention deficits caused by nicotine exposure during adolescence. Nat Neurosci. 2011b;14(4):417–419. doi: 10.1038/nn.2770. [DOI] [PubMed] [Google Scholar]
- Crews FT, Braun CJ, Hoplight B, Switzer RC, III, Knapp DJ. Binge ethanolconsumption causes differential brain damage in young adolescent rats compared with adult rats. Alcohol Clin Exp Res. 2000;24(11):1712–1723. [PubMed] [Google Scholar]
- Crews F, He J, Hodge C. Adolescent cortical development: A critical period of vulnerability for addiction. Pharmacol Biochem Behav. 2007;86(2):189–199. doi: 10.1016/j.pbb.2006.12.001. [DOI] [PubMed] [Google Scholar]
- Crews FT, Vetreno RP. Neuroimmune basis of alcohol brain damage. Int Rev Neurobiol. 2014;118:315–357. doi: 10.1016/B978-0-12-801284-0.00010-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dao JM, McQuown SC, Loughlin SE, Belluzzi JD, Leslie FM. Nicotine alters limbic function in adolescent rat by a 5-HT1A receptor mechanism. Neuropsychopharmacology. 2011;36(7):1319–1331. doi: 10.1038/npp.2011.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Day AM, Celio MA, Lisman SA, Johansen GE, Spear LP. Acute and chronic effects of alcohol on trail making test performance among underage drinkers in a field setting. J Stud Alcohol Drugs. 2013;74(4):635–641. doi: 10.15288/jsad.2013.74.635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Desikan A, Wills DN, Ehlers CL. Ontogeny and adolescent alcohol exposure in Wistar rats: open field conflict, light/dark box and forced swim test. Pharmacol Biochem Behav. 2014;122:279–285. doi: 10.1016/j.pbb.2014.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diaz-Granados JL, Graham D. The effects of continuous and intermittent ethanol exposure in adolescence on the aversive properties of ethanol during adulthood. Alcohol Clin Exp Res. 2007;31(12):2020–2027. doi: 10.1111/j.1530-0277.2007.00534.x. [DOI] [PubMed] [Google Scholar]
- Doreumus TL, Brunell SC, Rajendran P, Spear LP. Factors influencing elevated ethanol consumption in adolescent relative to adult rats. Alcohol Clin Exp Res. 2005;29(10):1796–1808. doi: 10.1097/01.alc.0000183007.65998.aa. [DOI] [PubMed] [Google Scholar]
- Doremus-Fitzwater TL, Varlinskaya EI, Spear LP. Motivational systems in adolescence: possible implications for age differences in substance abuse and other risk-taking behaviors. Brain Cogn. 2010;72(1):114–123. doi: 10.1016/j.bandc.2009.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doura MB, Gold AB, Keller AB, Perry DC. Adult and periadolescent rats differ in expression of nicotine cholinergic receptor subtypes and in the response of these subtypes to chronic nicotine exposure. Brain Res. 2008;1215:40–52. doi: 10.1016/j.brainres.2008.03.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doura MB, Luu TV, Lee NH, Perry DC. Persistent gene expression changes in ventral tegmental area of adolescent but not adult rats in response to chronic nicotine. Neuroscience. 2010;170(2):503–513. doi: 10.1016/j.neuroscience.2010.06.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dwyer JB, McQuown SC, Leslie FM. The dynamic effects of nicotine on the developing brain. Pharmacol Ther. 2009;122(2):125–139. doi: 10.1016/j.pharmthera.2009.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ehlers C, Criado J. Adolescent ethanol exposure: Does it produce long-lasting electrophysiological effects? Alcohol. 2010;44(1):27–37. doi: 10.1016/j.alcohol.2009.09.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ehlers C, Criado J, Wills D, Liu W, Crews F. Periadolescent ethanol exposure reduces adult forebrain ChAT+IR neurons: Correlation with behavioral pathology. Neuroscience. 2011;29(199):333–345. doi: 10.1016/j.neuroscience.2011.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ehlers CL, Liu W, Wills DM, Crews FT. Periadolescent ethanol vapor exposure persistently reduces measures of hippocampal neurogenesis that are associated with behavioral outcomes in adulthood. Neuroscience. 2013;244:1–15. doi: 10.1016/j.neuroscience.2013.03.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ehlinger DG, Bergstrom HC, Burke JC, Fernandez GM, McDonald CG, Smith RF. Adolescent nicotine-induced dendrite remodeling in the nucleus accumbens is rapid, persistent, and D1-dopamine receptor dependent. Brain Struct Funct. 2016;221(1):133–145. doi: 10.1007/s00429-014-0897-3. [DOI] [PubMed] [Google Scholar]
- Ellgren M, Artmann A, Tkaylych O, Gupta A, Hansen HS, Hansen SH, Devi LA, Hurd YL. Dynamic changes of the endogenous cannabinoid and opioid mesocorticolimbic systems during adolescence: THC effects. Eur Neuropharmacol. 2008;18(11):826–834. doi: 10.1016/j.euroneuro.2008.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ernst M, Fudge JL. A developmental neurobiological model of motivated behavior: anatomy, connectivity and ontogeny of the triadic nodes. Neurosci Biobehav Rev. 2009;33(3):367–382. doi: 10.1016/j.neubiorev.2008.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Faria R, Magalhäes A, Monteiro PR, Gomes-Da-Silva J, Amélia Tavares M, Summavielle T. MDMA in adolescent male rats: decreased serotonin in the amygdala and behavioral effects in the elevated plus-maze test. Ann NY Acad Sci. 2006;1074:643–649. doi: 10.1196/annals.1369.062. [DOI] [PubMed] [Google Scholar]
- Fleming R, Acheson S, Moore S, Wilson W, Swartzwelder H. In the rat, chronic intermittent ethanol exposure during adolescence alters the ethanol sensitivity of tonic inhibition in adulthood. Alcohol Clin Exp Res. 2012;36(2):279–285. doi: 10.1111/j.1530-0277.2011.01615.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fountain SB, Rowan JD, Kelley BM, Willey AR, Nolley EP. Adolescent exposure to nicotine impairs adult serial pattern learning in rats. Exp Brain Res. 2008;187(4):65–56. doi: 10.1007/s00221-008-1346-4. [DOI] [PubMed] [Google Scholar]
- Gass JT, Glen WB, Jr, McGonigal JT, Trantham-Davidson H, Lopez MF, Randall PK, Yaxley R, Floresco SB, Chandler LJ. Adolescent alcohol exposure reduces behavioral flexibility, promotes disinhibition, and increases resistance to extinction of ethanol self-administration in adulthood. Neuropsychopharmacology. 2014;39(11):2570–2583. doi: 10.1038/npp.2014.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilpin NW, Karanikas CA, Richardson HN. Adolescent binge drinking leads to changes in alcohol drinking, anxiety, and amygdalar corticotrophin releasing factor cells in adulthood in male rats. PLoS One. 2012;7(2):1–12. doi: 10.1371/journal.pone.0031466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hamilton DA, Brigman JL. Behavioral flexibility in rats and mice: contributions of distinct frontocortical regions. Genes Brain Behav. 2015;14(1):4–21. doi: 10.1111/gbb.12191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Higuera-Matas A, Soto-Montenegro ML, del Olmo N, Miquens M, Torres I, Vaquero JJ, Sanchez J, Garcia-Lecumberri C, Desco M, Ambrosio E. Augmented acquisition of cocaine self-administration and altered brain glucose metabolism in adult female but not male rats exposed to a cannabinoid agonist during adolescence. Neuropsychopharmacology. 2008;33(4):806–813. doi: 10.1038/sj.npp.1301467. [DOI] [PubMed] [Google Scholar]
- Higuera-Matas A, Botreau F, Miguens M, Del Olmo N, Borcel E, Perez-Alvarez L, Garcia-Lecumberri C, Ambrosio E. Chronic periadolescent cannabinoid treatment enhances adult hippocampal PSA-NCAM expression in male Wistar rats but only has marginal effects on anxiety, learning and memory. Pharmacol Biochem Behav. 2009;93(4):482–490. doi: 10.1016/j.pbb.2009.06.013. [DOI] [PubMed] [Google Scholar]
- Higuera-Matas A, Ucha M, Ambrosio E. Long-term consequences of perinatal and adolescent cannabinoid exposure on neural and psychological processes. Neurosci Biobehav Rev. 2015;55:119–146. doi: 10.1016/j.neubiorev.2015.04.020. [DOI] [PubMed] [Google Scholar]
- Ho A, Chin AJ, Dole VP. Early experience and the consumption of alcohol by adult C57BL/6J mice. Alcohol. 1989;6:511–515. doi: 10.1016/0741-8329(89)90060-8. [DOI] [PubMed] [Google Scholar]
- Jager G, Ramsey NF. Long-term consequences of adolescent cannabis exposure on the development of cognition, brain structure and function: an overview of animal and human research. Curr Drug Abuse Rev. 2008;1(2):114–123. doi: 10.2174/1874473710801020114. [DOI] [PubMed] [Google Scholar]
- Johnston LD, O’Malley PM, Miech RA, Bachman JG, Schulenberg JE. Monitoring the Future: national results on adolescent drug use: Overview of key findings. MI: Ann Arbor; 2016. [Google Scholar]
- Kandel DB, Kandel ER. A molecular basis for nicotine as a gateway drug. N Engl J Med. 2014;371(21):2038–2039. doi: 10.1056/NEJMc1411785. [DOI] [PubMed] [Google Scholar]
- Kantak KM, Barlow N, Tassin DH, Brisotti MF, Jordan CJ. Performance on a strategy set shifting task in rats following adult or adolescent cocaine exposure. Psychopharmacology (Berl) 2014;231(23):4489–4501. doi: 10.1007/s00213-014-3598-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karila L, Zarmdini R, Petit A, Lafaye G, Lowenstein W, Reynaud M. [Cocaine addiction: current data for the clinician] Presse Med. 2014;43(1):9–17. doi: 10.1016/j.lpm.2013.01.069. [DOI] [PubMed] [Google Scholar]
- Keeley RJ, Trow J, Bye C, McDonald RJ. Part II: Strain- and sex-specific effects of adolescent exposure to THC on adult brain and behavior: Variants of learning, anxiety and volumetric estimates. Behav Br Res. 2015;288:132–152. doi: 10.1016/j.bbr.2015.01.001. [DOI] [PubMed] [Google Scholar]
- Kelley BM, Rowan JD. Long-term low-level adolescent nicotine exposure produces dose-dependent changes in cocaine sensitivity and reward in adult mice. Int J Dev Neurosci. 2004;22(5-6):339–348. doi: 10.1016/j.ijdevneu.2004.04.002. [DOI] [PubMed] [Google Scholar]
- Kelly PA, Ritchie IM, McBean DE, Olverman HJ. Functional consequences of perinatal exposure to 3,4-methylenedioxymethamphetamine in rat brain. Br J Pharmacol. 2002;137(7):963–970. doi: 10.1038/sj.bjp.0704961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kindlundh-Högberg AM, Blomqvist A, Malki R, Schiöth HB. Extensve neuroadaptive changes in cortical gene-transcrdipt expressions of the glutamate system in response to repeated intermittent MDMA administration in adolescent rats. BMC Neurosci. 2008;9:39. doi: 10.1186/1471-2202-9-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klomp A, den Hollander B, de Bruin K, Booij J, Remenan L. The effects of ectasy (MDMA) on brain serotonin transporters are dependent on age-of-first exposure in recreational users and animals. PLoS One. 2012;7(10):e47524. doi: 10.1371/journal.pone.0047524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kokoshka JM, Fleckenstein AE, Wilkins DG, Hanson GR. Age-dependent differential responses of monoaminergic systems to high doses of methamphetamine. J Neurochem. 2000;75(5):2095–2102. doi: 10.1046/j.1471-4159.2000.0752095.x. [DOI] [PubMed] [Google Scholar]
- Koob GF, LeMoal M. Neurobiology of addiction. San Diego, CA: Elsevier; 2006. [Google Scholar]
- Kuhn CM, Wilson W, Swartzwelder S. Enduring effects of adolescent drug exposure. Biol Psychiatry. 2013;74(7):480–4881. doi: 10.1016/j.biopsych.2013.07.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee TT, Wainright SR, Hill MN, Galea LA, Gorzalka BB. Sex, drugs, and adult neurogenesis: sex-dependent effects of escalating adolescent cannabinoid exposure on adult hippocampal neurogenesis, stress reactivity, and amphetamine sensitization. Hippocampus. 2014;24(3):280–292. doi: 10.1002/hipo.22221. [DOI] [PubMed] [Google Scholar]
- Levin ED, Slade S, Wells S, Cauley M, Petro A, Vendittelli A, Johnson M, Williams P, Horton K, Rezvani AH. Behav Brain Res. 2011;225(2):473–481. doi: 10.1016/j.bbr.2011.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lopez-Gallardo M, Lopez-Rodriguez AB, Llorente-Berzal A, Rotllant D, Mackie K, Armario A, Nadal R, Viveros MP. Maternal deprivation and adolescent cannabinoid exposure impact hippocampal astrocytes, CB1 receptors and brain-derived neurotrophic factor in a sexually dimorphic fashion. Neuroscience. 2012;204:90–103. doi: 10.1016/j.neuroscience.2011.09.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lopez-Rodriguez AB, Llorente-Berzal A, Garcia-Segura LM, Viveros MP. Sex-dependent long-term effects of adolescent exposure to THC and/or MDMA on neuroinflammation and serotonergic and cannabinoid systems in rats. Br J Pharmacol. 2014;171(6):1435–1447. doi: 10.1111/bph.12519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lubman DI, Cheetham A, Yucel M. Cannabis and adolescent brain development. Pharmacol Ther. 2015;148:1–16. doi: 10.1016/j.pharmthera.2014.11.009. [DOI] [PubMed] [Google Scholar]
- Maldonado-Devincci AM, Badanich KA, Kirstein CL. Alcohol during adolescence selectively alters immediate and long-term behavior and neurochemistry. Alcohol. 2010;44:57–66. doi: 10.1016/j.alcohol.2009.09.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malone D, Hill M, Rubino T. Adolescent cannabis use and psychosis: Epidemiology and neurodevelopmental models. Br J Pharmacol. 2010;160(3):511–522. doi: 10.1111/j.1476-5381.2010.00721.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mateos-Garcia A, Roger-Sanchez C, Rodriguez-Arias M, Minarro J, Aguilar MA, Manzanedo C, Arenas MC. Higher sensitivity to the conditioned rewarding effects of cocaine and MDMA in High-Novelty-Seekers mice exposed to a cocaine binge during adolescence. Psychopharmacology (Berl) 2015;232(1):101–113. doi: 10.1007/s00213-014-3642-y. [DOI] [PubMed] [Google Scholar]
- Matthews DB, Tinsley KL, Diaz-Granados JL, Tokunaga S, Silvers JM. Chronic intermittent exposure to ethanol during adolescence produces tolerance to the hypnotic effects of ethanol in male rats: A dose-dependent analysis. Alcohol. 2008;42(8):617–621. doi: 10.1016/j.alcohol.2008.09.001. [DOI] [PubMed] [Google Scholar]
- McFadden LM, Hoonakker AJ, Vieira-Brock PL, Stout KA, Sawada NM, Ellis JD, Allen SC, Walters ET, Nielsen SM, Gibb JW, Alburges ME, Wilkins DG, Hanson GR, Fleckenstein AE. Methamphetamine treatment during development attenuates the dopaminergic deficits caused by subsequent high-dose methamphetamine administration. Synapse. 2011;65(8):771–777. doi: 10.1002/syn.20902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McFadden LM, Hadlock GC, Allen SC, Vieira-Brock PL, Stout KA, Ellis JD, Hoonaker AJ, Andrenyak DM, Nielsen SM, Wilkins DG, Hanson GR, Fleckenstein AE. J Pharmacol Exp Ther. 2012;340(2):295–303. doi: 10.1124/jpet.111.188433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McPherson CS, Lawrence AJ. Exposure to amphetamine in rats during periadolescence establishes behavioral and extrastriatal neural sensitization to adulthood. Int J Neuropsychopharmacol. 2006;9(4):377–392. doi: 10.1017/S1461145705005845. [DOI] [PubMed] [Google Scholar]
- McQuown SC, Belluzzi JD, Leslie FM. Low dose nicotine treatment during early adolescence increases subsequent cocaine reward. Neurotoxicol Teratol. 2007;29(1):66–73. doi: 10.1016/j.ntt.2006.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Montesinos J, Pascual M, Rodriguez-Aris M, Minarro J, Guerri C. Involvement of TLR4 in the long-term epigenetic changes, rewarding and anxiety effects induced by intermittent ethanol treatment in adolescence. Brain Behav Immun. 2015a doi: 10.1016/j.bbi.2015.12.006. [DOI] [PubMed] [Google Scholar]
- Montesinos J, Pascual M, Pla A, Maldonado C, Rodriguez-Aris M, Minarro J, Guerri C. TLR4 elimination prevents synaptic and myelin alterations and long-term cognitive dysfunctions in adolescent mice with intermittent ethanol treatment. Brain Behav Immun. 2015b doi: 10.1016/j.bbi.2014.11.015. [DOI] [PubMed] [Google Scholar]
- Nasrallah N, Yang T, Bernstein I. Long-term risk preference and suboptimal decision making following adolescent alcohol use. Proc Natl Acad Sci USA. 2009;106(41):17600–17604. doi: 10.1073/pnas.0906629106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nolley EP, Kelley BM. Adolescent reward system perseveration due to nicotine: studies with methylphenidate. Neurotoxicol Teratol. 2007;29(1):47–56. doi: 10.1016/j.ntt.2006.09.026. [DOI] [PubMed] [Google Scholar]
- O’Shea M, Singh M, McGregor I, Mallet P. Chronic cannabinoid exposure produces lasting memory impairment and increased anxiety in adolescent but not adult rats. J Psychopharmacol. 2004;18(4):502–508. doi: 10.1177/026988110401800407. [DOI] [PubMed] [Google Scholar]
- O’Shea M, McGregor I, Mallet P. Repeated cannabinoid exposure during perinatal, adolescent or early adult ages produces similar longlasting deficits in object recognition and reduced social interaction in rats. J Psychopharmacol. 2006;20(5):611–621. doi: 10.1177/0269881106065188. [DOI] [PubMed] [Google Scholar]
- O’Tuathaugh CM, Hryniewiecka M, Behan A, Tighe O, Coughian C, Desbonnet L, Cannon M, Karaviorgou M, Gogos JA, Colter DR, Waddington JL. Chronic adolescent exposure to delta-9-tetrahydrocannibol in COMT mutant mice: impact on psychosis-related and other phenotypes. Neuropscyhopharmacology. 2010;35(11):2262–2273. doi: 10.1038/npp.2010.100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pandey SC, Akharkar AJ, Tang L, Zhang H. Potential role of adolescent alcohol exposure-induced amygdaloid histone modifications in anxiety and alcohol intake during adulthood. Neurobiol Dis. 2015;82:607–619. doi: 10.1016/j.nbd.2015.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pascual M, Blanco A, Cauli O, Minarro J, Guerri C. Intermittent ethanol exposure induces inflammatory brain damage and causes long-term behavioural alterations in adolescent rats. Eur J Neurosci. 2007;25(2):541–550. doi: 10.1111/j.1460-9568.2006.05298.x. [DOI] [PubMed] [Google Scholar]
- Pascual M, Boix J, Felipo V, Guerri C. Repeated alcohol administration during adolescence causes changes in the mesolimbic dopaminergic and glutamatergic systems and promotes alcohol intake in the adult rat. J Neurochem. 2009;108(4):920–931. doi: 10.1111/j.1471-4159.2008.05835.x. [DOI] [PubMed] [Google Scholar]
- Pascual M, Do Couto BR, Alfonso-Loeches S, Aguilar MA, Rodriguez-Aris M, Guerri C. Changes in histone acetylation in the prefrontal cortex of ethanol-exposed adolescent rats are associated with ethanol-induced place conditioning. Neuropharmacology. 2012;62(7):2309–2319. doi: 10.1016/j.neuropharm.2012.01.011. [DOI] [PubMed] [Google Scholar]
- Patrick ME, Schulenberg JE, Martz ME, Maggs JL, O’Malley PM, Johnston LD. Extreme binge drinking among 12th-grade students in the United States: prevalence and predictors. JAMA Pediatr. 2013;167:1019–1025. doi: 10.1001/jamapediatrics.2013.2392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Philpot R, Wecker L, Kirstein C. Repeated ethanol exposure during adolescence alters the development trajectory of dopaminergic output from the nucleus accumbens septi. Int J Dev Neurosci. 2009;27(8):805–815. doi: 10.1016/j.ijdevneu.2009.08.009. [DOI] [PubMed] [Google Scholar]
- Piper BJ, Meyer JS. Memory deficit and reduced anxiety in young adult rats given repeated intermittent MDMA treatment during the periadolescent period. Pharmacol Biochem Behav. 2004;79(4):723–731. doi: 10.1016/j.pbb.2004.10.001. [DOI] [PubMed] [Google Scholar]
- Piper BJ, Vu HL, Safain MG, Oliver AJ, Meyer S. Repeated adolescent 3,4-methylenedioxymethamphetamine (MDMA) exposure in rats attenuates the effects of a subsequent challenge with MDMA or a 5-hydroxytryptamine (1A) receptor agonist. J Pharmacol Exp Ther. 2006;317(2):838–849. doi: 10.1124/jpet.105.095760. [DOI] [PubMed] [Google Scholar]
- Popovic M, Caballero-Bleds M, Puelles L, Guerri C. Multiple binge alcohol consumption during rat adolescence increases anxiety but does not impair retention in the passive avoidance task. Neurosci Lett. 2004;357(2):79–82. doi: 10.1016/j.neulet.2003.10.046. [DOI] [PubMed] [Google Scholar]
- Quinn HR, Matsumoto I, Callaghan PD, Long LE, Arnold JC, Gunasekaran N, Thompson MR, Dawson B, Mallet PE, Kashem MA, Matsuda-Matsumoto H, Iwazaki T, McGregor IS. Adolescent rats find repeated delta-9-THC less aversive than adult rats but display greater residual cognitive deficits and changes in hippocampal protein expression following exposure. Neuropsychopharmacology. 2008;33(5):1113–1126. doi: 10.1038/sj.npp.1301475. [DOI] [PubMed] [Google Scholar]
- Quoilin C, Didone V, Tirelli E, Quertemont E. Chronic ethanol exposure during adolescence alters the behavioral responsiveness to ethanol in adult mice. Behav Brain Res. 2012;299(1):1–9. doi: 10.1016/j.bbr.2011.12.039. [DOI] [PubMed] [Google Scholar]
- Rau KS, Truong JG, Wilkins DG, Fleckenstein AE, Hanson GR. Age-dependent effects of methamphetamine on VMAT-2. Ann NY Acad Sci. 2006;1074:154–159. doi: 10.1196/annals.1369.015. [DOI] [PubMed] [Google Scholar]
- Realini N, Vigano D, Guidali C, Zamberletti E, Rubino T, Parolaro D. Chronic URB597 treatment at adulthood reverted most depressive-like symptoms induced by adolescent exposure to THC in female rats. Neuropharmacology. 2011;60(2–3):235–243. doi: 10.1016/j.neuropharm.2010.09.003. [DOI] [PubMed] [Google Scholar]
- Renard J, Krebs MO, LePen G, Jay TM. Long-term consequences of adolescent cannabinoid exposure in adult psychopathology. Front Neurosci. 2014;8:361. doi: 10.3389/fnins.2014.00361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Renaud J, Rosen LG, Loureiro M, De Oliveira C, Schmid S, Rushlow WJ, Laviolette SR. Adolescent cannabinoid exposure induces a persistent sub-cortical hyper-dopaminergic state and associated molecular adaptations in the prefrontal cortex. Cereb Cortex. 2016;2016:1–14. doi: 10.1093/cercor/bhv335. [DOI] [PubMed] [Google Scholar]
- Riddle EL, Kokoshka JM, Wilkins DG, Hanson GR, Fleckenstein AE. Tolerance to the neurotoxic effects of methamphetamine in young rats. Eur J Pharmacol. 2002;435(2–3):181–185. doi: 10.1016/s0014-2999(01)01592-8. [DOI] [PubMed] [Google Scholar]
- Risher ML, Fleming RL, Boutros N, Semenova S, Wilson WA, Levin ED, Markou A, Swartzwelder HS, Acheson SK. Long-term effects of chronic intermittent ethanol exposure in adolescent and adult rats: radial-arm maze performance and operant food reinforced responding. PLoS One. 2013;8(5):e62940. doi: 10.1371/journal.pone.0062940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Risher ML, Sexton HG, Risher WC, Wilson WA, Fleming RL, Madison RD, Moore SD, Eroglu C, Swartzwelder HS. Adolescent intermittent alcohol exposure: dysregulation of thrombospondins and synapse formation are associated with decreased neuronal density in the adult hippocampus. Alcohol Clin Exp Res. 2015;39(12):2403–2413. doi: 10.1111/acer.12913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robinson G, Most D, Ferguson LB, Mayfield J, Harris RA, Blednov YA. Neuroimmune pathways in alcohol consumption: evidence from behavioral and genetic studies in rodents and humans. Int Rev Neurobiol. 2014;118:13–39. doi: 10.1016/B978-0-12-801284-0.00002-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodd-Henricks ZA, Bell RL, Kuc KA, Murphy JM, McBride WJ, Lumeng L, Li TK. Effects of ethanol exposure on subsequent acquisition and extinction of ethanol self-administration and expression of alcohol-seeking behavior in adult alcohol-preferring (P) rats: I. Periadolescent exposure. Alcohol Clin Exp Res. 2002a;26(11):1632–1641. doi: 10.1097/01.ALC.0000036301.36192.BC. [DOI] [PubMed] [Google Scholar]
- Rodd-Henricks ZA, Bell RL, Kuc KA, Murphy JM, McBride WJ, Lumeng L, Li TK. Effects of ethanol exposure on subsequent acquisition and extinction of ethanol self-administration and expression of alcohol-seeking behavior in adult alcohol-preferring (P) rats: II. Adult exposure. Alcohol Clin Exp Res. 2002b;26(11):1642–1652. doi: 10.1097/01.ALC.0000036302.73712.9D. [DOI] [PubMed] [Google Scholar]
- Rubino T, Vigano D, Realini N, Guidali C, Braida D, Capurro V, Castiglioni C, Cherubino F, Romauldi P, Candeletti S, Sala M, Parolaro D. Chronic delta 9-tetrahydrocannabinol during adolescence provokes sex-dependent changes in the emotional profile in adult rats: behavioral and biochemical correlates. Neuropyschopharmacology. 2008;33(11):2760–2771. doi: 10.1038/sj.npp.1301664. [DOI] [PubMed] [Google Scholar]
- Rubino T, Realini N, Braida D, Alberio T, Capurro V, Vigano D, Giudali C, Sala M, Fasano M, Pasolaro D. The depressive phenotype induced in adult female rats by adolescent exposure to THC is associated with cognitive impairment and altered neuroplasticity in the prefrontal cortex. Neurotox Res. 2009a;15(4):291–302. doi: 10.1007/s12640-009-9031-3. [DOI] [PubMed] [Google Scholar]
- Rubino T, Realini N, Braida D, Guidi S, Capurro V, Viagno D, Guidali C, Pinter M, Sala M, Bartesaghi R, Paroloro D. Changes in hippocampal morphology and neuroplasticity induced by adolescent THC treatment are associated with cognitive impairment in adulthood. Hippocampus. 2009b;19(8):763–772. doi: 10.1002/hipo.20554. [DOI] [PubMed] [Google Scholar]
- Saalfield J, Spear LP. Consequences of repeated ethanol exposure during early or late adolescence on conditioned taste aversions in rats. Dev Cogn Neurosci. 2015;16:174–182. doi: 10.1016/j.dcn.2015.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sahr AE, Thielen RJ, Lumeng L, Li T-K, McBride WJ. Long-lasting alterations of the mesolimbic dopamine system after periadolescent ethanol drinking by alcohol-preferring rats. Alcohol Clin Exp Res. 2004;28(5):702–711. doi: 10.1097/01.alc.0000125344.79677.1c. [DOI] [PubMed] [Google Scholar]
- Schindler AG, Tsutsui KT, Clark JJ. Chronic alcohol intake during adolescence, but not adulthood, promotes persistent deficits in risk-based decision making. Alcohol Clin Exp Res. 2014;38(6):1622–1629. doi: 10.1111/acer.12404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schneider M. Puberty as a highly vulnerable developmental period for the consequences of cannabis exposure. Addict Biol. 2008;13(2):253–263. doi: 10.1111/j.1369-1600.2008.00110.x. [DOI] [PubMed] [Google Scholar]
- Schneider M, Koch M. Deficient social and play behavior in juvenile and adult rats after neonatal cortical lesion: effects of chronic pubertal cannabinoid treatment. Neuropsychopharmacology. 2005;30(5):944–957. doi: 10.1038/sj.npp.1300634. [DOI] [PubMed] [Google Scholar]
- Schneider M, Koch M. The effect of chronic peripubertal cannabinoid treatment on deficient object recognition memory in rats after neonatal mPFC lesion. Eur Psychopharmacol. 2007;17(3):180–186. doi: 10.1016/j.euroneuro.2006.03.009. [DOI] [PubMed] [Google Scholar]
- Schneider M, Schomig E, Leweke FM. Acute and chronic cannabinoid treatment differentially affects recognition memory and social behavior in pubertal and adult rats. Addict Biol. 2008;13(3–4):345–357. doi: 10.1111/j.1369-1600.2008.00117.x. [DOI] [PubMed] [Google Scholar]
- Schulz S, Becker T, Nagel U, vonAmeln-Mayerhofer A, Koch M. Chronic co-administration of the cannabinoid receptor against WIN55, 212-2 during puberty or adulthood reverses 3,4 methylenedioxymetamphetamine (MDMA)-induced deficits in recognition memory but not in effort-based decision making. Pharmacol Biochem Behav. 2013;106:91–100. doi: 10.1016/j.pbb.2013.03.011. [DOI] [PubMed] [Google Scholar]
- Segev A, Rubin AS, Abush H, Richter-Levin G, Akirav I. Cannabinoid receptor activation prevents the effects of chronic mild stress on emotional learning and LTP in a rat model of depression. Neuropsychopharmacology. 2014;39(4):919–933. doi: 10.1038/npp.2013.292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Semple D, McIntosh A, Lawrie S. Cannabis as a risk factor for psychosis: systematic review. J Psychopharmacol. 2005;19(2):187–194. doi: 10.1177/0269881105049040. [DOI] [PubMed] [Google Scholar]
- Shanks RA, Ross JM, Doyle HH, Helton AK, Picou BN, Schulz J, Tavares C, Bryant S, Dawson BL, Lloyd SA. Adolescent exposure to cocaine, amphetamine, and methylphenidate cross-sensitizes adults to methamphetamine with drug- and sex-specific effects. Behav Brain Res. 2015;281:116–124. doi: 10.1016/j.bbr.2014.12.002. [DOI] [PubMed] [Google Scholar]
- Sherrill LK, Stanis JJ, Gulley JM. Age-dependent effects of repeated amphetamine exposure on working memory in rats. Behav Brain Res. 2013;242:84–94. doi: 10.1016/j.bbr.2012.12.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siciliano D, Smith R. Periadolescent alcohol alters adult behavioral characteristics in the rat. Physiol Behav. 2001;74(4–5):637–643. doi: 10.1016/s0031-9384(01)00623-0. [DOI] [PubMed] [Google Scholar]
- Siegel JA, Park BS, Raber J. Methamphetamine exposure during brain development alters the brain acetylcholine system in adolescent mice. J. Neurochem. 2011;119(1):89–99. doi: 10.1111/j.1471-4159.2011.07418.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sillivan SE, Black YD, Naydenov AV, Vassoler FR, Hanlin RP, Konradi C. Binge cocaine administration in adolescent rats affects amygdalar gene expression patterns and alters anxiety-related behavior in adulthood. Biol Psychiatry. 2011;70(6):583–592. doi: 10.1016/j.biopsych.2011.03.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Silvers JM, Tokunaga S, Mittleman G, O’Buckley T, Morrow AL, Matthews DB. Chronic intermittent ethanol exposure during adolescence reduces the effect of ethanol challenge on hippocampal allopregnanolone levels and Morris water maze task performance. Alcohol. 2006;39:151–158. doi: 10.1016/j.alcohol.2006.09.001. [DOI] [PubMed] [Google Scholar]
- Sircar R, Sircar D. Adolescent rats exposed to repeated ethanol treatments show lingering behavioral impairments. Alcohol Clin Exp Res. 2005;29(8):1402–1410. doi: 10.1097/01.alc.0000175012.77756.d9. [DOI] [PubMed] [Google Scholar]
- Sircar R, Sircar D. Repeated ethanol treatment in adolescent rats alters cortical NMDA receptor. Alcohol. 2006;39(1):51–58. doi: 10.1016/j.alcohol.2006.07.002. [DOI] [PubMed] [Google Scholar]
- Slawecki C, Betancourt M. Effects of adolescent ethanol exposure on ethanol consumption in adult rats. Alcohol. 2002;26(1):23–30. doi: 10.1016/s0741-8329(01)00192-6. [DOI] [PubMed] [Google Scholar]
- Slawecki C, Gilder A, Roth J, Ehlers E. Increased anxiety-like behavior in adult rats exposed to nicotine as adolescents. Pharmacol Biochem Behav. 2003;75(2):355–361. doi: 10.1016/s0091-3057(03)00093-5. [DOI] [PubMed] [Google Scholar]
- Slawecki CJ, Thorsell AK, Ehlers CL. Long-term neurobehavioral effects of alcohol or nicotine exposure in adolescent animal models. Ann NY Acad Sci. 2004;1021:448–452. doi: 10.1196/annals.1308.062. [DOI] [PubMed] [Google Scholar]
- Slawecki CJ, Thorsell AK, Khoury AE, Mathe AA, Ehlers CL. Increased CRF-like and NPY-like immunoreactivity in adult rats exposed to nicotine during adolescence: Relation to anxiety-like and depressive-like behavior. Neuropeptides. 2005;39(4):369–377. doi: 10.1016/j.npep.2005.06.002. [DOI] [PubMed] [Google Scholar]
- Slotkin TA. Nicotine and the adolescent brain: Insights from an animal model. Neurotoxicol Teratol. 2002;24(3):369–384. doi: 10.1016/s0892-0362(02)00199-x. [DOI] [PubMed] [Google Scholar]
- Slotkin TA, Cousins MM, Seidler FJ. Administration of nicotine to adolescent rats evokes regionally selective upregulation of CNS alpha 7 nicotinic acetylcholine receptors. Brain Res. 2004;1030(1):159–163. doi: 10.1016/j.brainres.2004.10.009. [DOI] [PubMed] [Google Scholar]
- Slotkin TA, MacKillop EA, Rudder CL, Ryde IT, Tate CA, Seidler FJ. Permanent, sex-selective effects of prenatal or adolescent nicotine exposure, separately or sequentially, in rat brain regions: Indices of cholinergic and serotonergic synaptic function, cell signaling, and neural cell number and size at 6 months of age. Neuropsychopharmacology. 2007;32(5):1082–1097. doi: 10.1038/sj.npp.1301231. [DOI] [PubMed] [Google Scholar]
- Smith LN, McDonald CG, Bergstrom HC, Brielmaier JM, Eppolito AK, Wheeler TL, Falco AM, Smith RF. Long-term changes in fear conditioning and anxiety-like behavior following nicotine exposure in adult versus adolescent rats. Pharmacol Biochem Behav. 2006;85(1):91–97. doi: 10.1016/j.pbb.2006.07.014. [DOI] [PubMed] [Google Scholar]
- Smith RF, McDonald CG, Bergstrom HC, Ehlinger DG, Brielmaier JM. Adolescent nicotine induces persisting changes in development of neural connectivity. Neurosci Biobehav Rev. 2015;55:432–443. doi: 10.1016/j.neubiorev.2015.05.019. [DOI] [PubMed] [Google Scholar]
- Spear LP. The adolescent brain and age-related behavioral manifestations. Neurosci Biobehav Rev. 2000;24(4):417–463. doi: 10.1016/s0149-7634(00)00014-2. [DOI] [PubMed] [Google Scholar]
- Spear LP. Rewards, aversions and affect in adolescence: Emerging convergences across laboratory animal and human data. Dev Cogn Neurosci. 2011;1:390–403. doi: 10.1016/j.dcn.2011.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spear LP. Adolescent alcohol exposure: Are there separable vulnerable periods within adolescence? Physiol Behav. 2015;148:122–130. doi: 10.1016/j.physbeh.2015.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spear LP. Alcohol consumption in adolescence: a translational perspective. Curr Addict Rep. 2016a;3(1):50–61. [Google Scholar]
- Spear LP. A developmental biological perspective of adolescent substance abuse: Animal models. In: Brown S, Zucker R, editors. The Oxford Handbook of Adolescent Substance Abuse. 2016b. [Google Scholar]
- Spear LP, Swartzwelder HS. Adolescent alcohol exposure and persistence of adolescent-typical pheontypes into adulthood: a mini-review. Neurosci Biobehav Rev. 2014;45:1–8. doi: 10.1016/j.neubiorev.2014.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stopponi S, Soverchia L, Ubaldi M, Cippitelli A, Serpelloni G, Ciccocioppo R. Chronic THC during adolescence increases the vulnerability to stress-induced relapse to heroin seeking in adult rats. Eur Neuropsychopharmacol. 2014;24(7):1037–1045. doi: 10.1016/j.euroneuro.2013.12.012. [DOI] [PubMed] [Google Scholar]
- Strong M, Yoneyama N, Fretwell A, Snelling C, Tanchuck M, Finn D. “Binge” drinking experience in adolescent mice shows sex differences and elevated ethanol intake in adulthood. Horm Behav. 2010;58(1):82–90. doi: 10.1016/j.yhbeh.2009.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Substance Abuse and Mental Health Services Administration (SAMHSA) Results from the 2012 national survey on drug use and health: summary of national findings, NSDUH Series H-46, HHS Publication No. (SMA) 13–4795. Rockville, MD: Substance Abuse and Mental Health Services Administration; 2013. [Google Scholar]
- Swartzwelder HS, Acheson SK, Miller KM, Sexton HG, Liu W, Crews FT, Risher ML. Adolescent intermittent alcohol exposure: Deficits in object recognition memory and forebrain cholinergic markers. PLoS One. 2015;10(11):e0140042. doi: 10.1371/journal.pone.0140042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tambour S, Brown LL, Crabbe JC. Gender and age at drinking onset affect voluntary alcohol consumption but neither the alcohol deprivation effect nor the response to stress in mice. Alcohol Clin Exp Res. 2008;32(12):2100–2106. doi: 10.1111/j.1530-0277.2008.00798.x. [DOI] [PubMed] [Google Scholar]
- Teixeira-Gomes A, Costa VM, Feio-Azevedo R, Bastos M, de Lourdes Carvalho F, Capela JP. The neurotoxicity of amphetamines during the adolescent period. Int J Dev Neurosci. 2015;41:44–62. doi: 10.1016/j.ijdevneu.2014.12.001. [DOI] [PubMed] [Google Scholar]
- Toalston JE, Deehan GA, Jr, Hauser SR, Engelman EA, Bell RL, Murphy JM, Truitt WA, McBride WJ, Rodd ZA. Reinforcing properties and neurochemical response of ethanol within the posterior ventral tegmental area are enhanced in adulthood by periadolescent ethanol consumption. J Pharmacol Exp Ther. 2014;351(2):317–326. doi: 10.1124/jpet.114.218172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trezza V, Cuomo V, Vanderschuren LJ. Cannabis and the developing brain: insights from behavior. Eur J Pharmacol. 2008;585(2–3):441–452. doi: 10.1016/j.ejphar.2008.01.058. [DOI] [PubMed] [Google Scholar]
- Truong JG, Wilkins DG, Baudys J, Crouch DJ, Johnson-Davis KL, Gibb JW, Hanson GR, Fleckenstein AE. Age-dependent methamphetamine-induced alterations in vesicular monoamine transporter-2 function: implications for neurotoxicity. J Pharmacol Exp Ther. 2005;314(3):1087–1092. doi: 10.1124/jpet.105.085951. [DOI] [PubMed] [Google Scholar]
- Varlinskaya EI, Spear LP. Social consequences of ethanol: Impact of age, stress, and prior history of ethanol exposure. Physiol Behav. 2015;148:145–150. doi: 10.1016/j.physbeh.2014.11.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Varlinskaya EI, Truxell EM, Spear LP. Chronic intermittent ethanol during adolescence: effects on social behavior and ethanol sensitivity in adulthood. Alcohol. 2014;48(5):433–444. doi: 10.1016/j.alcohol.2014.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vetreno RP, Broadwater M, Liu W, Spear LP, Crews FT. Adolescent, but not adult, binge ethanol exposure leads to persistent global reductions of choline acetyltransferase expressing neurons in brain. PLoS One. 2014;9(11):e113421. doi: 10.1371/journal.pone.0113421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vetreno RP, Crews FT. Binge ethanol exposure during adolescence leads to a persistent loss of neurogenesis in the dorsal and ventral hippocampus that is associated with impaired adult cognitive functioning. Front Neurosci. 2015;9:35. doi: 10.3389/fnins.2015.00035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vetter CS, Doremus-Fitzwater TL, Spear LP. Time-course of elevated ethanol intake in adolescent relative to adult rats under continuous, voluntary-access conditions. Alcohol Clin Exp Res. 2007;31(7):1159–1168. doi: 10.1111/j.1530-0277.2007.00417.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Viveros M-P, Marco EM, López-Gallardo M, Garcia-Segura LM, Wagner EJ. Framework for sex differences in adolescent neurobiology: A focus on cannabinoids. Neurosci Biobehav Rev. 2011;35:1740–1751. doi: 10.1016/j.neubiorev.2010.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Viveros M, Llorente R, Suarez J, Llorente-Berzal A, Lopez-Gallardo M, Rodriguez de Fonseca F. The endocannabinoid system in critical neurodevelopmental periods: Sex differences and neuropsychiatric implications. J Psychopharmacol. 2012;26(1):164–176. doi: 10.1177/0269881111408956. [DOI] [PubMed] [Google Scholar]
- Volz TJ, Farnsworth SJ, Fowley SD, Hanson GR, Fleckenstein AE. Age-dependent differences in dopamine transporter-2 function and their implications for methamphetamine neurotoxicity. Synapse. 2009;63(2):147–151. doi: 10.1002/syn.20580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vorhees CV, Reed TM, Morford LL, Fukumura M, Wood SL, Brown CA, Skelton MR, McCrea AE, Rock SL, Williams MT. Periadolescent rats (P41–50) exhibit increased *compared to juvenile (P21–30 or P31–40) or adult rats (P51–60) Neurotoxicol Teratol. 2005;27(1):117–134. doi: 10.1016/j.ntt.2004.09.005. [DOI] [PubMed] [Google Scholar]
- Walker BM, Ehlers CL. Appetitive motivational experience during adolescence results in enhanced alcohol consumption during adulthood. Behav Neurosci. 2009;124(4):926–935. doi: 10.1037/a0016002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, Lupica CR. Release of endogenous cannabinoids from ventral tegmental area dopamine neurons and the modulation of synaptic processes. Prog Neuropsychopharmacol Biol Psychiatry. 2014;52:24–27. doi: 10.1016/j.pnpbp.2014.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weaver M, Geier CF, Levin ME, Caggiula AR, Sved AF, Donny EC. Adolescent exposure to nicotine results in reinforcement enhancement but does not affect adult responding in rats. Drug Alcohol Depend. 2012;125(3):307–312. doi: 10.1016/j.drugalcdep.2012.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wegener N, Koch M. Behavioral disturbances and altered Fos protein expression in adult rats after chronic pubertal cannabinoid treatment. Brain Res. 2009;1253:81–91. doi: 10.1016/j.brainres.2008.11.081. [DOI] [PubMed] [Google Scholar]
- Wheeler AL, Lerch JP, Chakravarty MM, Friedel M, Sied JG, Fletcher PJ, Josselyn SA, Frankland PW. Adolescent cocaine exposure causes enduring macroscale changes in mouse brain structure. J Neurosci. 2013;33(5):1797–1803a. doi: 10.1523/JNEUROSCI.3830-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- White AM, Ghia AJ, Levin ED, Swartzwelder HS. Binge pattern ethanol exposure in adolescent and adult rats: Differential impact on subsequent responsiveness to ethanol. Alcohol Clin Exp Res. 2000;24(8):1251–1256. [PubMed] [Google Scholar]
- White AM, Bae JG, Truesdale MC, Ahmad S, Wilson WA, Swartzwelder HS. Chronic-intermittent ethanol exposure during adolescence prevents normal developmental changes in sensitivity to ethanol-induced motor impairments. Alcohol Clin Exp Res. 2002;26(7):960–968. doi: 10.1097/01.ALC.0000021334.47130.F9. [DOI] [PubMed] [Google Scholar]
- Wills T, Knapp DJ, Overstreet DH, Breese GR. Differential dietary ethanol intake and blood ethanol levels in adolescent and adult rats: Effects on anxiety-like behavior and seizure thresholds. Alcohol Clin Exp Res. 2008;32(8):1350–1360. doi: 10.1111/j.1530-0277.2008.00709.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wills T, Knapp D, Overstreet D, Breese GR. Sensitization, duration, and pharmacological blockade of anxiety-like behavior following repeated ethanol withdrawal in adolescent and adult rats. Alcohol Clin Exp Res. 2009;33(3):455–463. doi: 10.1111/j.1530-0277.2008.00856.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wong WC, Ford KA, Pagels NE, McCutcheon JE, Marinelli M. Adolescents are more vulnerable to cocaine addiction: behavioral and electrophysiological evidence. J Neurosci. 2013;33(11):4913–4922. doi: 10.1523/JNEUROSCI.1371-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yttri E, Burk J, Hunt P. Intermittent ethanol exposure in adolescent rats: Dose-dependent impairments in trace conditioning. Alcohol Clin Exp Res. 2004;28(10):1433–1436. doi: 10.1097/01.alc.0000147657.51745.a7. [DOI] [PubMed] [Google Scholar]
- Zamberletti E, Prini P, Speziali S, Gabaglio M, Solinas M, Parolaro D, Rubino T. Gender-dependent behavioral and biochemical effects of adolescent delta-9-tetrahydrocannabinol in adult maternally deprived rats. Neuroscience. 2012;204:245–257. doi: 10.1016/j.neuroscience.2011.11.038. [DOI] [PubMed] [Google Scholar]