Download as pdf or txt
Download as pdf or txt
You are on page 1of 28

Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

Contents lists available at ScienceDirect

Neuroscience and Biobehavioral Reviews


journal homepage: www.elsevier.com/locate/neubiorev

Review

Long-term consequences of perinatal and adolescent cannabinoid


exposure on neural and psychological processes
Alejandro Higuera-Matas ∗ , Marcos Ucha, Emilio Ambrosio
Department of Psychobiology, School of Psychology, National University of Distance Learning (UNED), C/ Juan del Rosal 10, 28040 Madrid, Spain

a r t i c l e i n f o a b s t r a c t

Article history: Marihuana is the most widely consumed illicit drug, even among adolescents and pregnant women. Given
Received 29 October 2014 the critical developmental processes that occur in the adolescent and fetal nervous system, marihuana
Received in revised form 30 March 2015 consumption during these stages may have permanent consequences on several brain functions in later
Accepted 29 April 2015
adult life. Here, we review what is currently known about the long-term consequences of perinatal and
Available online 8 May 2015
adolescent cannabinoid exposure. The most consistent findings point to long-term impairments in cogni-
tive function that are associated with structural alterations and disturbed synaptic plasticity. In addition,
Keywords:
several neurochemical modifications are also evident after prenatal or adolescent cannabinoid exposure,
Cannabinoid
Cognition
especially in the endocannabinoid, glutamatergic, dopaminergic and opioidergic systems. Important sex-
Emotion ual dimorphisms are also evident in terms of the long-lasting effects of cannabinoid consumption during
Drug addiction pregnancy and adolescence, and cannabinoids possibly have a protective effect in adolescents who have
Prenatal development suffered traumatic life challenges, such as maternal separation or intense stress. Finally, we suggest some
Adolescence future research directions that may encourage further advances in this exciting field.
Dopamine © 2015 Elsevier Ltd. All rights reserved.
Glutamate

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120
2. Epidemiology of marihuana use during critical developmental periods: pregnancy and adolescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120
3. The endocannabinoid system during perinatal development and adolescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
4. Role of the endocannabinoid system in the regulation of central nervous system development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
5. Perinatal cannabinoid exposure and long-lasting effects on neural and psychological processes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
5.1. Human studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
5.1.1. Cohort studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
5.1.2. Neuroimaging studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
5.2. Animal studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
5.2.1. Learning and memory . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
5.2.2. Emotional regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
5.2.3. Sensitivity to drugs of abuse later in life and potential addictive behavior. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 124
5.2.4. Monoaminergic systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 126
5.2.5. Glutamatergic and GABAergic systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 127
5.2.6. Endogenous opioid system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
5.2.7. Endogenous cannabinoid system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
6. Adolescent cannabinoid exposure and long-lasting effects on neural and psychological processes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
6.1. Human studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
6.1.1. Neuropsychological assessment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
6.1.2. Neuroimaging studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129

∗ Corresponding author at: Departamento de Psicobiología, Facultad de Psicología, UNED, C/ Juan del Rosal 10, 28040 Madrid, Spain. Tel.: +34 913989689;
fax: +34 913987491.
E-mail address: ahiguera@psi.uned.es (A. Higuera-Matas).

http://dx.doi.org/10.1016/j.neubiorev.2015.04.020
0149-7634/© 2015 Elsevier Ltd. All rights reserved.
120 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

6.2. Animal studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129


6.2.1. Learning and memory . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
6.2.2. Emotional regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
6.2.3. Sensitivity to drugs of abuse later in life and potential addictive behavior. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 134
6.2.4. Monoaminergic systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
6.2.5. Glutamatergic and GABAergic systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
6.2.6. Endogenous opioid system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
6.2.7. Endogenous cannabinoid system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
6.2.8. Cholinergic system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
6.2.9. Other signaling molecules and brain metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
6.2.10. Structural and synaptic plasticity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139
6.2.11. Cannabinoid exposure during adolescence as a normalizing agent against the deleterious effects of life challenges . . . . . . . . . 139
7. Concluding remarks and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 140

1. Introduction contributions into the long-lasting consequences of cannabinoid


exposure during adolescence. As for prenatal intake, there are also
Cannabis remains the most widely used illicit substance world- excellent reviews on cannabinoid exposure during adolescence and
wide, with an estimated annual prevalence of 3.9% of the adult the ensuing adult behavioral alterations (Chadwick et al., 2013;
population in 2013 (around 180 million users aged 15–64 years), Hurd et al., 2014; Realini et al., 2009; Renard et al., 2014; Rubino and
yet with marked regional differences (1.9% prevalence in Asia and Parolaro, 2008; Schneider, 2009, 2008; Trezza et al., 2012; Viveros
10.9% in Oceania for example: United Nations Office on Drugs and et al., 2011a,b), leading us to focus mainly on the most consistent or
Crime, 2013). The debate surrounding the possible legalization of interesting findings. We will first review the studies on long-term
cannabis has become quite lively, and with a growing number of cognitive alterations induced by adolescent cannabinoid exposure
countries and states in the USA having taken this step or contem- (see Table 5), before focusing on the emotional changes (including
plating doing so, it is crucial that such a decision is made based on anxiety or depressive-like behavior: Table 6), thereafter assessing
firm scientific grounds. the data available on the altered sensitivity to drugs of abuse found
Prenatal stages and adolescence are two crucial periods when in cannabinoid-exposed individuals. Subsequently, we will review
exposure to drugs of abuse affects the ongoing development of the the most important alterations found in the main neurotransmit-
central nervous system (CNS), altering the normal psychological ter systems (see Table 7), as well as focusing on the alterations
function of the individual. In this review, we will mainly focus on in morphological and synaptic plasticity induced by adolescent
the long-term effects of exposure to “cannabinoid agents” during cannabinoid exposure, and on some of the possible underlying
pregnancy and adolescence (see Table 1 for a list of the cannabinoid molecular mechanisms. Special attention will be paid to possible
agents cited in this review and their binding properties), while we protective actions of early cannabinoid exposure after traumatic
will not deal with acute effects of cannabinoids or those alterations life events, such as chronic stress or maternal deprivation.
studied immediately after chronic or repeated exposure. Sexual We believe that updating our knowledge of the long-term effects
dimorphism in the effects reported will be highlighted whenever of cannabis exposure during these two periods should help to clar-
studies were carried out on both sexes. Unless otherwise specified, ify how to cut the Gordian knot of marihuana legalization.
the sex of the animals tested is always masculine.
Although human studies provide the most relevant informa- 2. Epidemiology of marihuana use during critical
tion regarding the long-term influences of prenatal marihuana developmental periods: pregnancy and adolescence
exposure on later cognitive development and neural plasticity,
several confounding factors might limit the conclusions that can Marihuana is the illicit drug most widely consumed during preg-
be obtained from such studies, such as a possible malnutrition nancy (Brown and Graves, 2013; Calvigioni et al., 2014; Jaques
derived from the maternal consumption of cannabis, different 9 - et al., 2014), its consumption being strongly associated to cigarette
tetrahydrocannabinol (THC) consumption over time and between smoking (Gaalema et al., 2013) and to a lesser extent, to opi-
studies, or differences in educational background or socioeconomic oids, stimulants and alcohol (Burns et al., 2006). In the United
status. For this reason, animal studies are an invaluable tool when it States, approximately 4.6% of pregnant women reported having
comes to draw causal conclusions regarding the long-term effects consumed marihuana in the past month during the first trimester
of perinatal cannabinoid exposure. There are excellent reviews of pregnancy. This percentage went down to 2.9% during the second
on the behavioral consequences of prenatal THC and cannabinoid trimester and subsequently decreased to 1.4% in the third trimester
intake during pregnancy and lactation, summarizing our current (Substance Abuse and Mental Health Services Administration,
understanding of this field (Calvigioni et al., 2014; Campolongo 2009). In Brazil this percentage is similar, around 4.0% in the first
et al., 2011; Morris et al., 2011; Trezza et al., 2012; Wu et al., trimester (Mitsuhiro et al., 2007), as is the prevalence of its con-
2011). As such, we will only briefly review their most significant sumption in the general population of pregnant women in Europe.
findings. Along with these behavioral effects, prenatal exposure to For example, the prevalence of marihuana use during pregnancy
cannabinoids has consistently been associated with dysregulated in the UK was reported to be around 5% (Fergusson et al., 2002),
neurotransmission in the brain. Thus, we will also describe what is with a similar prevalence in Spain, where THC was detected in 5.3%
currently known about the neurochemical alterations induced by of newborns by meconium analysis (Lozano et al., 2007: although
in utero cannabis (see Table 4). this percentage rises to 10.3% in Ibiza, in the Balearic Islands
As for prenatal studies, human cohort designs of adolescent Friguls et al., 2012). In France, 1.2% of women reported having used
cannabis users and abusers might again include confounding fac- cannabis during pregnancy, a percentage that rose among younger
tors that affect the reliability of the results obtained. For this women, women living alone, or women who had a low level of
reason, the animal studies addressing this issue are crucial research education or low income (Saurel-Cubizolles et al., 2014). Notably,
A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146 121

Table 1
Ki values for ligands at human and rat cannabinoid receptors.

Human Rat

CB1 CB2 CB1 CB2

9 -THC 25.1 ± 5.54 35.2 ± 5.86 42.6 ± 5.01 13 ± 7.7


8 -THC NA NA 47.6 39.3
Cannabinol 525.3 ± 308.1 168.2 ± 32.16 368 ± 121.1 126.4
Cannabidiol 27542 2629.5 ± 230.5 2210.5 ± 558.08 1000
CP 55,940 3.1 ± 1.31 1.68 ± 0.54 0.94 ± 0.44 2.09 ± 0.72
WIN 55,512-2 62.4 ± 34.96 2.56 ± 1.16 7.17 ± 2.77 8.7 ± 7.5
Anandamide 239.2 ± 61.77 168.2 ± 32.16 368 ± 121.1 279
2-Arachidonoylglycerol 3423.6 ± 3288.24 1193.8 ± 327.71 1180.5 ± 538.59 1900 ± 1800

Adapted from Howlett et al. (2002), McPartland et al. (2007), Pertwee (2008).

when compared to the year prior to the survey, the reduction in play a role in embryonic stem-cell derived haematopoietic cell pro-
the prevalence of marihuana use during pregnancy in the UK is less liferation and lineage differentiation (Jiang et al., 2007).
than in the United States (see Moore et al., 2010), which is par- During brain development, the CB1R is expressed from very
ticularly interesting given that adolescent mothers are particularly early stages and its distribution within the brain resembles that
vulnerable and at risk for increased substance use when compared found in adults, as seen in rats (Berrendero et al., 1999, 1998;
with their nulliparous peers (reviewed in Chapman and Wu, 2013). Rodriguez de Fonseca et al., 1993) and humans (Glass et al., 1997;
Due to their young age, these mothers are likely to have subsequent Mato et al., 2003; Wang et al., 2003; Zurolo et al., 2010). The expres-
pregnancies that could also be adversely affected by substance use sion of the receptor in both species gradually increases during
(Cornelius et al., 2004). Indeed, even if the biological consequences development and it is found in areas related to the control of move-
of prenatal substance use are disregarded, postpartum use is a risk ment (e.g. the basal ganglia, cerebellum), cognition and attention
factor for substance abuse in children (U.S. Department of Health (e.g. the cerebral cortex), as well as emotions and memory (e.g. the
and Human Services, 1999). amygdala, hippocampus). Surprisingly, the CB1R is also present in
Marihuana is also the illicit drug most widely consumed dur- the white matter of the rat developing brain, where the receptor
ing adolescence both in the United States (Substance Abuse is not expressed significantly in the adult (Romero et al., 1997).
and Mental Health Services Administration, 2014a) and Europe This phenomenon has also been seen in humans (Mato et al., 2003),
(European Monitoring Centre for Drugs and Drug Addiction, 2014). where CB1R expression is elevated in the globus pallidus (Glass et al.,
In 2013, male adolescents showed a higher prevalence of last 1997; Wang et al., 2003) and substantia nigra pars reticulata (Glass
month use (approximately 8.4%) than female adolescents (approx- et al., 1997) when compared to the adult brain. During human
imately 5.6%) in the United States (Substance Abuse and Mental corticogenisis, the CB1R is expressed by Cajal-Retzius cells of the
Health Services Administration, 2014b). In Europe this same trend marginal zone, by migrating post-mitotic neurons, and by apical
is also observed with around 60% of the adolescents who reported and basal progenitors in the proliferative area of the ventricular
cannabis use in the last month being men (European Monitoring and subventricular zones (Saez et al., 2014; Zurolo et al., 2010). The
Centre for Drugs and Drug Addiction, 2014). In Spain, cannabis is expression of CB1R by projection neurons increases gradually as
also the illicit drug most widely consumed by adolescents (around differentiation proceeds, reaching a peak at the moment they reach
26.6% of people in this age segment report cannabis consumption in the cortical plate (Zurolo et al., 2010). All these findings suggest a
the last month), where male adolescents again show higher preva- specific role of the eCB system in brain development.
lence rates than females (Delegación del Gobierno para el Plan Although CB1R mRNA expression in the rat brain does not
Nacional sobre Drogas, 2013). change from the postnatal period to adulthood (McLaughlin et al.,
In the light of these data, it is crucial to understand what are the 1994), CB1R binding gradually increases in neonates, reaching a
potential long-term consequences of cannabis exposure at these peak around the pubertal period before decreasing to the adult lev-
crucial stages of development. In this review, we shall first outline els (Belue et al., 1995; McLaughlin et al., 1994; Rodriguez de Fonseca
the stages of nervous system development at which endogenous et al., 1993). There are some sexual dimorphisms in the develop-
cannabinoids (eCBs) play a major role. This will help us to under- ment expression of the CB1R. For example, subtle sex differences
stand how cannabis exposure can alter the normal developmental in the binding to CB1R were detected in the rat striatum and ven-
trajectories in the nervous system, thereby perturbing the neu- tral mesencephalon but not in the limbic forebrain (Rodriguez de
ral plasticity that constitutes the basis of cognitive processes and Fonseca et al., 1993). Similarly, neonatal male rats displayed signifi-
motivation. cantly higher levels of CB1Rs in CA3 while they had less CB2R in the
polymorphic layer of the dentate gyrus (DG) (Suarez et al., 2009).
3. The endocannabinoid system during perinatal In adolescence, subtle differences in the expression of hippocam-
development and adolescence pal CB1Rs were found with a lower density of CB1Rs in female rats
than in males (Marco et al., 2007). In humans, the CB1R increased
The endocannabinoid (eCB) system is present from the earli- from the postnatal period to adulthood (Mato et al., 2003), although
est phases of development (Maccarrone et al., 2014). Indeed, in CB1R mRNA expression peaks in neonates and toddlers, there-
rodents it has been reported that the CB1 receptor (CB1R) is present after decreasing gradually over the individual’s lifespan (Long et al.,
and functionally active in the preimplantation embryo and in the 2012).
uterus (Paria and Dey, 2000). Furthermore, eCB signaling at these Regarding the two major eCBs, 2-arachidonoylglycerol (2-AG)
stages is intimately associated with embryo–uterine interactions concentrations during brain development (2–8 nmol/g tissue)
during implantation. The deletion of the CB1R compromises normal exceed those of N-arachidonoylethanolamine (Anandamide or
cell proliferation and differentiation, leading to aberrant placenta- AEA) (3–6 pmol/g tissue) in whole rat brain lysates (Berrendero
tion and compromising embryo implantation (Paria and Dey, 2000; et al., 1999), the latter being detected at low concentrations at
Sun and Dey, 2008). Like the CB1R, the CB2 receptor (CB2R) is also mid-gestation. AEA levels rise gradually during the perinatal
expressed in mouse embryos from the blastocyst stage and it may period in the rat brain, reaching a peak during adolescence before
122 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

decreasing to the previous levels in most of the brain areas studied migratory stream (Oudin et al., 2011) and interneurons during cor-
up to date (Berrendero et al., 1999; Ellgren et al., 2008; Lee et al., ticogenesis (Berghuis et al., 2005). eCB signaling participates in the
2013; Rubino et al., 2014; Wenger et al., 2002). After that, AEA development of other neurotransmitter pathways, as seen in the
levels continue to rise until they reach adult levels (Lee et al., cholinergic system where 2-AG signaling promotes the differenti-
2013). Concomitantly, the AEA synthetic and hydrolytic enzymes, ation and synaptic connectivity of cholinergic projection neurons
N-acyl phosphatidylethanolamine phospholipase D (NAPE-PLD) (Keimpema et al., 2013). The development of glutamatergic neu-
and fatty acid amide hydrolase (FAAH), augment in the whole brain rons might also be regulated by the eCB system. Indeed, prenatal
as postnatal life progresses (Morishita et al., 2005), something exposure to the CB1 and CB2 receptor agonist WIN 55,212-2 (WIN)
also seen in the human dorsolateral prefrontal cortex (PFC) (Long alters migration of early-born glutamatergic neurons and GABAer-
et al., 2012). 2-AG concentrations also increase in mice and rats gic interneurons in the rat cerebral cortex, an effect that might
during development but by contrast, there is a peak in cortical and depend on the transcription factors Tbr1 and Tbr2 (Saez et al., 2014).
hippocampal 2-AG concentrations around the moment of birth Correct axon guidance in the retinothalamic pathway is another
(Berrendero et al., 1999; Keimpema et al., 2010). This is consistent developmental process that requires the presence of CB2Rs (Duff
with an increase in diacylglycerol lipase (DAGL) expression that et al., 2013), suggesting that both cannabinoid receptors are impor-
begins on GD14.5/GD16.5 (GD: Gestational Day) and that remains tant in shaping the developing nervous system in mammals. The
high thereafter until the moment of birth, when it decreases eCB system is also thought to fulfill an important role in synapto-
abruptly (Keimpema et al., 2010). In the rat PFC and nucleus genesis (Kim and Thayer, 2001) and in the modulation of synaptic
accumbens (NAcc) 2-AG levels were seen to be lower at later transmission in the developing hippocampus (Zhu and Lovinger,
stages than at the beginning of adolescence (Ellgren et al., 2008). In 2010). Taking into consideration all the data presented above, it is
humans, DAGL␣ mRNA is more strongly expressed in this period not surprising to find that repeated THC exposure disrupts eCB sig-
(between school age and young adulthood) than at any other stage, naling, particularly the temporal dynamics of the CB1R in rewiring
and monoacylglycerol lipase (MAGL) expression decreases after the fetal cortical circuitry. This phenomenon appears to have the
peaking in infancy (Long et al., 2012). Superior Cervical Ganglion 10 (SCG10)/stathmin-2 pathway as a
These differences in the expression of several elements of the substrate and it has been identified as the first cannabis-driven
eCB system during gestation and adolescence suggest a role for this molecular effector in the developing cerebrum (Tortoriello et al.,
system in the changes the brain undergoes during those stages, 2014).
reflecting periods of high vulnerability for the consequences of In the light of these findings, the consequences of in utero THC
exogenous cannabinoid administration. exposure on the development of the CNS in humans still remain
unclear. To our knowledge there is only one collection of human
fetal specimens with documented marihuana exposure (DiNieri
4. Role of the endocannabinoid system in the regulation of et al., 2011; Hurd et al., 2005; Wang et al., 2006, 2004). When
central nervous system development studied, early marihuana exposure was shown to impair growth in
mid-gestation fetuses and to diminish the mRNA expression of the
The question arises as to the role the eCB system fulfills dur- dopaminergic receptor D2 in the amygdala and NAcc (no changes
ing CNS development. Since the seminal work showing that eCBs were observed in the dorsal striatum: DiNieri et al., 2011; Hurd
regulate interneuron migration and morphogenesis by transacti- et al., 2005; Wang et al., 2004). Notably, in the amygdala, the differ-
vating the TrkB receptor (Berghuis et al., 2005), considerable effort ences reported were only found in male fetuses (Wang et al., 2006).
has focused on the influence of eCB signaling on neuronal develop- In addition, it was also shown that prenatal marihuana exposure
ment. It was shown that eCBs trigger CB1R internalization and its decreases the levels of preproenkephalin (PENK) gene expression
elimination from filopodia, and they induce chemorepulsion and in the fetal striatum (Wang et al., 2006), while the levels of prepro-
the collapse of GABAergic (GABA: ␥-aminobutyric acid) interneu- dynorphin (PDYN) transcripts remained unaffected (DiNieri et al.,
ron axonal growth cones (Berghuis et al., 2007). In addition, how the 2011; Wang et al., 2006). Opioid receptors were also affected in
subcellular distribution of MAGL in developing neurons establishes the fetal brain by in utero cannabis exposure, with increased ␮ opi-
2-AG microgradients that modulate both growth cone matura- oid receptors in the amygdale and decreased levels of the ␬ opioid
tion and axonal elongation was described (Keimpema et al., 2010). receptors in the mediodorsal thalamus (Wang et al., 2006). Lastly,
Moreover, the target selection of GABAergic interneurons lacking as mentioned before, maternal cannabis smoking was associated
the CB1R was shown to be impaired, highlighting the function of to reduced SCG10/stathmin-2 mRNA and protein expression in this
eCBs as axon guidance cues (Berghuis et al., 2007). Signaling at CB1 collection, a fact that was proposed to be a key molecular effector
and CB2 receptors also influenced neural progenitor proliferation mediating adverse effects of cannabis (Tortoriello et al., 2014).
(Aguado et al., 2005; Mulder et al., 2008) in part by its action on the
mTORC1/Pax6 pathway that drives Trb2 expression and basal pro-
genitor expansion (Díaz-Alonso et al., 2014). In the subventricular 5. Perinatal cannabinoid exposure and long-lasting effects
zone, intermediate progenitor cells that contribute to the gener- on neural and psychological processes
ation of pyramidal cells are targeted by CB1R, suggesting a role
of the eCB system in cell fate regulation (Kowalczyk et al., 2009). 5.1. Human studies
Moreover, eCB signaling controls the radial migration of pyrami-
dal cells, and the elongation and fasciculation of their long-range 5.1.1. Cohort studies
axons once they settle in a cortical layer (Mulder et al., 2008), such The issue of the long-term consequences of prenatal cannabis
as that of the corticothalamic projections (Wu et al., 2010). How- exposure has been widely studied in three human cohorts: the
ever, the pharmacological elevation of AEA through FAAH inhibitors Ottawa Prenatal Prospective Study (OPPS), the Maternal Health
during mid-gestation in mice did not affect neurogenesis or axonal Practices and Childhood Development Project (MHPCD) and more
development when measured at GD16.5 (Wu et al., 2014). One recently, the Generation R study. While the results of these stud-
of the main effectors of eCB’s influence on cortical development ies are reviewed elsewhere (Calvigioni et al., 2014; Huizink, 2014;
is the Slit2/Robo1 signaling pathway, with a critical involvement Jaques et al., 2014; Morris et al., 2011; Wu et al., 2011), let
of CB1R activation (Alpár et al., 2014). CB1Rs also influence the us just mention the most important findings. First, in both the
migration of other cells, like that of neuroblasts along the rostral OPPS and MHPCD studies the use of cannabis during pregnancy
A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146 123

was not associated with increased miscarriage rates, premature sites, which, together with the PFC, are important for performing
deliveries or any other complications. In the Generation R study, complex cognitive tasks (Smith et al., 2006).
another cohort was also studied in depth (El Marroun et al., 2010,
2011a,b, 2012; El Marroun et al., 2008, 2009), showing that mater- 5.2. Animal studies
nal cannabis use during pregnancy was associated with growth
restriction in mid and late pregnancy, and with lower birth weight. 5.2.1. Learning and memory
Since no such association was found for paternal cannabis use in In a wide variety of tests, several cognitive deficits have consis-
the same period, this would appear to represent a direct biological tently been reported in rats exposed to THC or other cannabinoid
effect of maternal intrauterine exposure to cannabis on fetal growth agents in utero (see Table 2). For example, a disruption of memory
(El Marroun et al., 2010). It is of more concern that the data from retention was found in a passive avoidance task in rats exposed
these OPPS and MHPCD cohorts suggest that newborns already dis- to WIN in utero (Mereu et al., 2003), while learning deficits in
play cannabis-induced neurobehavioral alterations (<1 week old), the Morris water maze were detected in juvenile rats that had
as do children later in development. The essential findings from been exposed to a cannabis resin during prenatal development
these cohorts indicate that prenatal marihuana exposure increases (Kawash et al., 1980). With regard to long-term memory, THC-
tremor and enhances the startle response in the newborn, and it exposed rats performed worse than controls in an avoidance test
is also associated with a wide variety of cognitive deficits (such and they also showed impaired short-term memory in an olfac-
as impaired sustained attention, self-control deficits and impaired tory memory task (Campolongo et al., 2007). In addition, there
executive function), as well as increased aggression and inattention is also evidence of impaired learning in an active avoidance task
(in this case only in girls: Huizink, 2014). Moreover, in a different set in WIN-exposed male rats (Antonelli et al., 2005). More recently,
of studies, prenatal cannabis exposure was also associated with an cannabinoid exposed animals showed no acquisition deficits in a
increased risk of nicotine addiction and marihuana use in adulthood passive avoidance test, while they exhibited deficits in the con-
(Day et al., 1994, 1993, 2006, 2011). Some of the aforementioned solidation process during retention testing (Silva et al., 2012). No
findings were confirmed in a smaller study (Noland et al., 2005), treatment-related effects on acquisition were evident in the active
suggesting that the severity of marihuana exposure during preg- place avoidance task performed on rats, although a significant treat-
nancy was positively correlated with impaired sustained attention ment effect was observed in reversal performance in males but not
in pre-school children. in females. Finally, in an attention task, a smaller percentage of
THC-exposed rats completed the test and they required more trials
to complete it (Silva et al., 2012). A recent study identified elevated
5.1.2. Neuroimaging studies AEA levels during perinatal development in mice due to FAAH inhi-
Neuroimaging data has been examined to assess the long-term bition, and working memory performance in the offspring of these
structural alterations in the white and gray matter associated with mice was impaired in the T-maze (Wu et al., 2014). WIN expo-
intrauterine marihuana exposure (Derauf et al., 2009). Although sure during prenatal development was seen failed to alter prepulse
studies are scarce, there is evidence of structural alterations in sub- inhibition of the startle response, or the startle response per se
jects prenatally exposed to marihuana. For example, a volumetric (Bortolato et al., 2006), ruling out a mediating role of sensory motor
magnetic resonance imaging (MRI) study of 10–14 year old children gating in the cognitive alterations summarized above.
found a reduced cortical gray matter after intrauterine marihuana
exposure, although the model used was not adjusted for age, gen- 5.2.2. Emotional regulation
der and other drug exposures (Rivkin et al., 2008). In another study There are conflicting results as to how prenatal cannabinoid
in which frontal white matter development was assessed using dif- exposure might affect anxiety-related behaviors (see Table 3).
fusion tensor imaging in children with prenatal cocaine exposure, While in one study prenatal THC exposure was associated with
intrauterine exposure to both cocaine and marihuana resulted in increased exploratory activity of male but not female offspring
higher (worse) mean diffusivity than exposure to cocaine alone in the elevated plus maze (EPM: Rubio et al., 1995), revealing
(Warner et al., 2006). decreased anxiety, the opposite trend was also found elsewhere
Two other functional MRI (fMRI) studies investigated the (Trezza et al., 2008). When using another anxiety index, ultrasonic
relationship between prenatal marihuana exposure and adoles- vocalizations (USVs) in infant rats, similar conflicting results were
cent/young adult neural functioning in tasks requiring cognitive obtained with one report showing a decrease in USVs (Antonelli
effort. In the first, subjects with and without prenatal mari- et al., 2005) and another showing the contrary (Trezza et al., 2008).
huana exposure underwent fMRI while performing a two condition If we examine social interactions, the results are again unclear.
Go/No-Go task. Although there were no significant group differ- In two different studies, evidence of reduced social interactions
ences for reaction time and errors of omission for either condition, was presented (O’Shea et al., 2006; Trezza et al., 2008), while in
the group exposed prenatally made more errors in the more diffi- another utero exposed rats displayed increased social interactions
cult task. The extent of prenatal marihuana exposure was positively (Newsom and Kelly, 2008). There are no clear reasons for these dis-
correlated to bilateral PFC and right premotor cortex activity, while crepant results, but it would appear that the different cannabinoids
left cerebellar activity was negatively correlated to the degree used are not the cause. Indeed, CP 55,940 (CP) or THC was employed
of marihuana exposure. This was interpreted as increased effort in the first two studies where decreases in social interaction were
needed to perform the task (Smith et al., 2004). In the second study, found. Perhaps, the specific tests used to measure social interac-
fMRI scans were performed on the same subjects during a visuospa- tion in each experiment might account for the differences in the
tial 2-back test. Although no significant differences in performance results reported in these three studies. In any case, further research
were observed between the prenatally exposed and unexposed is clearly needed to provide a definite answer to the question of
subjects, the amount of prenatal marihuana exposure was signifi- whether perinatal cannabinoid exposure indeed has a long-lasting
cantly and positively related to increased activity in the left inferior influence on emotional behavior.
and middle frontal gyri, the left parahippocampal gyrus, left mid- With regard to depressive behavior, one study that analyzed the
dle occipital gyrus and left cerebellum, and it was significantly and behavior of rats in the forced swimming test (FST) after prenatal
negatively related to activity in the right inferior and middle frontal THC treatment found no differences from control rats (Newsom
gyri. It was suggested that prenatal marihuana exposure may have and Kelly, 2008). By contrast, chronic elevation of AEA (induced by
altered the normal lateralization and connectivity of multiple brain the blockade of the FAAH enzyme) increased the immobility in this
124 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

Table 2
Behavioral and cognitive alterations found in adult animals after perinatal cannabinoid exposure.

Species Sex Treatment Test Test age Results Reference

Drug Dose Age

Rat ♂ CP 55,940 0.15, 0.2 and PND4-PND24 Novel object PND53 Memory O’Shea et al., 2006
0.3 mg/kg (i.p.) recognition impairment
for 7 days each
dose
Rat ♂ WIN 55,512-2 0.5 or 1 mg/kg GD5-GD20 Prepulse inhibition PND40, 60 and 80 No significant Bortolato et al., 2006
(s.c.) effects
Rat ♂ 9 -THC 5 mg/kg (p.o.) GD15-PND9 Social recognition PND80 Social memory Campolongo et al., 2007
impaired
Rat ♂–♀ 9 -THC 0.15 mg/kg (i.v.) GD1-GD21 Attention task PND55 Attention impaired Silva et al., 2012
Rat ♂ WIN 55,512-2 0.5 mg/kg (s.c.) GD5-GD20 Active place PND80 Impaired spatial Antonelli et al., 2005
avoidance learning and
memory
Rat ♂–♀ 9 -THC 0.15 mg/kg (i.v.) GD1-GD21 PND45-46 Impaired Silva et al., 2012
behavioral
flexibility ♂
Rat ♂–♀ 9 -THC 0.15 mg/kg (i.v.) GD1-GD21 Passive place PND22-23 Learning and Silva et al., 2012
avoidance memory
impairment
Rat ♂ WIN 55,512-2 0.5 mg/kg (s.c.) GD5-GD20 PND40 and 80 Memory Mereu et al., 2003
impairment
Rat ♂ 9 -THC 5 mg/kg (p.o.) GD15-PND9 Inhibitory PND81 Memory Campolongo et al., 2007
avoidance impairment
Rat ♂ WIN 55,512-2 0.5 mg/kg (s.c.) GD5-GD20 Homing PND6-12 Impaired memory Antonelli et al., 2005
Rat ♂ 9 -THC 5 mg/kg (p.o.) GD15-PND9 Olfactory memory PND80 Impaired memory Campolongo et al., 2007
task
Mouse ♂–♀ URB597(↑AEA) 10 mg/kg (i.p.) GD10-PND7 T Maze >PND60 Impaired working Wu et al., 2014
memory

test (Wu et al., 2014). However, the FST is contested as an animal later in life. Interestingly, this effect has been studied mainly for opi-
model of depression as it appears to be rather a test of the efficiency ates, with very little information regarding other classes of drugs.
of potential antidepressant drugs (Petit-Demouliere et al., 2005; For example, tolerance to the analgesic properties of morphine was
Yan et al., 2010). reported in male rats perinatally exposed to THC (Vela et al., 1995)
and enhanced conditioned place preference (CPP) was evoked by
5.2.3. Sensitivity to drugs of abuse later in life and potential a moderate dose of morphine in adult rats of both sexes exposed
addictive behavior to THC during perinatal development (Rubio et al., 1995). These
There is consensus in the literature that cannabis exposure dur- results have been replicated more recently in male rats (DiNieri
ing prenatal development affects the sensitivity to drugs of abuse et al., 2011). When we tested whether perinatal exposure to THC

Table 3
Emotional behavior alterations after perinatal cannabinoid exposure.

Species Sex Treatment Test Test age Results Reference

Drug Dose Age

Rat ♂–♀ 9 -THC 5 mg/kg (p.o.) GD5-PND24 Elevated plus maze PND70 Anxiety ↓♂ Rubio et al., 1995
Rat ♂ 9 -THC 2.5 mg/kg (p.o.) GD5-PND9 PND80 No significant effects Trezza et al., 2008
5 mg/kg (p.o.) Anxiety ↑
Mouse ♂–♀ URB597(↑AEA) 10 mg/kg (i.p.) GD10-PND7 >PND60 No significant effects Wu et al., 2014
Rat ♂ CP 55,940 0.15, 0.2 and PND4-PND24 Emergence test PND57 No significant effects O’Shea et al., 2006
0.3 mg/kg (i.p.) 7
days each dose
Rat ♂ 9 -THC 2 mg/kg (twice a GD1-PND10 Forced swimming PND99 No significant effects Newsom and Kelly, 2008
day) test
Mouse ♂–♀ URB597(↑AEA) 10 mg/kg (i.p.) GD10-PND7 >PND60 Depressive-like Wu et al., 2014
behavior ↑
Rat ♂ WIN 55,512-2 0.5 mg/kg (s.c.) GD5-GD20 Homing PND6-12 Emotional reactivity ↓ Antonelli et al., 2005
Rat ♂ 9 -THC 2 mg/kg (s.c.) twice GD1-PND10 Open field PND93 Anxiety ↑ Newsom and Kelly, 2008
a day
Mouse ♂–♀ URB597(↑AEA) 10 mg/kg (i.p.) GD10-PND7 >PND60 No significant effects Wu et al., 2014
Rat ♂ 9 -THC 2.5 mg/kg (p.o.) GD5-PND9 Social interaction PND35 No significant effects Trezza et al., 2008
5 mg/kg (p.o.) Social anxiety ↑
Rat ♂ CP 55,940 0.15, 0.2 and PND4-PND24 PND55 Social anxiety ↑ O’Shea et al., 2006
0.3 mg/kg (i.p.) 7
days each dose
Rat ♂ 9 -THC 2 mg/kg (twice a GD1-PND10 PND97 Social anxiety ↓ Newsom and Kelly, 2008
day)
Rat ♂ WIN 55,512-2 0.5 mg/kg (s.c.) GD5-GD20 Ultrasonic PND10 Emotional reactivity ↓ Antonelli et al., 2005
vocalizations
Rat ♂ 9 -THC 2.5 mg/kg (p.o.) GD5-PND9 PND12 No significant effects Trezza et al., 2008
5 mg/kg (p.o.) Emotional reactivity ↑
A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146 125

Table 4
Neurochemical alterations found in adult animals after perinatal cannabinoid exposure.

Species Sex Treatment Tests Reference

Drug Dose Age Age Results

Monoaminergic systems
Rat ♂ Hashish extract 20 mg/kg (p.o.) GD5-PND24 PND15, 20, 30, 40, TH mRNA: Ms PND15 ↑ Bonnin et al., 1994
70 PND70 ↓; TH levels: Ms
PND15 ↑
Mouse ♂ 9 -THC 50 mg/kg (p.o.) GD18 PND64-94 No significant alterations Dalterio et al., 1984a
found
CBN DA levels: Brain tissue ↓;
5-HT and 5HIAA levels: No
significant alterations found
CBD DA levels: Brain tissue ↓;
5-HT and 5HIAA levels: No
significant alterations found
Mouse ♂–♀ 9 -THC 50 mg/kg (p.o.) GD18 PND69♂/PND67- DA levels: No significant Dalterio et al., 1984b
77♀ alterations found; 5-HT
levels: HT ♀↓
CBN DA and 5-HT levels: No
significant alterations found
CBD DA and 5-HT levels: No
significant alterations found
Rat ♂ 9 -THC 0.15 mg/kg (i.v.) GD5-PND2 PND2 and 62 DRD2 mRNA and D2 receptor DiNieri et al., 2011
density: NAcc PND2 ↓,
PND62 ↓
Rat ♂–♀ 9 -THC 5 mg/kg (p.o.) GD5-PND24 >PND70 Ambulation (after SCH23390 Garcia et al., 1996
challenge): ↓
Rat ♂–♀ 9 -THC 5 mg/kg (p.o.) GD5-PND24 >PND70 DOPAC/DA ratio: NAcc ♀↓; Gonzalez et al., 2003
VTA ♀↓
Rat ♂  -THC
9
5 mg/kg (p.o.) GD13-PND7 PND90 SERT binding levels: NSt ↑; Molina-Holgado et al., 1993
aHT ↓; RphN ↓; LC ↓; 5-HIAA:
aHT ↑; SN ↑;
Rat ♂–♀ 9 -THC 5 mg/kg (p.o.) GD5-GD20/PND1 GD20 and PND1 5-HT levels: Di ♂↓↓–♀↓ Molina-Holgado et al., 1996
Rat ♂–♀ 9 -THC 0.1, 0.5 and GD5-PND24 >PND70 Motor response to Moreno et al., 2003
2 mg/kg (p.o.) quinpirole: Immobility ♂↑;
Stereotypes ↓♂ (2 mg/kg
dose)
Motor response to
Apomorphine: Locomotion
♂↓ (2 mg/kg dose)
Mouse ♂ WIN 55,512-2 0.5 mg/kg (s.c.) GD5-GD20 PND0-2 TH+ cells: No significant Tree et al., 2014
alterations found in any of
the areas studied
Rat ♂–♀ (mixed) 9 -THC 10 mg/kg (p.o.) GD0a -PND20 PND10, 20, 40 and No significant alterations Walters and Carr, 1988
60 found
8 -THC 1 mg/kg (p.o.) TH activity: St ↓ (PND60)
CBD 10 mg/kg (p.o.) TH activity: St↓ PND(60); D2
receptor Kd: St ↑ (PND10), D2
receptor density: St ↑
(PND10 and 20)

Glutamatergic and GABAergic systems


Rat ♂ WIN 55,512-2 0.5 mg/kg (s.c.) GD5-GD20 PND1 Basal and K+ evoked Glu: Cx Antonelli et al., 2005
↓(cc); NMDA receptor
function: Cx ↓(cc)
Rat ♂ WIN 55,512-2 0.5 mg/kg (s.c.) GD5-GD20 PND1 Basal and K+ evoked Glu: Cx Antonelli et al., 2005
↓(cc)
Rat ♂ WIN 55,512-2 0.5 mg/kg (s.c.) GD5-GD20 PND120-150 GABA+ and GAD 65/67+ cells: Benagiano et al., 2007
CbC ↑
Rat ♂ 9 -THC 5 mg/kg (p.o.) GD5-PND20 PND81 Glu levels: PFC ↓ Campolongo et al., 2007
Rat ♂ WIN 55,512-2 0.5 mg/kg (s.c.) GD5-GD20 PND40 Glu levels: FC ↓; Glu uptake: Castaldo et al., 2007
FC ↑; GLT1 levels: FC ↑;
EAAC1: FC ↑;
9 -THC 5 mg/kg (s.c.) GD5-GD20 Glu levels: FC↓
Rat ♂ 9 -THC 5 mg/kg (p.o.) GD15-PND9 PND40 Basal and K+ evoked Glu: HP Castaldo et al., 2010
↓; Glu uptake: HP ↓; GLT1
levels: HP ↓(syn); GLAST
levels:HP ↓(syn)
Rat ♂–♀ 9 -THC 5 mg/kg (p.o.) GD5-PND24 >PND70 Ambulation (after baclofen Garcia-Gil et al., 1999
challenge): ↓
Rat ♂ WIN 55,512-2 0.5 mg/kg (s.c.) GD5-GD20 PND40 and 80 Basal and K+ evoked Glu Mereu et al., 2003
levels: HP ↓
Rat ♂ WIN 55,512-2 0.5 or 0.1 mg/kg GD5-GD20 PND50 Firing pattern and intrinsic Shabani et al., 2011
(s.c.) electrophysiological activity
of Purkinje cells altered
Rat ♂ WIN 55,512-2 0.5 mg/kg (s.c.) GD5-GD20 PND50 Firing pattern and intrinsic Shabani et al., 2014
electrophysiological activity
of Purkinje cells altered
126 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

Table 4 (Continued)

Species Sex Treatment Tests Reference

Drug Dose Age Age Results

Rat ♂–♀ 9 -THC 5 mg/kg (p.o.) GD5-PND20 PND20, 30 and 70 Glutamine synthetase levels: Suarez et al., 2002
CbC PND20 ♂↓; PND30 ↓
Rat ♂–♀ 9 -THC 5 mg/kg (p.o.) GD5-PND20 PND20, 30 and 70 GluN1 levels: Cb PND30↓ Suarez et al., 2004a
PND70↓; GluN2/3 levels: Cb
PND20↓ PND30↓ PND70↓
Rat ♂–♀ 9 -THC 5 mg/kg (p.o.) GD5-PND20 PND20, 30 and 70 GLAST mRNA: Cb ↓; EAAC1 Suarez et al., 2004b
mRNA: Cb ↓; GLAST levels:
Cb ↓; EAAC1:PND20 and
30♂↓, PND20 ♀↓

Endogenous opioid system


Rat Not determined 9 -THC 5 mg/kg (p.o.) GD5-GD16 GD16 PENK mRNA: CPu ↑; Spt ↑; Th Perez-Rosado et al., 2000
↑; HT ↑, Mb ↑
GD5-GD18 GD18 PENK mRNA: Cx ↑; CPu ↑
♂–♀ GD5-GD21 GD21 PENK mRNA: Cx ♂↓; CPu
♂↓-♀↑; PvHN ♂↓-♀↑; VmHN
♂↓-♀↑
Rat Not determined 9 -THC 5 mg/kg (p.o.) GD5-GD16 GD16 No significant alterations Perez-Rosado et al., 2002
found
GD5-GD18 GD18 POMC mRNA: Arc↓; Cx ↓
♂–♀ GD5-GD21 GD21 POMC mRNA: Arc ♂↓-♀↑; Cx
♂↓-♀↑; HbN ♂↓-♀↑; PDYN
mRNA: Cx♀↑; HP ♀↑; PvHN
♀↑
Rat ♂–♀ 9 -THC 5 mg/kg (p.o.) GD5-PND24 >PND70 ␮ opioid receptor density: Vela et al., 1998
CPu ♂↓; frPaMC ♀↑; HP(CA3)
♀↑; LAmy ♀↓; AmyPmCN
♂↓-♀↑; VTA ♀↑; PAG ♀↑
Endogenous cannabinoid system
Rat ♂ WIN 55,512-2 0.5 mg/kg (s.c.) GD5-GD20 >PND80 CB1 receptor function: HP ↑; Castelli et al., 2007
St ↓; AEA levels: St ↑; Limb ↓;
FAAH activity: St ↓; Limb ↑;
NAPE-PLD activity: St ↑;
Limb ↓
Rat ♂–♀ 9 -THC 5 mg/kg (p.o.) GD5-PND24 >PND70 CB1 receptor density: Cx Garcia-Gil et al., 1999
(Superficial layers) ♂↓; Arc
♂↑; CB1 mRNA: HP(CA2)
♂↑; Vertical activity (after
SR141716A challenge): ♀↑

would lead to increased morphine self-administration, this was acute prenatal THC did not modify the concentration of brain bio-
only the case when a fixed ratio one schedule of reinforcement was genic amines (Dalterio et al., 1984a,b), it was later shown that
used, and only in female offspring (Vela et al., 1998). This effect does in perinatally THC-exposed females but not males had higher
not seem to be due to general changes in reward processing as there rates of dopamine synthesis in the limbic forebrain but not in the
were no differences in food-reinforced behavior. No differences striatum (Garcia et al., 1996). In addition, the DOPAC/dopamine
in self-administration were found when similar tests were per- (DOPAC: 3,4-dihydroxyphenylacetic acid) ratio in the NAcc and
formed for heroin, although THC exposed animals exhibited a lower ventral tegmental area (VTA) was lower in THC-exposed females
latency to press the active lever for heroin and more importantly, than in controls, suggesting decreased dopamine turnover (no such
an increased relapse rate after mild stress conditions or during effect was evident in the males: Gonzalez et al., 2003). By contrast,
extinction tests (Spano et al., 2007). As mentioned previously, most the expression of the tyrosine hydroxylase (TH) gene was weaker
of these studies focused on opiates and to our knowledge only two in the mesencephalon of exposed animals (Bonnin et al., 1994) or
studies examined the effects of perinatal cannabinoid exposure unchanged in the ventrolateral medulla oblongata (Tree et al., 2014).
on psychostimulant effects. In the first, both in male and female The aforementioned altered dopaminergic function could gener-
THC-exposed rats displayed a blunted locomotor response to an ate compensatory changes at the receptor level. For example, D2
amphetamine challenge in adulthood, although females were more receptor affinity was lower in the striatum of THC-exposed ani-
active overall (Silva et al., 2012). Similarly, there was a blunted rein- mals (Walters and Carr, 1988), and the amount of D2 receptors
forcing action of cocaine (measured in the CPP test) in the offspring in the NAcc was also reduced (DiNieri et al., 2011). More specifi-
of mice exposed to elevated levels of AEA during gestation (Wu cally, a decrease was observed in 3 H-raclopride binding in the NAcc
et al., 2014). Another study focused on alcohol self-administration, but not in the dorsal striatum of adult male rats exposed to THC
asking whether perinatally THC-exposed rats would self- in utero (DiNieri et al., 2011). This alteration was produced via a
administer more alcohol as adults and also, if it would alter gene decrease in the 3meH3K4 induction mark and an increase in the
expression patterns (Economidou et al., 2007). While THC affected 2meH3K9 repression mark in the offspring of THC-exposed moth-
the expression of 112 genes, similar alcohol self-administration ers (DiNieri et al., 2011). In functional terms, these changes in D2
rates were obtained in THC- and vehicle-exposed animals. receptor values seem to have a differential effect at pre- and post-
synaptic levels. Thus, perinatal exposure to THC in rats enhanced
5.2.4. Monoaminergic systems presynaptic dopamine D2 receptor mediated responses while post-
The effects of perinatal cannabinoid treatments on dopaminer- synaptic dopamine D2 receptor agonist-induced stereotypes were
gic function are controversial. While early reports suggested that dampened. Interestingly, these results were only observed in males
A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146 127

and not in females (Moreno et al., 2003; but see Garcia et al., prenatally exposed to the cannabinoid agonist. Indeed, prenatal
1996). Given the crucial role of D2 receptors in drug addiction, exposure to the cannabinoid receptor agonist WIN increased glu-
it is tempting to speculate that the reduction in these receptors tamate uptake through the overexpression of the GLT1 and EAAC1
might somehow contribute to the increased rewarding effects of glutamate transporter subtypes in the adult rat frontal cortex
opiates observed in animals exposed to cannabinoids during peri- (Castaldo et al., 2007). Cortical glutamate and norepinephrine (NE)
natal development. Indeed, imaging studies of adult subjects with were also diminished in the offspring of pregnant rats exposed
a history of drug abuse have reported reduced levels of D2 in the to THC during pregnancy, an effect that was accompanied by
striatum (Volkow et al., 2004), and animal models documented that long-lasting alterations to cortical genes related to glutamatergic
increasing NAcc D2 gene expression attenuates drug intake (Thanos and noradrenergic systems (Campolongo et al., 2007). In the
et al., 2008). cerebellum, THC exposure during prenatal development modu-
Only a few studies have focused on serotoninergic alterations lates glutamatergic nervous activity by diminishing glutamine
following prenatal cannabinoid exposure but in general, there synthetase (GS) expression (Suarez et al., 2002). GS expression
seems to be a decrease in 5-hydroxytryptamine (5-HT or sero- increased progressively after THC withdrawal and while in males
tonin) levels and an increase in 5-HT metabolism in animals that this effect was evident right after THC cessation, in females it only
were exposed to THC during perinatal development. The hypotha- appeared after withdrawal. These reduced GS levels were taken
lamus seems to show the most consistent results, with a decrease as an indication of reduced glutamate uptake in glial cells exposed
in serotonin uptake sites and increased 5-hydroxyindoleacetic acid to THC in utero, as confirmed by the lower levels of the GLAST and
(5-HIAA) levels (Molina-Holgado et al., 1993). This alteration to the EAAC1 protein in the cerebellum of prenatally exposed rats (Suarez
5-HT system seems to occur early in development since decreased et al., 2004b). There was sex-specific dynamics in this effect and
5-HT levels in the diencephalon were evident at the end of gestation indeed, the reductions lasted long into withdrawal in the males
and right after birth in animals exposed to THC, and they were more while they disappeared after THC cessation in the females (Suarez
prominent in males than in females (Molina-Holgado et al., 1996). et al., 2004b). Along with the dysregulated glutamatergic tone
Moreover, maternal exposure to THC decreased the levels of 5-HT found following in utero THC exposure, changes in glutamatergic
in the adult female hypothalamus but not in the male hypothala- receptors were also observed in these rats. For example, there is a
mus (Dalterio et al., 1984b). In addition, cannabinol or cannabidiol down-regulation of the cerebellar (␣-amino-3-hydroxy-5-methyl-
treatment during pregnancy reverted the decreased hypothalamic 4-isoxazolepropionic acid) AMPA glutamate receptor subunits
levels of the neurotransmitter and metabolite in the males after GluA1 and GluA2/3 following pre- and perinatal THC exposure.
castration (Dalterio et al., 1984a). In addition to the hypothala- This effect was stronger in males right after THC cessation and
mus, other brain areas also had altered 5-HT uptake sites, such as even lasted 50 days after THC withdrawal (Suarez et al., 2004a).
the neostriatum (NSt) (increased), Raphe nuclei (RphN) (decreased) These alterations in glutamatergic activity in cerebellar cells might
and locus coeruleus (LC) (decreased: Molina-Holgado et al., 1993). underlie the changes in intrinsic electrophysiological activity of
To our knowledge, no further studies have characterized the sero- Purkinje neurons observed in adult rats prenatally exposed to the
toninergic system in the adult brain after prenatal exposure to cannabinoid agonist WIN (Shabani et al., 2014, 2011). Impaired
cannabinoids. These alterations in serotoninergic system might be cortical N-methyl-d-aspartate (NMDA) function has also been
related to the effects in emotional regulation reviewed above, how- documented in rats exposed to THC in utero and the concentration-
ever, the lack of consistent results at the behavioral level makes it dependent increase in glutamate observed in cultured cortical cells
impossible to draw firm conclusions. If we try to compare those after incubation with NMDA is lost in cells obtained from WIN-
studies with similar doses and administration periods, we observe exposed pups (Antonelli et al., 2005). As discussed, the reduced
that animals show increased anxiety (Trezza et al., 2008) and glutamatergic tone found in these animals could be related to the
decreased 5-HT uptake sites in the RphN, which is reminiscent of decrease in cytosolic brain derived neurotrophic factor (BDNF), as
what is observed in human patients (Reimold et al., 2008). wells as to the decrease in extracellular signal-regulated kinase
(ERK) and calcium calmodulin-kinase II phosphorylation in the
5.2.5. Glutamatergic and GABAergic systems hippocampus and frontal cortex (Maj et al., 2007). Although the
Given the crucial role of glutamatergic transmission in learn- evidence for cognitive impairment after perinatal cannabinoid
ing, memory and synaptic plasticity, researchers have paid a exposure is not wholly consistent, decreased glutamate levels in
great deal of attention to changes in this neurotransmitter sys- the hypothalamus and impaired performance in a passive place
tem after prenatal cannabinoid treatment. There seems to be some avoidance task suggest a possible link between both effects. In
degree of consensus in the literature that perinatal cannabinoid addition, the decreased glutamate levels in the frontal cortex
exposure dampens glutamate function in the adult animal. Using (Castaldo et al., 2007) could be related to the poorer performance
in vivo microdialysis, a significant decrease in K+ -evoked and/or in an attentional task and in a reversal task, both dependent on
basal extracellular glutamate levels was found in the hippocam- frontal cortex function (Carli and Invernizzi, 2014; Robbins, 2007).
pus (Mereu et al., 2003) or frontal cortex (Castaldo et al., 2007) To our knowledge, only two studies have investigated the influ-
of juvenile and adult rats born to dams that received WIN. The ence of prenatal THC exposure on GABAergic regulation during
hippocampal alterations in glutamatergic transmission were trans- adulthood. In the first of these studies, THC exposure did not
lated into distorted synaptic plasticity and in this regard, the modify either glutamic acid decarboxylase (GAD) activity or GABA
induction of hippocampal long-term potentiation was impaired content in the VTA, NAcc, substantia nigra, caudate-putamen or
in cannabinoid-exposed animals (Mereu et al., 2003). A similar globus pallidus, suggesting no changes in basal presynaptic activ-
reduction in glutamate outflow was also observed in hippocampal ity of GABA-containing neurons (Garcia-Gil et al., 1999). The motor
slices obtained from pups born to mothers exposed to WIN. More- response of these animals was also tested in the open-field test
over, a reduced glutamate uptake was observed in synaptosomal after a single injection of muscimol, a GABAA receptor agonist, or
preparations, probably caused by the observed downregulation of baclofen, a GABAB receptor agonist. The motor inhibition caused
the glial glutamate transporters GLT1 and GLAST, and it was sug- by baclofen was more marked in magnitude in THC-exposed males
gested to develop as a compensatory mechanism in response to the and females, unlike the response to the GABAA receptor agonist,
decreased glutamate outflow (Castaldo et al., 2010). muscimol. The functional activity of GABAB receptors was also
These decreased cortical glutamate levels could be explained measured to track the origins of the reported effects of baclofen
by the increased glutamate transport in this area found in rats in THC-exposed animals. However, perinatal THC exposure did
128 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

not increase baclofen-stimulated [35S ]-GTP␥S binding in the areas not females exhibited a small decrease in binding in the superficial
studied. In the second study, GAD and GABA immunoreactivity was layers of the cerebral cortex. In the CA2 layer of Ammon’s horn,
quantified in the cerebellum of the offspring of rats exposed to there was an increase in CB1 mRNA in THC-exposed male animals,
WIN during pregnancy. Although these elements were distributed although the most marked and probably relevant changes were
similarly in WIN-exposed animals and their controls, there were seen in the arcuate nucleus of the hypothalamus. In this nucleus,
quantitative differences between them. WIN-treated rats had more THC-exposed males exhibited increased binding, whereas it tended
GAD and GABA immunoreactive cells in the molecular and Purk- to decrease in THC-exposed females. In the second study (Castelli
inje layers, with no changes in the granular layer (Benagiano et al., et al., 2007), CB1R density and affinity was not affected in any of
2007). the regions analyzed, yet there was an increased functional activa-
Clearly more research is needed to gain a deeper understanding tion of cannabinoid receptors in the hippocampus and decreased
of the alterations to the GABAergic systems caused by cannabi- activation in the striatum (as indicated by 35 S-GTP-␥-S binding
noid exposure during perinatal development and their potential autoradiography in WIN-exposed offspring). Endogenous cannabi-
functional implications. noid levels and cannabinoid-related enzyme activities were also
determined in this study, and the increased AEA levels in the stria-
tum were associated with reduced FAAH and enhanced NAPE-PLD
5.2.6. Endogenous opioid system
activities. Opposite changes in AEA levels and enzymatic activities
One of the first pieces of evidence that perinatal cannabinoid
were detected in the limbic areas of WIN-treated rats (specifically
exposure might affect the endogenous opioid system came from an
olfactory tubercles, NAcc and septal nuclei).
early report showing that perinatal exposure to THC in mice altered
The increased activity of CB1Rs in the hippocampus and the
enkephalin and NE sensitivity in the vas deferens (Dalterio et al.,
stronger transcription of its gene could suggest enhanced signal-
1980). Eighteen years later, we showed that perinatal exposure
ing through this system, which might contribute to explain some
to THC produces specific long-term regional changes in ␮ opioid
of the cognitive deficits reported above (Basavarajappa et al., 2014;
receptor binding that differed in function of the sex of the animal
Mereu et al., 2003).
(Vela et al., 1998). THC-exposed males exhibited a lower density
of these receptors in the dorsal striatum and posteromedial corti-
cal amygdala than their controls. Receptor binding in THC-exposed
6. Adolescent cannabinoid exposure and long-lasting
females was only lower than in controls in the lateral amygdala,
effects on neural and psychological processes
whereas they exhibited a higher density of these receptors in the
frontoparietal cortex, hippocampus (CA3 area), posteromedial cor-
6.1. Human studies
tical amygdala, VTA and the periaqueductal gray matter. Prenatal
exposure to THC also modified the expression of genes encoding
6.1.1. Neuropsychological assessment
several opioid peptides in the fetal brain, decreasing for example
As indicated, marihuana is the illegal drug most widely con-
PENK mRNA in the caudate-putamen, hypothalamic paraventric-
sumed during adolescence in Europe (especially Spain, France and
ular and ventromedial nuclei, and in the cerebral cortex of males
Italy: European Monitoring Centre for Drugs and Drug Addiction,
but increasing it in the same regions of females with the excep-
2014) and the United States (Substance Abuse and Mental Health
tion of the cerebral cortex (Perez-Rosado et al., 2000). Other opioid
Services Administration, 2013). Adolescence is a developmental
neuropeptides have also been analyzed and prenatal-THC exposure
period of intense neural remodeling and plasticity (Blakemore,
altered proopiomelanocortin (POMC) and PDYN mRNA expres-
2013; Lenroot and Giedd, 2006) with important sexual dimor-
sion in most brain areas studied at different fetal ages, although
phisms in the developmental trajectories of several brain regions
the effects were sex-dependent (Perez-Rosado et al., 2002). For
(Lenroot and Giedd, 2010; Lenroot et al., 2007; Raznahan et al.,
example, at GD21 there was more mRNA for POMC in the arcu-
2014). For example, male and female adolescents show diver-
ate nucleus, cerebral cortex and habenular nuclei of THC-exposed
gent structural maturation within the ventro-rostral putamen,
females and less in THC-exposed males. With regards PDYN, it was
medial rostral caudate, rostral pallidum, and lateral-dorsal tha-
more strongly expressed in the cerebral cortex, hippocampus and
lamus, which accumulate most of the functional abnormalities
paraventricular hypothalamic nucleus of THC-exposed females,
associated with depression, and also the ventral striatal systems
whereas no changes were observed in THC-exposed males. It is
involved in key aspects of risk-taking behavior (Raznahan et al.,
important to note that these changes were evident in the fetal
2014). Another brain region that develops substantially during
brain and no studies have been carried out to determine if these
the teenage years is the PFC, which is involved in executive
alterations are long-lasting (reaching the adult period) or transient.
functions, such as decision making, inhibitory control and plan-
With regards the functional implications of the long-term alter-
ning, social understanding and self-awareness (Blakemore, 2013).
ations of ␮ opioid receptors, the most relevant finding is that their
Although brain size is thought to stabilize at around five years of
levels decrease in the VTA of females alone, consistent with the
age, important neurodevelopmental processes continue through-
increased morphine self-administration observed in these animals
out adolescence, including myelinization (Benes, 1989), synaptic
(Vela et al., 1998). Indeed, VTA ␮ opioid receptors play a crucial role
refinement (Huttenlocher and Dabholkar, 1997) and a reduction in
in opiate reinforcement (Zhang et al., 2009).
gray matter volume (Blakemore, 2012). Indeed, longitudinal stud-
ies of subjects from 3 to 30 years of age show a general pattern of
5.2.7. Endogenous cannabinoid system childhood peaks of gray matter followed by adolescent declines,
The endogenous cannabinoid system is an important regulator structural and functional rises in connectivity and integrative pro-
of several physiological and psychological functions. However, only cessing, and a changing coordination between limbic/subcortical
two studies that we are aware of have assessed the alterations to and frontal lobe functions, extending well into the adult life (Giedd,
the endogenous cannabinoid system of the adult offspring of moth- 2008). As a result, insults received in adolescence might pervasively
ers exposed to THC during gestation. In the first (García-Gil et al., affect the normal course of CNS development.
1999), no changes in cannabinoid receptor binding were witnessed There has been a great deal of controversy regarding the pos-
in most of the regions analyzed (basal ganglia, cerebellum, limbic sible long-term consequences of adolescent marihuana abuse on
structures and most of the hippocampal regions), although some cognition. Several questions arise in this debate, for example, is
marginal effects were found. For example, THC-exposed males but the age of onset of marihuana abuse a determinant factor in its
A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146 129

detrimental effects? Second, are these deficits general or specific to the inferior posterior vermis were identified (smaller in cannabis
certain cognitive domains? Third, are these deficits restored after users), as were sex-dependent effects of cannabis use on the volume
a period of abstinence? With regard to the first question, it seems of the PFC (larger than the controls in female cannabis users and
that the age of onset is indeed an important factor. A recent study smaller in males). This gender-specific difference was also reported
highlighted the neuropsychological decline in persistent cannabis elsewhere (McQueeny et al., 2011), with larger right amygdala
intake from childhood to midlife (Meier et al., 2012). The aim of volumes in female but not male cannabis users compared to the
that study was to test the association between persistent cannabis control population. The hippocampus is also affected by adolescent
use and neuropsychological decline, and to determine whether this cannabis use and in a recent study, subjects with cannabis use dis-
decline is concentrated among cannabis users whose consumption order that remained abstinent for six months developed alterations
begins in adolescence. The subjects belonged to the Dunedin cohort, to hippocampal surface shape (Smith et al., 2015).
a birth cohort of 1037 individuals followed from birth (1972/1973) Chronic adolescent cannabis users also seem to adopt differ-
to 38 years of age, and the strength of this work is that it included ent strategies and engage alterative neural networks than controls
measures of cognitive functioning prior to any known cannabis when resolving cognitive tasks. For example, cannabis users show
exposure in the participants. Indeed, neuropsychological testing increased activity during a cognitive task in the basal ganglia and
was conducted at 13 years of age, before the initiation of cannabis postcentral gyrus of the right hemisphere and bilaterally in the
use, and again at 38 years. The study tried to rule out the most fre- precuneus and superior parietal lobe, suggesting a compensatory
quently cited confounding factors in such reports (acute or residual neural effort. Additionally, cannabis users demonstrated a positive
cannabis intoxication, tobacco dependence, hard-drug dependence association between performance in the cognitive task and brain
– e.g. heroin, cocaine, amphetamines, alcohol dependence, and activation in the left temporal gyrus, superior temporal lobe and
schizophrenia), and several deficits in neuropsychological func- anterior cingulate cortex, while controls showed a negative associa-
tions were observed across functional domains. Importantly, the tion in these areas. Conversely, marihuana users showed a negative
impairment was concentrated among adolescent-onset cannabis association between brain activation and cognitive performance in
users, with more persistent use associated with greater decline the right temporal gyrus, thalamus, pulvinar, left parahippocampal
(Meier et al., 2012). gyrus and bilateral uncus while the converse pattern was found in
Other groups have confirmed the importance of the age of onset controls (Padula et al., 2007). In a similar task, the performance of
of cannabis use showing, for example, that early-onset cannabis adolescent cannabis users with brief and sustained cannabis absti-
users performed worse in several neuropsychological tasks that nence was comparable, yet recent users displayed greater activity
focused on executive functioning (Fontes et al., 2011). This was con- in the medial and left superior prefrontal cortices and bilateral
firmed by relating earlier onset of marihuana use, and the increased insula. By contrast, an increased response in the right precentral
frequency and magnitude of marihuana use, with poorer cognitive gyrus was evident in abstinent users (Schweinsburg et al., 2010).
function relative to later onset users (Gruber et al., 2012). Another The activation detected by fMRI in a group of abstinent cannabis
study found that the age of onset of marihuana use was the only users on a Go/No-Go task has been compared with healthy subjects
factor predicting impaired visual attention (Ehrenreich et al., 1999). (Tapert et al., 2007). Although a similar level of task performance
Despite these findings, the issue of the importance of early onset was achieved by both groups, adolescent cannabis users exhibited
of cannabis abuse has been contested. It was reported that current stronger activation during inhibitory trials than healthy subjects
(but not past) heavy cannabis users performed worse than controls in the right dorsolateral prefrontal, bilateral medial frontal, bilat-
in terms of verbal memory, with no differences between men and eral inferior and superior parietal lobules, and the right occipital
women (Pope et al., 2001). Indeed, only current heavy cannabis gyrus when. In the non-inhibitory trials, differences were mainly
users showed a decline in IQ, memory and processing speed (but found in the right prefrontal, insular and parietal cortices, with
not other abilities) at the age of 17–20 years of age relative to their cannabis users showing greater activation in these areas than the
baseline performance at 9–12 years of age (Fried et al., 2002). controls. These results suggest a greater neurocognitive effort by
The second question has no clear answer. While Meier’s report cannabis users during the task, even after the abstinence period.
provides evidence for a general impairment, a meta-analysis of Indeed, these findings might reflect a sort of neuroadaptation, i.e.
the non-acute effects of cannabis use reported significant, yet the recruitment of additional regions as a compensatory mech-
very modest, adverse effects of cannabis on measures of learning anism to maintain normal cognitive performance in response to
and forgetting. Thus, the detrimental effects of cannabis may be chronic cannabis exposure, most prominently in the PFC (Batalla
specific to some neurocognitive functions as opposed to general et al., 2013). In addition to all these data, the specific importance
(Grant et al., 2003). of the age of onset of marijuana use has been recently highlighted
The data available do not provide a definite answer to the third in a fMRI study that showed similar structural alterations in recre-
question (i.e. are these deficits restored after abstinence?). For ational cannabis as those observed in heavy users (decreased gray
example, it appeared that cessation of cannabis use did not fully matter density in the left parahippocampal gyrus and the right tem-
restore neuropsychological function (Meier et al., 2012), as con- poral lobe) only when they began consumption early in their lives
firmed by a recent study (Smith et al., 2015). More recently, heavy (Battistella et al., 2014).
cannabis users were shown to perform worse than controls at base-
line as well as at 1 and 7 days after supervised abstinence, but not 6.2. Animal studies
after 28 days (Pope et al., 2001). Clearly, more research is needed
to provide a clear answer to these questions however one of the 6.2.1. Learning and memory
merits of Meier et al.’s report is to underscore the importance of In most of the studies assessing long-term cognitive con-
prevention and intervention efforts related to adolescent onset and sequences of adolescent cannabis exposure, the novel object
persistent regular use of cannabis (Gonzalez and Swanson, 2012). recognition task or the novel place recognition task have been used
(Abush and Akirav, 2013, 2012; Cadoni et al., 2013; Higuera-Matas
6.1.2. Neuroimaging studies et al., 2009; Llorente-Berzal et al., 2013; Mateos et al., 2011; O’Shea
A systematic review of the neuroimaging studies that focus et al., 2006, 2004; O’Tuathaigh et al., 2010; Quinn et al., 2008;
on adolescent chronic cannabis users identified two performed Realini et al., 2011; Renard et al., 2013; Schneider and Koch, 2003;
on chronic adolescent cannabis users after 28 days of abstinence Schneider et al., 2005; Schulz et al., 2013; Zamberletti et al., 2014,
(Batalla et al., 2013). In these reports, alterations in the volume of 2012), although the Y maze (Cadoni et al., 2013; Mateos et al., 2011;
130 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

Table 5
Behavioral and cognitive alterations found in adult animals after adolescent cannabinoid exposure.

Species Sex Treatment Test age Results Reference

Drug Dose Age

Novel object recognition


Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.) PND45-60 PND61, 70, 90 Memory impairment up to 10 Abush and Akirav, 2012
days after withdrawal
Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.) PND45-60 PND91 Memory impairment Abush and Akirav, 2013
Rat ♂ 9 -THC 2, 4 and 8 mg/kg (i.p.) PND40-42 PND70-80 No deficits shown Cadoni et al., 2013
twice a day for 1 day
each dose
Rat ♂–♀ CP 55,940 0.4 mg/kg/day (i.p.) PND28-38 PND103 No deficits shown Higuera-Matas et al., 2009
Rat ♂–♀ 9 -THC 2.5, 5 and 10 mg/kg PND28-45 PND75 Memory impairment ♀ Llorente-Berzal et al., 2013
(i.p.), for 7, 6 and 5
days, respectively
Rat ♂–♀ CP 55,940 0.4 mg/kg/day (i.p.) PND28-43 PND76 Memory impairment ♂ Mateos et al., 2011
Rat ♀ CP 55,940 0.15, 0.2 and 0.3 mg/kg PND30-50 PND72 Memory impairment O’Shea et al., 2004
(i.p.) for 3, 8 and 10
days, respectively
Rat ♂ CP 55,940 0.15, 0.2 and 0.3 mg/kg PND30-50 PND79, 81 Memory impairment O’Shea et al., 2006
(i.p.) for 7 days each
dose
Mouse ♂–♀ 9 -THC 4 or 8 mg/kg (s.c.) PND32-52 >PND73 Memory impairment O’Tuathaigh et al., 2010
Rat ♂ 9 -THC 1 mg/kg (i.p.) for 2 days PND32-48 PND67 Memory impairment Quinn et al., 2008
and 5 mg/kg on
alternate days (8 doses)
Rat ♀ 9 -THC 2.5, 5 and 10 mg/kg PND35-45 PND98-104 Memory impairment Realini et al., 2011
(i.p.), for 3, 4 and 4
days, respectively
(twice a day)
Rat ♂ CP 55,940 0.15, 0.2 and 0.3 mg/kg PND29-49 PND77, 84 Memory impairment Renard et al., 2013
(i.p.) for 7 days at each
dose
Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.). 1 dose: PND40-65 PND85-90 Memory impairment Schneider and Koch, 2003
10 days; 2 doses: 5
days; 0 doses: 10 days
(randomized)
Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.). 1 dose: PND15-40 PND80 No deficits shown Schneider et al., 2005
10 days; 2 doses: 5
days; 0 doses: 10 days
(randomized)
Rat ♂ WIN 55,212-2 7.5 mg/kg (i.p.). 1 dose: PND40-65 PND75 No deficits shown Schulz et al., 2013
10 days; 2 doses: 5
days; 0 doses: 10 days
(randomized)
Rat ♂–♀ 9 -THC 2.5, 5 and 10 mg/kg PND35-45 PND65 Memory impairment ♀ Zamberletti et al., 2012
(i.p.), for 3, 4 and 4
days, respectively
(twice a day)
Rat ♀ 9 -THC 2.5, 5 and 10 mg/kg PND35-45 >PND75 Memory impairment ♀ Zamberletti et al., 2014
(i.p.), for 3, 4 and 4
days, respectively
(twice a day)

Novel place recognition


Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.) PND45-60 PND61, 70, 90, 135 Impaired spatial memory up to Abush and Akirav, 2012
75 days after withdrawal
Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.) PND45-60 PND91 No deficits shown Abush and Akirav, 2013
Rat ♂–♀ CP 55,940 0.4 mg/kg/day (i.p.) PND28-43 PND70 Impaired spatial memory ♀ Mateos et al., 2011
Rat ♂ CP 55,940 0.15, 0.2 and 0.3 mg/kg PND29-49 PND90, 97 Impaired spatial memory Renard et al., 2013
(i.p.) for 7 days at each
dose
Rat ♀ 9 -THC 2.5, 5 and 10 mg/kg PND35-45 >PND75 Impaired spatial memory ♀ Zamberletti et al., 2014
(i.p.), for 3, 4 and 4
days, respectively
(twice a day)

Morris water maze


Rat ♂ WIN 55,212-2 1 mg/kg (i.p.) PND30-50 >PND70 Impaired spatial learning and Abboussi et al., 2014
memory
Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.) PND45-60 PND61, 70 Impaired spatial memory 24 h Abush and Akirav, 2012
after withdrawal
Rat ♂ 9 -THC 5 mg/kg (i.p.) PND35-55 PND83 No deficits shown Cha et al., 2006
Rat ♂–♀ 9 -THC 5 mg/kg (i.p.) PND35-55 PND83 No deficits shown Cha et al., 2007
Rat ♂–♀ CP 55,940 0.4 mg/kg/day (i.p.) PND28-38 PND107 No deficits shown Higuera-Matas et al., 2009

Morris water maze (no-spatial)


Rat ♂ 9 -THC 5 mg/kg (i.p.) PND35-55 PND85 No deficits shown Cha et al., 2006
Rat ♂–♀ 9 -THC 5 mg/kg (i.p.) PND35-55 PND85 No deficits shown Cha et al., 2007
A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146 131

Table 5 (Continued)

Species Sex Treatment Test age Results Reference

Drug Dose Age

Y maze
Rat ♂ 9 -THC 2, 4 and 8 mg/kg (i.p.) PND40-42 PND70-80 No deficits shown Cadoni et al., 2013
twice a day for 1 day
each dose
Rat ♂–♀ CP 55,940 0.4 mg/kg/day (i.p.) PND28-43 PND44 Enhanced spatial working Mateos et al., 2011
memory ♂
Mouse ♂–♀  -THC 9
4 or 8 mg/kg (s.c.) PND32-52 >PND73 Impaired spatial working O’Tuathaigh et al., 2010
memory ♀ (8 mg/kg)
Rat ♀ 9 -THC 2.5, 5 and 10 mg/kg PND35-45 Not specified Impaired spatial working Rubino et al., 2014
(i.p.), for 3, 4 and 4 memory
days, respectively
(twice a day)

Active place avoidance


Rat ♂ WIN 55,212-2 1 mg/kg (i.p.) PND30-50 >PND70 No deficits shown Abboussi et al., 2014
Rat ♂–♀ 9 -THC 2 mg/kg (i.p.) PND22-40 PND73 Impaired spatial learning and Harte and Dow-Edwards, 2010
memory
PND41-60 PND76 No deficits shown

Passive place avoidance


Rat ♂ 9 -THC 2.5, 5 and 10 mg/kg PND35-45 >PND75 No deficits shown Rubino et al., 2009
(i.p.), for 3, 4 and 4
days, respectively
(twice a day)

Radial maze
Rat ♂–♀ 9 -THC 2 mg/kg (i.p.) PND35-45 >PND75 Impaired spatial working Rubino et al., 2009
memory

Fear conditioning
Mouse ♂ WIN 55,212-2 2 mg/kg (i.p.) PND30-32, 34 and >PND120 Impaired fear conditioning Gleason et al., 2012
39

Holeboard
Mouse ♂ 9 -THC 7 mg/kg every other PND28-48 PND138 No deficits shown Tantra et al., 2014
day (i.p.)

Attentional Set-Shifting Task


Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.). 1 dose: PND40-65 PND85-100 Impaired behavioral flexibility Gomes et al., 2014
10 days; 2 doses: 5
days; 0 doses: 10 days
(randomized)

O’Tuathaigh et al., 2010; Rubino et al., 2014), the active avoidance have also been observed in the novel place recognition task, which
task (Abboussi et al., 2014; Harte and Dow-Edwards, 2010), pas- has a strong spatial component (see below).
sive avoidance (Rubino et al., 2009), the radial maze (Rubino et al., Studies using the novel object recognition task or the novel
2009), the holeboard (Tantra et al., 2014) and the Morris water maze place recognition task have shown that adolescent exposure to
(Abush and Akirav, 2012; Cha et al., 2007, 2006; Higuera-Matas cannabinoids can indeed produce cognitive sequelae in adulthood.
et al., 2009) have also been used. In addition, emotional mem- For example, recognition and/or place memories were impaired
ory was also examined in one study by assessing fear conditioning in female rats (males were not tested in these experiments) that
memory (Gleason et al., 2012: see Table 5). were exposed to THC as adolescents and that were tested as adults
When spatial learning of adolescent rats was examined in the (Realini et al., 2011; Zamberletti et al., 2014). Similarly, two addi-
water maze 28 days after chronic THC treatment, no long-lasting tional studies confirmed that adolescent exposure to THC was
cognitive alterations were observed in either male or female rats associated with impaired performance in the novel object recogni-
(Cha et al., 2007, 2006). Similarly, no significant alterations in tion task in adulthood, again in females (Llorente-Berzal et al., 2013;
water maze performance, neither in reference memory tests nor Zamberletti et al., 2012). Notably, in Llorente-Berzal’s experiments,
working memory tests, were detected in adult male or female the overall effects of THC in the novel object task were probably due
rats that had been treated with the cannabinoid agonist CP during to a potentiated amnesic action in one of the THC groups that was
adolescence (Higuera-Matas et al., 2009). This was further con- also exposed to 3,4-methylenedioxy-methamphetamine (MDMA).
firmed when water maze performance (acquisition, short-term, Independently, a deficit in novel object recognition was also found
long-term memory and reversal learning) was assessed in rats in adult male rats chronically exposed to THC as adolescents (Quinn
exposed to the cannabinoid agonist WIN during adolescence and et al., 2008). These results were found in both wild type mice
after a 10 days drug-free period (Abush and Akirav, 2012). It could and those carrying a deletion of the catechol-O-methyltransferase
be argued that the spatial component of learning and memory is (COMT) gene. Moreover, there was a general interaction between
not affected by adolescent cannabinoid exposure; however, adult sex and treatment showing that males were more affected than
rats of both sexes exposed to THC during adolescence appear to females, yet this included a mixture of heterozygous and KO mice
perform worse than control animals in the radial maze (Rubino making it difficult to establish a clear sexual dimorphism in the
et al., 2009). Indeed, adult rats that were exposed to WIN during effect of THC (O’Tuathaigh et al., 2010). Other studies also pro-
adolescence did show impaired spatial learning and memory in vided evidence for cognitive impairment in both male and female
the water maze (Abboussi et al., 2014). Moreover, some deficits adult rats (O’Shea et al., 2006, 2004). Notably, chronic exposure
132 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

to CP during adolescence induced deficits in the novel object when animals were tested as adults, suggesting no lasting changes
and place location tasks in adulthood, in a sex-dependent man- in anxiety (Higuera-Matas et al., 2009). This was confirmed else-
ner (CP-exposed males showed deficits in the novel object task where in both the EPM and open field tests (Abush and Akirav,
while CP-exposed females performed worse in the object loca- 2013, 2012; Bambico et al., 2010; Bortolato et al., 2014; Llorente-
tion task: Mateos et al., 2011), although we failed to find evidence Berzal et al., 2013; Mateos et al., 2011; O’Tuathaigh et al., 2010;
for such impairments with a more restricted treatment (Higuera- Rubino et al., 2008). However, a more recent report showed that
Matas et al., 2009). Deficits in both object location and novel object chronic THC administration during adolescence evoked an anx-
tasks were also found in males when the WIN was administered, iogenic phenotype in adult animals revealed by less time spent
even after several drug-free periods (1, 10, 30 and 75 days: Abush in the open arms of the EPM (Stopponi et al., 2014). In addition,
and Akirav, 2013, 2012). These deficits seem to be specific to early increased anxiety at adulthood after adolescent cannabinoid expo-
exposure, because when an equivalent WIN treatment was admin- sure was reported only when anxiety was measured indirectly
istered to prepubertal, adult or pubertal rats and they were tested in the form of anxiety-induced hyponeophagia, the inhibition of
as adults, only the latter displayed deficits in object recognition ingestion and the approach to food when an animal is exposed
memory (Schneider and Koch, 2003; Schneider et al., 2005; but to an anxiety-provoking novel environment. Using this behavioral
see Schulz et al., 2013). Similar results were obtained in female paradigm, enhanced anxiety was evident in cannabinoid-exposed
rats exposed to CP during adolescence but not adulthood (O’Shea rats (Bambico et al., 2010).
et al., 2004). It appears that the aforementioned cognitive deficits There seems to be a stronger agreement with regards social
may also depend on the genetic make-up of the individuals, in this anxiety, and adolescent cannabinoid treatment is reported to pro-
sense strain-specific effects have been reported in terms of the sen- duce anxiogenic responses when animals reach adulthood. For
sitivity to the long-lasting amnesic actions of chronic cannabinoid example, when social interaction was analyzed in female rats
exposure during adolescence, as measured in the object location as a measure of anxiety, CP treatment during adolescence but
test (Renard et al., 2013 but see Cadoni et al., 2013). not adulthood was hampered social interaction, a fact that was
Deficits in reversal learning have also been found using the interpreted as reflecting increased anxiety in CP-exposed animals
active avoidance task. While THC administration during early ado- (O’Shea et al., 2004). These results in the females were confirmed
lescence had no effect on acquisition, it hindered performance in elsewhere using THC administration instead of CP (Realini et al.,
the reversal trial of both male and female rats exposed to the 2011; but see Zamberletti et al., 2012). Interestingly, when male
cannabinoid during early but not late adolescence (Harte and Dow- rats were tested, both adolescent and adult exposure to CP gener-
Edwards, 2010). In a recent study, deficits in a reversal task were ated a similar decrease in social interaction (O’Shea et al., 2006).
also found using WIN as the cannabinoid (Gomes et al., 2014). Inter- In part, these conflicting results may be explained by the incidence
estingly, another study using WIN reported impaired performance of genetic alterations among individuals that may modulate the
in a two-way active avoidance task (Abboussi et al., 2014). How- organism’s response to cannabinoids. In fact, it has been shown
ever, no differences in the passive avoidance task were evident as that a strong-weak activity COMT polymorphism may modulate
a result of adolescent THC exposure (Rubino et al., 2009). Emo- the effects of adolescent THC exposure on the risk of developing
tional memory is also affected by adolescent cannabinoid exposure psychosis in adulthood. Indeed, while adolescent exposure to THC
and contextual fear conditioning was impaired in adult mice that has no effects on anxiety measured in the EPM in wild-type ani-
had been exposed to the cannabinoid agonist WIN during adoles- mals, a COMT knock-out mice display decreased anxiety in this
cence (Gleason et al., 2012). This effect was not evident when the test (O’Tuathaigh et al., 2010). Notably, this anxiolytic effect was
cannabinoid agonist was administered during adulthood. only evident when animals were treated as adolescents and expo-
From all these results summarized above, it can be concluded sure to THC as adults produces no overt anxiolytic phenotype.
that adolescent cannabinoid exposure produces deleterious effects Another study also showed the influence of genetic background
on recognition, emotional and spatial memory, although this is not on the effects of adolescent THC on anxiety at adulthood. By using
the case when cannabinoid exposure occurs during the prepubertal the Lewis and Fischer 344 rat strains as a model (see Kosten and
or adult periods. In addition, while the gender specificity of these Ambrosio (2002) for a review), adolescent THC exposure was asso-
effects is not clear, whenever a sex-difference was noted, females ciated with less anxiety in adulthood but only in the Lewis strain,
showed the impairments with only one exception (Mateos et al., with no such effects evident in Fischer 344 rats (Cadoni et al., 2013).
2011). In any case, further research should be carried out on this Hedonic responses are another kind of emotional responses
issue. affected by adolescent cannabinoid exposure. Anhedonia is defined
as a blunting of sensitivity to rewarding stimuli, and it is a core
6.2.2. Emotional regulation symptomatological feature of depression. One way to measure
There is evidence that adolescent cannabinoid exposure alters anhedonia is the sucrose preference test, where animals are given
emotional regulation in adulthood, although these results vary the opportunity to choose between a sucrose solution (a natu-
depending on the particular index or process examined (see ral reward in rodents) and tap water. A consistent finding is that
Table 6). Thus, if we look at anxiety responses, we can find reduced adolescent THC induces anhedonia at adulthood evident in this
anxiety-related behavior has been observed in adulthood after test. For example, both male and female animals exposed to THC
chronic adolescent cannabinoid exposure. For example, rats (both as adolescents exhibited decreased sucrose preference as adults
males and females) that had been exposed to the cannabinoid ago- (Rubino et al., 2008; Bambico et al., 2010; Realini et al., 2011).
nist CP during adolescence spent longer in the open arms of an EPM, This phenotype could be reverted by increasing eCB levels through
indicating reduced anxiety (Biscaia et al., 2003). Likewise, puber- the administration of URB597, a FAAH inhibitor (Realini et al.,
tal treatment with WIN (under an irregular administration regime) 2011). There is however, one report showing that sucrose pref-
induced an anxiolytic phenotype evident in the EPM (longer spent erence is not affected in adult female rats exposed to CP during
in the open arms) and in the open field (higher percentage of time adolescence (Chadwick et al., 2011), which may perhaps reflect dif-
spent in the central zone: Wegener and Koch, 2009). Conversely, no ferences in the pharmacodynamic profiles between THC, WIN and
alterations to measures of anxiety were evident in adulthood after CP.
adolescence cannabinoid exposure in other studies. For example, Although not a direct measure of depressive-like phenotype, the
we treated adolescent rats with the cannabinoid agonist CP and FST has also been used to characterize emotional responses in adult
found no change in the time spent in the open arms of the EPM rats exposed to cannabinoids as adolescents. Adolescent exposure
A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146 133

Table 6
Emotional behavior alterations after adolescent cannabinoid exposure.

Species Sex Treatment Test age Results Reference

Drug Dose Age

Elevated plus maze


Rat ♂ WIN 55,512-2 0.2 or 1 mg/kg (i.p.) PND70-100 >PND70 No significant Bambico et al., 2010
effects
Rat ♂–♀ CP 55,940 0.4 mg/kg/day (i.p.) PND35-45 PND75-84 Anxiety ↓ Biscaia et al., 2003
Rat ♂ WIN 55,212-2 1 mg/kg (i.p.) twice a day PND35-48 PND60-70 No significant Bortolato et al., 2014
effects
Rat ♂ 9 -THC 2, 4 and 8 mg/kg (i.p.) twice a PND40-42 PND70-80 Anxiety ↓ Cadoni et al., 2013
day for 1 day each dose
Rat ♂–♀ CP 55,940 0.4 mg/kg/day (i.p.) PND28-38 PND100 No significant Higuera-Matas et al., 2009
effects
Rat ♂–♀ 9 -THC 2.5, 5 and 10 mg/kg (i.p.), for 7, PND28-45 PND46 No significant Llorente-Berzal et al., 2013
6 and 5 days, respectively. effects
Rat ♂–♀ CP 55,940 0.4 mg/kg/day (i.p.) PND28-43 PND64 No significant Mateos et al., 2011
effects
Mouse ♂ 9-THC 4 or 8 mg/kg (s.c.) PND32-52 PND73 No significant O’Tuathaigh et al., 2010
effects
Rat ♂–♀ 9-THC 2.5, 5 and 10 mg/kg (i.p.), for 3, PND35-45 PND75-85 No significant Rubino et al., 2008
4 and 4 days, respectively effects
(twice a day)
Rat ♂ 9-THC 2.5, 5 and 10 mg/kg (i.p.), for 3, PND35-45 PND52 Anxiety ↑ Stopponi et al., 2014
4 and 4 days, respectively
(twice a day)
Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.). 1 dose: 10 PND40-65 PND90 Anxiety ↓ Wegener and Koch, 2009
days; 2 doses: 5 days; 0 doses:
10 days (randomized)

Open field
Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.) PND45-60 PND61, 70, 90 Anxiety ↓ 24 h after Abush and Akirav, 2012
withdrawal
Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.) PND45-60 PND91 No significant Abush and Akirav, 2013
effects
Rat ♂ WIN 55,512-2 0.2 or 1 mg/kg (i.p.) PND70-100 >PND70 No significant Bambico et al., 2010
effects
Rat ♂–♀ CP 55,940 0.4 mg/kg/day (i.p.) PND35-45 PND75-84 No significant Biscaia et al., 2003
effects
Rat ♂–♀ 9 -THC 2.5, 5 and 10 mg/kg (i.p.), for 7, PND28-45 PND41, 75 Anxiety ↑ Llorente-Berzal et al., 2013
6 and 5 days, respectively.
Rat ♂–♀ 9-THC 2.5, 5 and 10 mg/kg (i.p.), for 3, PND35-45 PND75-85 No significant Rubino et al., 2008
4 and 4 days, respectively effects
(twice a day)
Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.). 1 dose: 10 PND40-65 PND85 Anxiety ↓ Wegener and Koch, 2009
days; 2 doses: 5 days; 0 doses:
10 days (randomized)

Social interaction
Mouse ♂ WIN 55,212-2 2 mg/kg (i.p.) PND30-32, 34 and 39 PND120+ No significant Gleason et al., 2012
effects
Rat ♀ CP 55,940 0.15, 0.2 and 0.3 mg/kg (i.p.) for PND30-50 PND73 Social anxiety ↑ O’Shea et al., 2004
3, 8 and 10 days, respectively
Rat ♂ CP 55,940 0.15, 0.2 and 0.3 mg/kg (i.p.) for PND30-50 PND80 Social anxiety ↑ O’Shea et al., 2006
7 days each dose
Rat ♂  -THC
9
1 mg/kg (i.p.) for 2 days and PND32-48 PND63 Social anxiety ↑ Quinn et al., 2008
5 mg/kg on alternate days (8
doses)
Rat ♀ 9-THC 2.5, 5 and 10 mg/kg (i.p.), for 3, PND35-45 PND98-104 Social anxiety ↑ Realini et al., 2011
4 and 4 days, respectively
(twice a day)
Rat ♂–♀ 9-THC 2.5, 5 and 10 mg/kg (i.p.), for 3, PND35-45 PND68 No significant Zamberletti et al., 2012
4 and 4 days, respectively effects
(twice a day)

Forced swimming test


Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.) PND45-60 PND91 No significant Abush and Akirav, 2013
effects
Rat ♂ WIN 55,512-2 0.2 or 1 mg/kg (i.p.) PND70-100 >PND70 Depressive-like Bambico et al., 2010
behavior ↑
Rat ♀ 9-THC 2.5, 5 and 10 mg/kg (i.p.), for 3, PND35-45 PND98-104 Depressive-like Realini et al., 2011
4 and 4 days, respectively behavior ↑
(twice a day)
Rat ♂–♀ 9-THC 2.5, 5 and 10 mg/kg (i.p.), for 3, PND35-45 PND75-85 Depressive-like Rubino et al., 2008
4 and 4 days, respectively behavior ♀↑
(twice a day)
Rat ♂–♀ 9-THC 2.5, 5 and 10 mg/kg (i.p.), for 3, PND35-45 PND71 Depressive-like Zamberletti et al., 2012
4 and 4 days, respectively behavior ↑
(twice a day)
134 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

Table 6 (Continued)

Species Sex Treatment Test age Results Reference

Drug Dose Age

Sucrose preference
Rat ♂ WIN 55,512-2 0.2 or 1 mg/kg (i.p.) PND70-100 >PND70 Anhedonia ↑ Bambico et al., 2010
Rat ♀ CP 55,940 0.4 mg/kg/day (i.p.) PND35-45 PND97-99 No significant Chadwick et al., 2011
effects
Rat ♀ 9-THC 2.5, 5 and 10 mg/kg (i.p.), for 3, PND35-45 PND98-104 Anhedonia ↑ Realini et al., 2011
4 and 4 days, respectively
(twice a day)
Rat ♂–♀ 9-THC 2.5, 5 and 10 mg/kg (i.p.), for 3, PND35-45 PND75-85 Anhedonia ↑ Rubino et al., 2008
4 and 4 days, respectively
(twice a day)

Sucrose intake
Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.) PND45-60 PND61, 70 and 100 No significant Abush and Akirav, 2013
effects

Palatable food preference


Rat ♀ 9-THC 2.5, 5 and 10 mg/kg (i.p.), for 3, PND35-45 PND98-104 Anhedonia ↑ Realini et al., 2011
4 and 4 days, respectively
(twice a day)

Novelty suppressed feeding


Rat ♂ WIN 55,512-2 0.2 or 1 mg/kg (i.p.) PND70-100 >PND70 Anxiety ↑ (1 mg/kg Bambico et al., 2010
dose)

Emergence test
Rat ♂ CP 55,940 0.15, 0.2 and 0.3 mg/kg (i.p.) for PND30-50 PND82 No significant O’Shea et al., 2006
7 days each dose effects

to THC increased the immobility time in adult rats (although only deprivation)-induced reinstatement (Tomasiewicz et al., 2012).
in females: Rubino et al., 2008), and it was shown that this phe- However when a pharmacological stressor (yohimbine) was used
notype could be reverted by increasing eCBs levels (Realini et al., to reinstate heroin self-administration behavior, the response
2011). However, these sex-specific effects were not reproduced and of THC exposed animals was stronger than that of the controls
increased immobility was seen in both males and females (Bambico (Stopponi et al., 2014). In a follow-up study, PENK was shown to
et al., 2010; Zamberletti et al., 2012). By contrast, no changes in be a crucial factor mediating the enduring effects of adolescent
immobility time were observed in the FST on adults after late- cannabis exposure on adult opiate vulnerability (Tomasiewicz
adolescent exposure to WIN (Abush and Akirav, 2013). et al., 2012). Indeed, the enhanced heroin self-administration
induced by adolescent THC exposure was impaired when PENK
6.2.3. Sensitivity to drugs of abuse later in life and potential levels were decreased by viral-mediated transfer of a miRNA tar-
addictive behavior geting the PENK gene. The increased vulnerability to the rewarding
There is evidence that adolescent use of marihuana could actions of opiates induced by adolescent cannabinoid exposure has
facilitate later drug abuse, a phenomenon that has been termed also been observed with CPP (Cadoni et al., 2013; Morel et al., 2009).
the Gateway hypothesis (Kandel et al., 2006). Earlier animal There seems to be some sex-dependency in these effects.
studies on psychostimulant drugs did not seem to support this Indeed, when we exposed male and female adolescent rats to CP
hypothesis, with adolescent exposure to THC producing no long- during adolescence and tested their morphine self-administration
lasting modulatory effects on locomotor activity or dopaminergic as adults, increased self-administration was only evident in males
responses induced by amphetamine (Ellgren et al., 2004). How- (Biscaia et al., 2008). These effects of adolescent cannabinoid
ever, it was later shown that adolescent exposure to THC indeed exposure on the rewarding effects of morphine in adulthood might
affected heroin reward in rats. In a series of elegant experiments, be transgenerational. Female rats treated with WIN or its vehicle
increased heroin self-administration (30 mg/kg/infusion) was seen for three consecutive days during adolescence (30 days of age)
in THC-exposed rats as compared to control animals, and there was were subsequently mated in adulthood (60 days of age), and the
also an upwards shift in the dose-response curve (Ellgren et al., adolescent and adult male offspring of these females were tested
2007). This heightened opiate sensitivity in THC animals was also for their response to morphine using the CPP paradigm (Byrnes
associated to increased cue-induced relapse (Tomasiewicz et al., et al., 2012). Both adolescent and adult WIN-F1 offspring exhibited
2012). There is however, one report suggesting that adolescent greater sensitivity to morphine CPP than their vehicle-F1 counter-
THC is not associated to increased heroin self-administration parts. A similar effect was observed on morphine sensitization in
during adulthood (Stopponi et al., 2014). These conflicting results the adult female offspring of rats treated with WIN during adoles-
could be due to the heroin dose used during training. Indeed. Stop- cence (Vassoler et al., 2013). These transgenerational effects were
poni et al., used a dose of 20 ␮g/kg, and when Ellgren et al., first also evident in the heroin self-administration paradigm, whereby
tested a dose of 15 ␮g/kg, no differences were evident between adolescent exposure to THC resulted in compulsive heroin intake
animals receiving THC or the vehicle alone. However, signifi- in the subsequent generation of rats as a consequence of parental
cant differences were later seen in the range of 7.5–30 ␮g/kg, germline exposure to the drug (Szutorisz et al., 2014). The facili-
suggesting that these conflicting results may be the result of: tating effects of adolescent THC exposure on opiate reward during
the different THC treatment regimes administered (1.5 mg/kg; adulthood also depend on the genetic background of individuals.
Postnatal day (PND) 28–49 in Ellgren et al., and Tomasiewicz et al., Adolescent THC exposure in Lewis rats did not affect heroin CPP
and increasing doses from 2.5 to 10 mg/kg; PND 35–45 in Stopponi but potentiated the effect of heroin priming on drug seeking. By
et al.); the rat strain used (Wistar vs. Long–Evans); or an interac- contrast, adolescent THC exposure to Fischer 344 rats increased
tion of both factors. As mentioned above, THC exposure during heroin CPP and made it resistant to extinction. THC-treated Lewis
adolescence also increased cue-induced but not stress (24 h food rats also showed increased seeking reactions during extinction and
A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146 135

higher hedonic reactions in response to heroin priming (Cadoni dorsal striatum of the adult F1 offspring of rats exposed to THC in
et al., 2013). their adolescence (Szutorisz et al., 2014).
With regard to psychostimulants, although earlier reports Apart from the aforementioned changes, the responsiveness
showed that THC exposure during adolescence produced no long- of the dopaminergic system to pharmacological stimuli is also
lasting changes in the motor response to amphetamine, recent affected. For example, the dynamics of the dopaminergic response
studies provide conflicting data with an increased response to in the NAcc was not affected after THC or heroin challenges in Lewis
amphetamine challenge (only in females in this one case: Gomes and Fischer 344 rats that had been exposed to THC during their
et al., 2014; Lee et al., 2014). We also demonstrated a gateway adolescence (Cadoni et al., 2013). However, the responsiveness to
effect in female rats for cocaine self-administration in adulthood heroin was altered after adolescent THC such that the increase
following adolescent CP administration. Yet no differences were in dopamine after a heroin challenge was higher in THC-exposed
observed during the maintenance phase once the behavior had animals in the NAcc-core (Fischer 344 rats) or both NAcc-core
been stably acquired and this effect was not observed when a and NAcc–shell (Lewis rats). By contrast, single-unit extracellular
natural reward (food pellets) was used as a reinforcer (Higuera- recordings obtained from antidromically identified mesoaccum-
Matas et al., 2008). The altered sensitivity to the initial rewarding bens dopaminergic neurons, and from their target cells in the NAcc,
effects of cocaine could be explained by modifications to the brain’s show that dopaminergic neurons were significantly less responsive
response to acute cocaine in our animals. We used positron emis- to stimulation by WIN in animals pretreated for 3 days during ado-
sion tomography to monitor brain responses to an acute cocaine lescence or adulthood with WIN and allowed a 2-week washout
challenge and found that chronic cannabinoid treatment abolished (Pistis et al., 2004). However, in the group pretreated in adolescence
the decrease in metabolic activity in the dorsal striatum of males but not in adulthood, long-lasting cross-tolerance to morphine,
that was observed after cocaine was administered to control rats. In cocaine and amphetamine developed. They concluded that an
addition, septal metabolism at resting conditions was higher in CP- enduring form of neuronal adaptation occurs in dopaminergic neu-
exposed females but it reached the basal values observed in control rons after sub-chronic cannabinoid intake at a young age, affecting
female rats after cocaine injection (Higuera-Matas et al., 2011). the subsequent responses to drugs of abuse. Regarding this matter,
A recent cohort study in humans has provided evidence for a an increased number of spontaneously active VTA dopaminergic
so called “reverse-gateway effect” whereby cannabis consumption neurons were seen recently in adult rats treated with WIN during
during adolescence would be associated to increased tobacco con- puberty in a recent study by (Gomes et al., 2014).
sumption later in life (Badiani et al., 2015). Animal studies are It is tempting to speculate that the altered function of the DA sys-
needed to experimentally verify this association and further inves- tem (especially the increased dopamine uptake and turnover, the
tigate the underlying neurobiological mechanisms. increased number of spontaneously active VTA dopamine neurons
and the enhanced dopamine levels after heroin) in cannabinoid
exposed animals might be responsible for the enhanced reward
6.2.4. Monoaminergic systems properties of opiates and psychostimulants reported before. Stri-
Chronic adolescent cannabinoid exposure is associated to atal D2 receptors might also play an important role as indicated by
dampened dopaminergic activity in adulthood and chronic ado- perinatal studies, although the evidence is conflicting, with some
lescent THC treatment reduced TH cell density in the VTA of mice studies reporting increases (Zamberletti et al., 2012), others no
(Behan et al., 2012). Not only were there fewer dopaminergic cells changes (Higuera-Matas et al., 2010) or even decreases (Szutorisz
but there is also increased dopamine turnover in adult animals et al., 2014).
exposed to cannabinoids during adolescence. Indeed, when the Very little is known about the long-term effects of adolescent
dopamine, NE, and DOPAC levels as well as the DOPAC/dopamine cannabinoid exposure on the adult serotoninergic or adrenergic
ratio in the mPFC, and dorsal and ventral striatum were examined, systems. One study looked at the responses of serotoninergic and
evidence was found of disturbed dopaminergic regulation. Indeed, noradrenergic neurons in adult rats exposed to WIN during ado-
a higher DOPAC/dopamine ratio (indicating an increased dopamine lescence. Serotoninergic neural activity in the dorsal raphe was
turnover) was observed in both the dorsal and ventral striatum significantly attenuated in WIN-exposed rats in a manner that was
of adult rats chronically treated with WIN (Bortolato et al., 2014). independent of 5-HT1A receptors and that was probably caused
In addition to fewer dopamine cells and increased turnover, there by dysregulation of CB1Rs (Bambico et al., 2010). When consid-
also seems to be a heightened dopamine uptake. In this respect, ering the mutual innervation existing between the dorsal raphe
we found that adult female but not male rats treated with CP serotoninergic neurons and LC noradrenergic neurons, the neural
during adolescence had higher dopamine transporter levels in the activity of LC noradrenergic neurons was enhanced after chronic
dorsal striatum (Higuera-Matas et al., 2010). D1 receptors were up- adolescent (but not adult) cannabinoid treatment. This neurochem-
regulated in the NAcc-Shell of cannabinoid-exposed males (but not ical picture, i.e. dampened 5-HT function and increased NE tone, is
females), while D2 receptors were down-regulated in the CA1 hip- a good candidate to explain the increased anxiety found in some
pocampal field in animals exposed to cannabinoids, irrespective of studies (Bambico et al., 2010; Gordon and Hen, 2004; O’Shea et al.,
sex. 2006, 2004; Stopponi et al., 2014; Yamamoto et al., 2014). A more
In terms of the changes in receptor levels after adolescent recent set of experiments studied the levels of the serotonin trans-
cannabinoid administration, more D1 receptors were found in the porter (SERT) protein in the hippocampus and parietal cortex of
NAcc of female but not male animals, and more D2 receptor levels adult rats treated with THC during adolescence. In males, THC
in the PFC of male but not female rats (Zamberletti et al., 2012). increased the number of SERT positive fibers while no such effect
There was also evidence of higher D2 levels in the NAcc in both was evident in the females (Lopez-Rodriguez et al., 2014).
sexes (Zamberletti et al., 2012). The different results obtained in
the aforementioned studies may reflect the distinct cannabinoids 6.2.5. Glutamatergic and GABAergic systems
used, the different age at which treatment was administered or the When exposed to cannabinoid as adolescents, a profound sex-
differences in the technical details of the autoradiography studies dependent disturbance of the glutamate/GABA balance in the
(different ligands used or the lack of reported non-specific binding hippocampus of adult rats became evident (Higuera-Matas et al.,
controls). Some of the changes in the dopaminergic system might 2012). Indeed, CP-treated females exhibit increased GABAA recep-
also be transgenerational and in this respect, weaker expression tor levels, as well as decreased K+ -induced glutamate release and
of the gene encoding the D2 receptor (Drd2A) was reported in the NMDA receptor levels. These females also showed weaker GABA
136 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

transporter 1 gene expression (GAT-1), and increased K+ -induced disinhibition in adulthood comparable to that seen in the juvenile
GABA release, however this effect was also evident in CP-treated PFC (Cass et al., 2014). Such disinhibition could be rescued by local
males. These results suggest enhanced GABAergic and decreased infusion of a GABAA receptor agonist, suggesting that GABAA recep-
glutamatergic tone in the hippocampus of these female rats. Very tors may be downregulated in PFC neurons as a consequence of
similar results were reported for NMDA receptors, whereby female pubertal WIN exposure (Cass et al., 2014).
but not male rats exposed to THC during adolescence had fewer
NMDA receptors in the hippocampus as adults (Zamberletti et al., 6.2.6. Endogenous opioid system
2012). Similarly, fewer NMDA receptors were detected in the hip- Adolescent THC exposure leads to long-term discrete mesolim-
pocampus of adult male rats (females were not studied in this bic and nigrostriatal alterations in elements of the endogenous
work) exposed to cannabinoids during adolescence (Rubino et al., opioid system. Adult male rats treated with THC during adoles-
2009), an effect that was replicated in the dorsal striatum in cence displayed higher levels of PENK mRNA in the NAcc-Shell
the F1 offspring of the rats exposed to THC during adolescence (Ellgren et al., 2007; Tomasiewicz et al., 2012). In addition, these
(Szutorisz et al., 2014). Different subunits of the NMDA and AMPA authors found evidence for increased ␮ opioid receptor activity in
glutamate receptors were also studied in these rats and weaker the substantia nigra and VTA, although binding to ␮ opioid recep-
expression of the Grin1, Grin2A, Gria1 and Gria2 genes (encoding tors was unaffected in this study (Ellgren et al., 2007). All these
the GluN1, GluN2B, GluA1 and GluA2 glutamate receptor subunits, changes in the endogenous opioid systems could be correlated with
respectively) was found in the striatum of the offspring of the THC- the enhanced heroin self-administration and relapse observed in
exposed rats. When the protein expression of these receptors was THC-exposed rats (Ellgren et al., 2007; Tomasiewicz et al., 2012).
assayed in western blots, only GluN1 and GluN2B subunits were By contrast, no changes in ␮ opioid receptor levels or activity were
diminished in THC exposed animals (Szutorisz et al., 2014). The detected in a study that also reported decreased PENK gene expres-
effects of adolescent THC exposure on the glutamatergic system sion in the dorsal striatum and NAcc-Shell (Morel et al., 2009).
have also been studied in the PFC of female rats. In this case, GluN2B However, when the expression of the ␮ opioid receptor was tested
and GluA1 levels at adulthood were higher in THC-exposed rats after a morphine challenge in a sensitization paradigm, the female
(Rubino et al., 2014). In addition, mGlu5 protein levels were dimin- offspring of rats treated with WIN as adolescents expressed more ␮
ished in the hippocampus of adult mice exposed to WIN during opioid receptor (OPRM1) mRNA in the NAcc (Vassoler et al., 2013).
adolescence (Gleason et al., 2012), although we could not replicate An effect of adolescent THC on adult levels of the dynorphin peptide
these findings when CP was the cannabinoid agonist administered was also found in the NAcc of female (but not male) rats (Rubino
to adolescent rats (Higuera-Matas et al., 2012). PSD95, an impor- et al., 2008). Increased dynorphin levels were found in THC-exposed
tant scaffolding protein of the post-synaptic density (reviewed in rats, which were related to the emotional disturbances provoked by
Van Zundert et al., 2004), was diminished in the hippocampus of the cannabinoid treatment. We also analyzed the levels and activ-
THC-exposed male rats (Rubino et al., 2009), yet it increased in the ity of ␮ opioid receptors in several brain areas of male and female
PFC of female rats after THC treatment during adolescence (Rubino rats exposed to CP during adolescence. We found that CP-treated
et al., 2014) which is consistent with the increased levels of the rats (both males and females) displayed more ␮ opioid receptors
glutamatergic subunits mentioned above. in the sub-callosal streak. We also observed significant interactions
The decrease in glutamate function in the hippocampus (i.e. between the sex of the animals and cannabinoid exposure in several
decreased glutamate levels after depolarization, decreased NMDA brain regions, such as the cingulate cortex, CA2 and CA3 hippocam-
receptor levels, decreased PSD95 levels, etc.) is consistent with pal fields and several thalamic nuclei, whereby males exposed to
the learning and memory impairments found in these animals, CP showed an overall decrease while females exposed to CP dis-
although to our surprise, there were not consequences on the played an overall increase in the same regions. In addition, the
establishment of long-term potentiation (LTP) in the hippocampus activity of these receptors was lower in the NAcc-Shell of CP-male
(Higuera-Matas et al., 2009), perhaps due to the saturating pro- but not female rats which could be related to the increased mor-
tocol used for LTP induction. Indeed, other authors were able to phine self-administration evident in CP-exposed animals (Biscaia
observe LTP deficits in the ventral subicular-NAcc pathway (Abush et al., 2008).
and Akirav, 2013, 2012).
With regard GABAergic cells, increased parvalbumin-positive 6.2.7. Endogenous cannabinoid system
cell soma sizes have been detected in the PFC of adult animals The long-lasting effects of cannabinoid administration during
exposed to THC as adolescents with no changes in the density of adolescence on the endogenous cannabinoid system later in life
these cells, suggesting a disturbance of prefrontal GABA function remain controversial. While there are studies where no long-lasting
in THC-exposed animals (Behan et al., 2012). By contrast, fewer changes in immunoreactivity, binding to or functional activation of
parvalbumin- and cholecystokinin-positive GABAergic cells were cannabinoid are reported (Behan et al., 2012; Chadwick et al., 2011;
found in the PFC (Zamberletti et al., 2014), although the total par- Ellgren et al., 2007; Gleason et al., 2012; Higuera-Matas et al., 2012;
valbumin or cholecystokinin protein assayed in western blots was Lopez-Rodriguez et al., 2014; Morel et al., 2009), other reports
no different in THC and control animals. Differences in the kind of describe lower levels of these receptors in adult animals admin-
animals used (mice or rats), the sex (males in the former exper- istered a cannabinoid during adolescence in almost all the areas
iments and females in the latter report) or some combination of studied (Rubino et al., 2014, 2008; Zamberletti et al., 2012). For
both might account for the differences in the results between these these reason it is not possible to draw firm conclusions on the func-
experiments. The levels of GABA were also measured and in the tional relevance of these changes. Thus, in one study using CP as
PFC, lower basal GABA levels were obtained in female rats (males the cannabinoid agent, a down-regulation in the levels of CB1Rs
not tested) that had been exposed to THC during adolescence, a in some hippocampal areas was observed in males while an up-
fact that can be explained by the lower levels of GAD67 (the GABA regulation was detected in the females (López-Gallardo et al., 2012).
synthetic enzyme), in this structure (Zamberletti et al., 2014). The Likewise, a sustained increase in cannabinoid levels during ado-
functional correlate of this altered prefrontal GABA function is still lescence (generated by blocking the FAAH enzyme with URB597)
to be determined but some candidate events have been proposed. was also associated to lower CB1R levels in the hippocampus, cau-
For example, in a series of elegant experiments a history of WIN date putamen, NAcc, VTA and higher levels in the locus coeruleus
exposure during early or mid-adolescence, but not in late adoles- (Marco et al., 2009). However, in a later study where THC was the
cence or adulthood, was sufficient to yield a state of prefrontal cannabinoid agonist administered during adolescence, no changes
A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146 137

Table 7
Neurochemical alterations found in adult animals after adolescent cannabinoid exposure.

Species Sex Treatment Tests Reference

Drug Dose Age Age Results

Monoaminergic systems
Rat ♂ WIN 55,512-2 0.2 or 1 mg/kg (i.p.) PND30-50 >PND70 5-HT neurons activity: DRph ↓; NA Bambico et al., 2010
neurons activity: LC (1 mg/kg dose)

Mouse ♂ 9 -THC 8 mg/kg (s.c.) PND32-52 PND150- TH+ cells density: VTA ↓ Behan et al., 2012
160
Rat ♂ WIN 55,212-2 2 mg/kg (i.p.) PND35-48 PND60-70 DOPAC levels: CPu ↑; DOPAC/DA Bortolato et al., 2014
ratio: NAcc ↑, CPu ↑
Rat ♂ 9 -THC 2, 4 and 8 mg/kg (i.p.) twice PND40-42 PND63-77 DA response to heroin: NAccCo ↑; Cadoni et al., 2013
a day for 1 day each dose NAccSh ↑a
Rat ♂ WIN 55,212-2 1.2 mg/kg (i.p.). 1 dose: 10 PND40-65 PND85-100 Locomotor response to Gomes et al., 2014
days; 2 doses: 5 days; 0 amphetamine ↑
doses: 10 days
(randomized)
>PND100 Spontaneously active DA neurons:
VTA ↑
Rat ♂–♀ CP 55,940 0.4 mg/kg (i.p.) PND28-38 PND121 DAT levels: CPu ↑♀; D1 receptor Higuera-Matas et al., 2010
density: NAccSh ↑♂; D2 receptor
density: HP(CA1-rad) ↓
Rat ♂–♀ 9 -THC 2.5, 5 and 10 mg/kg, for 7, 6 PND28-45 PND89-92 SERT+ fibers: PaCx ↑♂ Lopez-Rodriguez et al., 2014
and 5 days, respectively
(i.p.)
Rat ♂ WIN 55,512-2 2, 4 and 8 mg/kg (i.p.) (one PND38-40 PND54 WIN and Morphine evoked firing Pistis et al., 2004
day each) rate and burst firing: MsLimbPth ↓;
Cocaine and Amphetamine evoked
firing rate: MsLimbPth ↓
Rat ♂ 9 -THC 1.5 mg/kg (i.p.) PND28-49 PND62 (F1 D2 receptor mRNA: CPu ↓(F1) Szutorisz et al., 2014
offspring)
Rat ♂–♀ 9 -THC 2.5, 5 and 10 mg/kg (i.p.), PND35-45 PND82 D1 receptor density: NAcc ↑♀; D2 Zamberletti et al., 2012
for 3, 4 and 4 days, receptor density: PFC ↑♂; NAcc ↑
respectively (twice a day)

Glutamatergic and GABAergic systems


Mouse ♂ WIN 55,512-2 8 mg/kg (s.c.) PND32-52 PND150- Parvalbumine cell soma size: PFC ↑ Behan et al., 2012
160
Rat ♂ WIN 55,512-2 2 mg/kg (i.p.) PND35-40 PND65-85 IPSC frequency: mPFC (layer V) ↓ Cass et al., 2014
Mouse ♂ WIN 55,512-2 2 mg/kg (i.p.) PND30- PND150 mGlu5 levels: HP ↓ Gleason et al., 2012
32/33/34/39
Rat ♂–♀ CP 55,940 0.4 mg/kg (i.p.) PND28-38 PND100- K+ evoked GABA levels: HP ↑; K+ Higuera-Matas et al., 2012
115 evoked Glu levels: HP ♀↓; GAT-1
mRNA: HP ↓;
GABAA receptor density: HP ♀↑;
GABAB receptor density: HP ↑;
NMDA receptor density: HP ♀↓
Rat ♂ 9 -THC 2.5, 5 and 10 mg/kg (i.p.), PND35-45 PND75 NMDA receptor density: HP ↓; Rubino et al., 2009
for 3, 4 and 4 days, PSD95 levels: HP ↓
respectively (twice a day)
Rat ♀ 9 -THC 2.5, 5 and 10 mg/kg (i.p.), PND35-45 PND46, 60 GluN2A levels: PFC ↑ (PND60); Rubino et al., 2014
for 3, 4 and 4 days, and 75 GluN2B and GluA1 levels: PFC ↑
respectively (twice a day) (PND75); PSD95 levels: PFC ↑
Rat ♂ 9 -THC 1.5 mg/kg (i.p.) PND28-49 PND35 (F1 Grin2A and Gria2 mRNA: NAcc ↑ Szutorisz et al., 2014
offspring)
PND62 (F1 Grin1, Grin2A, Gria1 and Gria2
offspring) mRNA: CPu ↓; GluN1 and GluN2B
levels: CPu ↓; NMDA receptor
density: CPu ↓
Rat ♂–♀ 9 -THC 2.5, 5 and 10 mg/kg (i.p.), PND35-45 PND82 NMDA receptor density: HP ↓♀ Zamberletti et al., 2012
for 3, 4 and 4 days,
respectively (twice a day)
Rat ♀ 9 -THC 2, 5, 5 and 10 mg/kg (i.p.) PND35-45 PND75 GAD67 levels and activity; PFC ↓; Zamberletti et al., 2014
for 3, 4 and 4 days, GABA levels: PFC ↓;
respectively (twice a day) GAD67+ /Parvalbumin+ cells: PFC ↓;
GAD67+/Cholecystokinin+ cells:
PFC ↓

Endogenous opioid system


Rat ♂–♀ CP 55,940 0.4 mg/kg (i.p.) PND35-45 PND84 ␮ opioid receptor density: SCst ↑; Biscaia et al., 2008
CinCx ♂↓♀↑; HC ♂↓♀↑; Th ♂↓♀↑;
␮ opioid receptor function: NAccSh
↓♂
Rat ♂ 9 -THC 1.5 mg/kg (i.p.) every third PND28-49 PND57 ␮ opioid receptor density: No Ellgren et al., 2007
day significant alterations found; ␮
opioid receptor function: SN ↑;
VTA ↑; PENK mRNA: NAccSh ↑
138 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

Table 7 (Continued)

Species Sex Treatment Tests Reference

Drug Dose Age Age Results

Rat ♂ Dronabinol (9 -THC) 5 or 10 mg/kg (i.p.). Days 1, PND35-48 PND104 PENK mRNA: NAccSh ↓; CPu ↓ Morel et al., 2009
3, 4 and 6: 1 dose; days 7, 9
10, 11, 13 and 14: 2 doses;
days 2, 5, 8 and 12: No dose
Rat ♂–♀ 9 -THC 2.5, 5 and 10 mg/kg (i.p.), PND35-45 PND46 Dynorphin A levels: NAcc ♀↑ Rubino et al., 2008
for 3, 4 and 4 days,
respectively (twice a day)
Rat ♂ 9 -THC 1.5 mg/kg (i.p.) every third PND28-49 PND79 PENK mRNA: NAccSh ↑ Tomasiewicz et al., 2012
day

Endogenous cannabinoid system


Mouse ♂ 9 -THC 8 mg/kg (s.c.) PND32-52 PND150- CB1 receptor density: No Behan et al., 2012
160 significant alterations found in any
of the areas studied
Rat ♀ CP 55,940 0.4 mg/kg (i.p.) PND35-45 PND46 and CB1 receptor density: No Chadwick et al., 2011
74 significant alterations found in any
of the areas studied
Rat ♂ 9 -THC 1.5 mg/kg (i.p.) every third PND28-49 PND57 CB1 receptor density and function: Ellgren et al., 2007
day No significant alterations found in
any of the areas studied
Mouse ♂ WIN 55,512-2 2 mg/kg PND30- PND150 MGL levels: HP ↑; FAAH levels: HP Gleason et al., 2012
32/33/34/39 ↑
Rat ♂–♀ CP 55,940 0.4 mg/kg (i.p.) PND28-38 PND100- CB1 receptor function: HP (CA1, Higuera-Matas et al., 2012
115 CA2, CA3 and DG) ↑
Rat ♂–♀ CP 55,940 0.4 mg/kg (i.p.) PND28-42 PND80 CB1 receptor density: HP(CA1-or) López-Gallardo et al., 2012
♂↓♀↑; HP(CA1-rad) ♂↓;
HP(CA1-lac) ♂↓; DG-pol ♂↓♀↑
Rat ♂–♀ Dronabinol (9 -THC) 2.5, 5 and 10 mg/kg, for 7, 6 PND28-45 PND89-92 CB1 receptor density: No Lopez-Rodriguez et al., 2014
and 5 days, respectively significant alterations found in any
(i.p.) of the areas studied
Rat ♂–♀ CP 55,940 0.4 mg/kg (i.p.) PND28-43 PND81 CB1 receptor function: HP (CA1 and Mateos et al., 2011
CA2) ♂↑
Rat ♂ 9
Dronabinol ( -THC) 5 or 10 mg/kg (i.p.). Days 1, PND35-48 PND104 CB1 receptor density and function: Morel et al., 2009
3, 4 and 6: 1 dose; days 7, 9 No significant alterations found in
10, 11, 13 and 14: 2 doses; any of the areas studied
days 2, 5, 8 and 12: No dose
Rat ♂–♀ 9 -THC 2.5, 5 and 10 mg/kg (i.p.), PND35-45 PND46 CB1 receptor density: PFC ↓; CPu Rubino et al., 2008
for 3, 4 and 4 days, ♀↓; NAcc ♂↓; HT ↓; HP ↓; Amy ↓;
respectively (twice a day) Th ↓; PAG ↓; SN ♀↓; VTA ↓; Cb ↓
CB1 receptor function: PFC ♀↓; CPu
♀↓; NAcc ♀↓; GP ♀↓; HP ↓; Amy↓;
SN ↓; VTA ♀↓; Cb ↓
Rat ♀ 9 -THC 2.5, 5 and 10 mg/kg (i.p.), PND35-45 PND46, 60 CB1 receptor density: PFC ↓; CB1 Rubino et al., 2014
for 3, 4 and 4 days, and 75 receptor function: PFC ↓(PND46)
respectively (twice a day) ↑(PND60); AEA levels: PFC ↓;
MAGL activity: PFC ↓(PND46)
Rat ♂ 9 -THC 1.5 mg/kg (i.p.) PND28-49 PND35 (F1 CB1 receptor mRNA: NAcc ↑ Szutorisz et al., 2014
offspring)
PND62 (F1 CB1 receptor mRNA: CPu ↓
offspring)
Rat ♂–♀ 9 -THC 2.5, 5 and 10 mg/kg (i.p.), PND35-45 PND82 CB1 receptor density: PFC ↓; CPu ↓; Zamberletti et al., 2012
for 3, 4 and 4 days, NAcc ↓; HT ↓; HP ↓; Th ↓; Amy ↓;
respectively (twice a day) GP ↓; SN ↓; VTA ↓; Cb ↓
CB1 receptor function: PFC ↓; CPu
↓; NAcc ↓; HT ↓; HP ↓; Th ↓; Amy
↓; GP ↓; SN ↓; VTA ↓; Cb ↓

Cholinergic system
Rat ♂–♀ CP 55,940 0.4 mg/kg (i.p.) PND28-43 PND81 nACh receptor density: No Mateos et al., 2011
significant alterations found

Other signaling molecules or brain metabolism


Rat ♂–♀ CP 55,940 0.4 mg/kg (i.p.) PND28-38 PND101 Basal metabolic activity: PFC ♀↑; Higuera-Matas et al., 2008
Amy-EntCx ♀↓
Rat ♂–♀ CP 55,940 0.4 mg/kg/day (i.p.) PND28-38 PND100 PSA-NCAM: HP ♂↑ Higuera-Matas et al., 2009
Rat ♂–♀ 9 -THC 2.5, 5 and 10 mg/kg (i.p.), PND28-45 PND89-92 Arc levels: PFC ♀↓; HP ↓ Llorente-Berzal et al., 2013
for 3, 4 and 4 days,
respectively (twice a day)
Rat ♂–♀ CP 55,940 0.4 mg/kg (i.p.) PND28-42 PND80 BDNF levels: HP(CA3): ♀↓ López-Gallardo et al., 2012
Rat ♂–♀ 9 -THC 2.5, 5 and 10 mg/kg (i.p.), PND35-45 PND46 CREB activity: PFC ♀↓, HP ♀↓; NAcc Rubino et al., 2008
for 3, 4 and 4 days, ♀↑
respectively (twice a day)
A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146 139

were detected and only when MDMA was co-administered during effects of adolescent cannabinoid treatments on NCAMs seem to
adolescence did THC reduce CB1R expression in the hippocam- be dependent on the cannabinoid agent used. In fact, when we
pus, an effect that was restricted to female rats (Lopez-Rodriguez analyzed the levels of the different isoforms of NCAM in the hip-
et al., 2014). The activity of cannabinoid receptors was also found to pocampus after chronic adolescent CP treatment, no differences
be down-regulated in two of the aforementioned studies (Rubino were observed (Higuera-Matas et al., 2009). However, the levels
et al., 2008; Zamberletti et al., 2012), and up-regulated in three of the polysialylated form of NCAM (PSA-NCAM) increased in CP-
other studies (in Mateos et al., who only found changes in the treated male rats, which are consistent with the increased levels of
hippocampus of males (Mateos et al., 2011), in Rubino et al., who polysialic acid observed in the DG of mice exposed to THC as adoles-
reported the alterations in the PFC of young adult female rats (males cents (Tantra et al., 2014). PSA-NCAM, through its anti-adhesive and
were not tested) (Rubino et al., 2014) and in Higuera-Matas et al., insulating properties, may participate in the remodeling of synaptic
who found increased functional activation in females but not males structure, and for this reason it is crucial for brain plasticity (Nacher
(Higuera-Matas et al., 2012)). Interestingly, increments in receptor et al., 2013).
density or function were reported in most of the studies using a syn- In an attempt to reveal the long-term alterations in brain func-
thetic cannabinoid instead of THC, while decreases were reported tion caused by adolescent cannabinoid exposure, we conducted a
when the cannabinoid agent used during adolescence was THC (see positron emission tomography study on adult males and females
Table 7). Adolescent WIN treatment also provoked an increment in treated with CP as adolescents. We found 18 [F]-fluorodeoxyglucose
hippocampal levels of the degrading enzymes of AEA (FAAH) and imaging revealed a higher prefrontal cortical activity and reduced
2-AG (MAGL) (Gleason et al., 2012) although their activity was not activity in the amygdalo-enthorinal cortex of CP-exposed females.
altered in the PFC of adult female rats exposed to THC as adolescents No changes were detected in the males (Higuera-Matas et al., 2008).
(Rubino et al., 2014). Surprisingly even though the activity of FAAH
was not altered, adolescent THC exposure induced a reduction in
6.2.10. Structural and synaptic plasticity
the levels of AEA in the PFC of female rats measured at different
Long-term reduction in dendritic length, total dendritic number
time points (PNDs 46, 60 and 75: Rubino et al., 2014). Recently,
and dendritic spine number have been described in the DG of adult
adolescent exposure to THC was shown to have transgenerational
male rats treated with THC during adolescence (Rubino et al., 2009).
effects on the expression of the CB1R gene. In these experiments,
Consistent with these findings, and with the changes described
CB1R gene expression was stronger in the NAcc of the adolescent
in the glutamatergic systems (see above), adolescent cannabinoid
offspring of rats treated with THC as adolescents. By contrast, the
exposure has been seen to produce long lasting impairments in
adult offspring of rats exposed to THC as adolescents had weaker
long-term potentiation of the ventral subicular-NAcc pathway (Hila
CB1R gene expression in the dorsal striatum (Szutorisz et al., 2014).
Abush and Akirav, 2012, 2013; but see Higuera-Matas et al., 2009).
Hippocampal neurogenesis is also impaired in male but not female
6.2.8. Cholinergic system
rats after chronic exposure to HU-210 during adolescence (Lee et al.,
To our knowledge there is only one study that examined the
2014), as confirmed using WIN (although only studied male rats
adult cholinergic system in rats of both sexes after chronic cannabi-
were studied) (Abboussi et al., 2014). This deficit in neurogenesis
noid exposure during adolescence (Mateos et al., 2011), failing to
could be due to the impaired LTP mentioned before. Indeed, it has
identify changes in nicotinic receptors in the hippocampus of these
been shown that an LTP-inducing protocol activated a latent pre-
animals.
cursor pool, leading to increased neurogenesis in the dentate gyrus
(Kameda et al., 2012). In adult female rats, THC exposure during
6.2.9. Other signaling molecules and brain metabolism
adolescence abolished eCB-mediated long-term depression in the
Two molecules that are very important for plasticity have been
PFC and a reduction in the spine density on distal basal dendrites
examined, phosphorylated cAMP response element-binding pro-
of pyramidal neurons in layer II/III in the PFC of these animals was
tein (CREB) and BDNF. In one such study, the levels of phosphoCREB
observed (Rubino et al., 2014).
were measured in the PFC, NAcc and hippocampus of adult male
Adolescent cannabinoid exposure not only affects basic aspects
and female animals administered THC as adolescents (Rubino et al.,
of neural plasticity, such as LTP or spine density but also, it pervades
2008), demonstrating a decrease in CREB phosphorylation in the
more general aspects of nervous function. Indeed, there is a perma-
PFC and hippocampus, coupled with increased CREB activation in
nent suppression of cortical oscillations in mice after adolescent
the NAcc. These changes only occurred in the females and they were
exposure to cannabinoids (Raver and Keller, 2014).
related to the depressive phenotype found in female rats chroni-
These deficiencies in both structural and synaptic plasticity as
cally exposed to THC as adolescents. In addition, the levels of BDNF
well as in cortical function might explain the aforementioned cog-
in CA3 were seen to decrease in adult female but not male rats
nitive sequelae induced by adolescent cannabinoid consumption.
exposed to CP during adolescence (López-Gallardo et al., 2012),
which may also be relevant for the depressive phenotype described
in these animals (Rubino et al., 2008). Another plasticity-related 6.2.11. Cannabinoid exposure during adolescence as a
molecule that was down-regulated in the frontal cortex (females) normalizing agent against the deleterious effects of life challenges
or hippocampus (males and females) after adolescent exposure to We would like to bring up one aspect of adolescent cannabi-
THC was activity-regulated cytoskeleton-associated protein (Arc) noid exposure that has not been covered by earlier reviews on this
(Llorente-Berzal et al., 2013), a protein that is rapidly modulated in topic. During the last six years, some evidence has accumulated
a synaptic activity-dependent manner (Korb and Finkbeiner, 2011). in favor of possible compensating effects of adolescent cannabi-
Finally, a recent report suggested that chronic adolescent THC expo- noid exposure on the negative consequences of previous negative
sure reduced the levels of the cell-adhesion molecule NCAM 180 events (such as maternal separation, chronic stress or chronic expo-
in the hippocampus of St8sia2 (a polysialyltransferase that trans- sure to ‘ecstasy’ (MDMA)). For instance, cannabinoids ameliorate
fers polysialic acid molecules to NCAMs) KO mice (Tantra et al., the impaired synaptic plasticity and short-term memory induced
2014). Given that cell adhesion molecules play a crucial role in by chronic stress (Abush and Akirav, 2013). The deleterious effects
learning and memory (Lopez-Fernandez et al., 2007; Ronn et al., of chronic immobilization stress during adolescence on spatial
2000; Venero et al., 2006), these effects might explain some of the memory were reverted by administration of WIN after each stress
long-term cognitive impairments found in adult animals with ado- episode, an effect that was paralleled by restoration of LTP in the
lescent cannabinoid exposure and a susceptible phenotype. The subiculum-NAcc pathway.
140 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

During adolescence cannabinoids could also be beneficial for From these studies, it can be concluded that while cannabinoid
subjects with predisposition to show psychotic disorders. In this exposure during adolescence or puberty might have pervasive and
regard, adolescent THC was able to compensate the increase in negative effects on cognition or sensitivity to the rewarding effects
aggressive behavior observed after maternal deprivation in female of drugs of abuse, it is also true that it might have protective effects
but not male rats. The adolescent exposure to cannabinoids was against the deleterious consequences of other (and some times
also able to normalize the disturbed levels of NMDA glutamater- common) challenges during adolescence or early life (such as stress,
gic and D2 dopaminergic receptors in the dorsal striatum (only in exposure to other drugs of abuse or maternal separation).
females), and those of D1 dopaminergic receptors in the PFC (only
in males) in adult animals subjected to a single episode of mater- 7. Concluding remarks and future directions
nal deprivation (Zamberletti et al., 2012). In addition, CP treatment
during adolescence also normalized the levels of CB1Rs receptors In this review we have summarized the current state-of-the-art
in the hippocampus of adult rats that suffered an episode of mater- regarding the long-lasting effects of cannabinoid exposure on cog-
nal deprivation (López-Gallardo et al., 2012). Similarly, chronic nition, emotion, neurochemistry and plasticity during two critical
treatment with WIN during adolescence attenuated the locomo- periods: prenatal development and adolescence. The most con-
tor response to amphetamine in rats treated in utero (GD17) with a sistent findings suggest that early cannabinoid exposure leads to
single dose of the DNA methylating agent methylazoxymethanol cognitive dysfunction and alterations in glutamatergic, dopamin-
acetate (Gomes et al., 2014). This treatment, as well as mater- ergic, cannabinoid and opioidergic transmission, with notable
nal deprivation, is currently regarded as valid animal models of sexual dimorphisms. In addition, a facilitating effect on later drug
schizophrenia (Moore et al., 2006). Cannabinoid exposure dur- self-administration has been observed. While the origin of such
ing adolescence also abolished the increased sensitivity to the dimorphism is not clear, it could be related to the sex differ-
rewarding effects of morphine induced by chronic stress during ences in the eCB system seen during development (Rodriguez
adolescence, an effect that was accompanied by a normalization of de Fonseca et al., 1993), especially in the studies of perinatal
the altered PENK gene expression in the striatum that results from exposure to cannabinoids. Notwithstanding, cannabinoid exposure
maternal deprivation (Morel et al., 2009). Finally, chronic treatment during adolescence may also have a protective role when individ-
with WIN during puberty was able to revert the long-term impair- uals have faced severe life challenges, such as intense stress or
ments in working memory (assessed in the novel object recognition maternal separation. Clearly more research is needed, employing
task) induced by chronic treatment with 3,4-methylenedioxy-N- more sophisticated behavioral measures to further define com-
methylamphetamine (MDMA) during puberty (Schulz et al., 2013). plex cognitive operations, assays of self-control and impulsivity,
These protective actions could be related to the fact that THC can and paradigms that better resemble drug addiction phenomena in
normalize the decreased SERT levels induced by adolescent MDMA humans (including the cardinal features of this disease, such as
exposure (Lopez-Rodriguez et al., 2014). compulsivity). Such studies would have greater external validity,
There are several questions that need to be addressed regard- thereby better supporting preventive approaches for marihuana
ing these protective effects. For example, is there a specific time consumption during pregnancy or adolescence. In addition, the
window for the administration of cannabinoids? What are the protective role of cannabinoids during puberty should be further
downstream molecular mechanisms through which cannabinoids explored so that we can better identify individuals who might ben-
exert their protective effects? What kind of insults might be efit from the positive effects of such treatments, or those who might
compensated by cannabinoids? There are no straight answers suffer from their deleterious consequences.
to these questions. For example, WIN administered right after
stress episodes during adolescence reverted memory deficits and
restored LTP tested 30 days after the last stress episode (Abush and Acknowledgements
Akirav, 2013). However, these protective effects are not restricted
to the adolescent period. Robust evidence for these protective We would like to thank all the past and present members of
effects was also provided when the stress episode and cannabinoid the Psychobiology of Drug Addiction laboratory for their work and
administration occur in the adult life (Ganon-Elazar and Akirav, insight, all of which has made this review possible. Part of the
2009; Korem and Akirav, 2014; Segev et al., 2014). With regard to research of our group described here was funded by: the Minis-
the neurobiological mechanisms involved, the protective effects of terio de Ciencia e Innovación (SAF2013-47520-P); the Ministerio
cannabinoids after stress during adolescence were prevented by de Sanidad, Servicios Sociales e Igualdad (Red de Trastornos Adic-
administering the CB1R antagonist AM251, indicative of the cru- tivos, RTA-RD12/028/0020 of Instituto de Salud Carlos III, and
cial involvement of the CB1R (Abush and Akirav, 2013). Another Plan Nacional sobre Drogas, 2012I057); the Dirección General de
plausible cannabinoid effector is the glucocorticoid receptor (GR) Investigación de la Comunidad de Madrid (S-2011/BMD-2308;
(Ganon-Elazar and Akirav, 2009). However, the administration of Programa de Actividades I+D+I CANNAB-CM); the UNED (Plan
chronic WIN together with stress exposure did not affect GRs in any de Promoción de la Investigación); and the European Union
of the brain areas examined, suggesting that the beneficial effects (JUST/2013/DPIP/AG/4823-EU MADNESS).
of WIN on memory and plasticity were not mediated by alterations
to GR in this paradigm (Abush and Akirav, 2013). References
Some maternal deprivation effects are also reverted by chronic
cannabinoid administration during adolescence, such as aggres- Abboussi, O., Tazi, A., Paizanis, E., El Ganouni, S., 2014. Chronic exposure to
WIN55,212-2 affects more potently spatial learning and memory in adolescents
sive behavior or NMDA, D2 or D1 receptor levels (López-Gallardo than in adult rats via a negative action on dorsal hippocampal neurogenesis.
et al., 2012; Zamberletti et al., 2012). Why these changes only Pharmacol. Biochem. Behav. 120, 95–102, http://dx.doi.org/10.1016/j.pbb.2014.
occur in maternally deprived rats is currently unknown, but 02.014
Abush, H., Akirav, I., 2013. Cannabinoids ameliorate impairments induced by chronic
it could be related to the long-lasting alterations of the eCB
stress to synaptic plasticity and short-term memory. Neuropsychopharmacol-
system observed in these animals and/or (in contrast to what ogy 38, 1521–1534, http://dx.doi.org/10.1038/npp.2013.51
is observed for chronic adolescent stress) the persistent alter- Abush, H., Akirav, I., 2012. Short- and long-term cognitive effects of chronic cannabi-
ations in the hypothalamic–pituitary–adrenal axis (see Marco noids administration in late-adolescence rats. PLOS ONE 7, e31731, http://dx.doi.
org/10.1371/journal.pone.0031731
et al., 2015 for an excellent review of the maternal deprivation Aguado, T., Monory, K., Palazuelos, J., Stella, N., Cravatt, B., Lutz, B., Marsicano, G.,
model).” Kokaia, Z., Guzman, M., Galve-Roperh, I., 2005. The endocannabinoid system
A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146 141

drives neural progenitor proliferation. FASEB J. 19, 1704–1706, http://dx.doi. mesencephalic catecholaminergic neurons of immature and adult male rats
org/10.1096/fj.05-3995fje perinatally exposed to cannabinoids. Brain Res. Dev. Brain Res. 81, 147–150.
Alpár, A., Tortoriello, G., Calvigioni, D., Niphakis, M.J., Milenkovic, I., Bakker, J., Bortolato, M., Bini, V., Frau, R., Devoto, P., Pardu, A., Fan, Y., Solbrig, M.V., 2014. Juve-
Cameron, G.A., Hanics, J., Morris, C.V., Fuzik, J., Kovacs, G.G., Cravatt, B.F., nile cannabinoid treatment induces frontostriatal gliogenesis in Lewis rats. Eur.
Parnavelas, J.G., Andrews, W.D., Hurd, Y.L., Keimpema, E., Harkany, T., 2014. Neuropsychopharmacol. 24, 974–985, http://dx.doi.org/10.1016/j.euroneuro.
Endocannabinoids modulate cortical development by configuring Slit2/Robo1 2013.12.011
signalling. Nat. Commun. 5, 4421, http://dx.doi.org/10.1038/ncomms5421 Bortolato, M., Frau, R., Orrù, M., Casti, A., Aru, G.N., Fà, M., Manunta, M., Usai, A.,
Antonelli, T., Tomasini, M.C., Tattoli, M., Cassano, T., Tanganelli, S., Finetti, S., Mazzoni, Mereu, G., Gessa, G.L., 2006. Prenatal exposure to a cannabinoid receptor agonist
E., Trabace, L., Steardo, L., Cuomo, V., Ferraro, L., 2005. Prenatal exposure to the does not affect sensorimotor gating in rats. Eur. J. Pharmacol. 531, 166–170,
CB1 receptor agonist WIN 55,212-2 causes learning disruption associated with http://dx.doi.org/10.1016/j.ejphar.2005.12.017
impaired cortical NMDA receptor function and emotional reactivity changes Brown, H.L., Graves, C.R., 2013. Smoking and marijuana use in pregnancy.
in rat offspring. Cereb. Cortex 15, 2013–2020, http://dx.doi.org/10.1093/cercor/ Clin. Obstet. Gynecol. 56, 107–113, http://dx.doi.org/10.1097/GRF.
bhi076 0b013e318282377d
Badiani, A., Boden, J.M., De Pirro, S., Fergusson, D.M., Horwood, L.J., Harold, G.T., Burns, L., Mattick, R.P., Cooke, M., 2006. The use of record linkage to examine illicit
2015. Tobacco smoking and cannabis use in a longitudinal birth cohort: evidence drug use in pregnancy. Addiction 101, 873–882, http://dx.doi.org/10.1111/j.
of reciprocal causal relationships. Drug Alcohol Depend., http://dx.doi.org/10. 1360-0443.2006.01444.x
1016/j.drugalcdep.2015.02.015 Byrnes, J.J., Johnson, N.L., Schenk, M.E., Byrnes, E.M., 2012. Cannabinoid exposure
Bambico, F.R., Nguyen, N.T., Katz, N., Gobbi, G., 2010. Chronic exposure to in adolescent female rats induces transgenerational effects on morphine condi-
cannabinoids during adolescence but not during adulthood impairs emotional tioned place preference in male offspring. J. Psychopharmacol. 26, 1348–1354,
behaviour and monoaminergic neurotransmission. Neurobiol. Dis. 37, 641–655, http://dx.doi.org/10.1177/0269881112443745
http://dx.doi.org/10.1016/j.nbd.2009.11.020, S0969-9961(09)00342-8 [pii]. Cadoni, C., Simola, N., Espa, E., Fenu, S., Di Chiara, G., 2013. Strain dependence of
Basavarajappa, B.S., Nagre, N.N., Xie, S., Subbanna, S., 2014. Elevation of endogenous adolescent Cannabis influence on heroin reward and mesolimbic dopamine
anandamide impairs LTP, learning, and memory through CB1 receptor signaling transmission in adult Lewis and Fischer 344 rats. Addict. Biol., http://dx.doi.
in mice. Hippocampus 24, 808–818, http://dx.doi.org/10.1002/hipo.22272 org/10.1111/adb.12085
Batalla, A., Bhattacharyya, S., Yucel, M., Fusar-Poli, P., Crippa, J.A., Nogue, S., Torrens, Calvigioni, D., Hurd, Y.L., Harkany, T., Keimpema, E., 2014. Neuronal substrates and
M., Pujol, J., Farre, M., Martin-Santos, R., 2013. Structural and functional imaging functional consequences of prenatal cannabis exposure. Eur. Child Adolesc. Psy-
studies in chronic cannabis users: a systematic review of adolescent and adult chiatry, http://dx.doi.org/10.1007/s00787-014-0550-y
findings. PLOS ONE 8, e55821, http://dx.doi.org/10.1371/journal.pone.0055821 Campolongo, P., Trezza, V., Cassano, T., Gaetani, S., Morgese, M.G., Ubaldi, M., Sover-
Battistella, G., Fornari, E., Annoni, J.-M., Chtioui, H., Dao, K., Fabritius, M., Favrat, chia, L., Antonelli, T., Ferraro, L., Massi, M., Ciccocioppo, R., Cuomo, V., 2007.
B., Mall, J.-F., Maeder, P., Giroud, C., 2014. Long-term effects of cannabis on Perinatal exposure to delta-9-tetrahydrocannabinol causes enduring cognitive
brain structure. Neuropsychopharmacology 39, 2041–2048, http://dx.doi.org/ deficits associated with alteration of cortical gene expression and neurotrans-
10.1038/npp.2014.67 mission in rats. Addict. Biol. 12, 485–495, http://dx.doi.org/10.1111/j.1369-
Behan, A.T., Hryniewiecka, M., O’Tuathaigh, C.M., Kinsella, A., Cannon, M., Karayior- 1600.2007.00074.x
gou, M., Gogos, J.A., Waddington, J.L., Cotter, D.R., 2012. Chronic adolescent Campolongo, P., Trezza, V., Ratano, P., Palmery, M., Cuomo, V., 2011. Developmental
exposure to delta-9-tetrahydrocannabinol in COMT mutant mice: impact on consequences of perinatal cannabis exposure: behavioral and neuroendocrine
indices of dopaminergic, endocannabinoid and GABAergic pathways. Neuropsy- effects in adult rodents. Psychopharmacology (Berl). 214, 5–15, http://dx.doi.
chopharmacology 37, 1773–1783, http://dx.doi.org/10.1038/npp.2012.24 org/10.1007/s00213-010-1892-x
Belue, R.C., Howlett, A.C., Westlake, T.M., Hutchings, D.E., 1995. The ontogeny of Carli, M., Invernizzi, R.W., 2014. Serotoninergic and dopaminergic modulation of
cannabinoid receptors in the brain of postnatal and aging rats. Neurotoxicol. cortico-striatal circuit in executive and attention deficits induced by NMDA
Teratol. 17, 25–30. receptor hypofunction in the 5-choice serial reaction time task. Front. Neural
Benagiano, V., Lorusso, L., Flace, P., Girolamo, F., Rizzi, A., Sabatini, R., Auteri, P., Circ. 8, 58, http://dx.doi.org/10.3389/fncir.2014.00058
Bosco, L., Cagiano, R., Ambrosi, G., 2007. Effects of prenatal exposure to the CB- Cass, D.K., Flores-Barrera, E., Thomases, D.R., Vital, W.F., Caballero, A., Tseng, K.Y.,
1 receptor agonist WIN 55212-2 or CO on the GABAergic neuronal systems of 2014. CB1 cannabinoid receptor stimulation during adolescence impairs the
rat cerebellar cortex. Neuroscience 149, 592–601, http://dx.doi.org/10.1016/j. maturation of GABA function in the adult rat prefrontal cortex. Mol. Psychiatry
neuroscience.2007.07.050 19, 536–543, http://dx.doi.org/10.1038/mp.2014.14
Benes, F.M., 1989. Myelination of cortical-hippocampal relays during late adoles- Castaldo, P., Magi, S., Cataldi, M., Arcangeli, S., Lariccia, V., Nasti, A.A., Ferraro, L.,
cence. Schizophr. Bull. 15, 585–593. Tomasini, M.C., Antonelli, T., Cassano, T., Cuomo, V., Amoroso, S., 2010. Altered
Berghuis, P., Dobszay, M.B., Wang, X., Spano, S., Ledda, F., Sousa, K.M., Schulte, G., regulation of glutamate release and decreased functional activity and expression
Ernfors, P., Mackie, K., Paratcha, G., Hurd, Y.L., Harkany, T., 2005. Endocannabi- of GLT1 and GLAST glutamate transporters in the hippocampus of adolescent
noids regulate interneuron migration and morphogenesis by transactivating the rats perinatally exposed to Delta(9)-THC. Pharmacol. Res. 61, 334–341, http://
TrkB receptor. Proc. Natl. Acad. Sci. U. S. A. 102, 19115–19120, http://dx.doi.org/ dx.doi.org/10.1016/j.phrs.2009.11.008, S1043-6618(09)00277-1 [pii].
10.1073/pnas.0509494102 Castaldo, P., Magi, S., Gaetani, S., Cassano, T., Ferraro, L., Antonelli, T., Amoroso,
Berghuis, P., Rajnicek, A.M., Morozov, Y.M., Ross, R.A., Mulder, J., Urban, G.M., S., Cuomo, V., 2007. Prenatal exposure to the cannabinoid receptor agonist
Monory, K., Marsicano, G., Matteoli, M., Canty, A., Irving, A.J., Katona, I., Yana- WIN 55,212-2 increases glutamate uptake through overexpression of GLT1 and
gawa, Y., Rakic, P., Lutz, B., Mackie, K., Harkany, T., 2007. Hardwiring the brain: EAAC1 glutamate transporter subtypes in rat frontal cerebral cortex. Neurophar-
endocannabinoids shape neuronal connectivity. Science 316, 1212–1216, http:// macology 53, 369–378, http://dx.doi.org/10.1016/j.neuropharm.2007.05.019,
dx.doi.org/10.1126/science.1137406 S0028-3908(07)00155-4 [pii].
Berrendero, F., García-Gil, L., Hernández, M.L., Romero, J., Cebeira, M., de Castelli, M.P., Paola Piras, A., D’Agostino, A., Pibiri, F., Perra, S., Gessa, G.L., Maccarrone,
Miguel, R., Ramos, J.A., Fernández-Ruiz, J.J., 1998. Localization of mRNA M., Pistis, M., 2007. Dysregulation of the endogenous cannabinoid system in
expression and activation of signal transduction mechanisms for cannabi- adult rats prenatally treated with the cannabinoid agonist WIN 55,212-2. Eur. J.
noid receptor in rat brain during fetal development. Development 125, Pharmacol. 573, 11–19, http://dx.doi.org/10.1016/j.ejphar.2007.06.047, S0014-
3179–3188. 2999(07)00763-7 [pii].
Berrendero, F., Sepe, N., Ramos, J.A., Di Marzo, V., Fernandez-Ruiz, J.J., 1999. Anal- Cha, Y.M., Jones, K.H., Kuhn, C.M., Wilson, W.A., Swartzwelder, H.S., 2007. Sex dif-
ysis of cannabinoid receptor binding and mRNA expression and endogenous ferences in the effects of delta9-tetrahydrocannabinol on spatial learning in
cannabinoid contents in the developing rat brain during late gestation and early adolescent and adult rats. Behav. Pharmacol. 18, 563–569.
postnatal period. Synapse 33, 181–191, http://dx.doi.org/10.1002/(SICI)1098- Cha, Y.M., White, A.M., Kuhn, C.M., Wilson, W.a, Swartzwelder, H.S., 2006. Dif-
2396(19990901)33:3<181::AID-SYN3>3.0.CO;2-R ferential effects of delta9-THC on learning in adolescent and adult rats.
Biscaia, M., Fernández, B., Higuera-Matas, A., Miguéns, M., Viveros, M.-P.P., García- Pharmacol. Biochem. Behav. 83, 448–455, http://dx.doi.org/10.1016/j.pbb.2006.
Lecumberri, C., Ambrosio, E., Fernandez, B., Miguens, M., Garcia-Lecumberri, C., 03.006, S0091-3057(06)00065-7 [pii].
2008. Sex-dependent effects of periadolescent exposure to the cannabinoid ago- Chadwick, B., Miller, M.L., Hurd, Y.L., 2013. Cannabis use during adolescent devel-
nist CP-55,940 on morphine self-administration behaviour and the endogenous opment: susceptibility to psychiatric illness. Front. Psychiatry 4, 129, http://dx.
opioid system. Neuropharmacology 54, 863–873, http://dx.doi.org/10.1016/j. doi.org/10.3389/fpsyt.2013.00129
neuropharm.2008.01.006, S0028-3908(08)00020-8 [pii]. Chadwick, B., Saylor, A.J., Lopez, H.H., 2011. Adolescent cannabinoid exposure
Biscaia, M., Marin, S., Fernandez, B., Marco, E.M., Rubio, M., Guaza, C., Ambrosio, attenuates adult female sexual motivation but does not alter adulthood CB1R
E., Viveros, M.P., 2003. Chronic treatment with CP 55,940 during the peri- expression or estrous cyclicity. Pharmacol. Biochem. Behav. 100, 157–164,
adolescent period differentially affects the behavioural responses of male and http://dx.doi.org/10.1016/j.pbb.2011.07.006, S0091-3057(11)00242-5 [pii].
female rats in adulthood. Psychopharmacology (Berl). 170, 301–308, http://dx. Chapman, S.L., Wu, L.T., 2013. Substance use among adolescent mothers: a review.
doi.org/10.1007/s00213-003-1550-7 Child Youth Serv. Rev. 35, 806–815, http://dx.doi.org/10.1016/j.childyouth.
Blakemore, S.J., 2013. Teenage kicks: cannabis and the adolescent brain. 2013.02.004
Lancet 381, 888–889, http://dx.doi.org/10.1016/S0140-6736(12)61578-5, Cornelius, M.D., Leech, S.L., Goldschmidt, L., 2004. Characteristics of persistent smok-
S0140-6736(12)61578-5 [pii]. ing among pregnant teenagers followed to young adulthood. Nicotine Tob. Res.
Blakemore, S.-J.J., 2012. Imaging brain development: the adolescent brain. 6, 159–169, http://dx.doi.org/10.1080/14622200310001656975
Neuroimage 61, 397–406, http://dx.doi.org/10.1016/j.neuroimage.2011.11.080, Dalterio, S., Blum, K., DeLallo, L., Sweeney, C., Briggs, A., Bartke, A., 1980.
S1053-8119(11)01362-0 [pii]. Perinatal exposure to delta 9-THC in mice: altered enkephalin and nore-
Bonnin, A., de Miguel, R., Rodriguez-Manzaneque, J.C., Fernandez-Ruiz, J.J., San- pinephrine sensitivity in vas deferens. Subst. Alcohol Actions. Misuse 1,
tos, A., Ramos, J.A., 1994. Changes in tyrosine hydroxylase gene expression in 467–471.
142 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

Dalterio, S., Steger, R., Mayfield, D., Bartke, A., 1984a. Early cannabinoid exposure Ellgren, M., Spano, S.M., Hurd, Y.L., 2007. Adolescent cannabis exposure alters opiate
influences neuroendocrine and reproductive functions in male mice: I. Prenatal intake and opioid limbic neuronal populations in adult rats. Neuropsychophar-
exposure. Pharmacol. Biochem. Behav. 20, 107–113. macology 32, 607–615, http://dx.doi.org/10.1038/sj.npp.1301127
Dalterio, S., Steger, R., Mayfield, D., Bartke, A., 1984b. Early cannabinoid exposure European Monitoring Centre for Drugs and Drug Addiction, 2014. European Drug
influences neuroendocrine and reproductive functions in mice: II. Postnatal Report 2014: Trends and Developments.
effects. Pharmacol. Biochem. Behav. 20, 115–123. Fergusson, D.M., Horwood, L.J., Northstone, K., 2002. Maternal use of cannabis and
Day, N.L., Cottreau, C.M., Richardson, G.A., 1993. The epidemiology of alcohol, mari- pregnancy outcome. BJOG 109, 21–27.
juana, and cocaine use among women of childbearing age and pregnant women. Fontes, M.A., Bolla, K.I., Cunha, P.J., Almeida, P.P., Jungerman, F., Laranjeira, R.R.,
Clin. Obstet. Gynecol. 36, 232–245. Bressan, R.A., Lacerda, A.L., 2011. Cannabis use before age 15 and subsequent
Day, N.L., Goldschmidt, L., Thomas, C.A., 2006. Prenatal marijuana exposure con- executive functioning. Br. J. Psychiatry 198, 442–447, http://dx.doi.org/10.1192/
tributes to the prediction of marijuana use at age 14. Addiction 101, 1313–1322, bjp.bp.110.077479
http://dx.doi.org/10.1111/j.1360-0443.2006.01523.x Fried, P., Watkinson, B., James, D., Gray, R., 2002. Current and former marijuana use:
Day, N.L., Leech, S.L., Goldschmidt, L., 2011. The effects of prenatal marijuana expo- preliminary findings of a longitudinal study of effects on IQ in young adults.
sure on delinquent behaviors are mediated by measures of neurocognitive CMAJ 166, 887–891.
functioning. Neurotoxicol. Teratol. 33, 129–136, http://dx.doi.org/10.1016/j.ntt. Friguls, B., Joya, X., Garcia-Serra, J., Gomez-Culebras, M., Pichini, S., Martinez, S.,
2010.07.006, S0892-0362(10)00157-1 [pii]. Vall, O., Garcia-Algar, O., 2012. Assessment of exposure to drugs of abuse during
Day, N.L., Richardson, G.A., Geva, D., Robles, N., 1994. Alcohol, marijuana, and pregnancy by hair analysis in a Mediterranean island. Addiction 107, 1471–1479,
tobacco: effects of prenatal exposure on offspring growth and morphology at http://dx.doi.org/10.1111/j.1360-0443.2012.03828.x
age six. Alcohol. Clin. Exp. Res. 18, 786–794. Gaalema, D.E., Higgins, S.T., Pepin, C.S., Heil, S.H., Bernstein, I.M., 2013. Illicit drug use
Delegación del Gobierno para el Plan Nacional sobre Drogas, 2013. Encuesta among pregnant women enrolled in treatment for cigarette smoking cessation.
estatal sobre uso de drogas en enseñanzas secundarias (ESTUDES) Nicotine Tob. Res. 15, 987–991, http://dx.doi.org/10.1093/ntr/nts220
2012/2013. Ganon-Elazar, E., Akirav, I., 2009. Cannabinoid receptor activation in the basolat-
Derauf, C., Kekatpure, M., Neyzi, N., Lester, B., Kosofsky, B., 2009. Neuroimaging of eral amygdala blocks the effects of stress on the conditioning and extinction of
children following prenatal drug exposure. Semin. Cell Dev. Biol. 20, 441–454, inhibitory avoidance. J. Neurosci. 29, 11078–11088, http://dx.doi.org/10.1523/
http://dx.doi.org/10.1016/j.semcdb.2009.03.001, S1084-9521(09)00039-1 [pii]. JNEUROSCI.1223-09.2009
Díaz-Alonso, J., Aguado, T., de Salas-Quiroga, A., Ortega, Z., Guzmán, M., Garcia, L., de Miguel, R., Ramos, J.A., Fernandez-Ruiz, J.J., 1996. Perinatal delta 9-
Galve-Roperh, I., 2014. CB1 cannabinoid receptor-dependent activation of tetrahydrocannabinol exposure in rats modifies the responsiveness of midbrain
mTORC1/Pax6 signaling drives Tbr2 expression and basal progenitor expansion dopaminergic neurons in adulthood to a variety of challenges with dopaminergic
in the developing mouse cortex. Cereb. Cortex, http://dx.doi.org/10.1093/cercor/ drugs. Drug Alcohol Depend. 42, 155–166.
bhu039 Garcia-Gil, L., de Miguel, R., Romero, J., Perez, A., Ramos, J.A., Fernandez-Ruiz, J.J.,
DiNieri, J.A., Wang, X., Szutorisz, H., Spano, S.M., Kaur, J., Casaccia, P., Dow-Edwards, 1999. Perinatal delta9-tetrahydrocannabinol exposure augmented the magni-
D., Hurd, Y.L., 2011. Maternal cannabis use alters ventral striatal dopamine D2 tude of motor inhibition caused by GABA(B), but not GABA(A), receptor agonists
gene regulation in the offspring. Biol. Psychiatry 70, 763–769, http://dx.doi.org/ in adult rats. Neurotoxicol. Teratol. 21, 277–283, S0892036298000580 [pii].
10.1016/j.biopsych.2011.06.027, S0006-3223(11)00672-X [pii]. García-Gil, L., Romero, J., Ramos, J.A., Fernández-Ruiz, J.J., 1999. Cannabinoid recep-
Duff, G., Argaw, A., Cecyre, B., Cherif, H., Tea, N., Zabouri, N., Casanova, C., Ptito, M., tor binding and mRNA levels in several brain regions of adult male and female
Bouchard, J.-F., 2013. Cannabinoid receptor CB2 modulates axon guidance. PLOS rats perinatally exposed to 9-tetrahydrocannabinol. Drug Alcohol Depend. 55,
ONE 8, e70849, http://dx.doi.org/10.1371/journal.pone.0070849 127–136, http://dx.doi.org/10.1016/S0376-8716(98)00189-6
Economidou, D., Mattioli, L., Ubaldi, M., Lourdusamy, A., Soverchia, L., Hardiman, Giedd, J.N., 2008. The teen brain: insights from neuroimaging. J. Adolesc. Health 42,
G., Campolongo, P., Cuomo, V., Ciccocioppo, R., 2007. Role of cannabinoider- 335–343, http://dx.doi.org/10.1016/j.jadohealth.2008.01.007
gic mechanisms in ethanol self-administration and ethanol seeking in rat adult Glass, M., Dragunow, M., Faull, R.L., 1997. Cannabinoid receptors in the human
offspring following perinatal exposure to Delta9-tetrahydrocannabinol. Toxi- brain: a detailed anatomical and quantitative autoradiographic study in
col. Appl. Pharmacol. 223, 73–85, http://dx.doi.org/10.1016/j.taap.2007.05.008, the fetal, neonatal and adult human brain. Neuroscience 77, 299–318,
S0041-008X(07)00238-4 [pii]. S0306452296004289 [pii].
Ehrenreich, H., Rinn, T., Kunert, H.J., Moeller, M.R., Poser, W., Schilling, L., Gleason, K.A., Birnbaum, S.G., Shukla, A., Ghose, S., 2012. Susceptibility of the ado-
Gigerenzer, G., Hoehe, M.R., 1999. Specific attentional dysfunction in adults lescent brain to cannabinoids: long-term hippocampal effects and relevance to
following early start of cannabis use. Psychopharmacology (Berl). (142), schizophrenia. Transl. Psychiatry 2, e199, http://dx.doi.org/10.1038/tp.2012.122
295–301. Gomes, F.V., Guimarães, F.S., Grace, A.A., 2014. Effects of pubertal cannabinoid
El Marroun, H., Hudziak, J.J., Tiemeier, H., Creemers, H., Steegers, E.A., Jaddoe, V.W., administration on attentional set-shifting and dopaminergic hyper-responsivity
Hofman, A., Verhulst, F.C., van den Brink, W., Huizink, A.C., 2011a. Intrauterine in a developmental disruption model of schizophrenia. Int. J. Neuropsychophar-
cannabis exposure leads to more aggressive behavior and attention problems in macol. 18, http://dx.doi.org/10.1093/ijnp/pyu018
18-month-old girls. Drug Alcohol Depend. (118), 470–474, http://dx.doi.org/10. Gonzalez, B., de Miguel, R., Martin, S., Perez-Rosado, A., Romero, J., Garcia-
1016/j.drugalcdep.2011.03.004, S0376-8716(11)00130-X [pii]. Lecumberri, C., Fernandez-Ruiz, J., Ramos, J.A., Ambrosio, E., 2003. Effects of
El Marroun, H., Jaddoe, V.W., Hudziak, J.J., Roza, S.J., Steegers, E.A., Hofman, A., perinatal exposure to delta 9-tetrahydrocannabinol on operant morphine-
Verhulst, F.C., White, T.J., Stricker, B.H., Tiemeier, H., 2012. Maternal use reinforced behavior. Pharmacol. Biochem. Behav. 75, 577–584.
of selective serotonin reuptake inhibitors, fetal growth, and risk of adverse Gonzalez, R., Swanson, J.M., 2012. Long-term effects of adolescent-onset and persis-
birth outcomes. Arch. Gen. Psychiatry 69, 706–714, http://dx.doi.org/10.1001/ tent use of cannabis. Proc. Natl. Acad. Sci. U. S. A. 109, 15970–15971, http://dx.
archgenpsychiatry.2011.2333 doi.org/10.1073/pnas.1214124109
El Marroun, H., Tiemeier, H., Jaddoe, V.W., Hofman, A., Mackenbach, J.P., Steegers, Gordon, J.A., Hen, R., 2004. The serotonergic system and anxiety. Neuromolecular
E.A., Verhulst, F.C., van den Brink, W., Huizink, A.C., 2008. Demographic, Med. 5, 27–40, http://dx.doi.org/10.1385/NMM:5:1:027
emotional and social determinants of cannabis use in early pregnancy: the Gen- Grant, I., Gonzalez, R., Carey, C.L., Natarajan, L., Wolfson, T., 2003. Non-acute
eration R study. Drug Alcohol Depend. 98, 218–226, http://dx.doi.org/10.1016/ (residual) neurocognitive effects of cannabis use: a meta-analytic study. J. Int.
j.drugalcdep.2008.05.010, S0376-8716(08)00201-9 [pii]. Neuropsychol. Soc. 9, 679–689, http://dx.doi.org/10.1017/S1355617703950016
El Marroun, H., Tiemeier, H., Jaddoe, V.W., Hofman, A., Verhulst, F.C., van den Brink, Gruber, S.A., Dahlgren, M.K., Sagar, K.A., Gonenc, A., Killgore, W.D., 2012. Age of
W., Huizink, A.C., 2011b. Agreement between maternal cannabis use during onset of marijuana use impacts inhibitory processing. Neurosci. Lett. 511, 89–94,
pregnancy according to self-report and urinalysis in a population-based cohort: http://dx.doi.org/10.1016/j.neulet.2012.01.039, S0304-39401200094-8 [pii].
the Generation R Study. Eur. Addict. Res. 17, 37–43, http://dx.doi.org/10.1159/ Harte, L.C., Dow-Edwards, D., 2010. Sexually dimorphic alterations in locomotion
000320550 and reversal learning after adolescent tetrahydrocannabinol exposure in the
El Marroun, H., Tiemeier, H., Steegers, E.A., Jaddoe, V.W., Hofman, A., Verhulst, F.C., rat. Neurotoxicol. Teratol. 32, 515–524, http://dx.doi.org/10.1016/j.ntt.2010.05.
van den Brink, W., Huizink, A.C., 2009. Intrauterine cannabis exposure affects 001, S0892-0362(10)00109-1 [pii].
fetal growth trajectories: the Generation R Study. J. Am. Acad. Child Adolesc. Higuera-Matas, A., Botreau, F., Del Olmo, N., Miguens, M., Olias, O., Montoya, G.L.,
Psychiatry 48, 1173–1181, http://dx.doi.org/10.1097/CHI.0b013e3181bfa8ee, Garcia-Lecumberri, C., Ambrosio, E., 2010. Periadolescent exposure to cannabi-
S0890-8567(09)66073-1 [pii]. noids alters the striatal and hippocampal dopaminergic system in the adult
El Marroun, H., Tiemeier, H., Steegers, E.A., Roos-Hesselink, J.W., Jaddoe, V.W., rat brain. Eur. Neuropsychopharmacol. 20, 895–906, http://dx.doi.org/10.1016/
Hofman, A., Verhulst, F.C., van den Brink, W., Huizink, A.C., 2010. A prospec- j.euroneuro.2010.06.017, S0924-977X(10)00144-6 [pii].
tive study on intrauterine cannabis exposure and fetal blood flow. Early Hum. Higuera-Matas, A., Botreau, F., Miguens, M., Del Olmo, N., Borcel, E., Perez-
Dev. 86, 231–236, http://dx.doi.org/10.1016/j.earlhumdev.2010.03.006, S0378- Alvarez, L., Garcia-Lecumberri, C., Ambrosio, E., Miguéns, M., Pérez-Alvarez,
3782(10)00079-4 [pii]. L., García-Lecumberri, C., 2009. Chronic periadolescent cannabinoid treat-
Ellgren, M., Artmann, A., Tkalych, O., Gupta, A., Hansen, H.S., Hansen, S.H., Devi, ment enhances adult hippocampal PSA-NCAM expression in male Wistar rats
L.A., Hurd, Y.L., 2008. Dynamic changes of the endogenous cannabinoid and but only has marginal effects on anxiety, learning and memory. Pharma-
opioid mesocorticolimbic systems during adolescence: THC effects. Eur. Neu- col. Biochem. Behav. 93, 482–490, http://dx.doi.org/10.1016/j.pbb.2009.06.013,
ropsychopharmacol. 18, 826–834, http://dx.doi.org/10.1016/j.euroneuro.2008. S0091-3057(09)00209-3 [pii].
06.009, S0924-977X(08)00170-3 [pii]. Higuera-Matas, A., Miguens, M., Coria, S.M., Assis, M.A., Borcel, E., Del Olmo, N.,
Ellgren, M., Hurd, Y.L., Franck, J., 2004. Amphetamine effects on dopamine lev- Ambrosio, E., 2012. Sex-specific disturbances of the glutamate/GABA balance
els and behavior following cannabinoid exposure during adolescence. Eur. in the hippocampus of adult rats subjected to adolescent cannabinoid exposure.
J. Pharmacol. 497, 205–213, http://dx.doi.org/10.1016/j.ejphar.2004.06.048, Neuropharmacology 62, 1975–1984, http://dx.doi.org/10.1016/j.neuropharm.
S0014-2999(04)00689-2 [pii]. 2011.12.028, S0028-3908(12)00004-4 [pii].
A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146 143

Higuera-Matas, A., Soto-Montenegro, M.L., del Olmo, N., Miguens, M., Torres, I., Llorente-Berzal, A., Puighermanal, E., Burokas, A., Ozaita, A., Maldonado, R., Marco,
Vaquero, J.J., Sanchez, J., Garcia-Lecumberri, C., Desco, M., Ambrosio, E., 2008. E.M., Viveros, M.-P., 2013. Sex-dependent psychoneuroendocrine effects of THC
Augmented acquisition of cocaine self-administration and altered brain glucose and MDMA in an animal model of adolescent drug consumption. PLOS ONE 8,
metabolism in adult female but not male rats exposed to a cannabinoid agonist e78386, http://dx.doi.org/10.1371/journal.pone.0078386
during adolescence. Neuropsychopharmacology 33, 806–813. Long, L.E., Lind, J., Webster, M., Weickert, C.S., 2012. Developmental trajectory of the
Higuera-Matas, A., Soto-Montenegro, M.L., Montoya, G.L., García-Vázquez, V., Pas- endocannabinoid system in human dorsolateral prefrontal cortex. BMC Neu-
cau, J., Miguéns, M., Del Olmo, N., Vaquero, J.J., García-Lecumberri, C., Desco, rosci. 13, 87, http://dx.doi.org/10.1186/1471-2202-13-87
M., Ambrosio, E., 2011. Chronic cannabinoid administration to periadolescent Lopez-Fernandez, M.A., Montaron, M.F., Varea, E., Rougon, G., Venero, C., Abrous,
rats modulates the metabolic response to acute cocaine in the adult brain. Mol. D.N., Sandi, C., 2007. Upregulation of polysialylated neural cell adhesion
Imaging Biol. 13, 411–415, http://dx.doi.org/10.1007/s11307-010-0388-8 molecule in the dorsal hippocampus after contextual fear conditioning is
Howlett, A.C., Barth, F., Bonner, T.I., Cabral, G., Casellas, P., Devane, W.A., Felder, involved in long-term memory formation. J. Neurosci. 27, 4552–4561, http://
C.C., Herkenham, M., Mackie, K., Martin, B.R., Mechoulam, R., Pertwee, R.G., dx.doi.org/10.1523/JNEUROSCI.0396-07.2007
2002. International Union of Pharmacology. XXVII. Classification of cannabinoid López-Gallardo, M., López-Rodríguez, A.B., Llorente-Berzal, Á., Rotllant, D., Mackie,
receptors. Pharmacol. Rev. 54, 161–202. K., Armario, A., Nadal, R., Viveros, M.-P.P., Lopez-Gallardo, M., Lopez-Rodriguez,
Huizink, A.C., 2014. Prenatal cannabis exposure and infant outcomes: overview of A.B., Llorente-Berzal, A., 2012. Maternal deprivation and adolescent cannabi-
studies. Prog. Neuropsychopharmacol. Biol. Psychiatry 52, 45–52, http://dx.doi. noid exposure impact hippocampal astrocytes, CB1 receptors and brain-derived
org/10.1016/j.pnpbp.2013.09.014 neurotrophic factor in a sexually dimorphic fashion. Neuroscience 204, 90–103,
Hurd, Y.L., Michaelides, M., Miller, M.L., Jutras-Aswad, D., 2014. Trajectory of ado- http://dx.doi.org/10.1016/j.neuroscience.2011.09.063, S0306-4522(11)01149-
lescent cannabis use on addiction vulnerability. Neuropharmacology 76 (Pt B), 3 [pii].
416–424, http://dx.doi.org/10.1016/j.neuropharm.2013.07.028 Lopez-Rodriguez, A.B., Llorente-Berzal, A., Garcia-Segura, L.M., Viveros, M.-P., 2014.
Hurd, Y.L., Wang, X., Anderson, V., Beck, O., Minkoff, H., Dow-Edwards, D., 2005. Sex-dependent long-term effects of adolescent exposure to THC and/or MDMA
Marijuana impairs growth in mid-gestation fetuses. Neurotoxicol. Teratol. 27, on neuroinflammation and serotoninergic and cannabinoid systems in rats. Br.
221–229, http://dx.doi.org/10.1016/j.ntt.2004.11.002, S0892-0362(04)00150-3 J. Pharmacol. 171, 1435–1447, http://dx.doi.org/10.1111/bph.12519
[pii]. Lozano, J., Garcia-Algar, O., Marchei, E., Vall, O., Monleon, T., Giovannandrea, R.D.,
Huttenlocher, P.R., Dabholkar, A.S., 1997. Regional differences in synaptogenesis Pichini, S., 2007. Prevalence of gestational exposure to cannabis in a Mediter-
in human cerebral cortex. J. Comp. Neurol. 387, 167–178, http://dx.doi.org/10. ranean city by meconium analysis. Acta Paediatr. 96, 1734–1737, http://dx.doi.
1002/(SICI)1096-9861(19971020)387:2<167::AID-CNE1>3.0.CO;2-Z org/10.1111/j.1651-2227.2007.00535.x
Jaques, S.C., Kingsbury, A., Henshcke, P., Chomchai, C., Clews, S., Falconer, J., Abdel- Maccarrone, M., Guzmán, M., Mackie, K., Doherty, P., Harkany, T., 2014. Programming
Latif, M.E., Feller, J.M., Oei, J.L., 2014. Cannabis, the pregnant woman and her of neural cells by (endo)cannabinoids: from physiological rules to emerging
child: weeding out the myths. J. Perinatol. 34, 417–424, http://dx.doi.org/10. therapies. Nat. Rev. Neurosci. 15, 786–801, http://dx.doi.org/10.1038/nrn3846
1038/jp.2013.180 Maj, P.F., Collu, M., Fadda, P., Cattaneo, A., Racagni, G., Riva, M.A., 2007. Long-
Jiang, S., Fu, Y., Williams, J., Wood, J., Pandarinathan, L., Avraham, S., Makriyannis, A., term reduction of brain-derived neurotrophic factor levels and signaling
Avraham, S., Avraham, H.K., 2007. Expression and function of cannabinoid recep- impairment following prenatal treatment with the cannabinoid receptor 1
tors CB1 and CB2 and their cognate cannabinoid ligands in murine embryonic receptor agonist (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinyl-methyl)
stem cells. PLoS ONE 2, e641, http://dx.doi.org/10.1371/journal.pone.0000641 pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl]-1-naphthalenylmethanone. Eur. J.
Kameda, M., Taylor, C.J., Walker, T.L., Black, D.M., Abraham, W.C., Bartlett, P.F., 2012. Neurosci. 25, 3305–3311, http://dx.doi.org/10.1111/j.1460-9568.2007.05565.
Activation of latent precursors in the hippocampus is dependent on long-term x
potentiation. Transl. Psychiatry, http://dx.doi.org/10.1038/tp.2011.70 Marco, E.M., Granstrem, O., Moreno, E., Llorente, R., Adriani, W., Laviola, G., Viveros,
Kandel, D.B., Yamaguchi, K., Klein, L.C., 2006. Testing the Gateway Hypothesis. Addic- M.P., 2007. Subchronic nicotine exposure in adolescence induces long-term
tion 101, 470–476. effects on hippocampal and striatal cannabinoid-CB1 and mu-opioid receptors
Kawash, G.F., Yeung, D.L., Berg, S.D., 1980. Effects of administration of cannabis resin in rats. Eur. J. Pharmacol. 557, 37–43, http://dx.doi.org/10.1016/j.ejphar.2006.
during pregnancy on emotionality and learning in rats’ offspring. Percept. Mot. 11.013, S0014-2999(06)01268-4 [pii].
Skills 50, 359–365. Marco, E.M., Llorente, R., López-Gallardo, M., Mela, V., Llorente-Berzal, Á., Prada, C.,
Keimpema, E., Barabas, K., Morozov, Y.M., Tortoriello, G., Torii, M., Cameron, G., Viveros, M.-P., 2015. The maternal deprivation animal model revisited. Neurosci.
Yanagawa, Y., Watanabe, M., Mackie, K., Harkany, T., 2010. Differential sub- Biobehav. Rev. 51, 151–163, http://dx.doi.org/10.1016/j.neubiorev.2015.01.015
cellular recruitment of monoacylglycerol lipase generates spatial specificity of Marco, E.M., Rubino, T., Adriani, W., Viveros, M.P., Parolaro, D., Laviola, G., 2009.
2-arachidonoyl glycerol signaling during axonal pathfinding. J. Neurosci. 30, Long-term consequences of URB597 administration during adolescence on
13992–14007, http://dx.doi.org/10.1523/JNEUROSCI.2126-10.2010 cannabinoid CB1 receptor binding in brain areas. Brain Res. 1257, 25–31, http://
Keimpema, E., Tortoriello, G., Alpár, A., Capsoni, S., Arisi, I., Calvigioni, D., Hu, S.S.-J., dx.doi.org/10.1016/j.brainres.2008.12.037
Cattaneo, A., Doherty, P., Mackie, K., Harkany, T., 2013. Nerve growth factor scales Mateos, B., Borcel, E., Loriga, R., Luesu, W., Bini, V., Llorente, R., Castelli, M.P., Viveros,
endocannabinoid signaling by regulating monoacylglycerol lipase turnover in M.P., 2011. Adolescent exposure to nicotine and/or the cannabinoid agonist
developing cholinergic neurons. Proc. Natl. Acad. Sci. U. S. A. 110, 1935–1940, CP 55,940 induces gender-dependent long-lasting memory impairments and
http://dx.doi.org/10.1073/pnas.1212563110 changes in brain nicotinic and CB1 cannabinoid receptors. J. Psychopharmacol.
Kim, D., Thayer, S.A., 2001. Cannabinoids inhibit the formation of new synapses 25, 1676–1690, http://dx.doi.org/10.1177/0269881110370503
between hippocampal neurons in culture. J. Neurosci. 21, RC146. Mato, S., Del Olmo, E., Pazos, A., 2003. Ontogenetic development of cannabinoid
Korb, E., Finkbeiner, S., 2011. Arc in synaptic plasticity: from gene to behavior. Trends receptor expression and signal transduction functionality in the human brain.
Neurosci. 34, 591–598, http://dx.doi.org/10.1016/j.tins.2011.08.007 Eur. J. Neurosci. 17, 1747–1754.
Korem, N., Akirav, I., 2014. Cannabinoids prevent the effects of a footshock followed McLaughlin, C.R., Martin, B.R., Compton, D.R., Abood, M.E., 1994. Cannabinoid recep-
by situational reminders on emotional processing. Neuropsychopharmacology tors in developing rats: detection of mRNA and receptor binding. Drug Alcohol
39, 2709–2722, http://dx.doi.org/10.1038/npp.2014.132 Depend. 36, 27–31.
Kosten, T.A., Ambrosio, E., 2002. HPA axis function and drug addictive behaviors: McPartland, J.M., Glass, M., Pertwee, R.G., 2007. Meta-analysis of cannabinoid lig-
insights from studies with Lewis and Fischer 344 inbred rats. Psychoneuroen- and binding affinity and receptor distribution: interspecies differences. Br. J.
docrinology 27, 35–69. Pharmacol. 152, 583–593, http://dx.doi.org/10.1038/sj.bjp.0707399
Kowalczyk, T., Pontious, A., Englund, C., Daza, R.A.M., Bedogni, F., Hodge, R., Attardo, McQueeny, T., Padula, C.B., Price, J., Medina, K.L., Logan, P., Tapert, S.F., 2011. Gen-
A., Bell, C., Huttner, W.B., Hevner, R.F., 2009. Intermediate neuronal progeni- der effects on amygdala morphometry in adolescent marijuana users. Behav.
tors (basal progenitors) produce pyramidal-projection neurons for all layers of Brain Res. 224, 128–134, http://dx.doi.org/10.1016/j.bbr.2011.05.031, S0166-
cerebral cortex. Cereb. Cortex 19, 2439–2450, http://dx.doi.org/10.1093/cercor/ 4328(11)00436-0 [pii].
bhn260 Meier, M.H., Caspi, A., Ambler, A., Harrington, H., Houts, R., Keefe, R.S.E., McDonald,
Lee, T.T.-Y., Hill, M.N., Hillard, C.J., Gorzalka, B.B., 2013. Temporal changes in K., Ward, A., Poulton, R., Moffitt, T.E., 2012. Persistent cannabis users show neu-
N-acylethanolamine content and metabolism throughout the peri-adolescent ropsychological decline from childhood to midlife. Proc. Natl. Acad. Sci. U. S. A.
period. Synapse 67, 4–10, http://dx.doi.org/10.1002/syn.21609 109, E2657–E2664, http://dx.doi.org/10.1073/pnas.1206820109
Lee, T.T.Y., Wainwright, S.R., Hill, M.N., Galea, L.A.M., Gorzalka, B.B., 2014. Sex, Mereu, G., Fa, M., Ferraro, L., Cagiano, R., Antonelli, T., Tattoli, M., Ghiglieri, V.,
drugs, and adult neurogenesis: sex-dependent effects of escalating adolescent Tanganelli, S., Gessa, G.L., Cuomo, V., 2003. Prenatal exposure to a cannabi-
cannabinoid exposure on adult hippocampal neurogenesis, stress reactivity, noid agonist produces memory deficits linked to dysfunction in hippocampal
and amphetamine sensitization. Hippocampus 24, 280–292, http://dx.doi.org/ long-term potentiation and glutamate release. Proc. Natl. Acad. Sci. U. S. A. 100,
10.1002/hipo.22221 4915–4920, http://dx.doi.org/10.1073/pnas.0537849100
Lenroot, R.K., Giedd, J.N., 2010. Sex differences in the adolescent brain. Brain Cogn. Mitsuhiro, S.S., Chalem, E., Barros, M.C., Guinsburg, R., Laranjeira, R., 2007. Prevalence
72, 46–55, http://dx.doi.org/10.1016/j.bandc.2009.10.008 of cocaine and marijuana use in the last trimester of adolescent preg-
Lenroot, R.K., Giedd, J.N., 2006. Brain development in children and adolescents: nancy: socio-demographic, psychosocial and behavioral characteristics. Addict.
insights from anatomical magnetic resonance imaging. Neurosci. Biobehav. Rev. Behav. 32, 392–397, http://dx.doi.org/10.1016/j.addbeh.2006.05.001, S0306-
30, 718–729, http://dx.doi.org/10.1016/j.neubiorev.2006.06.001 4603(06)00151-1 [pii].
Lenroot, R.K., Gogtay, N., Greenstein, D.K., Wells, E.M., Wallace, G.L., Clasen, L.S., Molina-Holgado, F., Amaro, A., Gonzalez, M.I., Alvarez, F.J., Leret, M.L., 1996. Effect
Blumenthal, J.D., Lerch, J., Zijdenbos, A.P., Evans, A.C., Thompson, P.M., Giedd, of maternal delta 9-tetrahydrocannabinol on developing serotonergic system.
J.N., 2007. Sexual dimorphism of brain developmental trajectories during child- Eur. J. Pharmacol. 316, 39–42.
hood and adolescence. Neuroimage 36, 1065–1073, http://dx.doi.org/10.1016/ Molina-Holgado, F., Molina-Holgado, E., Leret, M.L., González, M.I., Reader, T.A.,
j.neuroimage.2007.03.053 1993. Distribution of indoleamines and [3H]paroxetine binding in rat brain
144 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

regions following acute or perinatal delta 9-tetrahydrocannabinol treatments. Pistis, M., Perra, S., Pillolla, G., Melis, M., Muntoni, A.L., Gessa, G.L., 2004. Adolescent
Neurochem. Res. 18, 1183–1191. exposure to cannabinoids induces long-lasting changes in the response to drugs
Moore, D.G., Turner, J.D., Parrott, A.C., Goodwin, J.E., Fulton, S.E., Min, M.O., Fox, H.C., of abuse of rat midbrain dopamine neurons. Biol. Psychiatry 56, 86–94.
Braddick, F.M., Axelsson, E.L., Lynch, S., Ribeiro, H., Frostick, C.J., Singer, L.T., 2010. Pope Jr., H.G., Gruber, A.J., Hudson, J.I., Huestis, M.A., Yurgelun-Todd, D., 2001. Neu-
During pregnancy, recreational drug-using women stop taking ecstasy (3,4- ropsychological performance in long-term cannabis users. Arch. Gen. Psychiatry
methylenedioxy-N-methylamphetamine) and reduce alcohol consumption, but 58, 909–915, yoa20391 [pii].
continue to smoke tobacco and cannabis: initial findings from the Develop- Quinn, H.R., Matsumoto, I., Callaghan, P.D., Long, L.E., Arnold, J.C., Gunasekaran, N.,
ment and Infancy Study. J. Psychopharmacol. 24, 1403–1410, http://dx.doi.org/ Thompson, M.R., Dawson, B., Mallet, P.E., Kashem, M.A., Matsuda-Matsumoto,
10.1177/0269881109348165 H., Iwazaki, T., McGregor, I.S., 2008. Adolescent rats find repeated Delta(9)-
Moore, H., Jentsch, J.D., Ghajarnia, M., Geyer, M.A., Grace, A.A., 2006. A neurobehav- THC less aversive than adult rats but display greater residual cognitive deficits
ioral systems analysis of adult rats exposed to methylazoxymethanol acetate on and changes in hippocampal protein expression following exposure. Neuropsy-
E17: implications for the neuropathology of schizophrenia. Biol. Psychiatry 60, chopharmacology 33, 1113–1126.
253–264, http://dx.doi.org/10.1016/j.biopsych.2006.01.003 Raver, S.M., Keller, A., 2014. Permanent suppression of cortical oscillations in mice
Morel, L.J., Giros, B., Dauge, V., 2009. Adolescent exposure to chronic delta- after adolescent exposure to cannabinoids: receptor mechanisms. Neurophar-
9-tetrahydrocannabinol blocks opiate dependence in maternally deprived macology 86, 161–173, http://dx.doi.org/10.1016/j.neuropharm.2014.07.006
rats. Neuropsychopharmacology 34, 2469–2476, http://dx.doi.org/10.1038/npp. Raznahan, A., Shaw, P.W., Lerch, J.P., Clasen, L.S., Greenstein, D., Berman, R., Pipitone,
2009.70 J., Chakravarty, M.M., Giedd, J.N., 2014. Longitudinal four-dimensional mapping
Moreno, M., Trigo, J.M., Escuredo, L., Rodriguez de Fonseca, F., Navarro, M., 2003. Peri- of subcortical anatomy in human development. Proc. Natl. Acad. Sci. U. S. A. 111,
natal exposure to delta 9-tetrahydrocannabinol increases presynaptic dopamine 1592–1597, http://dx.doi.org/10.1073/pnas.1316911111
D2 receptor sensitivity: a behavioral study in rats. Pharmacol. Biochem. Behav. Realini, N., Rubino, T., Parolaro, D., 2009. Neurobiological alterations at adult age
75, 565–575, S0091305703001175 [pii]. triggered by adolescent exposure to cannabinoids. Pharmacol. Res. 60, 132–138.
Morishita, J., Okamoto, Y., Tsuboi, K., Ueno, M., Sakamoto, H., Maekawa, N., Realini, N., Vigano, D., Guidali, C., Zamberletti, E., Rubino, T., Parolaro, D.,
Ueda, N., 2005. Regional distribution and age-dependent expression of N- 2011. Chronic URB597 treatment at adulthood reverted most depressive-like
acylphosphatidylethanolamine-hydrolyzing phospholipase D in rat brain. J. symptoms induced by adolescent exposure to THC in female rats. Neurophar-
Neurochem. 94, 753–762, http://dx.doi.org/10.1111/j.1471-4159.2005.03234. macology 60, 235–243, http://dx.doi.org/10.1016/j.neuropharm.2010.09.003,
x S0028-3908(10)00224-8 [pii].
Morris, C.V., DiNieri, J.A., Szutorisz, H., Hurd, Y.L., 2011. Molecular mechanisms of Reimold, M., Batra, A., Knobel, A., Smolka, M.N., Zimmer, A., Mann, K., Solbach, C.,
maternal cannabis and cigarette use on human neurodevelopment. Eur. J. Neu- Reischl, G., Schwärzler, F., Gründer, G., Machulla, H.-J., Bares, R., Heinz, A., 2008.
rosci. 34, 1574–1583, http://dx.doi.org/10.1111/j.1460-9568.2011.07884.x Anxiety is associated with reduced central serotonin transporter availability in
Mulder, J., Aguado, T., Keimpema, E., Barabas, K., Ballester Rosado, C.J., Nguyen, L., unmedicated patients with unipolar major depression: a [11C]DASB PET study.
Monory, K., Marsicano, G., Di Marzo, V., Hurd, Y.L., Guillemot, F., Mackie, K., Lutz, Mol. Psychiatry 13, 606–613, http://dx.doi.org/10.1038/sj.mp.4002149, 557.
B., Guzman, M., Lu, H.C., Galve-Roperh, I., Harkany, T., 2008. Endocannabinoid Renard, J., Krebs, M., Le Pen, G., Jay, T.M., 2014. Long-term consequences of adoles-
signaling controls pyramidal cell specification and long-range axon patterning. cent cannabinoid exposure in adult psychopathology. Front. Neurosci. 8, 1–14,
Proc. Natl. Acad. Sci. U. S. A. 105, 8760–8765, http://dx.doi.org/10.1073/pnas. http://dx.doi.org/10.3389/fnins.2014.00361
0803545105 Renard, J., Krebs, M.O., Jay, T.M., Le Pen, G., 2013. Long-term cognitive impair-
Nacher, J., Guirado, R., Castillo-Gómez, E., 2013. Structural plasticity of interneurons ments induced by chronic cannabinoid exposure during adolescence in rats:
in the adult brain: role of PSA-NCAM and implications for psychiatric disorders. a strain comparison. Psychopharmacology (Berl). 225, 781–790, http://dx.doi.
Neurochem. Res. 38, 1122–1133, http://dx.doi.org/10.1007/s11064-013-0977- org/10.1007/s00213-012-2865-z
4 Rivkin, M.J., Davis, P.E., Lemaster, J.L., Cabral, H.J., Warfield, S.K., Mulkern, R.V., Rob-
Newsom, R.J., Kelly, S.J., 2008. Perinatal delta-9-tetrahydrocannabinol exposure dis- son, C.D., Rose-Jacobs, R., Frank, D.A., 2008. Volumetric MRI study of brain in
rupts social and open field behavior in adult male rats. Neurotoxicol. Teratol. 30, children with intrauterine exposure to cocaine, alcohol, tobacco, and marijuana.
213–219, http://dx.doi.org/10.1016/j.ntt.2007.12.007, S0892-0362(07)00367-4 Pediatrics 121, 741–750, http://dx.doi.org/10.1542/peds.2007-1399
[pii]. Robbins, T.W., 2007. Shifting and stopping: fronto-striatal substrates, neurochemical
Noland, J.S., Singer, L.T., Short, E.J., Minnes, S., Arendt, R.E., Kirchner, H.L., Bearer, modulation and clinical implications. Philos. Trans. R. Soc. Lond. Ser. B: Biol. Sci.
C., 2005. Prenatal drug exposure and selective attention in preschoolers. 362, 917–932, http://dx.doi.org/10.1098/rstb.2007.2097, H01V0404612T2871
Neurotoxicol. Teratol. 27, 429–438, http://dx.doi.org/10.1016/j.ntt.2005.02.001, [pii].
S0892-0362(05)00040-1 [pii]. Rodriguez de Fonseca, F., Ramos, J.A., Bonnin, A., Fernandez-Ruiz, J.J., 1993. Presence
O’Shea, M., McGregor, I.S., Mallet, P.E., 2006. Repeated cannabinoid exposure during of cannabinoid binding sites in the brain from early postnatal ages. Neuroreport
perinatal, adolescent or early adult ages produces similar longlasting deficits in 4, 135–138.
object recognition and reduced social interaction in rats. J. Psychopharmacol. Romero, J., Garcia-Palomero, E., Berrendero, F., Garcia-Gil, L., Hernandez, M.L.,
20, 611–621. Ramos, J.A., Fernandez-Ruiz, J.J., 1997. Atypical location of cannabinoid receptors
O’Shea, M., Singh, M.E., McGregor, I.S., Mallet, P.E., 2004. Chronic cannabi- in white matter areas during rat brain development. Synapse 26, 317–323.
noid exposure produces lasting memory impairment and increased Ronn, L.C., Berezin, V., Bock, E., 2000. The neural cell adhesion molecule in synaptic
anxiety in adolescent but not adult rats. J. Psychopharmacol. 18, plasticity and ageing. Int. J. Dev. Neurosci. 18, 193–199.
502–508. Rubino, T., Parolaro, D., 2008. Long lasting consequences of cannabis exposure in
O’Tuathaigh, C.M.P., Hryniewiecka, M., Behan, A., Tighe, O., Coughlan, C., Desbonnet, adolescence. Mol. Cell. Endocrinol. 286, S108–S113.
L., Cannon, M., Karayiorgou, M., Gogos, J.A., Cotter, D.R., Waddington, J.L., 2010. Rubino, T., Prini, P., Piscitelli, F., Zamberletti, E., Trusel, M., Melis, M., Sagheddu, C.,
Chronic adolescent exposure to -9-tetrahydrocannabinol in COMT mutant Ligresti, A., Tonini, R., Di Marzo, V., Parolaro, D., 2014. Adolescent exposure to
mice: impact on psychosis-related and other phenotypes. Neuropsychophar- THC in female rats disrupts developmental changes in the prefrontal cortex.
macology 35, 2262–2273, http://dx.doi.org/10.1038/npp.2010.100 Neurobiol. Dis., http://dx.doi.org/10.1016/j.nbd.2014.09.015 (in press).
Oudin, M.J., Gajendra, S., Williams, G., Hobbs, C., Lalli, G., Doherty, P., 2011. Endo- Rubino, T., Realini, N., Braida, D., Guidi, S., Capurro, V., Vigano, D., Guidali, C., Pinter,
cannabinoids regulate the migration of subventricular zone-derived neuroblasts M., Sala, M., Bartesaghi, R., Parolaro, D., 2009. Changes in hippocampal morphol-
in the postnatal brain. J. Neurosci. 31, 4000–4011, http://dx.doi.org/10.1523/ ogy and neuroplasticity induced by adolescent THC treatment are associated
jneurosci.5483-10.2011 with cognitive impairment in adulthood. Hippocampus 19, 763–772.
Padula, C.B., Schweinsburg, A.D., Tapert, S.F., 2007. Spatial working memory perfor- Rubino, T., Vigano’, D., Realini, N., Guidali, C., Braida, D., Capurro, V., Castiglioni,
mance and fMRI activation interaction in abstinent adolescent marijuana users. C., Cherubino, F., Romualdi, P., Candeletti, S., Sala, M., Parolaro, D., Vigano,
Psychol. Addict. Behav. 21, 478–487, http://dx.doi.org/10.1037/0893-164X.21. D., 2008. Chronic Delta(9)-tetrahydrocannabinol during adolescence provokes
4.478 sex-dependent changes in the emotional profile in adult rats: behavioral and
Paria, B.C., Dey, S.K., 2000. Ligand-receptor signaling with endocannabinoids in biochemical correlates. Neuropsychopharmacology 33, 2760–2771, http://dx.
preimplantation embryo development and implantation. Chem. Phys. Lipids doi.org/10.1038/sj.npp.1301664
108, 211–220, S0009308400001973 [pii]. Rubio, P., Rodriguez de Fonseca, F., Munoz, R.M., Ariznavarreta, C., Martin-Calderon,
Perez-Rosado, A., Gomez, M., Manzanares, J., Ramos, J.A., Fernandez-Ruiz, J., 2002. J.L., Navarro, M., 1995. Long-term behavioral effects of perinatal exposure to
Changes in prodynorphin and POMC gene expression in several brain regions of delta 9-tetrahydrocannabinol in rats: possible role of pituitary-adrenal axis. Life
rat fetuses prenatally exposed to Delta(9)-tetrahydrocannabinol. Neurotoxicol. Sci. 56, 2169–2176.
Res. 4, 211–218, http://dx.doi.org/10.1080/10298420290023936 Saez, T.M.M., Aronne, M.P., Caltana, L., Brusco, A.H., 2014. Prenatal exposure to the
Perez-Rosado, A., Manzanares, J., Fernandez-Ruiz, J., Ramos, J.A., 2000. Prenatal CB1 and CB2 cannabinoid receptor agonist WIN 55,212-2 alters migration of
Delta(9)-tetrahydrocannabinol exposure modifies proenkephalin gene expres- early-born glutamatergic neurons and GABAergic interneurons in the rat cere-
sion in the fetal rat brain: sex-dependent differences. Brain Res. Dev. Brain Res. bral cortex. J. Neurochem. 129, 637–648, http://dx.doi.org/10.1111/jnc.12636
120, 77–81. Saurel-Cubizolles, M.-J., Prunet, C., Blondel, B., 2014. Cannabis use during pregnancy
Pertwee, R.G., 2008. The diverse CB1 and CB2 receptor pharmacology of three in France in 2010. BJOG 121, 971–977, http://dx.doi.org/10.1111/1471-0528.
plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9- 12626
tetrahydrocannabivarin. Br. J. Pharmacol. 153, 199–215, http://dx.doi.org/10. Schneider, M., 2009. Cannabis use in pregnancy and early life and its consequences:
1038/sj.bjp.0707442 animal models. Eur. Arch. Psychiatry Clin. Neurosci. 259, 383–393, http://dx.doi.
Petit-Demouliere, B., Chenu, F., Bourin, M., 2005. Forced swimming test in mice: org/10.1007/s00406-009-0026-0
a review of antidepressant activity. Psychopharmacology (Berl). 177, 245–255, Schneider, M., 2008. Puberty as a highly vulnerable developmental period for the
http://dx.doi.org/10.1007/s00213-004-2048-7 consequences of cannabis exposure. Addict. Biol. 13, 253–263.
A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146 145

Schneider, M., Drews, E., Koch, M., 2005. Behavioral effects in adult rats of chronic compulsive heroin-seeking and altered striatal synaptic plasticity in the sub-
prepubertal treatment with the cannabinoid receptor agonist WIN 55,212-2. sequent generation. Neuropsychopharmacology 39, 1315–1323, http://dx.doi.
Behav. Pharmacol. 16, 447–454. org/10.1038/npp.2013.352
Schneider, M., Koch, M., 2003. Chronic pubertal, but not adult chronic cannabinoid Tantra, M., Kröcher, T., Papiol, S., Winkler, D., Röckle, I., Jatho, J., Burkhardt, H., Ron-
treatment impairs sensorimotor gating, recognition memory, and the perfor- nenberg, A., Gerardy-Schahn, R., Ehrenreich, H., Hildebrandt, H., 2014. St8sia2
mance in a progressive ratio task in adult rats. Neuropsychopharmacology 28, deficiency plus juvenile cannabis exposure in mice synergistically affect higher
1760–1769. cognition in adulthood. Behav. Brain Res., http://dx.doi.org/10.1016/j.bbr.2014.
Schulz, S., Becker, T., Nagel, U., von Ameln-Mayerhofer, A., Koch, M., 2013. Chronic 08.062
co-administration of the cannabinoid receptor agonist WIN55,212-2 during Tapert, S.F., Schweinsburg, A.D., Drummond, S.P.A., Paulus, M.P., Brown, S.A., Yang,
puberty or adulthood reverses 3,4 methylenedioxymetamphetamine (MDMA)- T.T., Frank, L.R., 2007. Functional MRI of inhibitory processing in abstinent ado-
induced deficits in recognition memory but not in effort-based decision making. lescent marijuana users. Psychopharmacology (Berl). 194, 173–183, http://dx.
Pharmacol. Biochem. Behav. 106, 91–100, http://dx.doi.org/10.1016/j.pbb.2013. doi.org/10.1007/s00213-007-0823-y
03.011, S0091-3057(13)00077-4 [pii]. Thanos, P.K., Michaelides, M., Umegaki, H., Volkow, N.D., 2008. D2R DNA transfer into
Schweinsburg, A.D., Schweinsburg, B.C., Medina, K.L., McQueeny, T., Brown, S.A., the nucleus accumbens attenuates cocaine self-administration in rats. Synapse
Tapert, S.F., 2010. The influence of recency of use on fMRI response during spa- 62, 481–486, http://dx.doi.org/10.1002/syn.20523
tial working memory in adolescent marijuana users. J. Psychoactive Drugs 42, Tomasiewicz, H.C., Jacobs, M.M., Wilkinson, M.B., Wilson, S.P., Nestler, E.J.,
401–412. Hurd, Y.L., 2012. Proenkephalin mediates the enduring effects of adolescent
Segev, A., Rubin, A.S., Abush, H., Richter-Levin, G., Akirav, I., 2014. Cannabinoid recep- cannabis exposure associated with adult opiate vulnerability. Biol. Psychi-
tor activation prevents the effects of chronic mild stress on emotional learning atry 72, 803–810, http://dx.doi.org/10.1016/j.biopsych.2012.04.026, S0006-
and LTP in a rat model of depression. Neuropsychopharmacology 39, 919–933, 3223(12)00409-X [pii].
http://dx.doi.org/10.1038/npp.2013.292 Tortoriello, G., Morris, C.V., Alpar, A., Fuzik, J., Shirran, S.L., Calvigioni, D., Keimpema,
Shabani, M., Hosseinmardi, N., Haghani, M., Shaibani, V., Janahmadi, M., 2011. Mater- E., Botting, C.H., Reinecke, K., Herdegen, T., Courtney, M., Hurd, Y.L., Harkany,
nal exposure to the CB1 cannabinoid agonist WIN 55212-2 produces robust T., 2014. Miswiring the brain: 9-tetrahydrocannabinol disrupts cortical devel-
changes in motor function and intrinsic electrophysiological properties of cere- opment by inducing an SCG10/stathmin-2 degradation pathway. EMBO J. 33,
bellar Purkinje neurons in rat offspring. Neuroscience 172, 139–152, http://dx. 668–685, http://dx.doi.org/10.1002/embj.201386035
doi.org/10.1016/j.neuroscience.2010.10.031, S0306-4522(10)01361-8 [pii]. Tree, K.C., Scotto di Perretolo, M., Peyronnet, J., Cayetanot, F., 2014. In utero cannabi-
Shabani, M., Mahnam, A., Sheibani, V., Janahmadi, M., 2014. Alterations in the intrin- noid exposure alters breathing and the response to hypoxia in newborn mice.
sic burst activity of Purkinje neurons in offspring maternally exposed to the CB1 Eur. J. Neurosci. 40, 2196–2204, http://dx.doi.org/10.1111/ejn.12588
cannabinoid agonist WIN 55212-2. J. Membr. Biol. 247, 63–72, http://dx.doi.org/ Trezza, V., Campolongo, P., Cassano, T., Macheda, T., Dipasquale, P., Carratu,
10.1007/s00232-013-9612-1 M.R., Gaetani, S., Cuomo, V., 2008. Effects of perinatal exposure to delta-9-
Silva, L., Zhao, N., Popp, S., Dow-Edwards, D., 2012. Prenatal tetrahydrocannabinol tetrahydrocannabinol on the emotional reactivity of the offspring: a longitudinal
(THC) alters cognitive function and amphetamine response from weaning to behavioral study in Wistar rats. Psychopharmacology (Berl). 198, 529–537,
adulthood in the rat. Neurotoxicol. Teratol. 34, 63–71, http://dx.doi.org/10.1016/ http://dx.doi.org/10.1007/s00213-008-1162-3
j.ntt.2011.10.006, S0892-0362(11)00205-4 [pii]. Trezza, V., Campolongo, P., Manduca, A., Morena, M., Palmery, M., Vanderschuren,
Smith, A.M., Fried, P.A., Hogan, M.J., Cameron, I., 2006. Effects of prenatal mar- L.J., Cuomo, V., 2012. Altering endocannabinoid neurotransmission at critical
ijuana on visuospatial working memory: an fMRI study in young adults. developmental ages: impact on rodent emotionality and cognitive performance.
Neurotoxicol. Teratol. 28, 286–295, http://dx.doi.org/10.1016/j.ntt.2005.12.008, Front. Behav. Neurosci. 6, 2, http://dx.doi.org/10.3389/fnbeh.2012.00002
S0892-0362(05)00180-7 [pii]. U.S. Department of Health and Human Services, 1999. Blending perspective and
Smith, A.M., Fried, P.A., Hogan, M.J., Cameron, I., 2004. Effects of prenatal marijuana building common ground. A report to Congress on substance abuse and
on response inhibition: an fMRI study of young adults. Neurotoxicol. Teratol. child protection [WWW Document]. URL: http://aspe.hhs.gov/hsp/subabuse99/
26, 533–542, http://dx.doi.org/10.1016/j.ntt.2004.04.004, S0892036204000662 subabuse.htm (accessed 10.06.14).
[pii]. United Nations Office on Drugs and Crime, 2013. World Drug Report 2013. Malta.
Smith, M.J., Cobia, D.J., Reilly, J.L., Gilman, J.M., Roberts, A.G., Alpert, K.I., Wang, L., Bre- Van Zundert, B., Yoshii, A., Constantine-Paton, M., 2004. Receptor compartmental-
iter, H.C., Csernansky, J.G., 2015. Cannabis-related episodic memory deficits and ization and trafficking at glutamate synapses: a developmental proposal. Trends
hippocampal morphological differences in healthy individuals and schizophre- Neurosci. 27, 428–437, http://dx.doi.org/10.1016/j.tins.2004.05.010, S0166-
nia subjects. Hippocampus, http://dx.doi.org/10.1002/hipo.22427 2236(04)00164-X [pii].
Spano, M.S., Ellgren, M., Wang, X., Hurd, Y.L., 2007. Prenatal cannabis exposure Vassoler, F.M., Johnson, N.L., Byrnes, E.M., 2013. Female adolescent exposure to
increases heroin seeking with allostatic changes in limbic enkephalin systems cannabinoids causes transgenerational effects on morphine sensitization in
in adulthood. Biol. Psychiatry 61, 554–563, http://dx.doi.org/10.1016/j.biopsych. female offspring in the absence of in utero exposure. J. Psychopharmacol. 27,
2006.03.073, S0006-3223(06)00557-9 [pii]. 1015–1022, http://dx.doi.org/10.1177/0269881113503504
Stopponi, S., Soverchia, L., Ubaldi, M., Cippitelli, A., Serpelloni, G., Ciccocioppo, R., Vela, G., Fuentes, J.A., Bonnin, A., Fernandez-Ruiz, J., Ruiz-Gayo, M., 1995.
2014. Chronic THC during adolescence increases the vulnerability to stress- Perinatal exposure to delta 9-tetrahydrocannabinol (delta 9-THC) leads
induced relapse to heroin seeking in adult rats. Eur. Neuropsychopharmacol. to changes in opioid-related behavioral patterns in rats. Brain Res. 680,
24, 1037–1045, http://dx.doi.org/10.1016/j.euroneuro.2013.12.012 142–147.
Suarez, I., Bodega, G., Fernandez-Ruiz, J., Ramos, J.A., Rubio, M., Fernandez, B., 2004a. Vela, G., Martin, S., Garcia-Gil, L., Crespo, J.A., Ruiz-Gayo, M., Javier Fernandez-
Down-regulation of the AMPA glutamate receptor subunits GluR1 and GluR2/3 Ruiz, J., Garcia-Lecumberri, C., Pelaprat, D., Fuentes, J.A., Ramos, J.A., Ambrosio,
in the rat cerebellum following pre- and perinatal delta9-tetrahydrocannabinol E., 1998. Maternal exposure to delta9-tetrahydrocannabinol facilitates mor-
exposure. Cerebellum 3, 66–74. phine self-administration behavior and changes regional binding to central mu
Suarez, I., Bodega, G., Fernandez-Ruiz, J.J., Ramos, J.A., Rubio, M., Fernandez, B., 2002. opioid receptors in adult offspring female rats. Brain Res. 807, 101–109, S0006-
Reduced glial fibrillary acidic protein and glutamine synthetase expression in 8993(98)00766-5 [pii].
astrocytes and Bergmann glial cells in the rat cerebellum caused by delta(9)- Venero, C., Herrero, A.I., Touyarot, K., Cambon, K., Lopez-Fernandez, M.A., Berezin,
tetrahydrocannabinol administration during development. Dev. Neurosci. 24, V., Bock, E., Sandi, C., 2006. Hippocampal up-regulation of NCAM expression
300–312. and polysialylation plays a key role on spatial memory. Eur. J. Neurosci. 23,
Suarez, I., Bodega, G., Rubio, M., Fernandez-Ruiz, J.J., Ramos, J.A., Fernandez, B., 1585–1595.
2004b. Prenatal cannabinoid exposure down-regulates glutamate transporter Viveros, M.P., Llorente, R., Suarez, J., Llorente-Berzal, A., Lopez-Gallardo, M., de
expressions (GLAST and EAAC1) in the rat cerebellum. Dev. Neurosci. 26, Fonseca, F.R., Rodriguez de Fonseca, F., 2011a. The endocannabinoid system
45–53. in critical neurodevelopmental periods: sex differences and neuropsychi-
Suarez, J., Llorente, R., Romero-Zerbo, S.Y., Mateos, B., Bermudez-Silva, F.J., de atric implications. J. Psychopharmacol. 26, 164–176, http://dx.doi.org/10.1177/
Fonseca, F.R., Viveros, M.P., 2009. Early maternal deprivation induces gender- 0269881111408956
dependent changes on the expression of hippocampal CB(1) and CB(2) Viveros, M.P., Marco, E.M., Lopez-Gallardo, M., Garcia-Segura, L.M., Wagner, E.J.,
cannabinoid receptors of neonatal rats. Hippocampus 19, 623–632, http://dx. 2011b. Framework for sex differences in adolescent neurobiology: a focus on
doi.org/10.1002/hipo.20537 cannabinoids. Neurosci. Biobehav. Rev. 35, 1740–1751, http://dx.doi.org/10.
Substance Abuse and Mental Health Services Administration, 2014a. Results from 1016/j.neubiorev.2010.09.005, S0149-7634(10)00145-4 [pii].
the 2013 National Survey on Drug Use and Health: Summary of National Find- Volkow, N.D., Fowler, J.S., Wang, G.J., Swanson, J.M., 2004. Dopamine in
ings. drug abuse and addiction: results from imaging studies and treatment
Substance Abuse and Mental Health Services Administration, 2014b. Behavioral implications. Mol. Psychiatry 9, 557–569, http://dx.doi.org/10.1038/sj.mp.
Health Barometer. 4001507 4001507
Substance Abuse and Mental Health Services Administration, 2013. Results from the Walters, D.E., Carr, L.A., 1988. Perinatal exposure to cannabinoids alters neuro-
2012 National Survey on Drug Use and Health. chemical development in rat brain. Pharmacol. Biochem. Behav. 29, 213–216,
Substance Abuse and Mental Health Services Administration, 2009. Substance Use S0091-3057(88)90300-0 [pii].
among Women During Pregnancy and Following Childbirth, The NSDUH Report. Wang, X., Dow-Edwards, D., Anderson, V., Minkoff, H., Hurd, Y.L., 2006. Discrete
Sun, X., Dey, S.K., 2008. Aspects of endocannabinoid signaling in periimplanta- opioid gene expression impairment in the human fetal brain associated with
tion biology. Mol. Cell. Endocrinol. 286, S3–S11, http://dx.doi.org/10.1016/j.mce. maternal marijuana use. Pharmacogenomics J. 6, 255–264, http://dx.doi.org/10.
2008.01.002, S0303-7207(08)00008-7 [pii]. 1038/sj.tpj.6500375
Szutorisz, H., DiNieri, J.a, Sweet, E., Egervari, G., Michaelides, M., Carter, J.M., Ren, Wang, X., Dow-Edwards, D., Anderson, V., Minkoff, H., Hurd, Y.L., 2004. In utero
Y., Miller, M.L., Blitzer, R.D., Hurd, Y.L., 2014. Parental THC exposure leads to marijuana exposure associated with abnormal amygdala dopamine D2 gene
146 A. Higuera-Matas et al. / Neuroscience and Biobehavioral Reviews 55 (2015) 119–146

expression in the human fetus. Biol. Psychiatry 56, 909–915, http://dx.doi.org/ Yamamoto, K., Shinba, T., Yoshii, M., 2014. Psychiatric symptoms of noradrener-
10.1016/j.biopsych.2004.10.015, S0006-3223(04)01095-9 [pii]. gic dysfunction: a pathophysiological view. Psychiatry Clin. Neurosci. 68, 1–20,
Wang, X., Dow-Edwards, D., Keller, E., Hurd, Y.L., 2003. Preferential limbic expression http://dx.doi.org/10.1111/pcn.12126
of the cannabinoid receptor mRNA in the human fetal brain. Neuroscience 118, Yan, H.C., Cao, X., Das, M., Zhu, X.H., Gao, T.M., 2010. Behavioral animal models of
681–694, S0306452203000204 [pii]. depression. Neurosci. Bull. 26, 327–337, http://dx.doi.org/10.1007/s12264-010-
Warner, T.D., Behnke, M., Eyler, F.D., Padgett, K., Leonard, C., Hou, W., Garvan, C.W., 0323-7
Schmalfuss, I.M., Blackband, S.J., 2006. Diffusion tensor imaging of frontal white Zamberletti, E., Beggiato, S., Steardo, L., Prini, P., Antonelli, T., Ferraro, L., Rubino,
matter and executive functioning in cocaine-exposed children. Pediatrics 118, T., Parolaro, D., 2014. Alterations of prefrontal cortex GABAergic transmission
2014–2024, http://dx.doi.org/10.1542/peds.2006-0003 in the complex psychotic-like phenotype induced by adolescent delta-9-
Wegener, N., Koch, M., 2009. Behavioural disturbances and altered Fos protein tetrahydrocannabinol exposure in rats. Neurobiol. Dis. 63, 35–47, http://dx.doi.
expression in adult rats after chronic pubertal cannabinoid treatment. Brain org/10.1016/j.nbd.2013.10.028
Res. 1253, 81–91, http://dx.doi.org/10.1016/j.brainres.2008.11.081, S0006- Zamberletti, E., Prini, P., Speziali, S., Gabaglio, M., Solinas, M., Parolaro, D., Rubino,
8993(08)02848-5 [pii]. T., 2012. Gender-dependent behavioral and biochemical effects of adolescent
Wenger, T., Gerendai, I., Fezza, F., Gonzalez, S., Bisogno, T., Fernandez-Ruiz, J., Di delta-9-tetrahydrocannabinol in adult maternally deprived rats. Neuroscience
Marzo, V., 2002. The hypothalamic levels of the endocannabinoid, anandamide, 204, 245–257, http://dx.doi.org/10.1016/j.neuroscience.2011.11.038, S0306-
peak immediately before the onset of puberty in female rats. Life Sci. 70, 4522(11)01323-6 [pii].
1407–1414. Zhang, Y., Landthaler, M., Schlussman, S.D., Yuferov, V., Ho, A., Tuschl, T., Kreek, M.J.,
Wu, C.S., Jew, C.P., Lu, H.C., 2011. Lasting impacts of prenatal cannabis exposure and 2009. Mu opioid receptor knockdown in the substantia nigra/ventral tegmental
the role of endogenous cannabinoids in the developing brain. Futur. Neurol. 6, area by synthetic small interfering RNA blocks the rewarding and locomo-
459–480. tor effects of heroin. Neuroscience 158, 474–483, http://dx.doi.org/10.1016/j.
Wu, C.-S., Morgan, D., Jew, C.P., Andrews, M.-J., Leishman, E., Spencer, C.M., Czyzyk, neuroscience.2008.09.039
T., Bradshaw, H., Mackie, K., Lu, H.-C., 2014. Long-term consequences of perinatal Zhu, P.J., Lovinger, D.M., 2010. Developmental alteration of endocannabinoid retro-
fatty acid amino hydrolase inhibition. Br. J. Pharmacol. 171, 1420–1434. grade signaling in the hippocampus. J. Neurophysiol. 103, 1123–1129, http://dx.
Wu, C.S., Zhu, J., Wager-Miller, J., Wang, S., O’Leary, D., Monory, K., Lutz, B., Mackie, K., doi.org/10.1152/jn.00327.2009
Lu, H.C., 2010. Requirement of cannabinoid CB(1) receptors in cortical pyramidal Zurolo, E., Iyer, A.M., Spliet, W.G., Van Rijen, P.C., Troost, D., Gorter, J.A., Aronica, E.,
neurons for appropriate development of corticothalamic and thalamocorti- 2010. CB1 and CB2 cannabinoid receptor expression during development and in
cal projections. Eur. J. Neurosci. 32, 693–706, http://dx.doi.org/10.1111/j.1460- epileptogenic developmental pathologies. Neuroscience 170, 28–41, http://dx.
9568.2010.07337.x doi.org/10.1016/j.neuroscience.2010.07.004, S0306-4522(10)00958-9 [pii].

You might also like