You are on page 1of 13

Alcohol 110 (2023) 1e13

Contents lists available at ScienceDirect

Alcohol
journal homepage: http://www.alcoholjournal.org/

Combined exposure to alcohol and cannabis during development:


Mechanisms and outcomes
cs a, Helenice Charchat-Fichman a, J. Landeira-Fernandez a,
Martina V. Kova
Alexandre E. Medina b, 1, *, Thomas E. Krahe a, **, 1
a
Departamento de Psicologia, Laboratorio de Neuroci^ lica do Rio de Janeiro, Rua Marqu^
encia do Comportamento, Pontifícia Universidade Cato ~o
es de Sa
vea e Rio de Janeiro, RJ, 22451-900, Brazil
Vicente, 225, Ga
b
Department of Pediatrics e School of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, United States

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

Article history: Exposure to substances of abuse during pregnancy can have long-lasting effects on offspring. Alcohol is
Received 6 December 2022 one of the most widely used substances of abuse that leads to the most severe consequences. Recent
Received in revised form studies in the United States, Canada, and the United Kingdom showed that between 1% and 7% of all
20 January 2023
children exhibit signs and symptoms of fetal alcohol spectrum disorder (FASD). Despite preventive
Accepted 25 January 2023
campaigns, the rate of children with FASD has not decreased during recent decades. Alcohol con-
sumption often accompanies exposure to such drugs as tobacco, cocaine, opioids, and cannabis. These
Keywords:
interactions can be synergistic and exacerbate the deleterious consequences of developmental alcohol
Cannabinoid receptors
Drug interactions
exposure. The present review focuses on interactions between alcohol and cannabis exposure and the
Endocannabinoid system potential consequences of these interactions.
Fetal alcohol spectrum disorders (FASD) © 2023 Elsevier Inc. All rights reserved.
Prenatal drug exposure
Simultaneous alcohol and cannabis
exposure

Introduction known about its effects on the maternal and fetal central nervous
systems. The cannabis legalization process for medical and recre-
Substance use during pregnancy endangers the fetus and may ational use in various countries globally requires a better under-
alter neurodevelopment, causing life-long consequences. Cannabis standing of the effects of cannabis during pregnancy.
products are the most frequently used illicit drugs among pregnant The consequences of alcohol exposure on the developing child
women, and recent legalization in many states in the United States are well-documented (Mattson, Bernes, & Doyle, 2019; May et al.,
increased their use during the last decade (Substance Abuse and 2020; Riley & McGee, 2005; Wozniak, Riley, & Charness, 2019) and
Mental Health Services Administration, 2020). Marijuana is are considered the leading cause of avoidable developmental dis-
commonly used as an anti-nausea remedy during early stages of abilities (Abel & Sokol, 1986). Physical, cognitive, and neurobiolog-
pregnancy (Dickson et al., 2018). Interestingly, half of pregnant ical consequences in the fetus following gestational alcohol
women who report drinking alcohol also report consuming consumption are collectively known as fetal alcohol spectrum dis-
cannabis (Center for Behavioral Health Statistics and Quality, 2015). order (FASD). The most devastating outcome of FASD is fetal alcohol
Despite the high prevalence of simultaneous drug use, little is syndrome (FAS), which is associated with craniofacial and ocular
dysmorphology and growth and cognitive deficits. An increasing
number of studies demonstrate FAS-like physical alterations
* Corresponding author. Department of Pediatrics, University of Maryland School after gestational cannabis exposure (Boa-Amponsem, Zhang,
of Medicine, 655 W. Baltimore St., MD, 21201, Baltimore Mukhopadhyay, Ardrey, & Cole, 2019; Carty, Thornton, Gledhill, &
** Corresponding author. Departamento de Psicologia, Laborato rio de Neuro-
Willett, 2018; Fish et al., 2019; Gilbert et al., 2016). This literature
^ncia do Comportamento, Pontifícia Universidade Cato
cie lica do Rio de Janeiro, Rua
^s de Sa
~o Vicente, 225, Ga
vea e Rio de Janeiro, RJ, 22451-900, Brazil.þ55 21
suggests common mechanisms and pathways by which both alcohol
Marque
3527-1187 and cannabis exert teratogenic effects on the developing central
E-mail addresses: amedina@som.umaryland.edu (A.E. Medina), tekrahe@puc- nervous system. Both alcohol and cannabis are known as neuro-
rio.br (T.E. Krahe). inhibitory drugs. When alcohol is consumed together with other
1
These authors share senior co-authorship.

https://doi.org/10.1016/j.alcohol.2023.01.004
0741-8329/© 2023 Elsevier Inc. All rights reserved.
cs, H. Charchat-Fichman, J. Landeira-Fernandez et al.
M.V. Kova Alcohol 110 (2023) 1e13

Abbreviations GPCR G protein-coupled receptors


GPR55 cannabinoid receptor, activated with the same
2-AG 2-arachidonoylglycerol potency to THC as CB1
ACEA Synthetic Cannabinoid, CB1 receptor agonist HU-210 Synthetic Cannabinoid, CB1 receptor agonist
AEA anandamide JNK JUN N-terminal kinases
AM251 Synthetic Cannabinoid, CB1 receptor antagonist JWH133 Synthetic Cannabinoid, CB2 agonist
AM630 Synthetic Cannabinoid, CB2 receptor antagonist JZL195 inhibitor of both fatty acid amide hydrolase (FAAH)
Arc activity-regulated cytoskeleton-associated protein and monoacylglycerol lipase (MAGL)
cAMP adenosine 30 , cyclic 50 -monophosphate MAGL monoacylglycerol lipase
Caspase 3 protein that interacts with caspase-8 and caspase-9 MAPK pathway mitogen-activated protein kinase
CB1 receptor cannabinoid receptor 1 NFkB nuclear factor kappa light chain enhancer of activated
CB2 receptor cannabinoid receptor 2 B cells
CBD Cannabidiol, a non-psychotropic cannabis NMDA receptor ionotropic receptor activated by glutamic acid
constituent pERK1/2 phosphorylated extracellular regulated kinase 1/2
CBP Nuclear factor, CREB binding protein PLA₂ phospholipase A₂
CBs cannabinoids Rac Ras-related C3 botulinum toxin substrate 1
CDK5 Cyclin-dependent kinase 5 Rho a member of the Ras superfamily of small GTPases
CP 55.940 Synthetic Cannabinoid, CB1 receptor agonist Shh pathway Sonic hedgehog pathway
CREB cellular transcription factor Smoothened (Smo) receptor a G-protein coupled receptor,
DAGL diacylglycerol lipase Sonic-hedgehog (Shh) binding
ECBs endocannabinoids SR141716A Synthetic Cannabinoid, CB1 receptor antagonist
FAAH fatty acid amide hydrolase 1 THC psychoactive compound of cannabis
GABA Gamma-AminoButyric Acid WIN 55.212e2 Synthetic Cannabinoid, CB1 receptor agonist

inhibitory drugs (e.g., cannabis, opioids, or g-hydroxybutyric acid ascertainment method estimated 1e5% of total FASD cases in
[GHBA]) it synergistically increases its effect (Singh, 2019). However, Canada, Croatia, and the United States among the population.
the mechanisms that underlie the interaction between these two However, this rate reaches 23e28% in South African communities
substances and how this interaction leads to specific physical and (May et al., 2020). A recent study estimated that the prevalence of
behavioral features remain to be elucidated. The results of the few FASD among children and youths in the general population is 7.7
studies on this subject indicate the involvement of the endocanna- per 1000 (Lange et al., 2017). Popova and co-workers (Popova,
binoid system with an important role of the cannabinoid-1 (CB1) Lange, Shield, Burd, & Rehm, 2019) reported that the prevalence
receptor and sonic hedgehog pathway. The present review synthe- of FASD in subpopulations (i.e., childcare, special care, correctional
sizes recent findings from animal models that were prenatally institutions, Aboriginal, and specialized clinical groups) was 10e40
exposed simultaneously to alcohol and cannabinoids, with a focus times higher than in the global general population.
on mechanisms and behavioral and physical outcomes. Few data exist in the literature on simultaneous alcohol and
cannabis use among pregnant women. However, cannabis is well
Epidemiology known to be commonly used as an anti-nausea remedy during
pregnancy (Fish et al., 2019). Studies have described higher craving
Despite preventive campaigns in recent decades, substance use for other drugs among marijuana users (Center for Behavioral Health
during pregnancy is still relatively frequent. According to the 2019 Statistics and Quality, 2015; Hausknecht, Shen, Wang, Haj-
National Survey on Drug Use and Health, 5.4% of pregnant women Dahmane, & Shen, 2017; Wang et al., 2019) . Interestingly, 46.1% of
declared past-month marijuana use and 9.5% declared past-month pregnant women who smoked marijuana in the past year also
alcohol use in the United States (Substance Abuse and Mental declared past-month heavy alcohol use. Other studies estimated
Health Services Administration, 2020). The survey reported a sig- that 15.3% of women of peak fertility age (18e29 years) simulta-
nificant increase in marijuana use but a slight decrease in alcohol neously used alcohol and marijuana (Subbaraman & Kerr, 2015).
use among pregnant women compared with past years. Ko and co- Age-specific changes were observed in simultaneous alcohol and
workers (Ko, Farr, Tong, Creanga, & Callaghan, 2015) reported that marijuana use in past decades, with a significant increase among
16.3% of past-year marijuana users were pregnant and daily users, young adults (Terry-McElrath & Patrick, 2018). The high prevalence
and more than 10% of pregnant women used marijuana in the past of simultaneous use in this age group is particularly concerning
year. Furthermore, the study reported that 70% of pregnant women because substance use increases unintended pregnancies (Egan
believe that marijuana use is harmless. et al., 2019) and consequently may seriously affect the unrecog-
Alcohol-induced physical, cognitive, and structural deficits are nized fetus.
commonly known as FASD, but marijuana-induced disabilities or
alterations of central nervous system development are not yet Neurodevelopmental consequences of alcohol and cannabis
characterized or described as a distinct disorder. However, accu- prenatal exposure
mulating evidence demonstrates the teratogenicity of D9-tetrahy-
drocannabinol (THC) and cannabidiol (CBD), the main constituents Alcohol
of the Cannabis sativa plant (Boa-Amponsem et al., 2019; Carty
et al., 2018; Fish et al., 2019; Gilbert et al., 2016). Alcohol-induced impairments are well documented in the
The prevalence of FASD varies among countries and subgroups literature, with various types of timing and frequency of alcohol
within populations. Cultural differences may play an important exposure (Sulik, Johnston, & Webb, 1981). Alcohol is highly
role in this variation. Studies that employed an active case neurotoxic and easily crosses the placenta and bloodebrain barrier,
2
cs, H. Charchat-Fichman, J. Landeira-Fernandez et al.
M.V. Kova Alcohol 110 (2023) 1e13

causing changes in gene expression, neuronal proliferation (Luo & Cannabis


Miller, 1998), migration (Delatour, Yeh, & Yeh, 2019), oligodendro-
cyte number, and white matter integrity (Newville, Valenzuela, Li, The effects of alcohol have been widely investigated in recent
Jantzie, & Cunningham, 2017). It also interferes with growth fac- decades. Studies of cannabis-induced impairments in the devel-
tors and synaptogenesis and disrupts neuronal plasticity (Medina, oping brain have encountered many limitations, mainly because
2011). Alcohol also alters the homeostasis of brain activity levels the human consumption of cannabis can occur in different ways
by activating both excitatory and inhibitory receptors. Depending and with different amounts of constituents. The higher potency of
on the gestational age at which exposure occurs, nutritional char- cannabis preparations is associated with mental illness, poor aca-
acteristics of the mother, the amount and frequency of alcohol demic performance, lung inflammation, and vascular diseases
consumption, and the metabolism in the fetus, different types of (ElSohly et al., 2016).
damage can occur to the fetal nervous system. Also, prenatal The chemical structure of cannabinoids consists of two main
alcohol exposure is associated with long-term health and resiliency groups: phytocannabinoids (which naturally occur in the cannabis
deficits to adult-onset neurological disease (Bake, Hurst, Miranda, plant) and synthetic cannabinoids (human-made substances that
& Sohrabji, 2022). Recent studies indicate that genetic back- are intended for laboratory use or designer drugs with manipu-
ground may also play a key role in the susceptibility to the tera- lated chemical structures, mimicking psychoactive effects of the
togenic consequences of prenatal alcohol exposure. Gene- cannabis plant; Fish et al., 2019; Musselman & Hampton, 2014).
dependent differences in alcohol sensitivity were found in the More than 500 different constituents, including 104 cannabinoids,
gastrulation stage of mouse embryos when comparing two C57BL/6 have been identified in the Cannabis sativa plant (ElSohly et al.,
strains. Alcohol-exposed C57BL/6J embryos exhibited a more pro- 2016). The most frequently investigated components are THC
nounced transcriptional effect on craniofacial dysmorphology, cell and CBD. These two main components exert their effects in
death, and proliferation compared to C57BL/6NHsd ones (Boschen, different ways and have opposing effects. THC acts as a CB1 re-
Ptacek, Berginski, Simon, & Parnell, 2021). The latter harbor a ceptor agonist, whereas CBD exerts its effects through various
spontaneous Dock2 mutation affecting B cell signaling and immune pharmacological mechanisms by acting as a CB1 receptor antag-
tolerance that is not found in C57BL/6J mice (Mahajan et al., 2016). onist, thereby protecting against THC-induced neurodegeneration
Similarly, the tumor suppressor protein Tp53 gene deletion pro- (Curran et al., 2016). Consequently, the effects of THC and CBD are
tected against alcohol-induced morphological changes in mice and different. THC exposure appears to cause anxiety-like behavior
zebrafish embryos (Fish, Tucker, Peterson, Eberhart, & Parnell, and impair learning and memory. Studies of CBD found that it can
2021). Furthermore, Bax gene has been identified to facilitate exert protective effects against these THC-induced impairments
fetal eye and face malformations of mouse embryos following (Curran et al., 2016). Cannabis products that are available in the
gastrulation-stage exposure to alcohol (Fish et al., 2022). Together illicit market can largely differ in potency. Potency is based on the
these findings highlight the importance of genetic influences and amount of THC, the main psychoactive constituent of cannabis,
gene/environment interactions associated with the teratogenic ef- that is necessary to produce a given effect. According to cannabis
fects of alcohol on the developing fetus. potency trends (ElSohly et al., 2016), THC concentrations have
Even brief alcohol exposure during the first trimester of preg- increased in various preparations in the United States in recent
nancy can alter normal development of the neural tube and neural decades while CBD concentrations have decreased.
crest and lead to specific craniofacial and ocular dysmorphology, Cannabis constituents may exert their effects in distinct ways
growth deficiency, and neurocognitive impairments, which are selectively binding to i) G-protein coupled cannabinoid receptors
frequently observed in FAS, the most severe type of FASD (Sulik CB1 and CB2 (Felder et al., 1995), ii) G-protein coupled receptor 55
et al., 1981). (GPR55) (Lauckner et al., 2008), and iii) transient receptor po-
Second-trimester alcohol exposure is associated with alterations tential ion and cation channels such as TRPV1, TRPV2, TRPM8,
of the proliferation and migration of cortical neurons (Delatour et al., and TRPA channels (De Petrocellis et al., 2011). Cannabinoid re-
2019; Luo & Miller, 1998) and abnormal oligodendrocyte differen- ceptor agonists induce the inhibition of the activity of adenyl
tiation (Darbinian et al., 2021). These alterations are thought to lead cyclase and N-type voltage-dependent calcium channels,
to neurocognitive and somatosensory impairments among FASD decreasing the activity of cAMP-dependent protein kinase (Felder
individuals. Furthermore, baboon experiments suggest that alcohol et al., 1995). THC acts mainly through CB1 receptors on presyn-
may cause cerebrovascular dysfunction through endocannabinoid aptic terminals as a partial agonist, resulting in inhibitory syn-
signaling, which plays a role in arterial contractility regulation. aptic transmission. CBD (cannabidiol) is a negative allosteric
Vasodilation was observed in midterm baboon fetuses via CB2 re- modulator of cannabinoid receptors, with a low binding affinity
ceptor activation after alcohol exposure (Seleverstov et al., 2017; to CB1R (Kathmann, Flau, Redmer, Tra €nkle, & Schlicker, 2006).
Simakova et al., 2018). However, cerebral vasodilation in baboon The increase of CB1R activity by CBD is due to its action on non-
fetuses during mid-gestation disappeared by late gestation, sug- CB1 receptor targets that in turn inhibit endocannabinoid inac-
gesting age-dependent crosstalk between alcohol and the endo- tivation (Alp ar, Di Marzo, & Harkany, 2016). While CBN (canna-
cannabinoid system (Simakova et al., 2018). Recently, Rouzer and binol) is a low-affinity agonist of cannabinoid receptors (Rhee
Diaz (2022) showed that alcohol exposure during the second et al., 1997), some studies reported its TRPA1 agonistic and
trimester of rat gestation induces functional changes in the CRF TPRM8 antagonistic effects (De Petrocellis et al., 2011; Jordt et al.,
system (the stress peptide corticotropin-releasing factor) activity in 2004). Synthetic cannabinoids are full CB1 receptor agonists and
a sex-age and concentration-specific manner and increases the mimic the effects of THC, with high binding affinity to CB1 and
expression of anxiety-like behavior. An in-depth and cohesive CB2 receptors. However, synthetic cannabinoids, such as
description of the endocannabinoid system is provided below. CP55,940, JWH-018, WIN55, and 212-2 are more potent than THC
Studies of third-trimester alcohol exposure reported alterations (Augustin & Lovinger, 2022).
of synaptogenesis, an increase in apoptosis, and impairments in This increase in potency can have adverse outcomes on the
neuronal plasticity that lead to impairments in learning and mem- developing child during fetal exposure. A growing number of
ory and anxiety-like behavior (Baculis, Diaz, & Valenzuela, 2015; studies have investigated physical and psychological alterations
Filgueiras, Krahe, & Medina, 2010; Medina, 2011; Newville et al., that are induced by prenatal marijuana exposure (de Salas-
2017). Quiroga et al., 2020; Gunn et al., 2018; Raghunathan et al., 2019;
3
cs, H. Charchat-Fichman, J. Landeira-Fernandez et al.
M.V. Kova Alcohol 110 (2023) 1e13

Scheyer, Melis, Trezza, & Manzoni, 2019), suggesting that it im- Actions of alcohol and cannabis on the endocannabinoid system in
pairs endocannabinoid system signaling in the developing central the developing central nervous system
nervous system, representing an early harmful event during
development. In the presence of environmental stressors, alter- Studies of animal models of FASD have been developed in recent
ations of endocannabinoid signaling may provoke the emergence decades to shed light on the mechanisms of alcohol-induced tera-
of a specific phenotype, but the reason why alterations are not togenic effects in the developing central nervous system. These
always present after prenatal cannabinoid exposure remains un- studies have reported epigenetic modifications (for review, see
known (Richardson, Hester, & McLemore, 2016). Nonetheless, Basavarajappa & Subbanna, 2016), alterations of neuroplasticity
prenatal cannabis exposure is associated with lower birth weight, (Granato & Dering, 2018; Medina, 2011), and alterations of
alterations of sleep patterns, problems in executive functions, signaling pathways (Kumar, Singh, DiPette, & Singh, 2010; Shukrun,
emotional problems, impulsivity, hyperactivity (El Marroun et al., Shabtai, Pillemer, & Fainsod, 2019; Zhang, Ojiaku, & Cole, 2013).
2018), poor verbal memory (Ellingson et al., 2021), and impair- Alcohol exerts effects on the endocannabinoid system
ments in spatial cognition (de Salas-Quiroga et al., 2020). (Basavarajappa, Cooper, & Hungund, 1998; Basavarajappa, Saito,
During development, a heightened expression of CB1 mRNA Cooper, & Hungund, 1997), a fundamental and widespread neuro-
was observed in the amygdala and hippocampus of human fetal modulatory system in the brain and peripheral cells. The endo-
specimens from mothers with and without history of cannabis use cannabinoid system was discovered while studying psychoactive
during pregnancy (Wang, Dow-Edwards, Keller, & Hurd, 2003). mechanisms of the cannabis plant. This system is activated by both
These areas are part of the mesocorticolimbic circuit, which plays endocannabinoids (endogenously produced ligands) and cannabi-
an important role in emotional regulation, motor function, and noids (constituents of the Cannabis sativa plant). Since its discovery,
cognition. As the mesocorticolimbic dopamine circuits are coupled the endocannabinoid system has been widely studied in various
to CB1 receptors (Glass & Felder, 1997), Wang and colleagues contexts, including substance use disorders.
(Wang, Dow-Edwards, Anderson, Minkoff, & Hurd, 2004) sought The endocannabinoid system consists of endogenous ligands
to know whether prenatal cannabis exposure affects CB1 and (e.g., anandamide [AEA] and 2-arachidonylglycerol [2-AG]),
dopamine receptors D1 and D2. The study investigated mid- cannabinoid receptors (CB1 and CB2), interacting proteins, and
gestation post-mortem brains and found that cannabis exposure metabolic enzymes that are responsible for endocannabinoid for-
significantly reduced D2 mRNA expression in the amygdala basal mation and degradation. Anandamide is synthesized from phos-
nucleus in male, but not female subjects. This reduction was pholipids, such as N-acylphosphatidylethanolamine-specific
positively correlated with the amount of maternal cannabis con- phospholipase D (NAPE-PLD) and degraded by fatty acid amide
sumption. No significant alterations were found in CB1, D1, or D2 hydrolase (FAAH). The biosynthesis of 2-AG occurs through diac-
mRNA expression in the amygdala and hippocampus. No ylglycerol lipase a (DAGLa) and DAGLb and is degraded by mono-
cannabisealcohol interactions have been found; however, alcohol acylglycerol lipase (MAGL). The endogenous ligands AEA and 2-AG
contributed to the decreased expression of D1/D2 receptors in the bind to both CB1 and CB2 receptors on the presynaptic cell mem-
striatum (Wang et al., 2004). These results suggest that cannabis brane to activate a cascade of negative feedback mechanisms
may affect brain organization differently in males and females. within neurotransmitter systems to maintain homeostasis in the
Neurobiological studies of first-trimester cannabinoid exposure organism. CB1 receptors are mainly expressed in the central ner-
have found dramatic changes in neurodevelopment. Chick em- vous system and are more abundant than CB2 receptors, which
bryos that were exposed to THC during gastrulation exhibited have been detected predominantly in the peripheral nervous sys-
aberrant neural plate formation and alterations of somite, spinal tem (Basavarajappa, Joshi, Shivakumar, & Subbanna, 2019; Bukiya,
cord, brain, and heart development (Psychoyos, Hungund, Cooper, 2019).
& Finnell, 2008). Prenatal THC administration in mouse embryos
acted on the development of g-aminobutyric acid (GABA)ergic Endocannabinoid signaling mechanisms
interneurons and pyramidal neurons and led to alterations of
hippocampal function and deficits in spatial cognition. Impair- The endocannabinoid signaling system plays distinct roles and
ments in adult mice after prenatal THC exposure showed sexual has different mechanisms during fetal development and in the
dimorphism, in which male rats were selectively affected by pre- adult brain. The endocannabinoid signaling system plays an
natal THC administration (de Salas-Quiroga et al., 2020). important role during neurodevelopment, including neurogenesis,
Tortoriello et al. (2014) reported alterations of axon morphology proliferation, migration, axonal pathfinding, and the formation of
and an imbalance in cytoskeletal dynamics after early THC expo- synaptic connections (Bukiya, 2019; Wu, Jew, & Lu, 2011). The
sure, leading to miswiring of the brain in human fetuses. The endocannabinoid system may exert effects even before conception
administration of CB1 receptor agonists in mouse and zebrafish because it is present in reproductive tissues and affects sperm
embryos (Boa-Amponsem et al., 2019; Fish et al., 2019; Gilbert fertilizing capacity, ovulation, and hormonal production (for re-
et al., 2016) during the gastrulation period led to anterior neural view, see Bukiya, 2019).
tube, ocular, and craniofacial abnormalities, similar to manifesta- Rodent and chick studies suggest that cannabinoid receptors are
tions of FASD. present at early embryonal stages of development. In chick em-
Second-trimester exposure to cannabinoids is associated with a bryos, CB1 receptor gene expression was detected in the earliest
decrease in brain vasculature. Vasoconstriction was observed mi- appearing neurons in the hindbrain as early as stage 10 (somito-
nutes after the mouse embryonic brain was exposed to CP55,940, a genesis; a process during embryogenesis, after 33e38 h of incu-
frequently used synthetic cannabinoid in preclinical studies bation; Hamburger & Hamilton, 1992). At stage 11 (after 40e45 h of
(Raghunathan et al., 2019). THC exposure during the third trimester incubation), receptors appeared in the peripheral nervous system
resulted in a smaller inner diameter of the aorta in human fetuses and ophthalmic trigeminal placode, followed by the vestibuloa-
(El Marroun et al., 2010), which may lead to insufficient oxygen and coustic system, epibranchial ganglions, dorsal root ganglia, the
nutrient supply for developing organs. This may explain growth ventral forebrain, and the mesoderm (Begbie, Doherty, & Graham,
deficiencies that are frequently observed after marijuana exposure 2004). In rats, Cb1r mRNA expression and CB1 receptor binding
in neonates (Calvigioni, Hurd, Harkany, & Keimpema, 2014; El were detected as early as gestational day 11 in some cells of the
Marroun et al., 2009). neural tube and were present in several distinct structures in later
4
cs, H. Charchat-Fichman, J. Landeira-Fernandez et al.
M.V. Kova Alcohol 110 (2023) 1e13

stages (gestational day 14e15; Buckley, Hansson, Harta, & Mezey, Alcohol and cannabis are both known as neuroinhibitory sub-
1998). In humans, evidence shows CB1 receptor immunoreactivity stances. Alcohol potentiates the GABAA receptor-operated chloride
in the cortical plate at gestation week 9, whereas CB2 receptor channels by enhancing GABA activity, which results in decreased
immunoreactivity was detected only at later stages of the micro- neural excitability (Allan & Harris, 1987). However, experimental
glia/macrophage lineage (Zurolo et al., 2010). During fetal devel- studies indicate that during development, activation of GABAA re-
opment, the distribution of CB1 receptors is atypical and transient ceptors can be excitatory because of the high intracellular con-
compared with the adult central nervous system (Buckley et al., centration of chloride that produces reversal potentials that are
1998; Mato, Del Olmo, & Pazos, 2003). more positive than the resting membrane potential (Ben-Ari,
The presence of the endocannabinoid signaling system at early 2002). The combined use of other neuroinhibitory drugs, such as
stages of fetal development indicates its involvement in ontogen- cannabis, synergistically augments the inhibition that is caused by
esis. During fetal development (endo) cannabinoid-sensing re- alcohol. Alcohol augments inhibitory neurotransmission through
ceptors and related enzymatic machinery regulate neural GABA and decreases excitatory neurotransmission through gluta-
progenitor cell proliferation, neurite outgrowth, and directional mate (Singh, 2019). Furthermore, perinatal alcohol and marijuana
guidance (Alpa r et al., 2016). In early stages of central nervous exposure acts on inhibitory interneurons in the adult hippocampal
system development, both synthesis and degradation activities formation. Alcohol acts mainly on postsynaptic GABA receptors
occur in the same cell, unlike in the adult brain where postsynaptic (e.g., GABAᴀ receptors become hyperexcitable), and N-methyl-D-
on-demand ligand release activates cannabinoid receptors that are aspartate (NMDA) receptors become blocked, whereas marijuana
expressed on the presynaptic membrane. During developmental mostly targets presynaptic CB1 receptors. Simultaneous exposure
stages of proliferation and progenitor cell differentiation, the to alcohol and marijuana dramatically impacts inhibitory processes
endogenous ligand AEA blocks extracellular signal-regulated ki- in the dentate gyrus during adult neurogenesis (for review, see
nase (ERK e its activation is required for cell differentiation) Reid, Lysenko-Martin, Snowden, Thomas, & Christie, 2020).
through CB1 receptors, consequently regulating neural cell fate Experimental studies of the mechanisms of neuro-
(Rueda, Navarro, Martınez-Serrano, Guzma n, & Galve-Roperh, developmental effects of co-exposure to alcohol and marijuana
2002). The endogenous ligand 2-AG and the pharmacological suggest that these two substances share some effects on the same
activation of cannabinoid receptors increase progenitor cell pro- signaling pathways during early brain development, including the
liferation and differentiation (Aguado et al., 2005, 2006; Galve- sonic hedgehog signaling pathway. This pathway plays an impor-
Roperh et al., 2013; Jin et al., 2004). During axonal growth, 2-AG tant role in closing the neural tube (i.e., the induction of floor plate
is abundantly present in growth cones, and MAGL, the metabolic differentiation in the anterior midline) and establishing neural
enzyme that is responsible for degrading 2-AG, is at a low-level identity in the ventral part of the spinal cord and hindbrain (motor
concentration. MAGL is unable to degrade exogenous cannabi- neurons and interneurons; Ericson, Morton, Kawakami, Roelink, &
noids, such as THC, that engage cannabinoid receptors and induce Jessell, 1996). This signal transduction requires the Sonic hedgehog
abnormal signaling in stabilized axons (Calvigioni et al., 2014). (Shh) ligand to bind to Patched (Ptch) and to inhibit its inhibition on
In the adult central nervous system, the endocannabinoid sys- Smoothened (Smo). Smo is then free to translocate to the primary
tem acts as a retrograde neuromodulator messenger (Calvigioni cilium where it associates with Gai proteins. The inhibition of Gai
et al., 2014). The endogenous cannabinoids AEA and 2-AG are on adenyl cyclase (AC) impedes the conversion of adenosine
synthesized from the postsynaptic membrane on demand and bind triphosphate (ATP) into cyclic adenosine monophosphate (cAMP),
to cannabinoid receptors on presynaptic neuron in a retrograde leading to the inhibition of the accumulation of protein kinase A
manner (Basavarajappa et al., 2019). The activation of cannabinoid (PKA). This, in turn, helps maintain the Gli activator (Gli A) state,
receptors induces several signaling effects, such as activating preventing its proteolytic processing into Gli repressor forms (Gli
mitogen-activated protein kinase (MAPK), inhibiting adenyl R). Gene transcription needed for normal cell proliferation is pro-
cyclase, and altering ion channel activity (Bukiya, 2019). moted by PKA inhibition. Recent studies show that alcohol and
cannabis exert its actions through the Shh signaling pathway and
Simultaneous alcohol and cannabis exposure: neurodevelopmental may alter normal embryonic development (Boa-Amponsem et al.,
consequences and mechanisms 2019; Boa-Amponsem, Zhang, Burton, Williams, & Cole, 2020;
Fish et al., 2019). Both alcohol and cannabis alter this pathway in
The first studies that investigated the combined effects of distinct ways. Alcohol inhibits sonic hedgehog and maintains the
alcohol and cannabis use observed cross-tolerance between the Patched repression of Smo (Burton et al., 2022; Hong & Krauss,
́
two substances. Heavy marijuana users became less intoxicated 2012; Kahn et al., 2017). Cannabinoids have two mechanisms of
from alcohol consumption than non-marijuana users. They also action: one through the direct inhibition of Smo and one mecha-
exhibited fewer alcohol-induced neuropsychological deficiencies nism through CB1 receptor stimulation that leads to the formation
after co-administration (Jones & Stone, 1970). Participants who of heteromers with Smo (Amin, Ahmed, & Ali, 2022; Khaliullina,
simultaneously used both substances exhibited alterations of sleep Bilgin, Sampaio, Shevchenko, & Eaton, 2015). This latter mecha-
patterns (Zarcone, 1973), deficits in cognitive and psychomotor nism inhibits Smo-Gai protein signaling and stimulates CB1 re-
function (Manno, Kiplinger, Scholz, & Forney, 1971), and deficits in ceptor-Gas protein signaling (Boa-Amponsem et al., 2020; Fish
divided attention tasks (MacAvoy & Marks, 1975). The simulta- et al., 2019). The presumptive model of the mechanism of action
neous administration of both substances also appears to show after both alcohol and cannabinoid exposure is presented in Figs. 1
amplifying effects (Hansen et al., 2008). Human studies of com- and 2.
bined use often report that these individuals use alcohol and
marijuana simultaneously for their amplifying effects (Subbaraman First-trimester co-exposure to alcohol and cannabis
& Kerr, 2015; Terry-McElrath & Patrick, 2018). In the developing
brain, the precise spatial and temporal coordination of synaptic Co-exposure to cannabinoids and alcohol is linked to hol-
communication is essential for effective information processing oprosencephaly spectrum defects during early developmental
among excitatory pyramidal neurons, inhibitory interneurons, and stages. Even low doses of alcohol that are administered together
subcortical afferents (Berghuis et al., 2007). Alterations of these with cannabinoids have similar negative outcomes on the central
processes may cause long-lasting deficits in the developing brain. nervous system as high doses of alcohol exposure alone. In
5
cs, H. Charchat-Fichman, J. Landeira-Fernandez et al.
M.V. Kova Alcohol 110 (2023) 1e13

zebrafish and mouse embryos, the teratogenic effects of alcohol and phenotype was rescued. In the same study, the administration of
cannabinoids converge through the sonic hedgehog signaling subthreshold ethanol (0.5%) and JZL195 (inhibitor of endocanna-
pathway and induce ocular and facial dysmorphology (Fish et al., binoid degradative enzymes) induced FASD phenotype. The rescue
2019; Gilbert et al., 2016). Another study found that this neuro- of sonic hedgehog mRNA prevented microphthalmia after alcohol
degeneration was age-dependent in rat pups, with a peak at post- and ACEA treatment. Similar effects were observed in another
natal day 7 and disappearance by postnatal day 14 (Hansen et al., study that used the same paradigm, showing that alcohol and
2008). cannabinoids disrupted the signaling of sonic hedgehog and
Fish et al. (2019) found that cannabinoids and alcohol collabo- fibroblast growth factor and altered the development of GABAergic
ration during neurulation in zebrafish and mouse embryos is linked neurons in the forebrain. The simultaneous exposure to a sub-
to holoprosencephaly spectrum defects. Cannabinoid exposure threshold ACEA and alcohol modified behavior in zebrafish during
occurred through use of the synthetic cannabinoid HU-210 CP the late fry juvenile stage at approximately 2 months of age, and the
55,940 and phytocannabinoids CBD and THC. The least teratogenic neurobehavioral alterations were reversible after an injection of
cannabinoid was CBD, which is not a CB1 receptor agonist, but high- fgf8 mRNA (Boa-Amponsem et al., 2020). Alterations have also been
dose exposure caused serious eye defects. During neurulation found in the rat hippocampal formation after exposure to a com-
(gestational day 8 in mice), the eyes and brain develop from the bination of THC and alcohol vapor during gestational days 5e20. In
same neuroepithelium. Alcohol and cannabinoid exposure in this the dorsal CA1 region of the hippocampus, an increase in the
developmental period causes craniofacial alterations and abnor- number of parvalbumin interneurons was observed, with a
malities of brain development (Fig. 3). Small-eye phenotype was decrease in parvalbumin interneurons in the ventral CA1 region. No
observed after combined CP 55,940 and subthreshold ethanol difference in the ventral dentate gyrus was observed. Parvalbumin
exposure in zebrafish, an effect observed in mouse embryos. interneurons play a role in lateral inhibition, neurogenesis, and
Similar exposure consequences across species (mouse and zebra- network synchrony. Consequently, changes in parvalbumin inter-
fish) suggest the same underlying pathogenetic mechanisms of neuron density may influence adult neurogenesis, spatial working
alcohol and cannabinoids. Cannabinoids are dose-dependently memory, novel object exploration, and novel object location
teratogenic and enhance alcohol-induced deficits (Table 1). recognition (Reid et al., 2021).
Boa-Amponsem and co-workers (2019) reported the involve- The administration of lower concentrations of substances may
ment of the sonic hedgehog pathway by administering ethanol (1%) not induce the same effects as discussed above. Breit and co-
and low-dose CB1 receptor agonist arachidonoyl-20 -chlor- workers (Breit, Rodriguez, Lei, & Thomas, 2020) used a model of
oethylamide (ACEA), which induced dysmorphogenesis in zebra- co-exposure to alcohol and THC vapor to mimic e-cigarettes, a
fish embryos. Subthreshold ethanol (0.5%) combined with low-dose popular route of administration among pregnant women. Their
ACEA (3 mg/L) exposure induced microphthalmia and micro- study indicated that THC may alter alcohol metabolism, and co-
cephaly in 49% of the exposed animals and significantly increased exposure can increase blood alcohol levels. Alcohol also alters
risk-taking behavior in the novel tank diving test. However, in THC metabolism. Higher THC-OH metabolite levels were observed
the presence of CB1 receptor antagonist SR141716A, the FASD after combined drug exposure in rat dams. Interestingly, co-

Fig. 1. Simplified representation of altered embryonic signaling in the embryo after alcohol and cannabinoid exposure. Alcohol inhibits sonic hedgehog and maintains the
Patched repression of Smo, inhibiting the shh signaling. Cannabinoids have a direct inhibition of Smo, while stimulating CB1 receptors that leads to the formation of heteromers
with Smo. The latter mechanism reduces Smo-Gai protein signaling and stimulates CB1 receptor-Gas protein signaling. (Figure adapted from Fish et al., 2019).

6
cs, H. Charchat-Fichman, J. Landeira-Fernandez et al.
M.V. Kova Alcohol 110 (2023) 1e13

Fig. 2. Local signals in the telencephalon. Fibroblast Growth Factor (Fgf) and Sonic Hedgehog (Shh) (Figure adapted from Geng & Oliver, 2009; Petryk, Graf, & Marcucio, 2015)

exposure did not affect the gestational length, litter size, sex ratio, Second-trimester co-exposure
or birth weight, although the alcohol-exposed group exhibited a
delay in eye-opening. Prenatal THC exposure was associated with The development of the fetal brain vasculature system, and the
lower body weights during adolescence among offspring (Breit neural stem cell self-renewal and differentiation occur during the
et al., 2020). second trimester of gestation. Emerging evidence shows that

Fig. 3. Ocular and craniofacial morphology are altered in normal, alcohol-exposed, cannabinoid-exposed, and co-exposed mouse embryos. A) Craniofacial features of a)
normal, b) alcohol-exposed, c) cannabinoids-exposed, and d) combined alcohol- and cannabinoids-exposed mice. b) Alcohol-exposed mice: narrow eyelid openings and forehead,
small midface, short nose (nostril deficiency), hair follicles closer to the midline, long upper lip, philtrum deficiency. c) Cannabinoids-exposed mice: minor to moderate coloboma,
small jaws, philtrum deficiency. d) Combined alcohol and cannabinoids exposure: severe eye anomalies (anophthalmia), narrow forehead, small jaws, philtrum deficiency, small
nose (nostril deficiency). B) Growth features of drug exposures. e) normal weight and length of control mice. f) Alcohol-exposed mice: reduced weight and length. g) Cannabinoid-
exposed mice: reduced weight, no significant change in length. h) Combined alcohol and cannabinoid exposure: reduced body weight and length. (Figure adapted from Fish et al.,
2019).

7
cs, H. Charchat-Fichman, J. Landeira-Fernandez et al.
M.V. Kova Alcohol 110 (2023) 1e13

Table 1
Morphological and behavioral alterations after combined ethanol and cannabinoid exposure.

Measure Canabinoid Alcohol System Species Method of Exposure (time) Area /Brain Effects References
(CB) dose (Alc) dose analysis Region

Moderate 0.25 mg/kg 1.4 g/kg In vivo mouse dysmorphology GD 8 (beginning Eye/Face Anophthalmia and Fish et al.,
CP-55,940 þ Low assessment of neurulation) philtrum deficiency, cleft 2019
Alcohol palate, holoprosencephaly,
septal deficiency
Moderate 0.25 mg/kg 2.8 g/kg In vivo mouse dysmorphology GD 8 (beginning Eye/Face Severe eye defects, Fish et al.,
CP-55,940 þ High assessment of neurulation) philtrum deficiency, cleft 2019
Alcohol palate, holoprosencephaly
Low HU-210 þ Low 0.03 mg/kg 1.4 g/kg In vivo mouse dysmorphology GD 8 (beginning Eye/Face Near anophthalmia, Fish et al.,
Alcohol assessment of neurulation) philtrum and nostril 2019
deficiency, cleft palate,
brain structural changes,
decreased body weight
and length
Low THC þ Low 0.56 mg/kg 1.4 g/kg In vivo mouse dysmorphology GD 8 (beginning Eye/Face Increased eye defect Fish et al.,
Alcohol assessment of neurulation) incidence, decreased body 2019
weight and length
Low CP-55,940 1.0 mg/L 0.5% In vivo zebrafish dysmorphology 5.25e48 hpf Eye/Face No significant craniofacial Fish et al.,
þ Low Alcohol assessment (1e2 cell stage) or ocular alteration was 2019
observed after the
administration of this dose
combination
Low CP 55,940 2.5 mg/L 0.5% Ex vivo zebrafish Tissue content, 5.25e48 hpf Neural tube Midbrain/hindbrain Fish et al.,
þ Low Alcohol real-time (1-2 cell stage) defects, small eyes, 2019
quantitative PCR reduced Shh and
Gli1 gene expression
Moderately high 3.8 mg/L 0.5% Ex vivo zebrafish Tissue content, 5.25e48 hpf Neural tube Midbrain/hindbrain Fish et al.,
CP 55,940 þ Low real-time (1-2 cell stage) defects, small eyes, 2019
Alcohol quantitative PCR reduced Shh and
Gli1 gene expression
Low ACEA þ low 3 mg/L 0.5% In vivo zebrafish dysmorphology 6e24 hpf (chronic Eye/Face microphthalmia and Boa-
Alcohol assessment exposure) microcephaly in 49% of Amponsem
exposed zebrafish et al., 2019
Low ACEA þ high 3 mg/L 1% In vivo zebrafish dysmorphology 6e24 hpf (chronic Eye/Face microphthalmia and Boa-
Alcohol assessment exposure) microcephaly Amponsem
et al., 2019
Low ACEA þ low 3 mg/L 0.5% In vivo zebrafish dysmorphology 5.25e6.25 hpf Eye No significant ocular Boa-
Alcohol assessment (acute alteration was observed Amponsem
administration) et al., 2019
Low ACEA þ low 3 mg/L 0.5% In vivo zebrafish dysmorphology 8e10 hpf (acute Eye microphthalmia Boa-
Alcohol assessment administration) Amponsem
et al., 2019
Low ACEA þ low 3 mg/L 0.5% In vivo zebrafish dysmorphology 24e27 hpf (acute Eye No significant ocular Boa-
Alcohol assessment administration) alteration was observed Amponsem
et al., 2019
Low ACEA þ high 3 mg/L 1% In vivo zebrafish dysmorphology 5.25e6.25 hpf Eye microphthalmia Boa-
Alcohol assessment (first hour of Amponsem
gastrulation) et al., 2019
Low ACEA þ high 3 mg/L 1% In vivo zebrafish dysmorphology 8e10 hpf Eye microphthalmia Boa-
Alcohol assessment (transition Amponsem
gastrulation- et al., 2019
neurulation)
Low ACEA þ high 3 mg/L 1% In vivo zebrafish dysmorphology 24e27 hpf Eye microphthalmia Boa-
Alcohol assessment (formation of Amponsem
five-vesicle brain) et al., 2019
Low JZL195 þ low 2.5 mg/L 0.5% In vivo zebrafish dysmorphology 6e24 hpf (chronic Eye microphthalmia Boa-
Alcohol assessment exposure) Amponsem
et al., 2019
Low ACEA þ Low 1 mg/L 0.5% In vivo zebrafish novel tank 5.25e6.25 hpf behavior Increased risk-taking Boa-
Alcohol diving test behavior Amponsem
et al., 2019
Low ACEA þ Low 1 mg/L 0.5% In vivo zebrafish novel tank 8e10 hpf behavior Increased risk-taking Boa-
Alcohol diving test behavior Amponsem
et al., 2019
Low ACEA þ Low 1 mg/L 0.5% In vivo zebrafish novel tank 24e27 hpf behavior Increased risk-taking Boa-
Alcohol diving test behavior Amponsem
et al., 2019
Low ACEA þ Low 3 mg/L 0.5% In vivo zebrafish novel tank 5.25e6.25 hpf behavior Increased anxiety-like Boa-
Alcohol diving test behavior, but no alteration Amponsem
in tank diving response et al., 2019
Low ACEA þ High 3 mg/L 1% In vivo zebrafish Eye 8e10 hpf eye Small eye phenotype Boa-
Alcohol dysmorphology Amponsem
assessment et al., 2020

8
cs, H. Charchat-Fichman, J. Landeira-Fernandez et al.
M.V. Kova Alcohol 110 (2023) 1e13

Table 1 (continued )

Measure Canabinoid Alcohol System Species Method of Exposure (time) Area /Brain Effects References
(CB) dose (Alc) dose analysis Region

Low ACEA þ High 3 mg/L 1% In vivo zebrafish Eye 24e27 hpf eye Small eye phenotype Boa-
Alcohol dysmorphology Amponsem
assessment et al., 2020
Low ACEA þ Low 3 mg/L 0.5% In vivo zebrafish Eye 8e10 hpf eye Did not produce Boa-
Alcohol dysmorphology microphthalmia Amponsem
assessment et al., 2020
Low ACEA þ Low 3 mg/L 0.5% In vivo zebrafish Eye 24e27 hpf eye Did not produce Boa-
Alcohol dysmorphology microphthalmia Amponsem
assessment et al., 2020
Low ACEA þ Low 1 mg/L 0.5% In vivo zebrafish novel tank 8e10 hpf behavior Increased risk-taking Boa-
Alcohol diving test behavior Amponsem
et al., 2020
Low ACEA þ Low 1 mg/L 0.5% In vivo zebrafish novel tank 24e27 hpf (late behavior Increased risk-taking Boa-
Alcohol diving test neurulation) behavior Amponsem
et al., 2020
Third-trimester equivalent
High CP-55,940 0.4 mg/kg/ 5.25 g/kg/ In vivo Sprague- Early motor PD (postnatal day) Behavior Initial motor development Breit et al.,
þ High Alcohol day day Dawley development 4e9, (chronic (motor delay, severe motor 2019
rat task: grip administration) development coordination deficit
strength trial and and (particularly among
Parallel bar motor coordination) females).
coordination No effect on grip strength
trial

Abbreviations: PD: postnatal day; BAC: Blood Alcohol Concentration; hpf: hours post fertilization.

simultaneous alcohol and cannabis (CP-55940, synthetic cannabi- combined exposure enhanced the ethanol-related motor deficits in
noid) exposure may induce premature neural stem cell growth, some tasks in female subjects, but in other tasks, the initial delay
alter cell proliferation, and has been associated with acute and was reduced over time and these rats showed performance similar
delayed decrease in fetal-directed blood flow (Rouzer, Craig, to the control group. CP exposure increased blood alcohol con-
Mavuri, Sreeram, & Miranda, 2022). centration (BAC) levels in the co-exposed group, particularly in
female subjects, which may also explain the high mortality among
Third-trimester co-exposure co-exposed subjects (Breit et al., 2019). In humans, co-exposure
effects on structural connectivity of the developing central ner-
During the third trimester of gestation, the central nervous vous system were assessed during adolescence, using the diffusion
system undergoes rapid growth (brain growth spurt) by making tensor imaging technique (DTI) (Wade et al., 2020). White matter
and breaking synaptic connections, which lasts until late adoles- integrity relies on healthy oligodendrocyte development, and CB1
cence. Exposure to drugs during this period may interrupt neuronal receptor activation protects progenitors from programmed cell
plasticity and alter the formation and refinement of neural circuits death (apoptosis). Alcohol and cannabis use alters CB1 activity,
(Medina, 2011). Developmental studies of mouse embryos reported disrupting healthy oligodendrocyte development. The results show
that alcohol increases AEA levels in postsynaptic neurons through that co-use of both substances may lead to worse white matter
transcriptional activation of the NAPE-PLD and glycer- integrity in three tracts, including the inferior longitudinal fascic-
ophosphodiester phosphodiesterase 1 (GDE1) enzymes. High levels ulus, anterior thalamic radiation, and cingulum cingulate gyrus
of AEA decrease glutamate release through CB1 receptors on pre- (Wade et al., 2020). Moreover, Hansen et al. (2008) observed that
synaptic neurons. The decrease in glutamate release results in the co-administration of a mildly intoxicating dose of alcohol
NMDA receptor hypofunction and cyclin-dependent kinase 5 (0.5e1.8 g/kg, 1e3.6 g/kg total dose) and THC (1e10 mg/kg) resulted
(CDK5), ERK 1/2, and cyclic adenosine monophosphate response in massive neurodegeneration in rat neonates e an effect similar to
element binding protein (CREB) hyperphosphorylation, which that observed after exposure to a high dose of alcohol alone, sug-
causes the inhibition of Arc and Rac1 expression (Fig. 4). These gesting an amplification of alcohol's effects by THC. This neuro-
events disturb neuronal circuit refinement and cause lasting defi- degeneration was age-dependent, with a maximum level on
cits in synaptic plasticity (Basavarajappa et al., 2019). postnatal day 7, but this effect was absent on postnatal day 14
Combined alcohol and cannabis-exposed rats during postnatal (Hansen et al., 2008). The observed neurodegenerative effect was
days 4e9 (the period of brain growth spurt e human 3rd-trimester reversible by administration of the CB1 receptor antagonist
equivalent) showed altered motor function in some tasks at later SR141716A.
stages of development (Breit, Zamudio, & Thomas, 2019). Early
motor development was assessed using a grip strength and hin- Potential therapeutic implications
dlimb coordination task on postnatal days 12e20, and a parallel bar
motor coordination paradigm was applied to examine motor co- Preclinical studies indicate that a CB1 receptor antagonist,
ordination during adolescence (postnatal days 30e32). Cannabi- SR141716A, may have a protective effect against the effects of
noid effect was observed using CP-55,940 (CP), a CB1 and CB2 combined exposure to alcohol and cannabinoids during develop-
receptor agonist frequently used in synthetic marijuana prepara- ment such as behavioral deficits (Boa-Amponsem et al., 2019),
tions as it mimics THC effects. The results of the experiment morphological phenotypes (Fish et al., 2019), and neuro-
showed that combined exposure decreased body weight even more degeneration (Hansen et al., 2008). Shh mRNA overexpression has a
than in rats treated only with ethanol. Interestingly, only the similar effect, as it rescues normal juvenile behavior (Boa-
ethanol-exposed rats showed delayed motor development, while Amponsem et al., 2019), blocks development of microphthalmia
only CP-exposed rats showed advanced motor development. The phenotype, and prevents abnormal midbrain/hindbrain border
9
cs, H. Charchat-Fichman, J. Landeira-Fernandez et al.
M.V. Kova Alcohol 110 (2023) 1e13

Fig. 4. Developmental exposure to alcohol and cannabinoids leads to neurobehavioral defects. a) Alcohol activates gene transcription of GDE1 and NAPE-PLD enzymes; b)
increased AEA levels; c) and d) AEA and cannabinoids act through CB1R at pre-synaptic cell membrane; e) enhanced level of CB1R mRNA expression; f) CB1-mediated Ca2þ channel
inhibition; g) decreased glutamate release; h) NMDAR hypofunction; i) CDK/pERK signaling deficit; j) decreased CREB phosphorylation; k) inhibition of Arc, Rac expression.
(Figure adapted from Basavarajappa et al., 2019).

formation (Fish et al., 2019). A more recent study in zebrafish available. For instance, there are methylphenidate, neuroleptics
revealed that fibroblast growth factor fgf8 mRNA overexpression and/or mood stabilizers, stimulants, and choline supplementation
mitigated the behavioral deficits caused by exposure to alcohol and to name a few (for a comprehensive review, see Ritfeld, Kable,
cannabinoids (Boa-Amponsem et al., 2020). These findings may Holton, & Coles, 2022). However, clinical studies use small sam-
help to elucidate neuronal mechanisms underlying the effects of ple sizes and show mixed results while describing several side ef-
early exposure to alcohol and cannabinoids, but could also help to fects. As alcohol disrupts the formation of main neurotransmitter
delineate novel therapeutical strategies to prevent long-term systems, some of the above-mentioned drugs may have the
developmental deficits. opposite effect compared to the general population. It is important
Human studies concerning the effects of developmental co- to highlight that no guidelines exist about the optimal psycho-
exposure later in life are scarce. However, data from the study of pharmacological treatments for the FASD population (Ritfeld et al.,
Wade et al. (2020) show that combined exposure to alcohol and 2022). Perhaps, some of these strategies may be effective in cases of
cannabis during adolescence leads to lower white matter integrity developmental co-exposure. Future studies are needed to shed
across frontolimbic and frontoparietal tracts (Wade et al., 2020). It more light on this issue.
is suggested that the two substances may disrupt normal myeli-
nation e through activation of CB1 receptors e and lead to Conclusions
abnormal white matter formation. Disrupted frontolimbic tracts
have been found in individuals suffering from mood disorders Substance use during pregnancy is associated with alterations in
(Hermens et al., 2018; Shollenbarger, Price, Wieser, & Lisdahl, neurodevelopment and long-lasting deficits in offspring. The most
2015). Developmental alcohol exposure in ferrets resulted in used substances by pregnant women are alcohol and cannabis
increased functional connectivity between the caudal and rostral (Substance Abuse and Mental Health Services Administration,
portions of the posterior parietal cortex e areas that play a role on 2020), which are frequently consumed together. Accumulating
multisensory integration (MSI) (Tang, Xu, Gullapalli, & Medina, evidence shows that alcohol and cannabis, when administered
2018). More recently, Keum and colleagues (Keum, Pultorak, together, synergistically alter neurodevelopment from the earliest
Meredith, & Medina, 2023) demonstrated that developmental stages of gestation (Boa-Amponsem et al., 2019, 2020; Breit et al.,
alcohol exposure disrupts MSI in the ferret. Thus, altered white 2020; Fish et al., 2019; Reid et al., 2021). Recent data suggest the
matter integrity should be taken into consideration during neuro- involvement of the sonic hedgehog signaling pathway in in-
psychological evaluation and/or special educational care should be teractions between the effects of alcohol and cannabinoids,
provided for individuals with a developmental history of co- resulting in growth deficits, craniofacial and ocular dysmorphology
exposure to alcohol and cannabis. Yet, several treatments for (Fish et al., 2019), alterations of hippocampal areas that are asso-
developmental disorders arising from prenatal use of alcohol are ciated with learning and memory (Reid et al., 2021), and increases
10
cs, H. Charchat-Fichman, J. Landeira-Fernandez et al.
M.V. Kova Alcohol 110 (2023) 1e13

in risk-taking behavior and anxiety-like behavior in animal models Basavarajappa, B. S., & Subbanna, S. (2016). Epigenetic mechanisms in develop-
mental alcohol-induced neurobehavioral deficits. Brain Sciences, 6(2), 12.
(Boa-Amponsem et al., 2019, 2020). Future studies are necessary to
Begbie, J., Doherty, P., & Graham, A. (2004). Cannabinoid receptor, CB1, expression
understand the effects of simultaneous cannabis and alcohol use in follows neuronal differentiation in the early chick embryo. Journal of Anatomy,
humans by comparing alcohol-only, alcohol þ THC, and THC-only 205(3), 213e218.
groups and the effects of CBD. Ben-Ari, Y. (2002). Excitatory actions of gaba during development: The nature of the
nurture. Nature reviews. Nature Reviews Neuroscience, 3(9), 728e739.
Berghuis, P., Rajnicek, A. M., Morozov, Y. M., Ross, R. A., Mulder, J., Urb an, G. M., et al.
Author contributions (2007). Hardwiring the brain: Endocannabinoids shape neuronal connectivity.
Science, 316(5828), 1212e1216.
Boa-Amponsem, O., Zhang, C., Burton, D., Williams, K. P., & Cole, G. J. (2020). Ethanol
AEM conceived, designed, and supervised the study. MVK and cannabinoids regulate zebrafish GABAergic neuron development and
collected and analyzed data, and wrote the manuscript. TEK su- behavior in a Sonic Hedgehog and fibroblast growth factor-dependent mech-
anism. Alcoholism: Clinical and Experimental Research, 44(7), 1366e1377.
pervised MVK on the preparation and writing of the manuscript,
Boa-Amponsem, O., Zhang, C., Mukhopadhyay, S., Ardrey, I., & Cole, G. J. (2019).
and made the figures of the manuscript. JLF assisted with the design Ethanol and cannabinoids interact to alter behavior in a zebrafish fetal alcohol
of the table of the manuscript. JLF, AEM, HCF, and TEK revised and spectrum disorder model. Birth Defects Research, 111(12), 775e788.
Boschen, K. E., Ptacek, T. S., Berginski, M. E., Simon, J. M., & Parnell, S. E. (2021).
edited the text of the manuscript. All authors revised and approved
Transcriptomic analyses of gastrulation-stage mouse embryos with differential
the final version of the manuscript. susceptibility to alcohol. Disease Models & Mechanisms, 14(6), Article
dmm049012.
Breit, K. R., Rodriguez, C. G., Lei, A., & Thomas, J. D. (2020). Combined vapor expo-
Funding sure to THC and alcohol in pregnant rats: Maternal outcomes and pharmaco-
kinetic effects. Neurotoxicology and Teratology, 82, Article 106930.
This work was supported by grants from Conselho Nacional de Breit, K. R., Zamudio, B., & Thomas, J. D. (2019). Altered motor development
following late gestational alcohol and cannabinoid exposure in rats. Neuro-
Desenvolvimento Científico e Tecnolo gico (CNPq), Fundaça
~o de
toxicology and Teratology, 73, 31e41.
Amparo a  Pesquisa do Estado do Rio de Janeiro (FAPERJ) and Buckley, N., Hansson, S., Harta, G., & Mezey, E. (1998). Expression of the CB1 and CB2
Coordenaça~o de Aperfeiçoamento de Pessoal de Nível Superior receptor messenger RNAs during embryonic development in the rat. Neuro-
science, 82(4), 1131e1149.
(Capes). AEM is supported by NIH National Institute on Alcohol
Bukiya, A. N. (2019). Physiology of the endocannabinoid system during develop-
Abuse and Alcoholism (NIAAA) Grant R01AA13023. ment. In A. N. Bukiya (Ed.), Recent advances in cannabinoid physiology and pa-
thology (pp. 13e37). Berlin: Springer.
Burton, D. F., Boa-Amponsem, O. M., Dixon, M. S., Hopkins, M. J., Herbin, T. A.,
Declaration of competing interest Toney, S., et al. (2022). Pharmacological activation of the Sonic hedgehog
pathway with a Smoothened small molecule agonist ameliorates the severity of
The authors declare no competing interests. alcohol-induced morphological and behavioral birth defects in a zebrafish
model of fetal alcohol spectrum disorder. Journal of Neuroscience Research,
100(8), 1585e1601.
Acknowledgments Calvigioni, D., Hurd, Y. L., Harkany, T., & Keimpema, E. (2014). Neuronal substrates
and functional consequences of prenatal cannabis exposure. European Child &
Adolescent Psychiatry, 23(10), 931e941.
We are thankful to Bianca Janssens and Rafael Carnavale for Carty, D. R., Thornton, C., Gledhill, J. H., & Willett, K. L. (2018). Developmental effects
their assistance in data collection and data curation. of cannabidiol and D9-tetrahydrocannabinol in zebrafish. Toxicological Sciences,
162(1), 137e145.
Center for Behavioral Health Statistics and Quality. (2015). Behavioral health trends
References in the United States: Results from the 2014 national survey on drug use and health
(HHS Publication no. SMA 15-4927, NSDUH Series H-50). Rockville: Substance
Abel, E., & Sokol, R. (1986). Fetal alcohol syndrome is now leading cause of mental Abuse and Mental Health Services Administration. Retrieved from https://
retardation. Lancet, 328(8517), 1222. www.samhsa.gov/data/sites/default/files/NSDUH-FRR1-2014/NSDUH-FRR1-
Aguado, T., Monory, K., Palazuelos, J., Stella, N., Cravatt, B., Lutz, B., et al. (2005). The 2014.pdf.
endocannabinoid system drives neural progenitor proliferation. Federation of Curran, H. V., Freeman, T. P., Mokrysz, C., Lewis, D. A., Morgan, C. J. A., &
American Societies for Experimental Biology Journal, 19(12), 1704e1706. Parsons, L. H. (2016). Keep off the grass? Cannabis, cognition and addiction.
Aguado, T., Palazuelos, J., Monory, K., Stella, N., Cravatt, B., Lutz, B., et al. (2006). The Nature Reviews Neuroscience, 17(5), 293e306.
endocannabinoid system promotes astroglial differentiation by acting on neural Darbinian, N., Darbinyan, A., Merabova, N., Bajwa, A., Tatevosian, G., Martirosyan, D.,
progenitor cells. Journal of Neuroscience, 26(5), 1551e1561. et al. (2021). Ethanol-mediated alterations in oligodendrocyte differentiation in
Allan, A. M., & Harris, R. A. (1987). Involvement of neuronal chloride channels in the developing brain. Neurobiology of Disease, 148, Article 105181.
ethanol intoxication, tolerance, and dependence. Recent Developments in Alco- , M., Bisogno, T., Petrosino, S., et al.
De Petrocellis, L., Ligresti, A., Moriello, A. S., Allara
holism, 5, 313e325. (2011). Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on
Alpar, A., Di Marzo, V., & Harkany, T. (2016). At the tip of an iceberg: Prenatal TRP channels and endocannabinoid metabolic enzymes. British Journal of
marijuana and its possible relation to neuropsychiatric outcome in the Pharmacology, 163(7), 1479e1494.
offspring. Biological Psychiatry, 79(7), e33ee45. Delatour, L. C., Yeh, P. W., & Yeh, H. H. (2019). Ethanol exposure in utero disrupts
Amin, M. R., Ahmed, K. T., & Ali, D. W. (2022). Cannabinoid receptor 2 (Cb2r) me- radial migration and pyramidal cell development in the somatosensory cortex.
diates cannabinol (CBN) induced developmental defects in zebrafish. Scientific Cerebral Cortex, 29(5), 2125e2139.
Reports, 12(1), Article 20251. Dickson, B., Mansfield, C., Guiahi, M., Allshouse, A. A., Borgelt, L. M., Sheeder, J., et al.
Augustin, S. M., & Lovinger, D. M. (2022). Synaptic changes induced by cannabinoid (2018). Recommendations from cannabis dispensaries about first-trimester
drugs and cannabis use disorder. Neurobiology of Disease, 167, Article 105670. cannabis use. Obstetrics & Gynecology, 131(6), 1031e1038.
Baculis, B. C., Diaz, M. R., & Valenzuela, C. F. (2015). Third trimester-equivalent Egan, K. L., Cox, M. J., Suerken, C. K., Reboussin, B. A., Song, E. Y., Wagoner, K. G., et al.
ethanol exposure increases anxiety-like behavior and glutamatergic trans- (2019). More drugs, more problems? Simultaneous use of alcohol and mari-
mission in the basolateral amygdala. Pharmacology Biochemistry and Behavior, juana at parties among youth and young adults. Drug and Alcohol Dependence,
137, 78e85. 202, 69e75.
Bake, S., Hurst, D. A., Miranda, R. C., & Sohrabji, F. (2022). Prenatal alcohol exposure El Marroun, H., Brown, Q. L., Lund, I. O., Coleman-Cowger, V. H., Loree, A. M.,
exacerbates acute sensorimotor deficits and impedes long-term behavioral Chawla, D., et al. (2018). An epidemiological, developmental and clinical over-
recovery from the effects of an adult-onset cerebrovascular ischemic stroke. view of cannabis use during pregnancy. Preventive Medicine, 116, 1e5.
Alcoholism: Clinical and Experimental Research, 46(12), 2267e2279. El Marroun, H., Tiemeier, H., Steegers, E. A. P., Jaddoe, V. W. V., Hofman, A.,
Basavarajappa, B. S., Cooper, T. B., & Hungund, B. L. (1998). Effect of chronic ethanol Verhulst, F. C., et al. (2009). Intrauterine cannabis exposure affects fetal growth
exposure on mouse brain arachidonic acid specific phospholipase A2. trajectories: The generation R study. Journal of the American Academy of Child &
Biochemical Pharmacology, 55(4), 515e521. Adolescent Psychiatry, 48(12), 1173e1181.
Basavarajappa, B. S., Joshi, V., Shivakumar, M., & Subbanna, S. (2019). Distinct El Marroun, H., Tiemeier, H., Steegers, E. A. P., Roos-Hesselink, J. W., Jaddoe, V. W. V.,
functions of endogenous cannabinoid system in alcohol abuse disorders. British Hofman, A., et al. (2010). A prospective study on intrauterine cannabis exposure
Journal of Pharmacology, 176(17), 3085e3109. and fetal blood flow. Early Human Development, 86(4), 231e236.
Basavarajappa, B. S., Saito, M., Cooper, T. B., & Hungund, B. L. (1997). Activation of Ellingson, J. M., Ross, J. M., Winiger, E., Stalli1ngs, M. C., Corley, R. P., Friedman, N. P.,
arachidonic acid-specific phospholipase A2 in human neuroblastoma cells after et al. (2021). Familial factors may not explain the effect of moderate-to-heavy
chronic alcohol exposure: Prevention by GM1 ganglioside. Alcoholism: Clinical cannabis use on cognitive functioning in adolescents: A sibling-comparison
and Experimental Research, 21(7), 1199e1203. study. Addiction, 116(4), 833e844.

11
cs, H. Charchat-Fichman, J. Landeira-Fernandez et al.
M.V. Kova Alcohol 110 (2023) 1e13

ElSohly, M. A., Mehmedic, Z., Foster, S., Gon, C., Chandra, S., & Church, J. C. (2016). fetal alcohol spectrum disorders. Alcoholism: Clinical and Experimental Research,
Changes in cannabis potency over the last 2 decades (1995e2014): Analysis of 34(5), 928e937.
current data in the United States. Biological Psychiatry, 79(7), 613e619. Lange, S., Probst, C., Gmel, G., Rehm, J., Burd, L., & Popova, S. (2017). Global preva-
Ericson, J., Morton, S., Kawakami, A., Roelink, H., & Jessell, T. M. (1996). Two critical lence of fetal alcohol spectrum disorder among children and youth: A sys-
periods of Sonic Hedgehog signaling required for the specification of motor tematic review and meta-analysis. JAMA Pediatrics, 171(10), 948e956.
neuron identity. Cell, 87(4), 661e673. Lauckner, J. E., Jensen, J. B., Chen, H. Y., Lu, H. C., Hille, B., & Mackie, K. (2008). GPR55
Felder, C. C., Joyce, K. E., Briley, E. M., Mansouri, J., Mackie, K., Blond, O., et al. (1995). is a cannabinoid receptor that increases intracellular calcium and inhibits M
Comparison of the pharmacology and signal transduction of the human current. Proceedings of the National Academy of Sciences of the United States of
cannabinoid CB1 and CB2 receptors. Molecular Pharmacology, 48(3), 443e450. America, 105(7), 2699e2704.
Filgueiras, C. C., Krahe, T. E., & Medina, A. E. (2010). Phosphodiesterase type 1 in- Luo, J., & Miller, M. (1998). Growth factor-mediated neural proliferation: Target of
hibition improves learning in rats exposed to alcohol during the third trimester ethanol toxicity. Brain Research. Brain Research Reviews, 27(2), 157e167.
equivalent of human gestation. Neuroscience Letters, 473(3), 202e207. MacAvoy, M. G., & Marks, D. F. (1975). Divided attention performance of cannabis
Fish, E. W., Mendoza-Romero, H. N., Love, C. A., Dragicevich, C. J., Cannizzo, M. D., users and non-users following cannabis and alcohol. Psychopharmacologia,
Boschen, K. E., et al. (2022). The pro-apoptotic Bax gene modifies susceptibility 44(2), 147e152.
to craniofacial dysmorphology following gastrulation-stage alcohol exposure. Mahajan, V. S., Demissie, E., Mattoo, H., Viswanadham, V., Varki, A., Morris, R., et al.
Birth Defects Research, 114(19), 1229e1243. (2016). Striking immune phenotypes in gene-targeted mice are driven by a
Fish, E. W., Murdaugh, L. B., Zhang, C., Boschen, K. E., Boa-Amponsem, O., Mendoza- copy-number variant originating from a commercially available C57BL/6 strain.
Romero, H. N., et al. (2019). Cannabinoids exacerbate alcohol teratogenesis by a Cell Reports, 15(9), 1901e1909.
CB1-Hedgehog interaction. Scientific Reports, 9(1), Article 16057. Manno, J. E., Kiplinger, G. F., Scholz, N., & Forney, R. B. (1971). The influence of
Fish, E. W., Tucker, S. K., Peterson, R. L., Eberhart, J. K., & Parnell, S. E. (2021). Loss of alcohol and marihuana on motor and mental performance. Clinical Pharma-
tumor protein 53 protects against alcohol-induced facial malformations in mice cology & Therapeutics, 12(2), 202e211.
and zebrafish. Alcoholism: Clinical and Experimental Research, 45(10), Mato, S., Del Olmo, E., & Pazos, A. (2003). Ontogenetic development of cannabinoid
1965e1979. receptor expression and signal transduction functionality in the human brain.
Galve-Roperh, I., Chiurchiù, V., Díaz-Alonso, J., Bari, M., Guzm an, M., & European Journal of Neuroscience, 17(9), 1747e1754.
Maccarrone, M. (2013). Cannabinoid receptor signaling in progenitor/stem cell Mattson, S. N., Bernes, G. A., & Doyle, L. R. (2019). Fetal alcohol spectrum disorders:
proliferation and differentiation. Progress in Lipid Research, 52(4), 633e650. A review of the neurobehavioral deficits associated with prenatal alcohol
Geng, X., & Oliver, G. (2009). Pathogenesis of holoprosencephaly. The Journal of exposure. Alcoholism: Clinical and Experimental Research, 43(6), 1046e1062.
Clinical Investigation, 119(6), 1403e1413. May, P. A., Hasken, J. M., Stegall, J. M., Mastro, H. A., Kalberg, W. O., Buckley, D., et al.
Gilbert, M. T., Sulik, K. K., Fish, E. W., Baker, L. K., Dehart, D. B., & Parnell, S. E. (2016). (2020). Fetal alcohol spectrum disorders in a southeastern county of the United
Dose-dependent teratogenicity of the synthetic cannabinoid CP-55,940 in mice. States: Child characteristics and maternal risk traits. Alcoholism: Clinical and
Neurotoxicology and Teratology, 58, 15e22. Experimental Research, 44(4), 939e959.
Glass, M., & Felder, C. C. (1997). Concurrent stimulation of cannabinoid CB1 and Medina, A. E. (2011). Fetal alcohol spectrum disorders and abnormal neuronal
dopamine D2 receptors augments cAMP accumulation in striatal neurons: Ev- plasticity. The Neuroscientist, 17(3), 274e287.
idence for a gs linkage to the CB1 receptor. Journal of Neuroscience, 17(14), Musselman, M. E., & Hampton, J. P. (2014). “Not for human consumption”: A review
5327e5333. of emerging designer drugs. Pharmacotherapy: The Journal of Human Pharma-
Granato, A., & Dering, B. (2018). Alcohol and the developing brain: Why neurons die cology and Drug Therapy, 34(7), 745e757.
and how survivors change. International Journal of Molecular Sciences, 19(10), Newville, J., Valenzuela, C. F., Li, L., Jantzie, L. L., & Cunningham, L. A. (2017). Acute
2992. oligodendrocyte loss with persistent white matter injury in a third trimester
Gunn, R. L., Norris, A. L., Sokolovsky, A., Micalizzi, L., Merrill, J. E., & Barnett, N. P. equivalent mouse model of fetal alcohol spectrum disorder. Glia, 65(8),
(2018). Marijuana use is associated with alcohol use and consequences across 1317e1332.
the first 2 years of college. Psychology of Addictive Behaviors, 32(8), 885e894. Petryk, A., Graf, D., & Marcucio, R. (2015). Holoprosencephaly: Signaling interactions
Hamburger, V., & Hamilton, H. L. (1951). A series of normal stages in the develop- between the brain and the face, the environment and the genes, and the
ment of the chick embryo, 1992 Developmental Dynamics, 195(4), 231e272. phenotypic variability in animal models and humans. Wiley Interdisciplinary
Hansen, H. H., Krutz, B., Sifringer, M., Stefovska, V., Bittigau, P., Pragst, F., et al. Reviews. Developmental Biology, 4(1), 17e32.
(2008). Cannabinoids enhance susceptibility of immature brain to ethanol Popova, S., Lange, S., Shield, K., Burd, L., & Rehm, J. (2019). Prevalence of fetal alcohol
neurotoxicity. Annals of Neurology, 64(1), 42e52. spectrum disorder among special subpopulations: A systematic review and
Hausknecht, K., Shen, Y. L., Wang, R. X., Haj-Dahmane, S., & Shen, R. Y. (2017). meta-analysis. Addiction, 114(7), 1150e1172.
Prenatal ethanol exposure persistently alters endocannabinoid signaling and Psychoyos, D., Hungund, B., Cooper, T., & Finnell, R. H. (2008). A cannabinoid
endocannabinoid-mediated excitatory synaptic plasticity in ventral tegmental analogue of Delta9-tetrahydrocannabinol disrupts neural development in chick.
area dopamine neurons. Journal of Neuroscience, 37(24), 5798e5808. Birth Defects Research Part B: Developmental and Reproductive Toxicology, 83(5),
Hermens, D. F., Hatton, S. N., Lee, R. S. C., Naismith, S. L., Duffy, S. L., Paul 477e488.
Amminger, G., et al. (2018). In vivo imaging of oxidative stress and fronto-limbic Raghunathan, R., Liu, C., Kouka, A., Singh, M., Miranda, R. C., & Larin, K. V. (2019).
white matter integrity in young adults with mood disorders. European Archives Assessing the acute effects of prenatal synthetic cannabinoid exposure on
of Psychiatry and Clinical Neuroscience, 268(2), 145e156. murine fetal brain vasculature using optical coherence tomography. Journal of
Hong, M., & Krauss, R. S. (2012). Cdon mutation and fetal ethanol exposure syner- Biophotonics, 12(8), Article e201900050.
gize to produce midline signaling defects and holoprosencephaly spectrum Reid, H. M. O., Lysenko-Martin, M. R., Snowden, T. M., Thomas, J. D., & Christie, B. R.
disorders in mice. PLoS Genetics, 8(10), Article e1002999. (2020). A systematic review of the effects of perinatal alcohol exposure and
Jin, K., Xie, L., Kim, S. H., Parmentier-Batteur, S., Sun, Y., Mao, X. O., et al. (2004). perinatal marijuana exposure on adult neurogenesis in the dentate gyrus.
Defective adult neurogenesis in CB1 cannabinoid receptor knockout mice. Mo- Alcoholism: Clinical and Experimental Research, 44(6), 1164e1174.
lecular Pharmacology, 66(2), 204e208. Reid, H. M. O., Snowden, T. M., Shkolnikov, I., Breit, K. R., Rodriguez, C., Thomas, J. D.,
Jones, R. T., & Stone, G. C. (1970). Psychological studies of marijuana and alcohol in et al. (2021). Prenatal alcohol and cannabis exposure can have opposing and
man. Psychopharmacologia, 18(1), 108e117. region-specific effects on parvalbumin interneuron numbers in the hippo-
Jordt, S. E., Bautista, D. M., Chuang, H. H., McKemy, D. D., Zygmunt, P. M., campus. Alcoholism: Clinical and Experimental Research, 45(11), 2246e2255.
Ho€ gesta
€tt, E. D., et al. (2004). Mustard oils and cannabinoids excite sensory Rhee, M. H., Vogel, Z., Barg, J., Bayewitch, M., Levy, R., Hanus, L., et al. (1997).
nerve fibres through the TRP channel ANKTM1. Nature, 427(6971), 260e265. Cannabinol derivatives: Binding to cannabinoid receptors and inhibition of
Kahn, B. M., Corman, T. S., Lovelace, K., Hong, M., Krauss, R. S., & Epstein, D. J. (2017). adenylylcyclase. Journal of Medicinal Chemistry, 40(20), 3228e3233.
Prenatal ethanol exposure in mice phenocopies Cdon mutation by impeding Richardson, K. A., Hester, A. K., & McLemore, G. L. (2016). Prenatal cannabis expo-
Shh function in the etiology of optic nerve hypoplasia. Disease Models & sure e the “first hit” to the endocannabinoid system. Neurotoxicology and
Mechanisms, 10(1), 29e37. Teratology, 58, 5e14.
Kathmann, M., Flau, K., Redmer, A., Tra €nkle, C., & Schlicker, E. (2006). Cannabidiol is Riley, E. P., & McGee, C. L. (2005). Fetal alcohol spectrum disorders: An overview
an allosteric modulator at mu- and delta-opioid receptors. Naunyn-Schmiede- with emphasis on changes in brain and behavior. Experimental Biology and
berg's Archives of Pharmacology, 372(5), 354e361. Medicine, 230(6), 357e365.
Keum, D., Pultorak, K., Meredith, M. A., & Medina, A. E. (2023). Effects of develop- Ritfeld, G. J., Kable, J. A., Holton, J. E., & Coles, C. D. (2022). Psychopharmacological
mental alcohol exposure on cortical multisensory integration. European Journal treatments in children with fetal alcohol spectrum disorders: A review. Child
of Neuroscience, 57(5), 784e795. Psychiatry and Human Development, 53(2), 268e277.
Khaliullina, H., Bilgin, M., Sampaio, J. L., Shevchenko, A., & Eaton, S. (2015). Endo- Rouzer, S. K., Craig, L., Mavuri, S., Sreeram, A., & Miranda, R. C. (2022). Simultaneous
cannabinoids are conserved inhibitors of the Hedgehog pathway. Proceedings of prenatal alcohol and cannabinoid exposure during the second trimester inhibits
the National Academy of Sciences of the United States of America, 112(11), neural stem cell growth and fetal directed blood flow. Alcoholism: Clinical and
3415e3420. Experimental Research, 46(S1), 14e69. https://doi.org/10.1111/acer.14831.
Ko, J. Y., Farr, S. L., Tong, V. T., Creanga, A. A., & Callaghan, W. M. (2015). Prevalence Speaker abstracts.
and patterns of marijuana use among pregnant and nonpregnant women of Rouzer, S. K., & Diaz, M. R. (2022). Moderate prenatal alcohol exposure modifies sex-
reproductive age. American Journal of Obstetrics and Gynecology, 213(2), specific CRFR1 activity in the central amygdala and anxiety-like behavior in
201.e1e201.e10. adolescent offspring. Neuropsychopharmacology, 47(12), 2140e2149.
Kumar, A., Singh, C. K., DiPette, D. D., & Singh, U. S. (2010). Ethanol impairs acti- Rueda, D., Navarro, B., Martınez-Serrano, A., Guzma n, M., & Galve-Roperh, I. (2002).
vation of retinoic acid receptors in cerebellar granule cells in a rodent model of The endocannabinoid anandamide inhibits neuronal progenitor cell

12
cs, H. Charchat-Fichman, J. Landeira-Fernandez et al.
M.V. Kova Alcohol 110 (2023) 1e13

differentiation through attenuation of the Rap1/B-Raf/ERK pathway. Journal of development by inducing an SCG10/stathmin-2 degradation pathway. EMBO
Biological Chemistry, 277(48), 46645e46650. Journal, 33(7), 668e685.
de Salas-Quiroga, A., García-Rinco n, D., Go
 mez-Domínguez, D., Valero, M., Simo  n- Wade, N. E., Thomas, A. M., Gruber, S. A., Tapert, S. F., Filbey, F. M., & Lisdahl, K. M.
nchez, S., Paraíso-Luna, J., et al. (2020). Long-term hippocampal interneur-
Sa (2020). Binge and cannabis co-use episodes in relation to white matter integrity
onopathy drives sex-dimorphic spatial memory impairment induced by pre- in emerging adults. Cannabis and Cannabinoid Research, 5(1), 62e72.
natal THC exposure. Neuropsychopharmacology, 45(5), 877e886. Wang, X., Dow-Edwards, D., Anderson, V., Minkoff, H., & Hurd, Y. L. (2004). In utero
Scheyer, A. F., Melis, M., Trezza, V., & Manzoni, O. J. J. (2019). Consequences of marijuana exposure associated with abnormal amygdala dopamine D2 gene
perinatal cannabis exposure. Trends in Neurosciences, 42(12), 871e884. expression in the human fetus. Biological Psychiatry, 56(12), 909e915.
Seleverstov, O., Tobiasz, A., Jackson, J. S., Sullivan, R., Ma, D., Sullivan, J. P., et al. Wang, X., Dow-Edwards, D., Keller, E., & Hurd, Y. L. (2003). Preferential limbic
(2017). Maternal alcohol exposure during mid-pregnancy dilates fetal cerebral expression of the cannabinoid receptor mRNA in the human fetal brain.
arteries via endocannabinoid receptors. Alcohol, 61, 51e61. Neuroscience, 118(3), 681e694.
Shollenbarger, S. G., Price, J., Wieser, J., & Lisdahl, K. (2015). Poorer frontolimbic Wang, R., Shen, Y. L., Hausknecht, K. A., Chang, L., Haj-Dahmane, S., Vezina, P., et al.
white matter integrity is associated with chronic cannabis use, FAAH genotype, (2019). Prenatal ethanol exposure increases risk of psychostimulant addiction.
and increased depressive and apathy symptoms in adolescents and young Behavioural Brain Research, 356, 51e61.
adults. NeuroImage. Clinical, 8, 117e125. Wozniak, J. R., Riley, E. P., & Charness, M. E. (2019). Clinical presentation, diagnosis,
Shukrun, N., Shabtai, Y., Pillemer, G., & Fainsod, A. (2019). Retinoic acid signaling and management of fetal alcohol spectrum disorder. The Lancet Neurology, 18(8),
reduction recapitulates the effects of alcohol on embryo size. Genesis, 57(7e8), 760e770.
Article e23284. Wu, C. S., Jew, C. P., & Lu, H. C. (2011). Lasting impacts of prenatal cannabis exposure
Simakova, M., Tobiasz, A., Sullivan, R. D., Bisen, S., Duncan, J., Sullivan, J. P., et al. and the role of endogenous cannabinoids in the developing brain. Future
(2018). Gestational age-dependent interplay between endocannabinoid re- Neurology, 6(4), 459e480.
ceptors and alcohol in fetal cerebral arteries. Journal of Drug and Alcohol Zarcone, V. P. (1973). Marijuana and ethanol: Effects on sleep. International Journal
Research, 8, Article 236068. of Psychiatry in Medicine, 4(2), 201e212.
Singh, A. K. (2019). Alcohol interaction with cocaine, methamphetamine, opioids, Zhang, C., Ojiaku, P., & Cole, G. J. (2013). Forebrain and hindbrain development in
nicotine, cannabis, and g-hydroxybutyric acid. Biomedicines, 7(1), 16. zebrafish is sensitive to ethanol exposure involving agrin, Fgf, and sonic
Subbaraman, M. S., & Kerr, W. C. (2015). Simultaneous versus concurrent use of hedgehog function. Birth Defects Research Part A: Clinical and Molecular Tera-
alcohol and cannabis in the National Alcohol Survey. Alcoholism: Clinical and tology, 97(1), 8e27.
Experimental Research, 39(5), 872e879. Zurolo, E., Iyer, A. M., Spliet, W. G. M., Van Rijen, P. C., Troost, D., Gorter, J. A., et al.
Sulik, K. K., Johnston, M. C., & Webb, M. A. (1981). Fetal alcohol syndrome: (2010). CB1 and CB2 cannabinoid receptor expression during development and
Embryogenesis in a mouse model. Science, 214(4523), 936e938. in epileptogenic developmental pathologies. Neuroscience, 170(1), 28e41.
Tang, S., Xu, S., Gullapalli, R. P., & Medina, A. E. (2018). Effects of early alcohol exposure Substance Abuse and Mental Health Services Administration. (2020). Key substance
on functional organization and microstructure of a visual-tactile integrative cir- use and mental health indicators in the United States: Results from the 2019 Na-
cuit. Alcoholism: Clinical and Experimental Research, 42(4), 727e734. tional Survey on Drug Use and Health (HHS Publication No. PEP20-07-01-001,
Terry-McElrath, Y. M., & Patrick, M. E. (2018). Simultaneous alcohol and marijuana NSDUH Series H-55). Rockville, MD: Center for Behavioral Health Statistics
use among young adult drinkers: Age-specific changes in prevalence from 1977 and Quality, Substance Abuse and Mental Health Services Administration.
to 2016. Alcoholism: Clinical and Experimental Research, 42(11), 2224e2233. Retrieved from https://store.samhsa.gov/product/key-substance-use-and-
Tortoriello, G., Morris, C. V., Alpar, A., Fuzik, J., Shirran, S. L., Calvigioni, D., et al. mental-health-indicators-in-the-united-states-results-from-the-2019-
(2014). Miswiring the brain: D9-tetrahydrocannabinol disrupts cortical national-survey-on-Drug-Use-and-Health/PEP20-07-01-001.

13

You might also like