Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Apr 1.
Published in final edited form as: Nat Rev Neurosci. 2009 Mar 11;10(4):303–312. doi: 10.1038/nrn2598

Prenatal exposure to drugs: effects on brain development and implications for policy and education

Barbara L Thompson 1, Pat Levitt 1, Gregg D Stanwood 1
PMCID: PMC2777887  NIHMSID: NIHMS155617  PMID: 19277053

Abstract

The effects of prenatal exposure to drugs on brain development are complex and are modulated by the timing, dose, and route of drug exposure. It is difficult to assess these effects in clinical cohorts, which are beset with multiple exposures and difficulties in documenting use patterns. This can lead to misinterpretation of research findings by the general public, the media and policy makers, who may mistakenly assume that the legal or illegal status of a drug correlates with its biological impact on fetal brain development and long-term clinical outcomes. It is important to close the gap between what science tells us about the impact of prenatal drug exposure on the fetus and the mother, and what we do programmatically with regard to at-risk populations.

Introduction

Chemical neurotransmitters serve important functions in the coordination of the development of neurons and brain circuits (Box 1). Psychoactive drugs modulate receptors, transporters and other components of neurotransmission, many of which are expressed during prenatal stages of brain development, although their expression patterns and functions are sometimes quite different from their more typical roles later in life. Thus, the presence of these proteins in the developing brain underlies the well-documented impact of prenatal drug exposure on brain architecture, chemistry and neurobehavioral function in clinical cohorts and in animal models.

Box 1. Effects of neurotransmitters on brain development.

Studies beginning almost 50 years ago demonstrated that the capacity to synthesize and degrade neurotransmitters, particularly biogenic amines, many of the receptors through which they signal, and circuits that utilized them emerged early in embryogenesis 100, 146, 188-192. Given the appearance of neurotransmitters well before synapse formation, a variety of roles in neurodevelopmental processes were postulated, but not demonstrated. Modern in vitro approaches and genetic engineering of mice has facilitated discoveries that identify the pleiotropic nature of neurotransmitter signaling in neurodevelopmental processes. As a few examples of non-synaptic functions, dopamine regulates progenitor cell cycle kinetics and dendritic growth 62, 193, serotonin modulates cell proliferation and the response of growing axons to classic guidance molecules 149, 194, GABA activates the migration of developing neurons 195, and glutamate regulates oligodendrocyte precursor survival 196. Key to these influences is the expression of subsets of receptors in transient developmental patterns that mediate early neurotransmitter signaling for developmental purposes. This is followed by spatial and temporal reorganization and the expression of other subtypes that may be involved in modulating or directly inducing synaptic transmission.

Many legal drugs such as nicotine and alcohol can produce more severe deficits on brain development than some illicit drugs such as cocaine. However, erroneous and biased interpretations of the scientific literature often affect educational programs and even legal proceedings. For example, a pregnant woman whose emergency room toxicology screen revealed cocaine use was recently jailed and accused of using a deadly weapon against her unborn child in Tennessee, United States 1. Such policy decisions may have unintended adverse consequences: for example, they might harm the fetus due to pathophysiological activation of the maternal stress response system 2 and they might cause pregnant women addicted to drugs to avoid prenatal medical care (Box 2).

Box 2. Criminal prosecution of pregnant women using drugs of abuse.

A study in 1985 reporting damaging effects of cocaine use during pregnancy 16 produced a massive media response. Based on the media reports, laws were enacted in the United States requiring health care professionals to report pregnant illicit drug users to child welfare authorities and legislation was pursued to make drug use during pregnancy a criminal offense. The desire to guard fetal health and outcomes is understandable; however, research shows that pregnant women who fear prosecution and the potential loss of their children as a result of using drugs of abuse are less likely to seek essential prenatal and medical care. Thus, the policies that were meant to deter illegal drug use among pregnant women had unintended consequences, resulting in even greater risk to the fetus 197, 198. Moreover, the threat of criminal punishment can foster fear and mistrust between doctors and their patients, increase maternal stress, and endanger the health of women and their future children. Based on the science, one could argue that this is considerably worse than the drug exposure itself.

South Carolina in the United States has been particularly aggressive in using the court system in an attempt to deter drug use during pregnancy. At least 90 women have been prosecuted for stillbirths after using drugs or alcohol. Between 1989 and 1994, a prominent hospital in Charleston adopted a policy of informing police of any positive test for cocaine in pregnant women. The patients were informed that an arrest would occur if they failed to successfully complete a drug program. If a positive test was obtained at delivery the woman would be immediately arrested and charged 199. This policy was discontinued in 1994 because of pressures applied by the Department of Health and Human Services 199, and was found to be constitutionally deficient in 2001 by the Supreme Court 200.

In May 2008, the South Carolina Supreme Court overthrew the homicide by child abuse conviction of one of these women, Regina McKnight 201. They ruled that Mrs. McKnight had not received a fair trial due to ineffective counsel and the inclusion of unsupported scientific evidence 201. It is not yet known whether she will be re-tried for the charge, but these events suggest that policy may be coming into better accordance with scientific data.

Research findings in humans and animal models should be used to inform better policy and program development to reduce the population of children who are exposed to drugs prenatally. Much of the human data is being generated by the National Children's Study, which aims to examine the effects of environmental influences on the health and development of 100,000 children across the United States. However, logistical issues have made it difficult to coordinate and retain participation of the mothers and children.

In this article, we describe findings from relevant animal models that illuminate the specific mechanisms by which drugs of abuse, both illegal and legal, act on the brain. It is not possible to adequately review all drugs and all models (see 3-6 for other relevant reviews), thus we have selected a few examples in which substantial data have been generated. We highlight clinical findings in humans that parallel many of the findings in model systems. Finally, we discuss the complex social and policy issues involved in providing appropriate support structures for children exposed to drugs in utero to help improve their neurodevelopmental trajectory.

Animal models of prenatal drug abuse

The utility of animal models for understanding typical and atypical human development is well established. Animal models have enabled us to begin to elucidate the complex neurodevelopmental consequences of prenatal drug exposure. Although basic behavior and some aspects of global structure and functional neural activity can be studied in humans exposed developmentally to drugs, the specific neuroanatomical, molecular, and cellular consequences underlying these behavioral changes are not accessible to study in vivo. Well-designed animal models can therefore be powerful tools for revealing the neurobiological alterations that underlie later disrupted behavior.

However, the challenges of developing appropriate animal models are substantial. For example, although the orderly assembly of the cellular elements that comprise the developing nervous system is highly conserved across vertebrate species, the timing and duration of histogenic events and maturation of neurotransmitter systems may differ substantially. Moreover, metabolic pathways differ based on routes of exposure and across species; this therefore represents another important variable in recapitulating drug exposure patterns in humans. There are several key experimental variables to be considered when designing an animal model of prenatal drug exposure: the species to be used, the age of the embryo during exposure, the frequency and duration of exposure, the route of administration and the drug concentration.

The timeline for neurodevelopmental milestones has been documented in humans and for most animals used as models (Figure 1) 7-9. Thus, designing animal models of drug exposure during particular stages of fetal animal development that correlate with the timing of exposure during human pregnancy is realistic, at least to a first approximation. There are, however, further complications when considering how to model human exposure. These include both nutritional issues and the use of two or more drugs in combination. In addition, providing an environment in which the mother feels comfortable in disclosing the length of exposure, amount used and combinations of drugs used is critical for determining clinical profiles and in subsequently using the data to establish accurate animal models. Furthermore, some processes that occur during the third trimester of in utero development in humans occur postnatally in rodents and thus might be missed by studies conducted during gestation only. However, further complicating matters, rodent maternal and environmental interactions with the offspring are crucial for proper social and emotional development during this postnatal period, a situation that differs from that of humans during late gestation.

Figure 1. Developmental Events and Ontogeny of Drug Targets.

Figure 1

The peak periods of key neurodevelopmental events are depicted. Reference to timing of neurotransmitter system development includes appearance of receptors, transporters and synthetic machinery. Note that some of these neurotransmitter elements may continue to mature through puberty. Gestational age for both human (black font) and mouse (red font) are aligned with the black arrow to define the timeline for these events. Emerging drug targets (receptors, transporters, etc) are depicted along this same timeline in purple. Aff, afferents; CP, cortical plate; CTX, cortex; DA, dopamine; E, embryonic; GE, ganglionic eminence; LC, locus coeruleus; M, months; MZ, marginal zone; P, postnatal; PZ, proliferative zone; SP, sub-plate; VTA, ventral tegmental area; W, weeks; 5-HT, serotonin. Adapted from 202, 203.

Different species may provide distinct advantages depending on the specific hypotheses to be tested. For example, if one wishes to investigate the roles of certain genes in mediating the impact of prenatal drug exposure, genetically engineered mice may be most advantageous. This advantage might be weighed against the difficulties in administering drugs repeatedly to pregnant mice. Self-administration drug paradigms during pregnancy may be more imitative of human abuse than experimenter administered paradigms and rats have been the primary model species for this approach. On the other hand, establishing self-administration in rodents is sometimes problematic and requires other reinforcers to be superimposed on drug administration. Non-human primates most closely model the temporal domains of human brain development and behavior. However, it is logistically far more difficult and expensive to use non-human primates for research purposes, and as such this is not the ideal approach for rapid mechanistic discoveries. The long history of animal model research has in fact employed many different models and comparison of fundamental discoveries across species has been difficult. Cautious interpretation, in which one attempts to identify convergent findings, is particularly necessary for further advancements in the field. Convergence is often found in translating across larger behavioral dimensions, rather than in detailed features of the models.

Lastly, patterns of administration of different drugs vary. Some drugs (such as cocaine) are typically used in discrete episodes, whereas others (such as nicotine and alcohol) tend to be used chronically. This issue is likely to be important in determining the effects of developmental drug exposures. Thus, the effects of various substances may be reflective of both their inherent neurotoxicity and also of the patterns with which they are (ab)used.

Effects of illegal drugs of abuse

Cocaine

Cocaine is a psychostimulant that binds to monoaminergic – preferably dopaminergic – transporters and prevents uptake of extracellular monamines into the pre-synaptic cell, resulting in excess neurotransmitter in the synaptic cleft and excess stimulation of dopamine receptors. There are five known dopamine receptors: D1-like receptors (D1 and D5), and D2- like receptors (D2, D3, and D4). The roles of dopamine and these receptors in reward pathways have been demonstrated in both human and animal model 10, 11. Dopamine receptors are also located in the prefrontal cortex, hypothalamus, brainstem and even outside the CNS 12-15. The dopamine system develops early in gestation in all vertebrate species, and might therefore be sensitive to exogenous manipulation early in gestation. In fact, some early reports suggested a very severe phenotype in children exposed to cocaine in utero 16-18. Children were thought to be emotionally disrupted, cognitively impaired, less likely to socially interact, and more likely to die from sudden infant death syndrome (SIDS). Thus, the term “crack-baby” was introduced to describe children exposed to cocaine prenatally. However, these original studies were confounded by very small sample sizes, polydrug use, nutritional status and other psychosocial problems.

We now know that, irrespective of species, the extent to which prenatal exposure to cocaine affects brain development varies significantly. The assumptions made by the public about the biological impact of prenatal cocaine exposure may be based on the severity of symptoms associated with cocaine addiction, and the illegal nature of the substance. In humans, longitudinal studies have shown that there are long-term consequences of prenatal cocaine exposure; however, the behavioral dysfunction appears to be mild 19-24. The recreational use of cocaine during pregnancy results in a subtle, though dominant developmental phenotype that resembles attention deficit disorder (ADHD) 19-22, 25-29. Detailed studies have demonstrated that prenatal cocaine exposure can have long-lasting negative effects on cognitive and attention systems, mediated via regions such as the prefrontal cortex, and other higher-order cortical areas that express dopamine receptors and receive rich dopaminergic projections from the midbrain (Table 1) 19, 24, 27, 30-32. Recent data also suggest that there is increased likelihood that children exposed to cocaine prenatally will require special needs programs 33, which, from both an individual and societal perspective, is expensive.

Table 1.

Neurodevelopmental consequences of prenatal drug exposure.

Age of
exposure
Drug Neurochemistry
involved
Neurodevelopmental
consequences
References
Late early to mid
gestation (primarily
based on animal
studies)
Cocaine DA > NE and 5-HT

Blocks
monoaminergic
transporters and
Increases synaptic
concentrations of
monoamines
Altered neuroanatomical
morphology, disrupted
cognition, altered cellular
signaling
18-37, 42-47,
54-59, 63-65,
203


Throughout
gestation


Alcohol


GABA and NMDA

Blocks NMDA
receptor activity and
increases
GABAergic activity
Craniofacial
dysmorphologies,
decreased birth weight,
hyperactivity, cognitive
deficits, cortical
dysgenesis, cell death,
reduced brain volume


113-115, 118-
120, 126-132
Throughout
gestation
Nicotine Acetylcholine

Activates nAChRs
Decreased birth weight,
hyperactivity, cognitive
disabilities, emotional
disruptions
82, 86-94, 96-
98, 100-105,
107, 108
Throughout
gestation and early
postnatal exposure
Amphetamine/
Methamphetamine
DA > NE and 5-HT

Reverses the action
of monoaminergic
transporters and
Increases synaptic
concentrations of
monoamines
Low birth weight,
decreased arousal,
deficits in learning,
decreased volume of
hippocampus and striatum
66, 67, 70-73,
76-81

Animal models confirm that prenatal cocaine exposure results in specific permanent behavioral, cellular, and molecular changes 34-37. An intravenous model of prenatal cocaine exposure in the rabbit closely models the pharmacokinetic profile of human abusers 38-40. It has been used to delineate a number of highly specific changes, including altered GABA content in the cortex, altered calcium binding protein expression, morphological changes in pyramidal cells, and decreased dopamine D1 receptor coupling to its cognate G-protein, G 41-46 (Table 1). The most surprising alteration was a permanent reduction in second messenger coupling, which was found to be due to abnormal internalization of the D1 receptor. This suggested that the D1 receptor is not trafficked properly to the cell membrane where it would then interact with G 47. These cellular and molecular findings were observed only in dopamine rich cortical areas, which are intimately involved in cognition and executive functioning tasks, including attention 48-53. These studies have also been able to directly relate the timing of in utero cocaine exposure to its deleterious effects in offspring and suggest that the second trimester may be a particularly vulnerable period 54.

Other animal models show that prenatal cocaine exposure alters basic processes of neocortical development, including cell production and migration 55-60. These findings are consistent with recent studies demonstrating that dopamine can regulate progenitor cell proliferation and neuronal migration during prenatal development 61, 62.

Behavioral changes have been reported in various animal models, including deficits in attention tasks, emotional reactivity, and the reinforcing properties of drugs of abuse 30-32, 63, 64. These findings correspond with the human clinical literature that reports disturbances in both attention and emotion regulation in children exposed prenatally to cocaine.

Amphetamine/Methamphetamine

Amphetamine and methamphetamine are also psychostimulants; however, they target the monoaminergic system in a mechanistically distinct fashion from cocaine. Both drugs reverse the actions of monoamine transporters and enhance release of dopamine, norepinephrine, and serotonin into the synaptic cleft, increasing their availability to act upon post-synaptic receptors. In addition, both drugs can block the re-uptake and degradation of these neurotransmitters, further increasing their concentrations in the synaptic cleft. The addition of a methyl group allows methamphetamine to move through lipid permeable membranes easily. Both amphetamine and methamphetamine are routinely ingested, snorted, and smoked. The frequency of use and abuse of amphetamine and methamphetamine has steadfastly risen in the human population, exceeding cocaine use in many regions of the United States 65.

Because methamphetamine and amphetamine use during pregnancy has become prolific only recently, there are few studies defining its long-term consequences. Children exposed to methamphetamine or amphetamine during prenatal development show decreased arousal, increased stress, decreased school achievements, movement disturbances and low birth weight (a high risk factor for special needs programs at school age) 66-70. Neurocognitive testing showed that these children score lower on sustained attention, long-term spatial and verbal memory, and visual motor integration 71. Neuroimaging has produced reports of smaller striatum and hippocampus volumes and decreased numbers of dopamine D2 receptors and dopamine transporter density 71, 72. Polydrug use and other psychosocial risks complicate these studies 70, 73, making it challenging to define the clinical impact of each drug individually. Animal models have helped delineate the specific neurodevelopmental consequences of prenatal amphetamine and methamphetamine exposure.

The neurobiological consequences of prenatal methamphetamine and amphetamine exposure have been studied in mice and rats. Animals exposed to methamphetamine have lower birth weights, increased incidence of microgyria (smaller and more numerous convolutions in the cerebral cortex than usual), and deficits in visual system development 74-76. They also display impaired postural motor movements, deficits in specific learning paradigms, increased startle reflexes and decreased pre-pulse inhibition 76, 77. Furthermore, alterations in the levels and activity of the noradrenergic and serotonergic systems have been demonstrated in animals exposed to either amphetamine or methamphetamine 78-80. Some of the functional consequences (such as impaired sensory-motor coordination) of prenatal methamphetamine exposure are transmitted to the next generation of offspring 81. These findings demonstrate that prenatal amphetamine or methamphetamine exposure leads to a complex behavioral phenotype with similarly complex molecular and cellular roots.

Effects of Legal Drugs of Abuse

Nicotine

Nicotine binds to nicotinic acetylcholine receptors (nAChRs), a class of ligand-gated ion channels that are widely expressed throughout the fetal nervous system. Nicotine is usually presented to the fetus through maternal smoking or environmental exposure to second-hand smoke. Research has provided unequivocal evidence that active smoking and passive exposure to second-hand smoke can be teratogenic (for review, see 82), can lead to decreased birth weight and can increase the risks of preterm birth and SIDS (Table 1), all of which are high risk factors for behavioral impairment 69, 83-85. Numerous studies support a robust relationship between developmental tobacco smoke exposure and attention deficit disorders, hyperactivity, antisocial behavior and learning disabilities 86-94. However, differences in the amount of nicotine and other chemicals in different tobacco products make it challenging to compare findings across ethnic or age groups. Furthermore, the presence of additional potential neurotoxicants and biomodulators in tobacco smoke, make it difficult to determine the specific effects of nicotine exposure. This presents a challenge for clinicians who may wish to recommend nicotine replacement therapy (NRT) to pregnant patients who have difficulty terminating smoking. Although the additional chemicals are absent in NRT, high amounts of nicotine are still presented to the fetus. Based on animal studies (see below), such exposure is likely to lead to long-term disruption of brain architecture and chemistry. In fact, transdermal patch NRT may actually worsen birth outcomes relative to active smoking 95, 96. One possibility as reviewed recently 97, 98, is that this may reflect the continuous-delivery provided by the patch, which in animal models has been shown to increase fetal brain nicotine levels up to three times maternal blood levels 99.

Animal studies have provided the most compelling evidence that nicotine has substantial negative neurodevelopmental impact. Activation of nAChRs affects morphogenesis, spontaneous neural activity, and neuronal survival in rodents 100. In animals, plasma nicotine levels similar to those of pregnant women smoking “moderate” numbers of cigarettes produce potent neurobehavioral effects – including changes in locomotor activity, reward systems, anxiety, and cognition – in offspring 101, 102,100, 103-105. Higher doses produce fetal hypoxia and substantial growth retardation 106. Cortical cholinergic systems modulate sensory cognitive processing 107, and prenatal nicotine exposure impairs cognitive functions in animal models and in children 100, 108. Intriguingly, nicotine inhibits aromatase, an enzyme involved in estrogen synthesis in the placenta 109; this may interfere with sexual differentiation of the brain in males and changes have been observed in the timing of puberty onset in gestationally exposed male adolescents 110. Based on these findings, reducing smoking by pregnant women and their immediate family members will be very beneficial; whereas the value of NRT in pregnant women is questionable and requires in depth evaluation through prospective studies 98.

Alcohol

Alcohol use spans all socioeconomic classes, gender, race, education, and cultural groups. It is a pharmacological depressant, blocking NMDA receptor activity and increasing GABAergic activity, thereby decreasing cortical and sub-cortical activity 111, 112. Alcohol can also disrupt growth factor receptor signaling by disrupting plasma membrane integrity 113, 114. GABA and its receptors are present early in neuronal development, and can modulate progenitor cell proliferation, cell migration and neurite growth 115-117. Alcohol crosses the placental barrier at any point during pregnancy and thus can severely impact numerous histogenic processes (see below and Table 1).

There is overwhelming basic and clinical evidence regarding the negative neurodevelopmental consequences of prenatal alcohol exposure. These comprise an ICD/DSM recognized disorder, fetal alcohol syndrome, characterized by growth deficiencies, craniofacial dysmorphologies, and CNS damage 3, 118-121. Prenatal alcohol exposure can cause intellectual disability, deficits in learning, attention and motor development and hyperactivity 119, 120, 122-125. Although concomitant use of other drugs with alcohol is not unusual, alcohol is often used as a solitary drug. This clinical population provides a less mechanistically complex picture of the neurodevelopmental consequences of prenatal alcohol exposure. In addition, animal models have provided further understanding of the cellular and molecular consequences of prenatal ethanol exposure.

Animal models of prenatal ethanol exposure have confirmed an increase in birth defects including neurological dysfunction 3, 126-130. Exposure during all gestational periods has dramatic teratogenic consequences 131-133. There are reports of a decrease in spinal and cranial motor neuron production and size, neocortical and hippocampal dysgenesis, increased neuronal cell death, reduced or delayed neuronal migration and a decrease in myelination 134-137. Ethanol exposure also disrupts the integrity of plasma membrane receptors 138, 139. Thus, in addition to direct antagonism of neurotransmitter receptors, receptor tyrosine kinase signaling is also disrupted, leading to altered neurotrophic factor modulation of multiple histogenic events.

Elegant studies have begun to prove the impact of prenatal alcohol on endocrine function 140, 141. As well as alcohol's direct effects on the fetus, it also acts directly on the hypothalamic-pituitary-adrenal (HPA) axis of both the fetus and the mother. Modulating the HPA axis during development can permanently alter its responsiveness to later stressors, setting off a cascade of behavioral, cognitive, and cellular alterations in the adult 142. Similarly, alterations in maternal HPA activity could underlie some of the long-lasting consequences following prenatal alcohol. As such, intervention and behavioral therapy for these children might consider methods used in clinical populations of children exposed to severe stress in utero.

Effects of Prescription Neurotherapeutics

Antidepressant Medications

A report of a higher rate of cardiac abnormalities in the children of women using selective serotonin reuptake inhibitors (SSRIs) during pregnancy 143, 144 has led to enormous public and scientific interest. Animal studies have long implicated serotonin, the target of many antidepressants, in the development of the brain and peripheral organs 145-147. Serotonin and its receptors are expressed in the brain early in prenatal development 12, 148-151, and can modify dendritic and axonal differentiation 149, 152 (Figure 1). Although the mechanism remains unknown, early postnatal disruption of serotonin signaling in the rodent (at stages roughly equivalent to 3rd trimester and neonatal human periods), can permanently increase anxiety behaviors and disrupt learning 153, 154.

The animal studies are compelling, but epidemiological studies to date do not provide evidence of an increased risk of birth defects following SSRI use during pregnancy 155-159. It is important to consider that even though the rate of overt birth defects compared to the general population does not increase at typical dose levels, subtle yet significant behavioral dysfunctions in offspring could occur similar to prenatal cocaine exposure. The issues are thus complex and require additional, detailed studies to inform medical decisions. Health care professionals must weigh the relative neurodevelopmental impact of drug treatment of a psychiatric disorder against the potentially negative outcomes from untreated illnesses (including increased maternal stress due to depression or untreated neurochemical imbalances). Thus, when behavioral therapy alone is not effective in treating depression in a pregnant woman, the potential risk of drug therapy must be weighed against the risk for relapse of the disorder if pharmacological therapy is interrupted.

Other Prescription Medications

Several prescription medications are used for treatment of psychiatric, neurological and maternal pregnancy disorders. Little research has examined the long-term functional implications of these drug treatments, but new studies are beginning to highlight a complex clinical picture.

Concerns have been raised regarding the effects of antipsychotic medications (potent antagonists of dopamine and serotonin receptors) during pregnancy. Animal studies point to serious potential neurodevelopmental risks 160-164. However, there have been few clinical studies 165, 166, making it difficult to draw accurate conclusions regarding long-term impact on fetal development. As with other psychoactive drugs, an individualized calculation of relative risk is very important to balance the potential risk of drug exposure against the risk of a potential psychotic episode if pharmacotherapy is suspended.

Another commonly prescribed drug is valproate, an anti-convulsant and anti-mania therapeutic which blocks voltage-gated sodium and T-type calcium channels and inhibits the transamination of GABA 167. As a histone deacetylase inhibitor, valproate also can regulate the epigenetic modulation of gene transcription 168. Clinical data demonstrates high risk for autism spectrum disorder (ASD), teratogenic (neural tube defects; cranio-facial dysmorphia) and neurotoxic (apoptosis, reduced cell proliferation in multiple brain areas) effects of valproate exposure 169, 170. Rats exposed prenatally have lower sensitivity to pain, increased repetitive/stereotypic-like activity, higher anxiety, decreased level of social interaction and an increased basal level of corticosterone 171, 172. Alterations in NMDA receptor expression, enhanced long-term potentiation and neocortical hyperconnectivity also have been reported 173, 174.

Terbutaline and other related drugs were previously used commonly in late term pregnancies to reduce premature uterine contractions via stimulation of the β2-adrenergic receptor 175. Terbutaline exposure to neonatal animals results in neuroinflammation and long-lasting behavioral and cellular deficits 176-178. Furthermore, children exposed to terbutaline or related adrenoceptor stimulants during late gestation may have an increased incidence of learning and neuropsychiatric disorders 179, 180. Recent clinical studies have revealed an increased risk for the development of ASD in exposed offspring, consistent with a report of a modest increase in risk due to an allelic variant in the gene encoding the β2-adrenergic receptor 181; whether this genetic predisposition and environmental risk interact with one another is not yet known. In fact, the efficacy of these drugs in preventing preterm delivery when used for maintenance tocolysis is suspect 182, and their recognition for such uses has been withdrawn.

Balancing Prenatal Drug Exposure

Basic and clinical research unequivocally demonstrates that recreational or prescription use of drugs during pregnancy can be viewed as an anathema to healthy development. There is a clear conundrum, however, with regard to the need to balance exposure (in a clinically prescribed population) versus non-exposure when considering the long-term functional impact on brain development. The difficulties relate to the fact that high prenatal stress, malnutrition and untreated maternal psychiatric disorders can themselves increase risk for developmental disabilities in children.

With regard to recreational drugs, human addictive behavior makes this issue far more complicated than a simple public policy approach of ‘just say no’. Furthermore, it is clear that the idea that illegal drugs are more harmful to the unborn fetus than legal drugs is a misnomer; this concept, which strongly influences public policy, is not supported by findings from carefully designed and controlled research studies. For pregnant women who abuse drugs prior to conception, withdrawal of these drugs post-conception is not without risk to the fetus. Maternal stress can be severe during withdrawal, and general health status may decline; both severely impacting brain development of the fetus.

We now realize that prescription drug treatments for pregnant women with psychiatric and neurological disorders can have negative effects on fetal brain development and long-term behavioral outcomes. However, the underlying maternal pathophysiology of untreated disorders can lead to high risk nutritional and stress status for both the mother and the fetus. Perhaps underscoring the level of complexity of the maternal-fetal relationship, balancing these issues makes formulation of public and medical policies less straightforward and far more difficult than generally appreciated.

Proposals to Reduce Bias

When considering particular conditions or states, people generally default to frames that are most familiar to them. Frames are principles that are socially shared and persistent over time and which work symbolically to meaningfully structure the social world 183. It is essential for scientists to understand this concept, because the inadequate communication of information causes people to default to these frames. Framing has been defined as the process of selecting a few elements of a perceived reality and assembling a narrative that highlights connections among them to promote a particular interpretation for an individual 184. We have already mentioned the misnomers that illustrate this concept. The term ‘crack baby’ evolved from social perceptions regarding individuals who use crack cocaine, the assumed severity of impact of fetal exposure to an illegal drug, and a lack of understanding of the complex maternal status that included polydrug use and malnutrition. The press and scientists alike were culpable in promoting this image in the absence of documented biological and behavioral impact. By the time science caught up, the frame had been cemented, and remains an influential factor in defining public policy (Box 2).

Fully developed frames perform four functions: problem definition, causal analysis, moral judgment, and remedy promotion 184, 185. In order to provide more accurate information for policy makers the original concept must be reframed. Initially, the science must be explained in such a way that it redirects attention away from the default positions. In part, redefining frames involves identifying values and explanations that make the societal, not individual, goals, obvious. This can be done best by creating and subsequently using simplifying models to explain brain development, which in turn will assist in explaining the impact of drugs and stressors on the process. This provides an opportunity for science to explain how current policies surrounding drug use may have even more negative effects on the fetus than the drug itself, under certain circumstances. These models thus become vehicles through which science can positively influence policy thinking and decisions.

The Frameworks Institute has developed a metaphorical frame of brain development that captures the essence of the scientific concept 186. According to this model, the early years of life matter because early experiences affect the architecture of the maturing brain. The quality of that architecture establishes either a sturdy or a fragile foundation for the development and behavior that follows. Therefore, similar to building a house, getting things right the first time is easier than trying to fix them later 187. This explanation of brain development reduces the complexity of the problem to a simple, concrete analogy that helps policymakers, members of the media and the public organize information into a clear picture. Thus, understanding that the assembly of brain architecture starts early, from the bottom up, paralleling the process of skill development in a child makes the connection between the time dependent relationship between structure and function. This facilitates, and stimulates interest in understanding factors that can influence the process. For example, discussions of the more potent influence of alcohol and nicotine on developing brain architecture, compared to cocaine leads directly to the conclusion that such exposures also must have more profound functional consequences. This reduces the impact of the default frame (that illegal drugs are more detrimental to fetal development than legal drugs). Likewise, explanations of the impact of ‘toxic stress’, such as severe malnutrition, drug exposure, poor healthcare or lengthy incarceration, on fetal brain architecture can lead to discussions between scientists and policy makers that evaluate the factors that may have the greatest impact on child development 187 (Box 3). There is thus an attainable goal of eliminating default frames and introducing more scientifically-based simplifying models that can alter the course of social and legal policies to best manage the difficulties of drug exposure during pregnancy (Box 3).

Box 3. Closing the gap – Using what we know to inform what we do.

Looking to the future

Scientists often find it bewildering that what appear to be basic concepts of development are misrepresented or misinterpreted when policy decisions are being made. Policies are then established that are not based on the scientifically recognized factors that regulate development. Instead, policies reflect default frames of child development that typically focus on family, individual responsibility and safety - illegal equals ‘more dangerous’; legal equals ‘less dangerous’. Moreover, experts may unwittingly cross the line from information-providers to advocates. Science experts could thus be viewed as biased responders to queries from policy-makers, moving beyond their perceived area of expertise. Scientific findings need to be shared with the community in such a way that we avoid loading information into default frames so that science can inform the way that we, and other members of society, think about problems.

There are still significant questions that will require additional investigations. For example, what are the consequences on fetal brain development of suspending drug treatment of a psychiatric disorder in a pregnant woman? Are there sensitive and critical periods of fetal brain development that are more or less vulnerable to exposures of specific agents? Can we better define the biological interactions of multiple drug exposures, nutritional status and maternal stress that influence fetal brain development? Creating simple developmental models that clearly communicate these influences will be essential. Such models will assist policy makers in developing sound strategies for reducing the incidence of drug exposure during pregnancy and for implementing more effective treatments that maximize healthy development of the fetus, when drug exposure during pregnancy does occur.

Maternal Drug Abuse Policies
Factors that Influence Policy Decisions Strategies to Affect Public Policy
  • Public / Media Perception (free will; legality; excesses)

  • What Science Tells Us (maternal health; fetal-maternal interactions; brain architecture and chemistry)

  • Policy Makers (public perception; legality; child welfare)

  • Develop simplifying frames of factors that impact child development (e.g. drugs, stress, nutrition)

  • Develop key working partnerships (National Conference of State Legislatures; childhood-focused private foundations)

  • Engage scientists in providing impartial testimony. (“What science tells us”, “Just the facts”)

  • Scientists work with video and print media to tell a core story of the impact of drugs of abuse on fetal development.

Acknowledgements

We thank Susan Bales of Frameworks Institute and Jack Shonkoff, Gillian Najarian, Al Race, and all the members of the National Scientific Council on the Developing Child for insightful discussions with regard to how scientists and policy makers can work together to solve public problems.

Footnotes

Further information

The National Children's Study http://www.nationalchildrensstudy.gov/

National Scientific Council on the Developing Child http://www.developingchild.net/

Translating time website http://www.translatingtime.net/

Frameworks Institute http://www.frameworksinstitute.org/sfa.html

References

  • 1.Pinto C. Medical officials question arrest of pregnant patient. The Tennessean. 2008 [Google Scholar]
  • 2.Seckl JR, Meaney MJ. Glucocorticoid “programming” and PTSD risk. Ann N Y Acad Sci. 2006;1071:351–78. doi: 10.1196/annals.1364.027. [DOI] [PubMed] [Google Scholar]
  • 3.Levitt P. Prenatal effects of drugs of abuse on brain development. Drug and Alcohol Dependence. 1998;51:109–125. doi: 10.1016/s0376-8716(98)00070-2. [DOI] [PubMed] [Google Scholar]
  • 4.Thadani PV. The intersection of stress, drug abuse and development. Psychoneuroendocrinology. 2002;27:221–30. doi: 10.1016/s0306-4530(01)00046-4. [DOI] [PubMed] [Google Scholar]
  • 5.Randall CL. Alcohol and pregnancy: highlights from three decades of research. J Stud Alcohol. 2001;62:554–61. doi: 10.15288/jsa.2001.62.554. [DOI] [PubMed] [Google Scholar]
  • 6.Malanga CJ, 3rd, Kosofsky BE. Mechanisms of action of drugs of abuse on the developing fetal brain. Clinics in Perinatology. 1999;26:17–37. v-vi. [PubMed] [Google Scholar]
  • 7.Clancy B, Darlington RB, Finlay BL. Translating developmental time across mammalian species. Neuroscience. 2001;105:7–17. doi: 10.1016/s0306-4522(01)00171-3. [DOI] [PubMed] [Google Scholar]
  • 8.Clancy B, Finlay BL, Darlington RB, Anand KJ. Extrapolating brain development from experimental species to humans. Neurotoxicology. 2007;28:931–7. doi: 10.1016/j.neuro.2007.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Clancy B, et al. Web-based method for translating neurodevelopment from laboratory species to humans. Neuroinformatics. 2007;5:79–94. doi: 10.1385/ni:5:1:79. [DOI] [PubMed] [Google Scholar]
  • 10.Volkow ND, Fowler JS, Wang GJ, Swanson JM, Telang F. Dopamine in drug abuse and addiction: results of imaging studies and treatment implications. Arch Neurol. 2007;64:1575–9. doi: 10.1001/archneur.64.11.1575. [DOI] [PubMed] [Google Scholar]
  • 11.Wise RA. Dopamine, learning and motivation. Nat Rev Neurosci. 2004;5:483–94. doi: 10.1038/nrn1406. [DOI] [PubMed] [Google Scholar]
  • 12.Goldman-Rakic PS, Lidow MS, Gallager DW. Overlap of dopaminergic, adrenergic, and serotoninergic receptors and complementarity of their subtypes in primate prefrontal cortex. Journal of Neuroscience. 1990;10:2125–38. doi: 10.1523/JNEUROSCI.10-07-02125.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Djamgoz MB, Wagner HJ. Localization and function of dopamine in the adult vertebrate retina. Neurochem Int. 1992;20:139–91. doi: 10.1016/0197-0186(92)90166-o. [DOI] [PubMed] [Google Scholar]
  • 14.De Souza EB, Kuhar MJ. Dopamine receptors in the anterior lobe of the human pituitary gland: autoradiographic localization. Brain Res. 1984;306:391–5. doi: 10.1016/0006-8993(84)90394-9. [DOI] [PubMed] [Google Scholar]
  • 15.Murrin LC, Gale K, Kuhar MJ. Autoradiographic localization of neuroleptic and dopamine receptors in the caudate-putamen and substantia nigra: effects of lesions. Eur J Pharmacol. 1979;60:229–35. doi: 10.1016/0014-2999(79)90222-x. [DOI] [PubMed] [Google Scholar]
  • 16.Chasnoff IJ, Burns WJ, Schnoll SH, Burns KA. Cocaine use in pregnancy. New England Journal of Medicine. 1985;313:666–9. doi: 10.1056/NEJM198509123131105. [DOI] [PubMed] [Google Scholar]
  • 17.Chasnoff IJ, Burns KA, Burns WJ. Cocaine use in pregnancy: perinatal morbidity and mortality. Neurotoxicology & Teratology. 1987;9:291–3. doi: 10.1016/0892-0362(87)90017-1. [DOI] [PubMed] [Google Scholar]
  • 18.Bauchner H, Zuckerman B, Amaro H, Frank DA, Parker S. Teratogenicity of cocaine. J Pediatr. 1987;111:160–1. doi: 10.1016/s0022-3476(87)80385-2. [DOI] [PubMed] [Google Scholar]
  • 19.Dow-Edwards D, Mayes L, Spear L, Hurd Y. Cocaine and development: clinical, behavioral, and neurobiological perspectives--a symposium report. Neurotoxicol Teratol. 1999;21:481–90. doi: 10.1016/s0892-0362(99)00008-2. [DOI] [PubMed] [Google Scholar]
  • 20.Gingras JL, O'Donnell KJ. State control in the substance-exposed fetus. I. The fetal neurobehavioral profile: an assessment of fetal state, arousal, and regulation competency. Annals of the New York Academy of Sciences. 1998;846:262–76. [PubMed] [Google Scholar]
  • 21.Karmel BZ, Gardner JM. Prenatal cocaine exposure effects on arousal-modulated attention during the neonatal period. Developmental Psychobiology. 1996;29:463–80. doi: 10.1002/(SICI)1098-2302(199607)29:5<463::AID-DEV5>3.0.CO;2-M. [DOI] [PubMed] [Google Scholar]
  • 22.Mayes LC, Grillon C, Granger R, Schottenfeld R. Regulation of arousal and attention in preschool children exposed to cocaine prenatally. Annals of the New York Academy of Sciences. 1998;846:126–43. doi: 10.1111/j.1749-6632.1998.tb09731.x. [DOI] [PubMed] [Google Scholar]
  • 23.Richardson GA, Hamel SC, Goldschmidt L, Day NL. The effects of prenatal cocaine use on neonatal neurobehavioral status. Neurotoxicology & Teratology. 1996;18:519–28. doi: 10.1016/0892-0362(96)00062-1. [DOI] [PubMed] [Google Scholar]
  • 24.Singer LT, et al. Cognitive outcomes of preschool children with prenatal cocaine exposure. Jama. 2004;291:2448–56. doi: 10.1001/jama.291.20.2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Mayes LC. Exposure to cocaine: behavioral outcomes in preschool and school-age children. NIDA Research Monograph. 1996;164:211–29. [PubMed] [Google Scholar]
  • 26.Mayes LC, Bornstein MH, Chawarska K, Granger RH. Information processing and developmental assessments in 3-month-old infants exposed prenatally to cocaine. Pediatrics. 1995;95:539–45. [PubMed] [Google Scholar]
  • 27.Mayes LC, Cicchetti D, Acharyya S, Zhang H. Developmental trajectories of cocaine-and-other-drug-exposed and non-cocaine-exposed children. J Dev Behav Pediatr. 2003;24:323–35. doi: 10.1097/00004703-200310000-00003. [DOI] [PubMed] [Google Scholar]
  • 28.Richardson GA, Conroy ML, Day NL. Prenatal cocaine exposure: effects on the development of school-age children. Neurotoxicology & Teratology. 1996;18:627–34. doi: 10.1016/s0892-0362(96)00121-3. [DOI] [PubMed] [Google Scholar]
  • 29.Richardson GA. Prenatal cocaine exposure. A longitudinal study of development. Annals of the New York Academy of Sciences. 1998;846:144–52. [PubMed] [Google Scholar]
  • 30.Gabriel M, Taylor C, Burhans L. In utero cocaine, discriminative avoidance learning with low-salient stimuli and learning-related neuronal activity in rabbits (Oryctolagus cuniculus) Behav Neurosci. 2003;117:912–26. doi: 10.1037/0735-7044.117.5.912. [DOI] [PubMed] [Google Scholar]
  • 31.Morrow BA, Elsworth JD, Roth RH. Prenatal cocaine exposure disrupts non-spatial, short-term memory in adolescent and adult male rats. Behav Brain Res. 2002;129:217–23. doi: 10.1016/s0166-4328(01)00338-2. [DOI] [PubMed] [Google Scholar]
  • 32.Thompson BL, Levitt P, Stanwood GD. Prenatal cocaine exposure specifically alters spontaneous alternation behavior. Behav Brain Res. 2005;164:107–16. doi: 10.1016/j.bbr.2005.06.010. [DOI] [PubMed] [Google Scholar]
  • 33.Levine TP, et al. Effects of prenatal cocaine exposure on special education in school-aged children. Pediatrics. 2008;122:e83–91. doi: 10.1542/peds.2007-2826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Harvey JA. Cocaine effects on the developing brain: current status. Neurosci Biobehav Rev. 2004;27:751–64. doi: 10.1016/j.neubiorev.2003.11.006. [DOI] [PubMed] [Google Scholar]
  • 35.Lidow MS. Consequences of prenatal cocaine exposure in nonhuman primates. Brain Res Dev Brain Res. 2003;147:23–36. doi: 10.1016/j.devbrainres.2003.09.001. [DOI] [PubMed] [Google Scholar]
  • 36.Mayes LC. A behavioral teratogenic model of the impact of prenatal cocaine exposure on arousal regulatory systems. Neurotoxicol Teratol. 2002;24:385–95. doi: 10.1016/s0892-0362(02)00200-3. [DOI] [PubMed] [Google Scholar]
  • 37.Stanwood GD, Levitt P. Drug exposure early in life: functional repercussions of changing neuropharmacology during sensitive periods of brain development. Current Opinion in Pharmacology. 2004;4:65–71. doi: 10.1016/j.coph.2003.09.003. [DOI] [PubMed] [Google Scholar]
  • 38.Parlaman JP, Thompson BL, Levitt P, Stanwood GD. Pharmacokinetic profile of cocaine following intravenous administration in the female rabbit. Eur J Pharmacol. 2007;563:124–9. doi: 10.1016/j.ejphar.2007.02.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Evans SM, Cone EJ, Henningfield JE. Arterial and venous cocaine plasma concentrations in humans: relationship to route of administration, cardiovascular effects and subjective effects. J Pharmacol Exp Ther. 1996;279:1345–56. [PubMed] [Google Scholar]
  • 40.Jenkins AJ, Keenan RM, Henningfield JE, Cone EJ. Correlation between pharmacological effects and plasma cocaine concentrations after smoked administration. J Anal Toxicol. 2002;26:382–92. doi: 10.1093/jat/26.7.382. [DOI] [PubMed] [Google Scholar]
  • 41.Friedman E, Yadin E, Wang HY. Effect of prenatal cocaine on dopamine receptor-G protein coupling in mesocortical regions of the rabbit brain. Neuroscience. 1996;70:739–47. doi: 10.1016/s0306-4522(96)83011-9. [DOI] [PubMed] [Google Scholar]
  • 42.Jones LB, et al. In utero cocaine-induced dysfunction of dopamine D1 receptor signaling and abnormal differentiation of cerebral cortical neurons. Journal of Neuroscience. 2000;20:4606–14. doi: 10.1523/JNEUROSCI.20-12-04606.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wang HY, Runyan S, Yadin E, Friedman E. Prenatal exposure to cocaine selectively reduces D1 dopamine receptor-mediated activation of striatal Gs proteins. Journal of Pharmacology & Experimental Therapeutics. 1995;273:492–8. [PubMed] [Google Scholar]
  • 44.Stanwood GD, Parlaman JP, Levitt P. Anatomical abnormalities in dopaminoceptive regions of the cerebral cortex of dopamine D(1) receptor mutant mice. J Comp Neurol. 2005;487:270–82. doi: 10.1002/cne.20548. [DOI] [PubMed] [Google Scholar]
  • 45.Stanwood GD, Washington RA, Shumsky JS, Levitt P. Prenatal cocaine exposure produces consistent developmental alterations in dopamine-rich regions of the cerebral cortex. Neuroscience. 2001;106:5–14. doi: 10.1016/s0306-4522(01)00256-1. [DOI] [PubMed] [Google Scholar]
  • 46.Murphy EH, et al. Cocaine administration in pregnant rabbits alters cortical structure and function in their progeny in the absence of maternal seizures. Experimental Brain Research. 1997;114:433–41. doi: 10.1007/pl00005652. [DOI] [PubMed] [Google Scholar]
  • 47.Stanwood GD, Levitt P. Prenatal exposure to cocaine produces unique developmental and long-term adaptive changes in dopamine D1 receptor activity and subcellular distribution. J Neurosci. 2007;27:152–7. doi: 10.1523/JNEUROSCI.4591-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Clark L, Cools R, Robbins TW. The neuropsychology of ventral prefrontal cortex: decision-making and reversal learning. Brain Cogn. 2004;55:41–53. doi: 10.1016/S0278-2626(03)00284-7. [DOI] [PubMed] [Google Scholar]
  • 49.Collette F, Van der Linden M. Brain imaging of the central executive component of working memory. Neurosci Biobehav Rev. 2002;26:105–25. doi: 10.1016/s0149-7634(01)00063-x. [DOI] [PubMed] [Google Scholar]
  • 50.Elliott R. Executive functions and their disorders. Br Med Bull. 2003;65:49–59. doi: 10.1093/bmb/65.1.49. [DOI] [PubMed] [Google Scholar]
  • 51.Elston GN. Cortex, cognition and the cell: new insights into the pyramidal neuron and prefrontal function. Cereb Cortex. 2003;13:1124–38. doi: 10.1093/cercor/bhg093. [DOI] [PubMed] [Google Scholar]
  • 52.Goldman-Rakic PS. Regional and cellular fractionation of working memory. Proceedings of the National Academy of Sciences of the United States of America. 1996;93:13473–80. doi: 10.1073/pnas.93.24.13473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Goldman-Rakic PS. The prefrontal landscape: implications of functional architecture for understanding human mentation and the central executive. Philosophical Transactions of the Royal Society of London - Series B: Biological Sciences. 1996;351:1445–53. doi: 10.1098/rstb.1996.0129. [DOI] [PubMed] [Google Scholar]
  • 54.Stanwood GD, Washington RA, Levitt P. Identification of a sensitive period of prenatal cocaine exposure that alters the development of the anterior cingulate cortex. Cerebral Cortex. 2001;11:430–40. doi: 10.1093/cercor/11.5.430. [DOI] [PubMed] [Google Scholar]
  • 55.Crandall JE, Hackett HE, Tobet SA, Kosofsky BE, Bhide PG. Cocaine exposure decreases GABA neuron migration from the ganglionic eminence to the cerebral cortex in embryonic mice. Cereb Cortex. 2004;14:665–75. doi: 10.1093/cercor/bhh027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Gressens P, Kosofsky BE, Evrard P. Cocaine-induced disturbances of corticogenesis in the developing murine brain. Neuroscience Letters. 1992;140:113–6. doi: 10.1016/0304-3940(92)90694-3. [DOI] [PubMed] [Google Scholar]
  • 57.Lidow MS. Prenatal cocaine exposure adversely affects development of the primate cerebral cortex. Synapse. 1995;21:332–41. doi: 10.1002/syn.890210408. [DOI] [PubMed] [Google Scholar]
  • 58.Lidow MS, Song ZM. Effect of cocaine on cell proliferation in the cerebral wall of monkey fetuses. Cereb Cortex. 2001;11:545–51. doi: 10.1093/cercor/11.6.545. [DOI] [PubMed] [Google Scholar]
  • 59.Ren JQ, Malanga CJ, Tabit E, Kosofsky BE. Neuropathological consequences of prenatal cocaine exposure in the mouse. Int J Dev Neurosci. 2004;22:309–20. doi: 10.1016/j.ijdevneu.2004.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lidow MS, Song ZM. Primates exposed to cocaine in utero display reduced density and number of cerebral cortical neurons. J Comp Neurol. 2001;435:263–75. doi: 10.1002/cne.1028. [DOI] [PubMed] [Google Scholar]
  • 61.Crandall JE, et al. Dopamine receptor activation modulates GABA neuron migration from the basal forebrain to the cerebral cortex. J Neurosci. 2007;27:3813–22. doi: 10.1523/JNEUROSCI.5124-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ohtani N, Goto T, Waeber C, Bhide PG. Dopamine modulates cell cycle in the lateral ganglionic eminence. J Neurosci. 2003;23:2840–50. doi: 10.1523/JNEUROSCI.23-07-02840.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Harvey JA, et al. Effects of prenatal exposure to cocaine on the developing brain: anatomical, chemical, physiological and behavioral consequences. Neurotox Res. 2001;3:117–43. doi: 10.1007/BF03033234. [DOI] [PubMed] [Google Scholar]
  • 64.Stanwood GD, Levitt P. Repeated i.v. cocaine exposure produces long-lasting behavioral sensitization in pregnant adults, but behavioral tolerance in their offspring. Neuroscience. 2003;122:579–83. doi: 10.1016/j.neuroscience.2003.08.029. [DOI] [PubMed] [Google Scholar]
  • 65.Johnston LD, O'Malley PM, Bachman JG, Schulenberg JE. Monitoring the Future national survey results on drug use, 1975-2007. Volume I: Secondary school students. NIH Publication No. 08-6418A. 2008 [Google Scholar]
  • 66.Smith LM, et al. Prenatal methamphetamine use and neonatal neurobehavioral outcome. Neurotoxicol Teratol. 2008;30:20–8. doi: 10.1016/j.ntt.2007.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Smith LM, et al. The infant development, environment, and lifestyle study: effects of prenatal methamphetamine exposure, polydrug exposure, and poverty on intrauterine growth. Pediatrics. 2006;118:1149–56. doi: 10.1542/peds.2005-2564. [DOI] [PubMed] [Google Scholar]
  • 68.Petrou S, Sach T, Davidson L. The long-term costs of preterm birth and low birth weight: results of a systematic review. Child Care Health Dev. 2001;27:97–115. doi: 10.1046/j.1365-2214.2001.00203.x. [DOI] [PubMed] [Google Scholar]
  • 69.Chaikind S, Corman H. The impact of low birthweight on special education costs. J Health Econ. 1991;10:291–311. doi: 10.1016/0167-6296(91)90031-h. [DOI] [PubMed] [Google Scholar]
  • 70.Cernerud L, Eriksson M, Jonsson B, Steneroth G, Zetterstrom R. Amphetamine addiction during pregnancy: 14-year follow-up of growth and school performance. Acta Paediatr. 1996;85:204–8. doi: 10.1111/j.1651-2227.1996.tb13993.x. [DOI] [PubMed] [Google Scholar]
  • 71.Chang L, et al. Smaller subcortical volumes and cognitive deficits in children with prenatal methamphetamine exposure. Psychiatry Res. 2004;132:95–106. doi: 10.1016/j.pscychresns.2004.06.004. [DOI] [PubMed] [Google Scholar]
  • 72.Chang L, Alicata D, Ernst T, Volkow N. Structural and metabolic brain changes in the striatum associated with methamphetamine abuse. Addiction. 2007;102(Suppl 1):16–32. doi: 10.1111/j.1360-0443.2006.01782.x. [DOI] [PubMed] [Google Scholar]
  • 73.Derauf C, et al. Demographic and psychosocial characteristics of mothers using methamphetamine during pregnancy: preliminary results of the infant development, environment, and lifestyle study (IDEAL) Am J Drug Alcohol Abuse. 2007;33:281–9. doi: 10.1080/00952990601175029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Melo P, Rodrigues LG, Silva MC, Tavares MA. Effects of prenatal exposure to methamphetamine on the development of the rat retina. Ann N Y Acad Sci. 2006;1074:590–603. doi: 10.1196/annals.1369.058. [DOI] [PubMed] [Google Scholar]
  • 75.Melo P, Moreno VZ, Vazquez SP, Pinazo-Duran MD, Tavares MA. Myelination changes in the rat optic nerve after prenatal exposure to methamphetamine. Brain Res. 2006;1106:21–9. doi: 10.1016/j.brainres.2006.05.020. [DOI] [PubMed] [Google Scholar]
  • 76.Slamberova R, Pometlova M, Charousova P. Postnatal development of rat pups is altered by prenatal methamphetamine exposure. Prog Neuropsychopharmacol Biol Psychiatry. 2006;30:82–8. doi: 10.1016/j.pnpbp.2005.06.006. [DOI] [PubMed] [Google Scholar]
  • 77.Slamberova R, Pometlova M, Syllabova L, Mancuskova M. Learning in the Place navigation task, not the New-learning task, is altered by prenatal methamphetamine exposure. Brain Res Dev Brain Res. 2005;157:217–9. doi: 10.1016/j.devbrainres.2005.04.005. [DOI] [PubMed] [Google Scholar]
  • 78.Nasif FJ, Cuadra GR, Ramirez OA. Permanent alteration of central noradrenergic system by prenatally administered amphetamine. Brain Res Dev Brain Res. 1999;112:181–8. doi: 10.1016/s0165-3806(98)00166-7. [DOI] [PubMed] [Google Scholar]
  • 79.Gomes-da-Silva J, et al. Prenatal exposure to methamphetamine in the rat: ontogeny of tyrosine hydroxylase mRNA expression in mesencephalic dopaminergic neurons. Ann N Y Acad Sci. 2002;965:68–77. [PubMed] [Google Scholar]
  • 80.Cabrera TM, Levy AD, Li Q, van de Kar LD, Battaglia G. Prenatal methamphetamine attenuates serotonin mediated renin secretion in male and female rat progeny: evidence for selective long-term dysfunction of serotonin pathways in brain. Synapse. 1993;15:198–208. doi: 10.1002/syn.890150305. [DOI] [PubMed] [Google Scholar]
  • 81.Slamberova R, Pometlova M, Rokyta R. Effect of methamphetamine exposure during prenatal and preweaning periods lasts for generations in rats. Dev Psychobiol. 2007;49:312–22. doi: 10.1002/dev.20203. [DOI] [PubMed] [Google Scholar]
  • 82.Rogers JM. Tobacco and pregnancy: overview of exposures and effects. Birth Defects Res C Embryo Today. 2008;84:1–15. doi: 10.1002/bdrc.20119. [DOI] [PubMed] [Google Scholar]
  • 83.Hollins K. Consequences of antenatal mental health problems for child health and development. Curr Opin Obstet Gynecol. 2007;19:568–72. doi: 10.1097/GCO.0b013e3282f1bf28. [DOI] [PubMed] [Google Scholar]
  • 84.Hack M. Young adult outcomes of very-low-birth-weight children. Semin Fetal Neonatal Med. 2006;11:127–37. doi: 10.1016/j.siny.2005.11.007. [DOI] [PubMed] [Google Scholar]
  • 85.Gianni ML, et al. Twelve-month neurofunctional assessment and cognitive performance at 36 months of age in extremely low birth weight infants. Pediatrics. 2007;120:1012–9. doi: 10.1542/peds.2006-3364. [DOI] [PubMed] [Google Scholar]
  • 86.Lambe M, Hultman C, Torrang A, Maccabe J, Cnattingius S. Maternal smoking during pregnancy and school performance at age 15. Epidemiology. 2006;17:524–30. doi: 10.1097/01.ede.0000231561.49208.be. [DOI] [PubMed] [Google Scholar]
  • 87.George L, Granath F, Johansson AL, Anneren G, Cnattingius S. Environmental tobacco smoke and risk of spontaneous abortion. Epidemiology. 2006;17:500–5. doi: 10.1097/01.ede.0000229984.53726.33. [DOI] [PubMed] [Google Scholar]
  • 88.Cnattingius S. The epidemiology of smoking during pregnancy: smoking prevalence, maternal characteristics, and pregnancy outcomes. Nicotine Tob Res. 2004;6(Suppl 2):S125–40. doi: 10.1080/14622200410001669187. [DOI] [PubMed] [Google Scholar]
  • 89.DiFranza JR, Aligne CA, Weitzman M. Prenatal and postnatal environmental tobacco smoke exposure and children's health. Pediatrics. 2004;113:1007–15. [PubMed] [Google Scholar]
  • 90.Fried PA, Watkinson B, Gray R. Differential effects on cognitive functioning in 13- to 16-year-olds prenatally exposed to cigarettes and marihuana. Neurotoxicol Teratol. 2003;25:427–36. doi: 10.1016/s0892-0362(03)00029-1. [DOI] [PubMed] [Google Scholar]
  • 91.Fried PA, Watkinson B. Differential effects on facets of attention in adolescents prenatally exposed to cigarettes and marihuana. Neurotoxicol Teratol. 2001;23:421–30. doi: 10.1016/s0892-0362(01)00160-x. [DOI] [PubMed] [Google Scholar]
  • 92.Makin J, Fried PA, Watkinson BA. comparison of active and passive smoking during pregnancy: long-term effects. Neurotoxicol Teratol. 1991;13:5–12. doi: 10.1016/0892-0362(91)90021-n. [DOI] [PubMed] [Google Scholar]
  • 93.Eskenazi B, Prehn AW, Christianson RE. Passive and active maternal smoking as measured by serum cotinine: the effect on birthweight. Am J Public Health. 1995;85:395–8. doi: 10.2105/ajph.85.3.395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Langley K, Rice F, van den Bree MB, Thapar A. Maternal smoking during pregnancy as an environmental risk factor for attention deficit hyperactivity disorder behaviour. A review. Minerva Pediatr. 2005;57:359–71. [PubMed] [Google Scholar]
  • 95.Gaither KH, Huber LR, Thompson ME, Huet-Hudson YM. Does the Use of Nicotine Replacement Therapy During Pregnancy Affect Pregnancy Outcomes? Matern Child Health J. 2008 doi: 10.1007/s10995-008-0361-1. [DOI] [PubMed] [Google Scholar]
  • 96.Schroeder DR, et al. Nicotine patch use in pregnant smokers: smoking abstinence and delivery outcomes. J Matern Fetal Neonatal Med. 2002;11:100–7. doi: 10.1080/jmf.11.2.100.107. [DOI] [PubMed] [Google Scholar]
  • 97.Pauly JR, Slotkin TA. Maternal tobacco smoking, nicotine replacement and neurobehavioural development. Acta Paediatr. 2008 doi: 10.1111/j.1651-2227.2008.00852.x. [DOI] [PubMed] [Google Scholar]
  • 98.Slotkin TA. If nicotine is a developmental neurotoxicant in animal studies, dare we recommend nicotine replacement therapy in pregnant women and adolescents? Neurotoxicol Teratol. 2008;30:1–19. doi: 10.1016/j.ntt.2007.09.002. [DOI] [PubMed] [Google Scholar]
  • 99.Sarasin A, et al. Adrenal-mediated rather than direct effects of nicotine as a basis of altered sex steroid synthesis in fetal and neonatal rat. Reprod Toxicol. 2003;17:153–62. doi: 10.1016/s0890-6238(02)00119-3. [DOI] [PubMed] [Google Scholar]
  • 100.Dwyer JB, Broide RS, Leslie FM. Nicotine and brain development. Birth Defects Res C Embryo Today. 2008;84:30–44. doi: 10.1002/bdrc.20118. [DOI] [PubMed] [Google Scholar]
  • 101.Navarro HA, et al. Prenatal exposure to nicotine impairs nervous system development at a dose which does not affect viability or growth. Brain Res Bull. 1989;23:187–92. doi: 10.1016/0361-9230(89)90146-9. [DOI] [PubMed] [Google Scholar]
  • 102.Roy TS, Seidler FJ, Slotkin TA. Prenatal nicotine exposure evokes alterations of cell structure in hippocampus and somatosensory cortex. J Pharmacol Exp Ther. 2002;300:124–33. doi: 10.1124/jpet.300.1.124. [DOI] [PubMed] [Google Scholar]
  • 103.Paz R, Barsness B, Martenson T, Tanner D, Allan AM. Behavioral teratogenicity induced by nonforced maternal nicotine consumption. Neuropsychopharmacology. 2007;32:693–9. doi: 10.1038/sj.npp.1301066. [DOI] [PubMed] [Google Scholar]
  • 104.Levin ED, et al. Increased nicotine self-administration following prenatal exposure in female rats. Pharmacol Biochem Behav. 2006;85:669–74. doi: 10.1016/j.pbb.2006.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Vaglenova J, Birru S, Pandiella NM, Breese CR. An assessment of the long-term developmental and behavioral teratogenicity of prenatal nicotine exposure. Behav Brain Res. 2004;150:159–70. doi: 10.1016/j.bbr.2003.07.005. [DOI] [PubMed] [Google Scholar]
  • 106.Slotkin TA. Fetal nicotine or cocaine exposure: which one is worse? Journal of Pharmacology & Experimental Therapeutics. 1998;285:931–45. [PubMed] [Google Scholar]
  • 107.Sarter M, Hasselmo ME, Bruno JP, Givens B. Unraveling the attentional functions of cortical cholinergic inputs: interactions between signal-driven and cognitive modulation of signal detection. Brain Res Brain Res Rev. 2005;48:98–111. doi: 10.1016/j.brainresrev.2004.08.006. [DOI] [PubMed] [Google Scholar]
  • 108.Liang K, et al. Neonatal nicotine exposure impairs nicotinic enhancement of central auditory processing and auditory learning in adult rats. Eur J Neurosci. 2006;24:857–66. doi: 10.1111/j.1460-9568.2006.04945.x. [DOI] [PubMed] [Google Scholar]
  • 109.Barbieri RL, Gochberg J, Ryan KJ. Nicotine, cotinine, and anabasine inhibit aromatase in human trophoblast in vitro. J Clin Invest. 1986;77:1727–33. doi: 10.1172/JCI112494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Fried PA, James DS, Watkinson B. Growth and pubertal milestones during adolescence in offspring prenatally exposed to cigarettes and marihuana. Neurotoxicol Teratol. 2001;23:431–6. doi: 10.1016/s0892-0362(01)00161-1. [DOI] [PubMed] [Google Scholar]
  • 111.Treiman DM. GABAergic mechanisms in epilepsy. Epilepsia. 2001;42(Suppl 3):8–12. doi: 10.1046/j.1528-1157.2001.042suppl.3008.x. [DOI] [PubMed] [Google Scholar]
  • 112.Huang ZJ, Di Cristo G, Ango F. Development of GABA innervation in the cerebral and cerebellar cortices. Nat Rev Neurosci. 2007;8:673–86. doi: 10.1038/nrn2188. [DOI] [PubMed] [Google Scholar]
  • 113.Feng MJ, Yan SE, Yan QS. Effects of prenatal alcohol exposure on brain-derived neurotrophic factor and its receptor tyrosine kinase B in offspring. Brain Res. 2005;1042:125–32. doi: 10.1016/j.brainres.2005.02.017. [DOI] [PubMed] [Google Scholar]
  • 114.Miller MW. Expression of transforming growth factor-beta in developing rat cerebral cortex: effects of prenatal exposure to ethanol. J Comp Neurol. 2003;460:410–24. doi: 10.1002/cne.10658. [DOI] [PubMed] [Google Scholar]
  • 115.Borodinsky LN, et al. GABA-induced neurite outgrowth of cerebellar granule cells is mediated by GABA(A) receptor activation, calcium influx and CaMKII and erk1/2 pathways. J Neurochem. 2003;84:1411–20. doi: 10.1046/j.1471-4159.2003.01638.x. [DOI] [PubMed] [Google Scholar]
  • 116.Schwartz JP. Neurotransmitters as neurotrophic factors: a new set of functions. Int Rev Neurobiol. 1992;34:1–23. doi: 10.1016/s0074-7742(08)60096-3. [DOI] [PubMed] [Google Scholar]
  • 117.Schwartz ML, Meinecke DL. Early expression of GABA-containing neurons in the prefrontal and visual cortices of rhesus monkeys. Cereb Cortex. 1992;2:16–37. doi: 10.1093/cercor/2.1.16. [DOI] [PubMed] [Google Scholar]
  • 118.Walker A, Rosenberg M, Balaban-Gil K. Neurodevelopmental and neurobehavioral sequelae of selected substances of abuse and psychiatric medications in utero. Child Adolesc Psychiatr Clin N Am. 1999;8:845–67. [PubMed] [Google Scholar]
  • 119.Kosofsky BE. Specificity of neurobehavioral outcomes associated with prenatal alcohol exposure. J Womens Health. 1998;7:603–4. [PubMed] [Google Scholar]
  • 120.Chiriboga CA. Fetal alcohol and drug effects. Neurologist. 2003;9:267–79. doi: 10.1097/01.nrl.0000094941.96358.d1. [DOI] [PubMed] [Google Scholar]
  • 121.Bada HS, et al. Low birth weight and preterm births: etiologic fraction attributable to prenatal drug exposure. J Perinatol. 2005;25:631–7. doi: 10.1038/sj.jp.7211378. [DOI] [PubMed] [Google Scholar]
  • 122.Loebstein R, Koren G. Pregnancy outcome and neurodevelopment of children exposed in utero to psychoactive drugs: the Motherisk experience. J Psychiatry Neurosci. 1997;22:192–6. [PMC free article] [PubMed] [Google Scholar]
  • 123.Fried PA, Watkinson B, Gray R. A follow-up study of attentional behavior in 6-year-old children exposed prenatally to marihuana, cigarettes, and alcohol. Neurotoxicol Teratol. 1992;14:299–311. doi: 10.1016/0892-0362(92)90036-a. [DOI] [PubMed] [Google Scholar]
  • 124.Linnet KM, et al. Maternal lifestyle factors in pregnancy risk of attention deficit hyperactivity disorder and associated behaviors: review of the current evidence. Am J Psychiatry. 2003;160:1028–40. doi: 10.1176/appi.ajp.160.6.1028. [DOI] [PubMed] [Google Scholar]
  • 125.Williams JH, Ross L. Consequences of prenatal toxin exposure for mental health in children and adolescents: a systematic review. Eur Child Adolesc Psychiatry. 2007;16:243–53. doi: 10.1007/s00787-006-0596-6. [DOI] [PubMed] [Google Scholar]
  • 126.Snow ME, Keiver K. Prenatal ethanol exposure disrupts the histological stages of fetal bone development. Bone. 2007;41:181–7. doi: 10.1016/j.bone.2007.04.182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Simpson ME, Duggal S, Keiver K. Prenatal ethanol exposure has differential effects on fetal growth and skeletal ossification. Bone. 2005;36:521–32. doi: 10.1016/j.bone.2004.11.011. [DOI] [PubMed] [Google Scholar]
  • 128.Johnston MC, Bronsky PT. Prenatal craniofacial development: new insights on normal and abnormal mechanisms. Crit Rev Oral Biol Med. 1995;6:368–422. doi: 10.1177/10454411950060040601. [DOI] [PubMed] [Google Scholar]
  • 129.Randall CL, Taylor WJ. Prenatal ethanol exposure in mice: teratogenic effects. Teratology. 1979;19:305–11. doi: 10.1002/tera.1420190305. [DOI] [PubMed] [Google Scholar]
  • 130.Miller MW, Dow-Edwards DL. Structural and metabolic alterations in rat cerebral cortex induced by prenatal exposure to ethanol. Brain Res. 1988;474:316–26. doi: 10.1016/0006-8993(88)90445-3. [DOI] [PubMed] [Google Scholar]
  • 131.Miller MW. Effect of prenatal exposure to ethanol on glutamate and GABA immunoreactivity in macaque somatosensory and motor cortices: critical timing of exposure. Neuroscience. 2006;138:97–107. doi: 10.1016/j.neuroscience.2005.10.060. [DOI] [PubMed] [Google Scholar]
  • 132.Miller MW. Effect of early exposure to ethanol on the protein and DNA contents of specific brain regions in the rat. Brain Res. 1996;734:286–94. [PubMed] [Google Scholar]
  • 133.Mooney SM, Miller MW. Episodic exposure to ethanol during development differentially affects brainstem nuclei in the macaque. J Neurocytol. 2001;30:973–82. doi: 10.1023/a:1021832522701. [DOI] [PubMed] [Google Scholar]
  • 134.Barrow Heaton MB, et al. Prenatal ethanol exposure reduces spinal cord motoneuron number in the fetal rat but does not affect GDNF target tissue protein. Dev Neurosci. 1999;21:444–52. doi: 10.1159/000017412. [DOI] [PubMed] [Google Scholar]
  • 135.Shetty AK, Phillips DE. Effects of prenatal ethanol exposure on the development of Bergmann glia and astrocytes in the rat cerebellum: an immunohistochemical study. J Comp Neurol. 1992;321:19–32. doi: 10.1002/cne.903210103. [DOI] [PubMed] [Google Scholar]
  • 136.Redila VA, et al. Hippocampal cell proliferation is reduced following prenatal ethanol exposure but can be rescued with voluntary exercise. Hippocampus. 2006;16:305–11. doi: 10.1002/hipo.20164. [DOI] [PubMed] [Google Scholar]
  • 137.Ozer E, Sarioglu S, Gure A. Effects of prenatal ethanol exposure on neuronal migration, neuronogenesis and brain myelination in the mice brain. Clin Neuropathol. 2000;19:21–5. [PubMed] [Google Scholar]
  • 138.Honse Y, Nixon KM, Browning MD, Leslie SW. Cell surface expression of NR1 splice variants and NR2 subunits is modified by prenatal ethanol exposure. Neuroscience. 2003;122:689–98. doi: 10.1016/s0306-4522(03)00603-1. [DOI] [PubMed] [Google Scholar]
  • 139.Hughes PD, Wilson WR, Leslie SW. Effect of gestational ethanol exposure on the NMDA receptor complex in rat forebrain: from gene transcription to cell surface. Brain Res Dev Brain Res. 2001;129:135–45. doi: 10.1016/s0165-3806(01)00192-4. [DOI] [PubMed] [Google Scholar]
  • 140.Zhang X, Sliwowska JH, Weinberg J. Prenatal alcohol exposure and fetal programming: effects on neuroendocrine and immune function. Exp Biol Med (Maywood) 2005;230:376–88. doi: 10.1177/15353702-0323006-05. [DOI] [PubMed] [Google Scholar]
  • 141.Wilcoxon JS, Kuo AG, Disterhoft JF, Redei EE. Behavioral deficits associated with fetal alcohol exposure are reversed by prenatal thyroid hormone treatment: a role for maternal thyroid hormone deficiency in FAE. Mol Psychiatry. 2005;10:961–71. doi: 10.1038/sj.mp.4001694. [DOI] [PubMed] [Google Scholar]
  • 142.Champagne F, Meaney MJ. Like mother, like daughter: evidence for non-genomic transmission of parental behavior and stress responsivity. Prog Brain Res. 2001;133:287–302. doi: 10.1016/s0079-6123(01)33022-4. [DOI] [PubMed] [Google Scholar]
  • 143.Kallen B, Otterblad Olausson P. Antidepressant drugs during pregnancy and infant congenital heart defect. Reprod Toxicol. 2006;21:221–2. doi: 10.1016/j.reprotox.2005.11.006. [DOI] [PubMed] [Google Scholar]
  • 144.Kallen BA, Otterblad Olausson P. Maternal drug use in early pregnancy and infant cardiovascular defect. Reprod Toxicol. 2003;17:255–61. doi: 10.1016/s0890-6238(03)00012-1. [DOI] [PubMed] [Google Scholar]
  • 145.Buznikov GA, Shmukler YB, Lauder JM. From oocyte to neuron: do neurotransmitters function in the same way throughout development? Cellular & Molecular Neurobiology. 1996;16:537–59. doi: 10.1007/BF02152056. [DOI] [PubMed] [Google Scholar]
  • 146.Lauder JM. Hormonal and humoral influences on brain development. Psychoneuroendocrinology. 1983;8:121–55. doi: 10.1016/0306-4530(83)90053-7. [DOI] [PubMed] [Google Scholar]
  • 147.Whitaker-Azmitia PM, Druse M, Walker P, Lauder JM. Serotonin as a developmental signal. Behavioural Brain Research. 1996;73:19–29. doi: 10.1016/0166-4328(96)00071-x. [DOI] [PubMed] [Google Scholar]
  • 148.Bonnin A, Peng W, Hewlitt W, Levitt P. Expression mapping of 5-HT1 serotonin receptor subtypes during fetal and early postnatal mouse forebrain development. Neuroscience. 2006 doi: 10.1016/j.neuroscience.2006.04.036. in press. [DOI] [PubMed] [Google Scholar]
  • 149.Bonnin A, Torii M, Wang L, Rakic P, Levitt P. Serotonin modulates the response of embryonic thalamocortical axons to netrin-1. Nat Neurosci. 2007;10:588–97. doi: 10.1038/nn1896. [DOI] [PubMed] [Google Scholar]
  • 150.Lambe EK, Krimer LS. Goldman-Rakic, P.S. Differential postnatal development of catecholamine and serotonin inputs to identified neurons in prefrontal cortex of rhesus monkey. J Neurosci. 2000;20:8780–7. doi: 10.1523/JNEUROSCI.20-23-08780.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Whitaker-Azmitia PM, Lauder JM, Shemmer A, Azmitia EC. Postnatal changes in serotonin receptors following prenatal alterations in serotonin levels: further evidence for functional fetal serotonin receptors. Brain Res. 1987;430:285–9. doi: 10.1016/0165-3806(87)90161-1. [DOI] [PubMed] [Google Scholar]
  • 152.Persico AM, Di Pino G, Levitt P. Multiple receptors mediate the trophic effects of serotonin on ventroposterior thalamic neurons in vitro. Brain Res. 2006;1095:17–25. doi: 10.1016/j.brainres.2006.04.006. [DOI] [PubMed] [Google Scholar]
  • 153.Gross C, et al. Serotonin1A receptor acts during development to establish normal anxiety-like behaviour in the adult. Nature. 2002;416:396–400. doi: 10.1038/416396a. [DOI] [PubMed] [Google Scholar]
  • 154.Ansorge MS, Zhou M, Lira A, Hen R, Gingrich JA. Early-life blockade of the 5-HT transporter alters emotional behavior in adult mice. Science. 2004;306:879–81. doi: 10.1126/science.1101678. [DOI] [PubMed] [Google Scholar]
  • 155.Maschi S, et al. Neonatal outcome following pregnancy exposure to antidepressants: a prospective controlled cohort study. Bjog. 2008;115:283–9. doi: 10.1111/j.1471-0528.2007.01518.x. [DOI] [PubMed] [Google Scholar]
  • 156.Andrade SE, et al. Use of antidepressant medications during pregnancy: a multisite study. Am J Obstet Gynecol. 2008;198:e1–5. doi: 10.1016/j.ajog.2007.07.036. 194. [DOI] [PubMed] [Google Scholar]
  • 157.Pearson KH, et al. Birth outcomes following prenatal exposure to antidepressants. J Clin Psychiatry. 2007;68:1284–9. doi: 10.4088/jcp.v68n0817. [DOI] [PubMed] [Google Scholar]
  • 158.Oberlander TF, et al. Infant serotonin transporter (SLC6A4) promoter genotype is associated with adverse neonatal outcomes after prenatal exposure to serotonin reuptake inhibitor medications. Mol Psychiatry. 2008;13:65–73. doi: 10.1038/sj.mp.4002007. [DOI] [PubMed] [Google Scholar]
  • 159.Einarson A, et al. Evaluation of the risk of congenital cardiovascular defects associated with use of paroxetine during pregnancy. Am J Psychiatry. 2008;165:749–52. doi: 10.1176/appi.ajp.2007.07060879. [DOI] [PubMed] [Google Scholar]
  • 160.Zuo J, et al. Distinct neurobehavioral consequences of prenatal exposure to sulpiride (SUL) and risperidone (RIS) in rats. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:387–97. doi: 10.1016/j.pnpbp.2007.09.005. [DOI] [PubMed] [Google Scholar]
  • 161.Singh Y, Jaiswal AK, Singh M, Bhattacharya SK. Effect of prenatal haloperidol administration on anxiety patterns in rats. Indian J Exp Biol. 1997;35:1284–90. [PubMed] [Google Scholar]
  • 162.Castro R, Brito B, Segovia J, Martin-Trujillo JM, Notario V. Prenatal haloperidol induces a selective reduction in the expression of plasticity-related genes in neonate rat forebrain. Brain Res Mol Brain Res. 1994;26:74–80. doi: 10.1016/0169-328x(94)90076-0. [DOI] [PubMed] [Google Scholar]
  • 163.Leonard BE. Effect of psychotropic drugs administered to pregnant rats on the behaviour of the offspring. Neuropharmacology. 1981;20:1237–42. [PubMed] [Google Scholar]
  • 164.Miller JC, Friedhoff AJ. Prenatal neurotransmitter programming of postnatal receptor function. Prog Brain Res. 1988;73:509–22. doi: 10.1016/S0079-6123(08)60523-3. [DOI] [PubMed] [Google Scholar]
  • 165.Trixler M, Gati A, Fekete S, Tenyi T. Use of antipsychotics in the management of schizophrenia during pregnancy. Drugs. 2005;65:1193–206. doi: 10.2165/00003495-200565090-00002. [DOI] [PubMed] [Google Scholar]
  • 166.Gentile S. Clinical utilization of atypical antipsychotics in pregnancy and lactation. Ann Pharmacother. 2004;38:1265–71. doi: 10.1345/aph.1D485. [DOI] [PubMed] [Google Scholar]
  • 167.Landmark CJ. Targets for antiepileptic drugs in the synapse. Med Sci Monit. 2007;13:RA1–7. [PubMed] [Google Scholar]
  • 168.Gottlicher M. Valproic acid: an old drug newly discovered as inhibitor of histone deacetylases. Ann Hematol. 2004;83(Suppl 1):S91–2. doi: 10.1007/s00277-004-0850-2. [DOI] [PubMed] [Google Scholar]
  • 169.Carrim ZI, McKay L, Sidiki SS, Lavy TE. Early intervention for the ocular and neurodevelopmental sequelae of Fetal Valproate Syndrome. J Paediatr Child Health. 2007;43:643–5. doi: 10.1111/j.1440-1754.2007.01176.x. [DOI] [PubMed] [Google Scholar]
  • 170.Duncan S. Teratogenesis of sodium valproate. Curr Opin Neurol. 2007;20:175–80. doi: 10.1097/WCO.0b013e32805866fb. [DOI] [PubMed] [Google Scholar]
  • 171.Schneider T, et al. Gender-specific behavioral and immunological alterations in an animal model of autism induced by prenatal exposure to valproic acid. Psychoneuroendocrinology. 2008;33:728–40. doi: 10.1016/j.psyneuen.2008.02.011. [DOI] [PubMed] [Google Scholar]
  • 172.Schneider T, Przewlocki R. Behavioral alterations in rats prenatally exposed to valproic acid: animal model of autism. Neuropsychopharmacology. 2005;30:80–9. doi: 10.1038/sj.npp.1300518. [DOI] [PubMed] [Google Scholar]
  • 173.Rinaldi T, Kulangara K, Antoniello K, Markram H. Elevated NMDA receptor levels and enhanced postsynaptic long-term potentiation induced by prenatal exposure to valproic acid. Proc Natl Acad Sci U S A. 2007;104:13501–6. doi: 10.1073/pnas.0704391104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Rinaldi T, Silberberg G, Markram H. Hyperconnectivity of local neocortical microcircuitry induced by prenatal exposure to valproic acid. Cereb Cortex. 2008;18:763–70. doi: 10.1093/cercor/bhm117. [DOI] [PubMed] [Google Scholar]
  • 175.Travis BE, McCullough JM. Pharmacotherapy of preterm labor. Pharmacotherapy. 1993;13:28–36. [PubMed] [Google Scholar]
  • 176.Zerrate MC, et al. Neuroinflammation and behavioral abnormalities after neonatal terbutaline treatment in rats: implications for autism. J Pharmacol Exp Ther. 2007;322:16–22. doi: 10.1124/jpet.107.121483. [DOI] [PubMed] [Google Scholar]
  • 177.Meyer A, Seidler FJ, Aldridge JE, Slotkin TA. Developmental exposure to terbutaline alters cell signaling in mature rat brain regions and augments the effects of subsequent neonatal exposure to the organophosphorus insecticide chlorpyrifos. Toxicol Appl Pharmacol. 2005;203:154–66. doi: 10.1016/j.taap.2004.08.005. [DOI] [PubMed] [Google Scholar]
  • 178.Rhodes MC, et al. Terbutaline is a developmental neurotoxicant: effects on neuroproteins and morphology in cerebellum, hippocampus, and somatosensory cortex. J Pharmacol Exp Ther. 2004;308:529–37. doi: 10.1124/jpet.103.060095. [DOI] [PubMed] [Google Scholar]
  • 179.Pitzer M, Schmidt MH, Esser G, Laucht M. Child development after maternal tocolysis with beta-sympathomimetic drugs. Child Psychiatry Hum Dev. 2001;31:165–82. doi: 10.1023/a:1026419720410. [DOI] [PubMed] [Google Scholar]
  • 180.Hadders-Algra M, Touwen BC, Huisjes HJ. Long-term follow-up of children prenatally exposed to ritodrine. Br J Obstet Gynaecol. 1986;93:156–61. doi: 10.1111/j.1471-0528.1986.tb07880.x. [DOI] [PubMed] [Google Scholar]
  • 181.Connors SL, et al. beta2-adrenergic receptor activation and genetic polymorphisms in autism: data from dizygotic twins. J Child Neurol. 2005;20:876–84. doi: 10.1177/08830738050200110401. [DOI] [PubMed] [Google Scholar]
  • 182.Thornton JG. Maintenance tocolysis. Bjog. 2005;112(Suppl 1):118–21. doi: 10.1111/j.1471-0528.2005.00599.x. [DOI] [PubMed] [Google Scholar]
  • 183.Reese S, Gandy O, Grant A, editors. Framing Public Life: Perspectives on Media and Our Understanding of the Social World. Lawrence Erlbaum Assoc; Philadelphia: 1993. [Google Scholar]
  • 184.Entman RM. Framing: Toward Clarification of a Fractured Paradigm. Journal of Communication. 1993;43:51–58. [Google Scholar]
  • 185.Entman RM. Projections of Power. University of Chicago Press; 2004. [Google Scholar]
  • 186.Bales SN. Communicating Early Childhood Education: Using Strategic Frame Analysis to Shape Dialogue. Bulletin of Zero to Three. 1999;19 [Google Scholar]
  • 187.National Advisory Mental Health Council Transformative Neurodevelopmental Research in Mental Illness. Report of the National Advisory Mental Health Council's Workgroup. 2008 [Google Scholar]
  • 188.Whitaker-Azmitia PM. Serotonin and brain development: role in human developmental diseases. Brain Res Bull. 2001;56:479–85. doi: 10.1016/s0361-9230(01)00615-3. [DOI] [PubMed] [Google Scholar]
  • 189.Represa A, Ben-Ari Y. Trophic actions of GABA on neuronal development. Trends Neurosci. 2005;28:278–83. doi: 10.1016/j.tins.2005.03.010. [DOI] [PubMed] [Google Scholar]
  • 190.Nguyen L, et al. Neurotransmitters as early signals for central nervous system development. Cell Tissue Res. 2001;305:187–202. doi: 10.1007/s004410000343. [DOI] [PubMed] [Google Scholar]
  • 191.Lauder JM, Schambra UB. Morphogenetic roles of acetylcholine. Environ Health Perspect. 1999;107(Suppl 1):65–9. doi: 10.1289/ehp.99107s165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Levitt P, Harvey JA, Friedman E, Simansky K, Murphy EH. New evidence for neurotransmitter influences on brain development. Trends in Neurosciences. 1997;20:269–74. doi: 10.1016/s0166-2236(96)01028-4. [DOI] [PubMed] [Google Scholar]
  • 193.Song ZM, et al. D1 dopamine receptor regulation of microtubule-associated protein-2 phosphorylation in developing cerebral cortical neurons. J Neurosci. 2002;22:6092–105. doi: 10.1523/JNEUROSCI.22-14-06092.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Lauder JM, Wallace JA, Krebs H. Roles for serotonin in neuroembryogenesis. Adv Exp Med Biol. 1981;133:477–506. doi: 10.1007/978-1-4684-3860-4_28. [DOI] [PubMed] [Google Scholar]
  • 195.Behar TN, Schaffner AE, Scott CA, Greene CL, Barker JL. GABA receptor antagonists modulate postmitotic cell migration in slice cultures of embryonic rat cortex. Cerebral Cortex. 2000;10:899–909. doi: 10.1093/cercor/10.9.899. [DOI] [PubMed] [Google Scholar]
  • 196.Brazel CY, Nunez JL, Yang Z, Levison SW. Glutamate enhances survival and proliferation of neural progenitors derived from the subventricular zone. Neuroscience. 2005;131:55–65. doi: 10.1016/j.neuroscience.2004.10.038. [DOI] [PubMed] [Google Scholar]
  • 197.Budetti PP, et al. George Washington, University; Washington, D.C.: 1993. [Google Scholar]
  • 198.Poland ML, Dombrowski MP, Ager JW, Sokol RJ. Punishing pregnant drug users: enhancing the flight from care. Drug and Alcohol Dependence. 1993;31:199–203. doi: 10.1016/0376-8716(93)90001-7. [DOI] [PubMed] [Google Scholar]
  • 199.Annas GJ. Testing poor pregnant patients for cocaine--physicians as police investigators. N Engl J Med. 2001;344:1729–32. doi: 10.1056/NEJM200105313442219. [DOI] [PubMed] [Google Scholar]
  • 200.Ferguson v., City of Charleston. 2001 121 S.Ct. 1281. [Google Scholar]
  • 201.McKnight v., State of South Carolina. 2008 26484. [Google Scholar]
  • 202.Rakic P. A small step for the cell, a giant leap for mankind: a hypothesis of neocortical expansion during evolution. Trends Neurosci. 1995;18:383–8. doi: 10.1016/0166-2236(95)93934-p. [DOI] [PubMed] [Google Scholar]
  • 203.Stanwood GD, Levitt P. In: Handbook of Developmental Cognitive Neuroscience. 2nd Nelson CA, Luciana M, editors. MIT Press; 2008. pp. 83–94. [Google Scholar]

RESOURCES