Abstract
The past decades witnessed a surge of interest in neuroimaging study of normal and abnormal early brain development. Structural and functional studies of normal early brain development revealed massive structural maturation as well as sequential, coordinated, and hierarchical emergence of functional networks during the infancy period, providing a great foundation for the investigation of abnormal early brain development mechanisms. Indeed, studies of altered brain development associated with either genetic or environmental risks emerged and thrived. In this paper, we will review selected studies of genetic and environmental risks that have been relatively more extensively investigated-familial risks, candidate risk genes, and genome-wide association studies (GWAS) on the genetic side; maternal mood disorders and prenatal drug exposures on the environmental side. Emerging studies on environment-gene interactions will also be reviewed. Our goal was not to perform an exhaustive review of all studies in the field but to leverage some representative ones to summarize the current state, point out potential limitations, and elicit discussions on important future directions.
Keywords: Early brain development, MRI, Infancy, Risk Genes, Prenatal drug exposure, Maternal Disorders, gene-environment interactions
INTRODUCTION
Complex interactions between genes and environment during development determine the structural and functional growth of the brain and behavior. Directly linking particular genetic and environmental risk factors to specific behaviors has proven to be extremely challenging due to the lack of one-to-one links. Notably, numerous studies on neurodevelopmental and/or psychiatric brain disorders confirm that a disorder is usually associated with a variety of genetic and/or environmental risk factors (Abrahams and Geschwind, 2008; Walsh et al., 2008) while a known genetic and/or environmental risk may result in different variants of behavioral phenotypes (Hessl et al., 2001; Hoffbuhr et al., 2002; Warren and Li, 2005; Yehuda et al., 2006). Part of the reasons for this lack of specificity arises from the fact that neither genes nor the environment directly code for/influence behavior but work together to determine the building blocks of different cells in the brain, the collective effort of which ultimately produces behavior. Therefore, brain measures represent a critical intermediate step between risk factors and behavioral output and have the potential to reveal more specific mechanisms since brain-related phenotypes (e.g., volume, cortical thickness, white matter diffusivity, and functional connectivity, etc.) can be determined for specific regions, networks, and systems, most of which can be more objectively quantified than behavioral phenotypes (Gao et al., 2016; Graham et al., 2014; Raschle et al., 2012). Together with the advancement of various neuroimaging techniques, many studies have turned to brain measures as a stepping stone to bridge the gap between genes/environment and behavior (Lenroot and Giedd, 2008).
Existing studies of gene-brain and environment-brain relationships in the adult brain shed lights on different risk factors that are likely associated with different brain disorders (Caspi and Moffitt, 2006; Chiang et al., 2009; Glahn et al., 2010; Thompson et al., 2001). However, these relationships reflect a long history of convolved interactions between genes/environment and the brain that spans across a prolonged period of postnatal development. Therefore, the translational value is limited since it is extremely challenging, if at all possible, to reverse the long-accumulated effects through medical, behavioral, or other interventions. Together with the increasing consensus that most, if not all, brain disorders have developmental origins (Insel, 2010; O’Donnell and Meaney, 2017; Swanson and Wadhwa, 2008), focus has been increasingly shifted to the identification of genetic and/or environmental risk factors that affect early brain development. Particularly, studies during the neonatal/infancy period have the best potential to minimize postnatal influences that could potentially mask early impacts. Therefore, such studies tend to offer a better depiction of genetic/environmental influences on early brain development. Moreover, this early window hosts one of the most active phase of brain development when many of the brain’s structural, functional, and behavioral processes are unfolding, thus the early impacts are likely amplified with age. The idea that small disturbances during early development spread over time to produce large deviations from typical growth trajectories has been largely agreed on and gone by many names including developmental cascades, chain reactions, and butterfly, snowball, amplification, spillover, or progressive effects (Masten and Cicchetti, 2010). Therefore, the study of pre- and perinatal risks during early infancy period represents a priority from both scientific and clinical perspectives (Gao et al., 2016; Masten and Cicchetti, 2010).
Given the relatively limited behavioral repertoire and practical difficulties associated with infant behavioral evaluation, studying the brain directly is of particular importance. However, accurate delineation of effects of risks in the infant brain has previously been limited by technical difficulties on non-invasive imaging with high spatial fidelity. Basically, electroencephalogram (EEG) (Anderson et al., 1985; Dreyfus-Brisac and Larroche, 1971; Videman et al., 2016; Wen et al., 2017b) and functional near-infrared spectroscopy (fNIRS) (Grossmann and Johnson, 2010; Nakano et al., 2009; Wilcox et al., 2010; Wilcox et al., 2012) represent some of the most widely applied techniques to explore infant brain development. However, their poor spatial resolution (centimeters) and low depth penetration (limited to cortical regions) make them less ideal tools to examine potentially subtle and region-specific effects. In contrast, non-invasive magnetic resonance imaging (MRI) represents a versatile tool that is known to have high spatial resolution (millimeters), provide whole brain coverage, and be able to explore both the structural and functional aspects of infant brain development (Cao et al., 2017; Deoni et al., 2011; Gao et al., 2009a; Gilmore et al., 2007; Gilmore et al., 2012; Hazlett et al., 2017; Huang et al., 2015; Huang et al., 2006). However, due to low tolerance to movement artifacts, most MRI studies of the infant brain are conducted during either natural (Gao et al., 2016; Gao et al., 2009b) or sedation-induced sleeping state(Allievi et al., 2016; Arichi et al., 2012). The sleeping state likely would not affect structural brain examinations but investigations of active functional responses requiring awake state is largely prohibited. Fortunately, recent advancements in resting-state fMRI technique (Biswal et al., 1995) that is capable of delineating the brain’s intrinsic functional architecture in sleeping infants opened a promising new window to peek into functional alterations in the infant brain associated with different genetic and/or environmental risks (Doria et al., 2011; Fransson et al., 2007; Gao et al., 2014; Gao et al., 2016; Gao et al., 2009b; Smyser et al., 2010).
Admittedly, neuroimaging study of gene-/environment-brain relationships during infancy is itself in its infancy. In this preliminary review, we sought to summarize findings from the first set of such studies to better understand the current state of the field, point out potential limitations, and elicit discussions on future directions. We will focus on selected categories of genetic and environmental risk studies that have been relatively more extensively investigated. Specifically, we will cover familial risks, candidate risk genes, and genome-wide association studies (GWAS) on the genetic side and maternal mood disorders and prenatal drug exposures on the environmental side. Studies on potential gene-environment interaction mechanisms will also be touched upon. We fully appreciate that there are other categories of risks (e.g., poverty, maternal obesity, pollution, etc.) and variants of studies but they are beyond the scope of this review. This review is organized as follows: in section 1 we describe the potential mechanisms and experimental findings on the effects of genetic risks on early brain development; in section 2, we introduce the potential mechanisms and experimental findings on the effects of maternal mood disorders and prenatal drug exposures on early brain development; in section 3, we will discuss potential gene-environment interaction effects on early brain development; in section 4 we briefly summarizes the practical considerations in neonatal neuroimaging; finally, we point out some limitations of current studies and discuss future directions moving forward. The selectively discussed neuroimaging studies of genetic and environmental influences on early brain development in this review are listed in Table 1.
Table 1.
1. Studies of infants at high familial risk for psychiatric disorders | |||
---|---|---|---|
Article | Population | N | Main Findings |
Gilmore et al., 2010 | Neonatal offspring of mothers with schizophrenia or schizoaffective disorder and matched comparison mothers without psychiatric illness | 26 offspring of mothers with schizophrenia or schizoaffective disorder and 26 matched controls | The high-risk neonates had non-significantly larger intracranial, cerebral spinal fluid (CSF), and lateral ventricle volumes. Subgroup analysis revealed that male high-risk infants had significantly larger intracranial, CSF, total gray matter, and lateral ventricle volumes; the female high-risk neonates were similar to the female comparison subjects. |
Shi et al., 2012 | Same as Gilmore et al., 2010 | Same as Gilmore et al., 2010 | The brain structural associations of the high-risk neonates tended to have globally lower efficiency, longer connection distance, and less number of hub nodes and edges with relatively higher betweenness. Subgroup analysis showed that male neonates were significantly disease-affected, while the female neonates were not. |
Li et al., 2016 | Same as Gilmore et al., 2010 | 21 offspring of mothers with schizophrenia or schizoaffective disorder and 26 matched controls | Female high-genetic-risk neonates had significantly thinner cortical thickness in the right lateral occipital cortex than the female control neonates. High-genetic-risk neonates had marginally different cortical thickness in a number of other brain areas. |
Wolf et al., 2015 | Infants at high risk for autism spectrum disorder (ASD, having an older sibling with a community diagnosis of ASD) and control groups. | 270 infants at high familial risk for ASD and 108 low-risk controls at 6, 12 and 24 months of age | Significantly increased corpus callosum area and thickness in children with ASD starting at 6 months of age followed by a declining in growth rate, resulting diminished differences at 2 years of age. |
Shen et al., 2017 | Same as Wolf et al., 2015 | 221 infants at high risk for ASD and 122 low risk controls at 6, 12 and 24 months of age | Infants who developed ASD had significantly greater extra-axial CSF volume at 6 months compared with comparison groups |
Lewis et al., 2017 | Same as Wolf et al., 2015 | 260 infants at 6 and 12 months of age with our without known risk for ASD | Inefficiencies in high-risk infants later classified as ASD were detected from 6 months onward in regions involved in low-level sensory processing. |
Hazlett et al., 2017 | Same as Wolf et al., 2015 | 106 infants at high familial risk of ASD and 42 low-risk infants | Hyperexpansion of the cortical surface area between 6 and 12 months of age precedes brain volume overgrowth observed between 12 and 24 months in 15 high-risk infants who were diagnosed with autism at 24 months. Brain surface area information of 6–12-month-old individuals predicted the diagnosis of autism in individual high-risk children at 24 months. |
Emerson et al., 2017 | Same as Wolf et al., 2015 | 59 6-month-old infants with a high familial risk for ASD | Functional connectivity magnetic resonance imaging features measured at 6 months of age correctly identified which individual children would receive a research clinical best-estimate diagnosis of ASD at 24 months of age. |
2. Studies of candidate genes | |||
Knickmeyer et al., 2014 | Neonates with or without parental psychiatric history. | 272 neonates. | Local variation in gray matter volume was significantly associated with polymorphisms in DISC1 (rs821616), COMT, NRG1, APOE, ESR1 (rs9340799), and BDNF. Neonates homozygous for the DISC1 (rs821616) serine allele exhibited reduced GM in the frontal lobes, and neonates homozygous for the COMT valine allele exhibited reduced GM in the temporal cortex and hippocampus. |
Dean et al., 2014 | Healthy, typically developing 2- to 25-month-old infants with no family history of Alzheimer disease or other neurological or psychiatric disorders | 162 infants at 2–25 months of age. | Infant apolipoprotein E (APOE) ε4 allele carriers had lower myelin water fraction (MWF) and gray matter volume (GMV) measurements than non-carriers in areas preferentially affected by AD. They also showed greater MWF and GMV measurements in extensive frontal regions. |
Krishnan et al., 2017 | Preterm infants | Two independent cohorts of preterm infants (cohort 1: n = 70; cohort 2: n = 271) | Common genetic variation in DLG4 (rs17203281) is associated with fractional anisotropy in preterm infants |
3. Genome-wide association studies (GWAS) | |||
Xia et al., 2017 | Infants (300 male, 261 female) between 0 and 24 weeks of age, including 295 singletons or unpaired twins, 17 sibling pairs and 232 twins. | 561 infants | An intronic single-nucleotide polymorphism (SNP) in IGFBP7 (rs114518130) achieved genome-wide significance for gray matter volume (P=4.15 × 10–10). An intronic SNP in WWOX (rs10514437) neared genome-wide significance for white matter volume (P=1.56 × 10–8). |
Krishnan et al., 2016 | Preterm infants (mean gestational age (GA) 28 + 4 weeks, mean postmenstrual age (PMA) at scan 40 + 3 weeks) | 72 preterm infants. | Significant relationships between lipid pathways, peroxisome proliferator-activated receptor (PPAR) signaling particularly, and variability in preterm white matter development, measured by fractional anisotropy were detected. Five genes were found to be highly associated with the phenotype: aquaporin 7 (AQP7), malic enzyme 1, NADP(+)-dependent, cytosolic (ME1), perilipin 1 (PLIN1), solute carrier family 27 (fatty acid transporter), member 1 (SLC27A1), and acetyl-CoA acyltransferase 1 (ACAA1). |
4. Studies of prenatal maternal mood disorders | |||
Qiu et al., 2013 | Full term infants | 175 neonates with 35 of them having repeated MRI scans at 6 months | Children of mothers reporting increased anxiety during pregnancy showed slower growth of both the left and right hippocampus over the first 6 months of life. |
Qiu et al., 2015a | Full term infants | 24 infants at 6 months of age | Infants born to mothers with higher prenatal maternal depressive symptoms showed greater functional connectivity of the amygdala with the left temporal cortex and insula, as well as the bilateral anterior cingulate, medial orbitofrontal and ventromedial prefrontal cortices. |
Rifkin-Graboi et al., 2013 | Full term neonates | 157 neonates | Significantly lower fractional anisotropy and axial diffusivity, but not volume, were detected in the right amygdala in the infants of mothers with high compared with those with low depression scores. |
Wen et al., 2017a | 4.5-year-old children | 235 children | Greater prenatal maternal depressive symptoms were associated with larger right amygdala volume in girls, but not in boys. Increased postnatal maternal depressive symptoms were associated with higher right amygdala FA in the overall sample and girls, but not in boys. |
5. Studies of prenatal drug exposures | |||
Grewen et al., 2014 | Full term neonates with or without prenatal drug exposure | 33 with PCE co-morbid with other drugs, 46 drug-free controls and 40 with prenatal exposure to other drugs (nicotine, alcohol, marijuana, opiates, SSRIs) but without cocaine. | Reduced prefrontal and frontal gray matter volume and enhanced whole brain CSF volumes in neonates with prenatal cocaine exposure (PCE) compared with drug-free newborns and those with exposure to similar other drugs but not cocaine. |
Salzwedel et al., 2015 | Same as Grewen et al., 2014 | Same as Grewen et al., 2014 | Drug-common effects were detected within the amygdala–frontal, insula–frontal, and insula–sensorimotor circuits. A prenatal cocaine exposure (PCE)-specific effect was detected within a sub-region of the amygdala–frontal network. |
Grewen et al., 2015 | Same as Grewen et al., 2014 | 20 with prenatal marijuana exposure (PME) co-morbid with other drugs, 20 drug-free controls and 20 with prenatal exposure to other drugs (nicotine, alcohol, marijuana, opiates, SSRIs) but without marijuana. | Both marijuana-specific and drug-common alterations in functional connectivity were detected among a range of functional circuits associated with the amygdala, hippocampus, putamen, anterior/posterior insula, caudate, and anterior/posterior thalamus. |
Salzwedel et al., 2016 | Same as Grewen et al., 2014 | Same as Grewen et al., 2014 | PCE-related hyper-connectivity between the thalamus and frontal regions and a drug-common hypo-connective signature between the thalamus and motor-related regions were detected. PCE-specific neonatal thalamo-frontal connectivity was inversely related to cognitive and fine motor scores and thalamo-motor connectivity showed a positive relationship with composite motor scores. Moreover, cocaine by selective-serotonin-reuptake-inhibitor (SSRI) interactions were detected, suggesting the combined use of these drugs during pregnancy could have additional consequences on fetal development. |
6. Studies of interactions between environmental and genetic effects | |||
Qiu et al., 2015b | Same as Qiu et al., 2013 | 146 neonates | Individual COMT SNPs modulated the association between antenatal maternal anxiety and the prefrontal and parietal cortical thickness in neonates. Specifically, the A-val-G (AGG) haplotype probabilities modulated positive associations of antenatal maternal anxiety with cortical thickness in the right ventrolateral prefrontal cortex and the right superior parietal cortex and precuneus. |
Qiu et al., 2017 | Same as Qiu et al 2013 | 168 and 85 mother–infant dyads from Asian and United States of America cohorts, respectively | A genomic profile risk score for major depressive disorder (GPRSMDD) moderated the association between antenatal maternal depressive symptoms and the right amygdala volume in neonates. |
1. Neuroimaging study on the effects of genetic risks on early brain development
Although single-gene disorders can produce complex behavioral phenotypes as in Rett syndrome, Lesch–Nyhan Syndrome, and Fragile X, modern genetic research indicates that common behavioral conditions such as autism and attention-deficit-hyperactivity disorder reflect the action of multiple genes acting in concert with non-genetic factors (Goldstein and Reynolds, 2011). While potential mechanisms and preliminary studies of gene-environment interactions will be discussed in section 3, this section will focus on studies that specifically examine genetic risks without substantial consideration of environmental variables.
Expression of disease-related genes during pre- and perinatal development
Precise regulation of gene expression has been well documented in human brain development. Importantly, it has been shown that 90 per cent of genes expressed in the brain are differentially regulated both spatially and temporally and the bulk of this spatio-temporal regulation occurred during prenatal development (Kang et al., 2011). For example, for DCX, a gene expressed in neuronal progenitor cells and immature migrating neurons, its expression increases until early mid-fetal development and then gradually declines, consistent with the growth of immature neurons. In contrast, genes expressed in dendrites (MAP1A, MAPT, CAMK2A) and synapses (SYP, SYPL1, SYPL2, SYN1) show steep increases between the late mid-fetal period and one year of postnatal age, in line with the documented growth trajectories of dendritic arborization and synaptogenesis (Tau and Peterson, 2010). Notably, many genes associated with autism spectrum disorder (ASD), schizophrenia, and intellectual disability also exhibit elevated expression in fetal and early postnatal life (Birnbaum et al., 2015; Birnbaum et al., 2014). Specific examples include CNTNAP2, which encodes a neurexin family protein implicated in ASD and is highly enriched in orbital and dorsal lateral prefrontal cortices during fetal development while its expression increases in other cortical areas during early infancy, and NRGN, a gene encoding a postsynaptic protein kinase substrate associated with schizophrenia which is highly expressed in all cortical areas starting from birth and continuously increases in expression until late childhood (Kang et al., 2011). The documented high level of expression of disease-related genes during pre- and perinatal development suggests that the fetal and neonatal brain may be particularly susceptible to risk-related genetic variation. Indeed, emerging neuroimaging studies of infant brain development have provided solid evidence for this hypothesis. These includes studies of infants at high familial risk for psychiatric disorders, candidate gene studies, and genome-wide association studies.
Studies of infants at high familial risk for psychiatric disorders
Regarding high familial risk, the majority of work in this area has focused on infant siblings of children with autism(Hazlett et al., 2017; Lewis et al., 2017; Shen et al., 2017; Wolff et al., 2015). Because autism can be diagnosed as early as two-years of age, these studies also allow comparisons between high-genetic risk individuals who progress to an autism diagnosis and those who do not. These studies indicate that children who go on to a diagnosis of ASD are distinguished by atypical longitudinal brain changes, from 6–24 months of age. In particular, these infants exhibit a greater increase in brain volume from 12–24 months of age (Hazlett et al., 2017), increased surface area expansion from 6 to 12 months of age (Hazlett et al., 2017), an increase in size of the genu of the corpus callosum at 6 month of age, followed by a declining growth trajectory (Wolff et al., 2015), and expanding regions of decreased efficiency (weighted distance between two nodes using number and distance of white matter fiber tracts) beginning in the right temporal lobe at 6 months and expanding to temporal, parietal and occipital lobes by 24 months (Lewis et al., 2017). These children also show increased extra-axial cerebral spinal fluid (CSF) volume at 6 months of age (Shen et al., 2017). A study of genetic risk for schizophrenia (i.e., maternal diagnosis of schizophrenia or schizoaffective disorder)(Gilmore et al., 2010) found that male neonates at genetic risk for schizophrenia had larger intracranial, total gray matter, and CSF volumes than control neonates. In a follow-up study from the same group, Shi et al (Shi et al., 2012) went on to show that male neonates at genetic risk for schizophrenia also demonstrated lower efficiency, less hubs, and longer connection distance according to graph theory-based analysis of morphological brain networks. A preliminary study (Li et al., 2016) of cortical thickness and surface area, also be the same group, suggested that the widespread cortical thinning observed in adults with schizophrenia was not present in neonates, but region-specific alterations are detectable. Notably, male and female at-risk neonates showed very different profiles in comparison to typical controls (Li et al., 2016).
Studies of candidate genes
The first candidate gene study of infant neuroimaging phenotypes was carried out by Knickmeyer et al (Knickmeyer et al., 2014). In this study of ~1 month old infants, carriers of the apolipoprotein E (APOE) ε4 variant, a major susceptibility factor for late onset Alzheimer’s disease (AD), had reduced gray matter volume in medial temporal cortex, suggesting the ε4 variant’s contribution to brain phenotypes associated with Alzheimer’s risk is present before birth. Brain volume differences were also observed for other risk genes, including disrupted-in-schizophrenia-1 (DISC1), catechol-O-methyltransferase (COMT), neuregulin (NRG), brain-derived neurotrophic factor (BDNF), and glutamate decarboxylase 1 (GAD1) (Knickmeyer et al., 2014). Consistent with these findings, BDNF polymorphism has recently been shown to be associated with individual differences in temperament in 4-month-old infants (Giusti et al., 2017). A subsequent study of healthy infant (2~25 months of age) carriers and non-carriers of the APOE ε4 allele observed lower white matter myelin water fraction and gray matter volume in infant ε4 carriers (Dean et al., 2014) in precuneus, posterior/middle cingulate, lateral temporal, and medial occipitotemporal regions, areas preferentially affected by AD. Finally, a recent study by Krishnan et al (Krishnan et al., 2017) has reported that common genetic variation in DLG4 (rs17203281) is associated with fractional anisotropy in preterm infants. In addition to being required for synaptic plasticity associated with NMDA receptor signaling, DLG4 is a hub protein in the microglial inflammatory response. The authors hypothesize that this particular genetic variant may modulate responses to neuroinflammation in children born preterm (Krishnan et al., 2017).
Although fascinating, these results must be considered in light of known pitfalls and limitations of candidate gene studies. First, candidate gene studies only investigate a few a-priori variants of interest; these genes likely represent a very small fraction of all variants involved in human brain development. Second, sample sizes are relatively modest (272 for Knickmeyer et al. 2014; 162 for Dean et al. 2014; and 70 and 271 (2 cohorts) for Krishnan et al., 2017). They are thus well powered to detect large and moderate effects sizes, but would be underpowered to detect genetic effects typical for genome-wide association studies (GWAS) of human disease (Collins et al., 2012). It was originally hoped that effect sizes for brain phenotypes would be larger than those for psychiatric disease itself, but recent GWAS of global and subcortical brain volumes in adults suggest this might not be the case (Hibar et al., 2017; Stein et al., 2012). Third, as in all areas of science, replication is critical for delineating between true-positive and false-positive effects. Most of the variants studied in Knickmeyer et al. (2014) have not been evaluated in independent neonate samples. There is some overlap between Knickmeyer et al. (2014) and Dean et al. (2014) in terms of brain areas effected by APOE ε4. Krishnan et al. (2017) investigated two cohorts; significant effects of DLG4 (rs17203281) were observed in both. Finally, it should be noted that, with the exception of APOE, genes investigated in Knickmeyer et al (2014) have not emerged as significant psychiatric risk genes in subsequent GWAS studies. While they clearly play important roles in brain development, they may not be directly relevant to mental health.
Genome-wide association studies (GWAS)
For all the above-mentioned reasons, focus has increasingly shifted from candidate gene approaches to genome-wide association studies (GWAS). The first GWAS on structural brain development in a population cohort of infants (Xia et al., 2017) revealed an intronic single-nucleotide polymorphism (SNP) in IGFBP7 (rs114518130) which achieved genome-wide significance for gray matter volume and an intronic SNP in WWOX (rs10514437) which neared genome-wide significance for white matter volume. The former locus is also within 100kb of REST, a master negative regular of neurogenesis which binds at thousands of locations across the genome (Johnson et al. 2007). Additional loci with small p-values tagged psychiatric GWAS associations, transcription factors expressed in developing brain, and other genes with strong biological plausibility including HTR1B, which encodes the 5-hydroxytryptamine (serotonin) receptor 1B, and RBFOX1, an RNA splicing factor which regulates expression of large genetic networks during early neuronal development. Krishnan et al (2016) have also used genome wide data to assess whether common genetic variation influenced white matter microstructure in preterm infants. As they had a very small sample size (72 infants), they focused on pathway and network-based approaches. Results suggest a possible role for peroxisome proliferator-activated receptor signaling (Krishnan et al., 2016). These early results suggest that GWAS studies of infant neuroimaging phenotypes will be fruitful in terms of identifying genes and molecular pathways involved in prenatal and infant brain development. However, GWAS studies come with their own methodological pitfalls and limitations. These have been reviewed in detail elsewhere (Karlsen et al., 2010; Riancho, 2012), but we will highlight three issues in particular here. First, GWAS is most effective when traits are underpinned by a small number of loci with large effect sizes. Most human traits examined thus far do not follow this pattern; instead they are underpinned by many loci with small effect sizes. GWAS requires thousands or tens of thousands of individuals to detect these effects. Such sample sizes are not yet available for infant neuroimaging phenotypes. Second, the large number of statistical tests performed in a GWAS study means that a high number of associations arise by chance. Even with stringent thresholds, replication is key. This has not yet been possible for infant neuroimaging studies, due to the lack of multiple, large cohorts with GWAS genotypes. This situation is likely to change in the near future as infant neuroimaging becomes more common and independent groups start working together as in the ENIGMA (Enhancing Neuro Imaging Genetics through Meta-Analysis) consortium (Bearden and Thompson, 2017). Finally, spurious associations may arise due to technical or complex genetic reasons (such as population stratification: the presence of a systematic difference in allele frequencies between subpopulations in a population due to different ancestry). Replication will be important for ruling out such effects.
Overall, these studies corroborate the rich gene expression during pre-/perinatal development and indicate that brain alterations associated with different risk genes can be detected during early infancy. Further investigations are needed to explore the potential to develop robust imaging-based biomarkers for later onset of behavioral problems and/or disorders among infants at risk. Promising results have emerged from studies of infants at high genetic risk for autism where cortical surface features and resting-state connectivity measured in the first year of life distinguish children who go on to develop autism from those who do not (Emerson et al., 2017; Hazlett et al., 2017). However, as most cases of autism (and other serious mental illnesses) are sporadic, similar studies in population cohorts are urgently needed.
2. Neuroimaging study on the effects of environmental risks on early brain development
Environmental factors have been shown to exert vital effects on early brain development(Booij et al., 2015; Thompson et al., 2009). Similar as section 1, while its interactions with genes are robustly documented and will be discussed later, this section will focus on studies of environmental risks including maternal mental disorders and prenatal drug exposures, without substantial consideration of genetic variants.
Environmental risk factors for mental disorders are diverse in nature but could be categorized in three main categories based on timing: 1) prenatal risks, including maternal mood disorders (e.g., depression/anxiety), substance abuse (e.g., prenatal drug exposure) during pregnancy, among others; 2) perinatal risks, including low birth weight, birth complications, and deprivation of normal parental care during early infancy, among others; 3) risks in childhood and beyond, including childhood abuse, childhood neglect, premature parental loss, exposure to family conflict and violence, trauma, substance abuse, and toxic exposure, among others. In this review, we will highlight selected studies of the first two categories of risks.
Studies of prenatal maternal mood disorders
Prenatal maternal mood disorders are associated with an increased risk for neurobehavioral, cognitive/socio-emotional problems (Waters et al., 2014), and depression in the offspring (Field et al., 2004; Pawlby et al., 2009; Pearson et al., 2013). Although the precise mechanisms underlying the effects of maternal mood status on fetal development remains to be determined, evidences accumulate on alterations in hormones and neurotransmitters in the uterine environment (Van den Bergh et al., 2005). For example, studies have shown that levels of cortisol are highly correlated between paired maternal and fetal plasma samples (Gitau et al., 1998). Moreover, newborns of prenatally depressed mothers exhibit biochemical profiles similar to those observed in depressed mothers, including elevated cortisol and norepinephrine and lower dopamine and serotonin (Field et al., 2004). Notably, proper functioning of these hormones and neurotransmitters is critical for the timing and guidance of neurogenesis, neuronal differentiation/migration, apoptosis, synaptogenesis and myelination (Nowakowski and Hayes, 2002). Therefore, disturbances of the delicate balance between these hormones and neurotransmitters in the fetus related to maternal depression would likely influence critical prenatal programing of different neural circuits thus shedding far reaching effects on offspring behavior. Of particular importance are the potential impacts on the hypothalamic–pituitary–adrenal (HPA) axis (Challis et al., 2001), the limbic system, and the prefrontal cortex, which may collectively contribute to cognitive/emotional regulation problems of children born to mothers with mood disorders.
Neuroimaging studies of effects of prenatal maternal mood disorders are emerging. In one study of maternal anxiety, Qiu et al (Qiu et al., 2013) reported that children of mothers with elevated anxiety levels showed slower growth of bilateral hippocampal volume during the first six months of life. Moreover, the left and right hippocampal growths show differential reactivity to postnatal maternal anxiety levels. Maternal anxiety in pregnancy was also associated with variation in fractional anisotropy (FA) of brain regions important to cognitive-emotional responses to stress (i.e., the right insula and dorsolateral prefrontal cortex), sensory processing (e.g., right middle occipital), and socio-emotional function (e.g., the right angular gyrus, uncinate fasciculus, posterior cingulate, and parahippocampus), which in turn predicted infant internalizing but not externalizing behavior 1 year later (Rifkin-Graboi et al., 2015). The same group also reported reduced water diffusivity and fractional anisotropy properties of the right amygdala in neonates born with mothers with higher depressive symptoms (Rifkin-Graboi et al., 2013) and altered functional connectivity patterns of the amygdala in 6-month-old infants born from mothers with greater maternal depressive symptoms (Qiu et al., 2015a). A follow-up study in 4.5-year-old children(Wen et al., 2017a) shows that greater prenatal maternal depressive symptoms were associated with larger right amygdala volume in girls, but not in boys. Likewise, postnatal maternal depression promotes forms of parenting (Fleming et al., 1988) that enhance stress reactivity, social withdrawal, and inattention (Bruder-Costello et al., 2007; Degnan et al., 2010; Moffitt et al., 1996), which in turn predicts an increased risk for depression and behavioral problems in the offspring (Mars et al., 2015; Matijasevich et al., 2015). Increased postnatal maternal depressive symptoms were associated with higher right amygdala FA in the overall sample and girls, but not in boys (Wen et al., 2017a). Interestingly, in a general population, the infant spent at least 50% of his/her day time hours with his/her mother, both lower maternal sensitivity and higher maternal depression predicted greater relative right frontal EEG asymmetry (Wen et al., 2017b). Furthermore, greater relative right frontal EEG asymmetry of 6-month-old infants predicted their greater negative emotionality at 12 months of age. This suggested that amongst infants with sufficient postnatal maternal exposure, both maternal sensitivity and mental health, are important influences on early brain development (Wen et al., 2017b). These studies provide compelling evidence that maternal mood disorders, both prenatal and postnatal, could affect in-utero and/or postnatal brain development, especially within limbic structures involved in arousal and emotion regulation, presenting some initial clues on the brain mechanisms of transgenerational transmission of vulnerability for mood disorders during pre- and postnatal development.
Studies of prenatal drug exposures
Prenatal drug exposures comprise another category of environmental risk that has received considerable research interest in recent years(Behnke et al., 2013; Brady et al., 2003; Chasnoff et al., 1986; Ching and Tang, 1986; Derauf et al., 2009), partly due to the alarming rate of increase in substance abuse during pregnancy (Behnke et al., 2013; Ross et al., 2015). Prenatal drug exposure affects fetal brain development both directly (most drugs readily cross both the placenta(Behnke and Eyler, 1993) and blood-brain-barriers(Schou et al., 1977) to bind with endogenous receptors in fetal brain during critical periods) and indirectly through actions on the placenta (Bhide and Kosofsky, 2009). In fetal brain, different drugs act via shared and unique pathways but the most important ones relate to the disruptions of endogenous neurotransmitter systems which play important roles in brain development during critical periods of gestation(Nowakowski and Hayes, 2002). For example, the primary psychoactive compound in marijuana, ∆9-tetrahydrocannabinol (THC), is an exogenous cannabinoid which binds to type 1 cannabinoid receptors. This likely disrupts endogenous cannabinoid signaling which plays a critical role in control of neurogenesis, phenotypic specification of immature neurons, and establishment of the normal fetal neuronal network architecture (Gaffuri et al., 2012; Harkany et al., 2008). On the other hand, prenatal cocaine and amphetamines act primarily within the mesolimbic dopaminergic pathway to increase volume and duration of monoamines (dopamine, serotonin, norepinephrine) within the synapse. Opioid mechanisms may include altered fetal brain opioid receptor (OR) expression, opioid-linked decreases in myelin volume and maturation(Eschenroeder et al., 2012; Sanchez et al., 2008; Vestal-Laborde et al., 2014), and enhanced activity of locus coeruleus neurons (Aghajanian et al., 1992; Duman et al., 1988; Nestler et al., 1994, 1999; Selley et al., 1997; Van Bockstaele et al., 2010; Van Bockstaele and Valentino, 2013) that would naturally restrain noradrenergic activation throughout the brain which may then contribute to greater stress reactivity and arousal(Kinney et al., 1990). Opioid-related epigenetic effects include hypermethylation to silence not only (mu)OR, but also global DNA expression(Doehring et al., 2013; Knothe et al., 2016). Moreover, interactions between OR gene variants and epigenetic effects may yield differential responses to similar exposures(Oertel et al., 2012).
In human children and adolescents, prenatal drug exposures are linked to abnormal brain volumes(Dow-Edwards et al., 2006), impaired white matter microstructures(Derauf et al., 2009; Warner et al., 2006), and different cognitive/behavioral deficits (Connor et al., 2006; Leech et al., 1999; Walhovd et al., 2007; Walhovd et al., 2010). While studies in children and older populations may be confounded by postnatal influences, emerging neuroimaging studies in the neonatal brain provide initial confirmation of the prenatal effects of drug exposure. Specifically, Grewen et al (Grewen et al., 2014) conducted one of the first MRI studies of effects of prenatal drug exposures on neonatal structural brain development. They reported reduced prefrontal and frontal gray matter volume and enhanced whole brain CSF volumes in neonates with prenatal cocaine exposure (PCE, comorbid with other drugs) compared with drug-free newborns and those with exposure to similar other drugs but not cocaine, indicating PCE-specific effects on structural brain growth are evident in neonates. Subsequently, Salzwedel et al (Salzwedel et al., 2016; Salzwedel et al., 2015) published the first studies on the effects of PCE on neonatal functional brain development using resting-state fMRI. They reported aberrant amygdala-prefrontal (Salzwedel et al., 2015) and thalamus-frontal (Salzwedel et al., 2016) functional connectivity specifically associated with PCE. Drug-common alterations in thalamus-motor area connectivity were also observed (Salzwedel et al., 2016). In addition to cocaine, the same group also examined effects of prenatal marijuana exposure on neonatal functional brain organization and reported reductions in connectivity of insula and striatal seeds with multiple visual and cerebellar regions (Grewen et al., 2015). In another study of prenatal exposure to antiepileptic drugs using EEG, Videman et al (Videman et al., 2016) showed significant differences in several features of neonatal cortical electrical activity, including alpha bursts, frequency spectrum, and spatial distribution of bilateral synchrony. King et al., also reported disruption of auditory gating measured by EEG in 4–6 month old infants exposed to prenatal nicotine (King et al., 2017). Importantly, Salzwedel et al (Salzwedel et al., 2016) went on to show that degrees of functional connectivity disruption in neonates associated with prenatal drug exposure significantly predicted cognitive and motor-related outcome measures at 3 months of age. Although preliminary and requiring independent validation, these findings provide exciting new directions in the search of imaging-based biomarkers for later behavioral outcomes in these at-risk infants. Another important issue/limitation of drug exposure research relates to the fact that drug-using mothers rarely use a single drug so it’s difficult to tease out drug-specific effects. That being said, the potential drug-drug interaction effects are also critical research questions. Indeed, a preliminary examination of the potential interaction effects between cocaine and selective-serotonin-reuptake-inhibitor (SSRI) (Salzwedel et al., 2016) indicated that the combined use of both cocaine and SSRI during pregnancy resulted in the worst outcomes on thalamocortical functional connectivity, highlighting the necessity of further research on this important topic. Overall, existing studies provide convergent evidence on the effects of prenatal drug exposure on neonatal brain structural and functional development but important limitations associated with multi-drug usage has to be better addressed in future studies.
3. Interactions between genetic risks, prenatal drug exposures, and other family environment factors
Long debates between nature and nurture have gradually converged on the idea that they both have vital influences on brain and behavioral development while most, if not all, parts of their influences are unfolded in a time-dependent manner through complex interactions rather than single-domain actions. This hypothesis implies that individual differences in genetic makeup will modulate individual differences in their resilience or vulnerability to environmental adversities and vice versa- individual differences in environmental exposures may modulate subject-specific gene expression along time (i.e., epigenetics). This gene-environment bi-directional conversation was observed in cases of both early adverse life events and prenatal drug exposure. For example, Caspi et al (Caspi et al., 2002) reported that a functional polymorphism of the gene encoding the neurotransmitter-metabolizing enzyme monoamine oxidyase A (MAOA) moderated the effect of childhood maltreatment in violence behaviors. For prenatal drug exposure, both animal models (Downing et al., 2009; Goodlett et al., 1989) and human studies (Jacobson et al., 2006; Streissguth and Dehaene, 1993) have convincingly demonstrated that genetic background can confer either susceptibility or resilience to risks of prenatal drug exposure (Goodlett et al., 1989). From the other direction, the effects of early life adversities have been well studied and findings suggest that early life adverse exposures can disrupt gene expressions not only in specific candidate genes but also on the whole genome at both brain and peripheral level (Bick et al., 2012). Similarly, different fetal drug exposures were shown to alter gene expression in the brain in both animal and human studies (Gangisetty et al., 2014; Lester and Padbury, 2009; Lussier et al., 2015).
While relatively extensive gene-environment interaction studies have been conducted during childhood and beyond as mentioned above, little was done to characterize such interactions during early brain development, especially those using brain measures as the target phenotype. Among the few existing studies, Qiu et al (Qiu et al., 2015b) showed that individual COMT SNPs modulated the association between antenatal maternal anxiety and the prefrontal and parietal cortical thickness in neonates. Specifically, the A-val-G (AGG) haplotype probabilities modulated positive associations of antenatal maternal anxiety with cortical thickness in the right ventrolateral prefrontal cortex and the right superior parietal cortex and precuneus. In contrast, the G-met-A (GAA) haplotype probabilities modulated negative associations of antenatal maternal anxiety with cortical thickness in bilateral precentral gyrus and the dorsolateral prefrontal cortex. In another study of maternal depression (Qiu et al., 2017), the same group found that a genomic profile risk score for major depressive disorder (GPRSMDD) moderated the association between antenatal maternal depressive symptoms and the right amygdala volume in neonates, further underscoring the gene-environment interdependence in fetal brain development. The study also identified that glutamate receptor activity played an important role in the relationship between prenatal maternal depressive symptoms and neonatal right amygdala volume. Multiple approaches, including genome-wide analyses, implicate glutamatergic synaptic transmission in the etiology of depression (Lee et al., 2012; Skolnick et al., 2009) and patients with major depressive disorder (MDD) exhibit abnormal glutamate concentrations in the amygdala (Michael et al., 2003). Post-mortem studies with MDD suggest altered glutamatergic synaptic signaling (Hashimoto et al., 2007) and ketamine, a glutamatergic intervention, is a treatment for MDD (Caddy et al., 2014). Interestingly, the direction of the interaction between maternal depressive symptoms and infant genotype on right amygdala volume in the US sample is in the opposite direction to that observed in the Asian sample, suggesting that despite common underlying biological processes, polymorphisms may operate in an ethnicity-specific manner(Qiu et al., 2017). Taken together, encouraging preliminary results on the gene-environment interaction effects on early brain development have emerged but more systematic studies and independent validations are desperately needed to further our understanding of this complex issue.
4. Practical considerations in neonatal/infant imaging
In clinical settings, neuroimaging of infants are routinely conducted during sedation but for ethical considerations, sedation is not an option for most developmental neuroimaging research. In contrast, imaging naturally sleeping infants represents the most common practice in developmental neuroimaging studies (Gao et al., 2016). Many challenges exist for the preparation and actual imaging of non-sedated, naturally sleeping infants and certain procedures have been developed to cope with such challenges (Raschle et al., 2012). First, careful preparation and proper environment should be given to help caregivers to put the infant to sleep. Examples include scheduling the session during the infant’s nap time, pre-exposure to the scanner noise through a CD for at least a week before appointment, feeding and swaddling in a dimmed room before imaging (or inside the scanner room), and reserving at least a two-hour time slot to accommodate sleep preparation time, among others. To prepare for actual imaging after the infant fall asleep, proper ear protection (e.g., ear plugs, noise-cancelling headphones) must be in place, the infant need to be wrapped and secured on the scanner bed (preferably with a warm blanket on top of a secured mattress), and proper monitoring devices for vital signals (e.g., heart rate, blood pressure, and pulse oximetry) are connected, for imaging. During image acquisition, besides the caregiver, a pediatric nurse should stay in the imaging suite to monitor the vital signals for safety. If the infant begins to wake, MRI acquisition can be paused and the caregiver can try to soothe the infant back to sleep. However, if the infant cries or becomes distressed, the scan needs to be stopped and the infant be removed from the scanner. For a more detailed discussion on the challenges, procedures, and ethical considerations related to infant neuroimaging, please refer to (Raschle et al., 2012).
Conclusions, Limitations and Future Directions
Overall, the field of early brain development witnessed a surge of interest in characterizing the effects of genetic and environmental risks using different neuroimaging techniques during the past decades. In this review, we have summarized findings from selected studies on familial risks for psychiatric disorders, candidate gene studies, and GWAS studies on the genetic side; and maternal mood disorders and prenatal drug exposures on the environmental side. Moreover, preliminary studies on the gene-environment interactions in infants are also discussed. Overall, as summarized above, existing studies have consistently conveyed the idea that structural and functional brain alterations associated with both genetic and environmental risks, as well as their interactions, could be detected as early as in neonates. Moreover, the abnormality profiles may change with age and differ between boys and girls. By further linking early brain alterations to behavioral outcomes, these studies provide the exciting first set of evidences demonstrating the potential of neuroimaging in the early identification of risks for later development problems and/or disorders.
As encouraging as these previous studies are, the field is likely still in its infancy. As partly mentioned above in separate sections, there are many issues calling for more future studies to address. For example, most studies of infants at high familial risk (Gilmore et al., 2010; Hazlett et al., 2017; Li et al., 2016; Wolff et al., 2015) have not integrated neuroimaging with molecular genetics, so the specific genes and pathways underlying the observed associations are unclear. Although machine learning-based approaches (Hazlett et al., 2017) show promising classification results but these findings need to be validated in independent samples. For candidate gene studies, known risk genes represent a very small fraction of all variants involved in the development of human brain. Furthermore, many of the ‘classic’ candidate genes in psychiatry (with the exception of the APOE ε4 allele) have not been supported by recent large-scale GWAS studies. For GWAS studies, sample size is a major limiting factor so large-scale collaborations as exemplified by ENIGMA (Bearden and Thompson, 2017) are needed in the infant population to overcome this barrier. For studies of maternal mood disorders, active medication use represents a confounding factor and should be better controlled in future studies. For prenatal drug exposure studies, multiple drug usage acts as a major limitation for characterization of brain changes associated with a specific drug. Future studies simultaneously modeling all drugs, as well as their interactions, are needed to tease our drug-specific effects but much larger sample sizes are likely needed to offer enough power for such multivariate estimation. Overall, for both genetic and environmental risk studies, it is critical to consider the other part of the equation and emphasize their interactive effects on early brain and behavioral development. Better understandings of the mechanisms leading to adverse developmental outcomes can only be achieved through combined examination of both the genetic makeup and environmental exposures since each of the two domains of factors can offer either protection against or catalyst for the expression of the effects from the other domain. However, the issue of sample size becomes even more challenging in these studies so multi-sites collaborations are critical in future attempts of systematic delineation of gene-environment interaction effects on early brain and behavioral development. Therefore, it is essential that the field can embrace the open-science concept and move forward through more data sharing and closer collaboration.
In the future, two of the most important and overarching questions deserve the whole field to rally around and tackle. The first question is “What do the observed early brain alterations tell us about future behavior”? Answering this question requires not only early imaging during infancy but also longitudinal follow-up of behavioral outcomes. More importantly, successful prediction would have to rely on the development of advanced analytical algorithms capable of handling the multimodal, large-scale neuroimaging features, including the increasingly popular deep-learning and artificial intelligence techniques. Encouraging preliminary predictions have been reported (Emerson et al., 2017; Hazlett et al., 2017; Rifkin-Graboi et al., 2015; Salzwedel et al., 2016) but much more efforts are needed to build and independently test different prediction models to achieve the goal of deriving imaging-based biomarkers for early identification of risks. The second question is “What can we do to modify the aberrant brain growth thus improving behavioral outcomes”? Existing studies already provided clues on the environmental and/or genetic risk factors that affect early brain development but much more work need to be done for these findings to guide meaningful interventions (e.g., environmental modification, behavioral therapy, medication, or new generation gene editing techniques). Examples include the establishment and independent validation of causal pathways of action, development of new behavioral/environmental/genetic intervention strategies, and deriving image-based quantitative biomarkers to gauge the effects of intervention trials, among others. Apparently, answers to these two questions represent some of the most important long-term goals in the developmental research field and many challenges need to be overcome before their advent. Importantly, such a monumental endeavor requires collective efforts from different fields including but not limited to imaging, developmental psychology, genetics, neuroscience, and advanced computation. Therefore, interdisciplinary collaboration is a must in the long journey from imaging to imaging-based prediction and intervention design, the advent of which, hopefully, could provide desperately needed help to the affected children and their families.
Acknowledgments
This work is supported by the National Institutes of Health: NS088975 to WG, DA042988, DA043171, and DA036645 to WG and KG, MH070890 and HD053000 to JHG, MH092335 and MH104330 to RKS, U01MH110274 (UNC/UMN Baby Connectome Project) to WL, and by National Medical Research Council: NMRC/TCR/012-NUHS/2014 to AQ.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Abrahams BS, Geschwind DH. Advances in autism genetics: on the threshold of a new neurobiology. Nat Rev Genet. 2008;9:341–355. doi: 10.1038/nrg2346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aghajanian GK, Alreja M, Nestler EJ, Kogan JH. Dual mechanisms of opiate dependence in the locus coeruleus. Clin Neuropharmacol. 1992;15(Suppl 1 Pt A):143a–144a. doi: 10.1097/00002826-199201001-00077. [DOI] [PubMed] [Google Scholar]
- Allievi AG, Arichi T, Tusor N, Kimpton J, Arulkumaran S, Counsell SJ, Edwards AD, Burdet E. Maturation of Sensori-Motor Functional Responses in the Preterm Brain. Cereb Cortex. 2016;26:402–413. doi: 10.1093/cercor/bhv203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anderson CM, Torres F, Faoro A. The EEG of the early premature. Electroencephalogr Clin Neurophysiol. 1985;60:95–105. doi: 10.1016/0013-4694(85)90015-x. [DOI] [PubMed] [Google Scholar]
- Arichi T, Fagiolo G, Varela M, Melendez-Calderon A, Allievi A, Merchant N, Tusor N, Counsell SJ, Burdet E, Beckmann CF, Edwards AD. Development of BOLD signal hemodynamic responses in the human brain. Neuroimage. 2012;63:663–673. doi: 10.1016/j.neuroimage.2012.06.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bearden CE, Thompson PM. Emerging Global Initiatives in Neurogenetics: The Enhancing Neuroimaging Genetics through Meta-analysis (ENIGMA) Consortium. Neuron. 2017;94:232–236. doi: 10.1016/j.neuron.2017.03.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Behnke M, Eyler FD. The consequences of prenatal substance use for the developing fetus, newborn, and young child. Int J Addict. 1993;28:1341–1391. doi: 10.3109/10826089309062191. [DOI] [PubMed] [Google Scholar]
- Behnke M, Smith VC, Committee on Substance, A., Committee on, F., Newborn Prenatal substance abuse: short- and long-term effects on the exposed fetus. Pediatrics. 2013;131:e1009–1024. doi: 10.1542/peds.2012-3931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhide PG, Kosofsky BE. Neuro-developmental consequences of prenatal drug exposure. Preface Dev Neurosci. 2009;31:5. doi: 10.1159/000209397. [DOI] [PubMed] [Google Scholar]
- Bick J, Naumova O, Hunter S, Barbot B, Lee M, Luthar SS, Raefski A, Grigorenko EL. Childhood adversity and DNA methylation of genes involved in the hypothalamus-pituitary-adrenal axis and immune system: whole-genome and candidate-gene associations. Dev Psychopathol. 2012;24:1417–1425. doi: 10.1017/S0954579412000806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Birnbaum R, Jaffe AE, Chen Q, Hyde TM, Kleinman JE, Weinberger DR. Investigation of the prenatal expression patterns of 108 schizophrenia-associated genetic loci. Biol Psychiatry. 2015;77:e43–51. doi: 10.1016/j.biopsych.2014.10.008. [DOI] [PubMed] [Google Scholar]
- Birnbaum R, Jaffe AE, Hyde TM, Kleinman JE, Weinberger DR. Prenatal expression patterns of genes associated with neuropsychiatric disorders. Am J Psychiatry. 2014;171:758–767. doi: 10.1176/appi.ajp.2014.13111452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Biswal B, Yetkin FZ, Haughton VM, Hyde JS. Functional connectivity in the motor cortex of resting human brain using echo-planar MRI. Magn Reson Med. 1995;34:537–541. doi: 10.1002/mrm.1910340409. [DOI] [PubMed] [Google Scholar]
- Booij L, Tremblay RE, Szyf M, Benkelfat C. Genetic and early environmental influences on the serotonin system: consequences for brain development and risk for psychopathology. J Psychiatry Neurosci. 2015;40:5–18. doi: 10.1503/jpn.140099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brady TM, Visscher W, Feder M, Burns AM. Maternal drug use and the timing of prenatal care. J Health Care Poor Underserved. 2003;14:588–607. doi: 10.1353/hpu.2010.0700. [DOI] [PubMed] [Google Scholar]
- Bruder-Costello B, Warner V, Talati A, Nomura Y, Bruder G, Weissman M. Temperament among offspring at high and low risk for depression. Psychiatry research. 2007;153:145–151. doi: 10.1016/j.psychres.2007.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caddy C, Giaroli G, White TP, Shergill SS, Tracy DK. Ketamine as the prototype glutamatergic antidepressant: pharmacodynamic actions, and a systematic review and meta-analysis of efficacy. Ther Adv Psychopharmacol. 2014;4:75–99. doi: 10.1177/2045125313507739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao M, He Y, Dai Z, Liao X, Jeon T, Ouyang M, Chalak L, Bi Y, Rollins N, Dong Q, Huang H. Early Development of Functional Network Segregation Revealed by Connectomic Analysis of the Preterm Human Brain. Cereb Cortex. 2017;27:1949–1963. doi: 10.1093/cercor/bhw038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caspi A, McClay J, Moffitt TE, Mill J, Martin J, Craig IW, Taylor A, Poulton R. Role of genotype in the cycle of violence in maltreated children. Science. 2002;297:851–854. doi: 10.1126/science.1072290. [DOI] [PubMed] [Google Scholar]
- Caspi A, Moffitt TE. Gene-environment interactions in psychiatry: joining forces with neuroscience. Nat Rev Neurosci. 2006;7:583–590. doi: 10.1038/nrn1925. [DOI] [PubMed] [Google Scholar]
- Challis JR, Sloboda D, Matthews SG, Holloway A, Alfaidy N, Patel FA, Whittle W, Fraser M, Moss TJ, Newnham J. The fetal placental hypothalamic-pituitary-adrenal (HPA) axis, parturition and post natal health. Mol Cell Endocrinol. 2001;185:135–144. doi: 10.1016/s0303-7207(01)00624-4. [DOI] [PubMed] [Google Scholar]
- Chasnoff IJ, Burns KA, Burns WJ, Schnoll SH. Prenatal drug exposure: effects on neonatal and infant growth and development. Neurobehav Toxicol Teratol. 1986;8:357–362. [PubMed] [Google Scholar]
- Chiang MC, Barysheva M, Shattuck DW, Lee AD, Madsen SK, Avedissian C, Klunder AD, Toga AW, McMahon KL, de Zubicaray GI, Wright MJ, Srivastava A, Balov N, Thompson PM. Genetics of brain fiber architecture and intellectual performance. J Neurosci. 2009;29:2212–2224. doi: 10.1523/JNEUROSCI.4184-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ching M, Tang L. Neuroleptic drug-induced alterations on neonatal growth and development. I. Prenatal exposure influences birth size, mortality rate, and the neuroendocrine system. Biol Neonate. 1986;49:261–269. doi: 10.1159/000242540. [DOI] [PubMed] [Google Scholar]
- Collins AL, Kim Y, Sklar P, International Schizophrenia, C. O’Donovan MC, Sullivan PF. Hypothesis-driven candidate genes for schizophrenia compared to genome-wide association results. Psychol Med. 2012;42:607–616. doi: 10.1017/S0033291711001607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Connor PD, Sampson PD, Streissguth AP, Bookstein FL, Barr HM. Effects of prenatal alcohol exposure on fine motor coordination and balance: A study of two adult samples. Neuropsychologia. 2006;44:744–751. doi: 10.1016/j.neuropsychologia.2005.07.016. [DOI] [PubMed] [Google Scholar]
- Dean DC, 3rd, Jerskey BA, Chen K, Protas H, Thiyyagura P, Roontiva A, O’Muircheartaigh J, Dirks H, Waskiewicz N, Lehman K, Siniard AL, Turk MN, Hua X, Madsen SK, Thompson PM, Fleisher AS, Huentelman MJ, Deoni SC, Reiman EM. Brain differences in infants at differential genetic risk for late-onset Alzheimer disease: a cross-sectional imaging study. JAMA Neurol. 2014;71:11–22. doi: 10.1001/jamaneurol.2013.4544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Degnan KA, Almas AN, Fox NA. Temperament and the environment in the etiology of childhood anxiety. Journal of Child Psychology and Psychiatry. 2010;51:497–517. doi: 10.1111/j.1469-7610.2010.02228.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deoni SC, Mercure E, Blasi A, Gasston D, Thomson A, Johnson M, Williams SC, Murphy DG. Mapping infant brain myelination with magnetic resonance imaging. J Neurosci. 2011;31:784–791. doi: 10.1523/JNEUROSCI.2106-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Derauf C, Kekatpure M, Neyzi N, Lester B, Kosofsky B. Neuroimaging of children following prenatal drug exposure. Semin Cell Dev Biol. 2009;20:441–454. doi: 10.1016/j.semcdb.2009.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doehring A, Oertel BG, Sittl R, Lotsch J. Chronic opioid use is associated with increased DNA methylation correlating with increased clinical pain. Pain. 2013;154:15–23. doi: 10.1016/j.pain.2012.06.011. [DOI] [PubMed] [Google Scholar]
- Doria V, Beckmann CF, Arichi T, Merchant N, Groppo M, Turkheimer FE, Counsell SJ, Murgasova M, Aljabar P, Nunes RG, Larkman DJ, Rees G, Edwards AD. Emergence of resting state networks in the preterm human brain. Proc Natl Acad Sci U S A. 2011;107:20015–20020. doi: 10.1073/pnas.1007921107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dow-Edwards DL, Benveniste H, Behnke M, Bandstra ES, Singer LT, Hurd YL, Stanford LR. Neuroimaging of prenatal drug exposure. Neurotoxicol Teratol. 2006;28:386–402. doi: 10.1016/j.ntt.2006.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Downing C, Balderrama-Durbin C, Broncucia H, Gilliam D, Johnson TE. Ethanol teratogenesis in five inbred strains of mice. Alcohol Clin Exp Res. 2009;33:1238–1245. doi: 10.1111/j.1530-0277.2009.00949.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dreyfus-Brisac C, Larroche JC. Discontinuous electroencephalograms in the premature newborn and at term. Electro-anatomo-clinical correlations. Rev Electroencephalogr Neurophysiol Clin. 1971;1:95–99. doi: 10.1016/s0370-4475(71)80022-9. [DOI] [PubMed] [Google Scholar]
- Duman RS, Tallman JF, Nestler EJ. Acute and chronic opiate-regulation of adenylate cyclase in brain: specific effects in locus coeruleus. J Pharmacol Exp Ther. 1988;246:1033–1039. [PubMed] [Google Scholar]
- Emerson RW, Adams C, Nishino T, Hazlett HC, Wolff JJ, Zwaigenbaum L, Constantino JN, Shen MD, Swanson MR, Elison JT, Kandala S, Estes AM, Botteron KN, Collins L, Dager SR, Evans AC, Gerig G, Gu H, McKinstry RC, Paterson S, Schultz RT, Styner M, Network, I. Schlaggar BL, Pruett JR, Jr, Piven J. Functional neuroimaging of high-risk 6-month-old infants predicts a diagnosis of autism at 24 months of age. Sci Transl Med. 2017;9 doi: 10.1126/scitranslmed.aag2882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eschenroeder AC, Vestal-Laborde AA, Sanchez ES, Robinson SE, Sato-Bigbee C. Oligodendrocyte responses to buprenorphine uncover novel and opposing roles of mu-opioid- and nociceptin/orphanin FQ receptors in cell development: implications for drug addiction treatment during pregnancy. Glia. 2012;60:125–136. doi: 10.1002/glia.21253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Field T, Diego M, Dieter J, Hernandez-Reif M, Schanberg S, Kuhn C, Yando R, Bendell D. Prenatal depression effects on the fetus and the newborn. Infant Behavior and Development. 2004;27:216–229. [Google Scholar]
- Fleming AS, Flett GL, Ruble DN, Shaul DL. Postpartum adjustment in first-time mothers: Relations between mood, maternal attitudes, and mother-infant interactions. Developmental psychology. 1988;24:71–81. [Google Scholar]
- Fransson P, Skiold B, Horsch S, Nordell A, Blennow M, Lagercrantz H, Aden U. Resting-state networks in the infant brain. Proc Natl Acad Sci U S A. 2007;104:15531–15536. doi: 10.1073/pnas.0704380104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaffuri AL, Ladarre D, Lenkei Z. Type-1 cannabinoid receptor signaling in neuronal development. Pharmacology. 2012;90:19–39. doi: 10.1159/000339075. [DOI] [PubMed] [Google Scholar]
- Gangisetty O, Bekdash R, Maglakelidze G, Sarkar DK. Fetal alcohol exposure alters proopiomelanocortin gene expression and hypothalamic-pituitary-adrenal axis function via increasing MeCP2 expression in the hypothalamus. PLoS One. 2014;9:e113228. doi: 10.1371/journal.pone.0113228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao W, Alcauter S, Elton A, Hernandez-Castillo CR, Smith JK, Ramirez J, Lin W. Functional Network Development During the First Year: Relative Sequence and Socioeconomic Correlations. Cereb Cortex. 2014 doi: 10.1093/cercor/bhu088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao W, Lin W, Chen Y, Gerig G, Smith JK, Jewells V, Gilmore JH. Temporal and spatial development of axonal maturation and myelination of white matter in the developing brain. AJNR Am J Neuroradiol. 2009a;30:290–296. doi: 10.3174/ajnr.A1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao W, Lin W, Grewen K, Gilmore JH. Functional Connectivity of the Infant Human Brain: Plastic and Modifiable. Neuroscientist. 2016 doi: 10.1177/1073858416635986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao W, Zhu H, Giovanello KS, Smith JK, Shen D, Gilmore JH, Lin W. Evidence on the emergence of the brain’s default network from 2-week-old to 2-year-old healthy pediatric subjects. Proc Natl Acad Sci U S A. 2009b;106:6790–6795. doi: 10.1073/pnas.0811221106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilmore JH, Kang C, Evans DD, Wolfe HM, Smith JK, Lieberman JA, Lin W, Hamer RM, Styner M, Gerig G. Prenatal and neonatal brain structure and white matter maturation in children at high risk for schizophrenia. Am J Psychiatry. 2010;167:1083–1091. doi: 10.1176/appi.ajp.2010.09101492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilmore JH, Lin W, Prastawa MW, Looney CB, Vetsa YS, Knickmeyer RC, Evans DD, Smith JK, Hamer RM, Lieberman JA, Gerig G. Regional gray matter growth, sexual dimorphism, and cerebral asymmetry in the neonatal brain. J Neurosci. 2007;27:1255–1260. doi: 10.1523/JNEUROSCI.3339-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilmore JH, Shi F, Woolson SL, Knickmeyer RC, Short SJ, Lin W, Zhu H, Hamer RM, Styner M, Shen D. Longitudinal development of cortical and subcortical gray matter from birth to 2 years. Cereb Cortex. 2012;22:2478–2485. doi: 10.1093/cercor/bhr327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gitau R, Cameron A, Fisk NM, Glover V. Fetal exposure to maternal cortisol. Lancet. 1998;352:707–708. doi: 10.1016/S0140-6736(05)60824-0. [DOI] [PubMed] [Google Scholar]
- Glahn DC, Winkler AM, Kochunov P, Almasy L, Duggirala R, Carless MA, Curran JC, Olvera RL, Laird AR, Smith SM, Beckmann CF, Fox PT, Blangero J. Genetic control over the resting brain. Proc Natl Acad Sci U S A. 2010;107:1223–1228. doi: 10.1073/pnas.0909969107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goldstein S, Reynolds C. Handbook of neurodevelopmental and genetic disorders in children. Guilford Press; New York: 2011. [Google Scholar]
- Goodlett CR, Gilliam DM, Nichols JM, West JR. Genetic influences on brain growth restriction induced by development exposure to alcohol. Neurotoxicology. 1989;10:321–334. [PubMed] [Google Scholar]
- Graham AM, Pfeifer JH, Fisher PA, Lin W, Gao W, Fair DA. The potential of infant fMRI research and the study of early life stress as a promising exemplar. Dev Cogn Neurosci. 2014;12C:12–39. doi: 10.1016/j.dcn.2014.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grewen K, Burchinal M, Vachet C, Gouttard S, Gilmore JH, Lin W, Johns J, Elam M, Gerig G. Prenatal cocaine effects on brain structure in early infancy. Neuroimage. 2014;101:114–123. doi: 10.1016/j.neuroimage.2014.06.070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grewen K, Salzwedel AP, Gao W. Functional Connectivity Disruption in Neonates with Prenatal Marijuana Exposure. Front Hum Neurosci. 2015;9:601. doi: 10.3389/fnhum.2015.00601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grossmann T, Johnson MH. Selective prefrontal cortex responses to joint attention in early infancy. Biol Lett. 2010 doi: 10.1098/rsbl.2009.1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harkany T, Keimpema E, Barabas K, Mulder J. Endocannabinoid functions controlling neuronal specification during brain development. Mol Cell Endocrinol. 2008;286:S84–90. doi: 10.1016/j.mce.2008.02.011. [DOI] [PubMed] [Google Scholar]
- Hashimoto K, Sawa A, Iyo M. Increased Levels of Glutamate in Brains from Patients with Mood Disorders. Biological psychiatry. 2007;62:1310–1316. doi: 10.1016/j.biopsych.2007.03.017. [DOI] [PubMed] [Google Scholar]
- Hazlett HC, Gu H, Munsell BC, Kim SH, Styner M, Wolff JJ, Elison JT, Swanson MR, Zhu H, Botteron KN, Collins DL, Constantino JN, Dager SR, Estes AM, Evans AC, Fonov VS, Gerig G, Kostopoulos P, McKinstry RC, Pandey J, Paterson S, Pruett JR, Schultz RT, Shaw DW, Zwaigenbaum L, Piven J, Network, I., Clinical, S., Data Coordinating, C., Image Processing, C., Statistical, A. Early brain development in infants at high risk for autism spectrum disorder. Nature. 2017;542:348–351. doi: 10.1038/nature21369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hessl D, Dyer-Friedman J, Glaser B, Wisbeck J, Barajas RG, Taylor A, Reiss AL. The influence of environmental and genetic factors on behavior problems and autistic symptoms in boys and girls with fragile X syndrome. Pediatrics. 2001;108:E88. doi: 10.1542/peds.108.5.e88. [DOI] [PubMed] [Google Scholar]
- Hibar DP, Adams HHH, Jahanshad N, Chauhan G, Stein JL, Hofer E, Renteria ME, Bis JC, Arias-Vasquez A, Ikram MK, Desrivieres S, Vernooij MW, Abramovic L, Alhusaini S, Amin N, Andersson M, Arfanakis K, Aribisala BS, Armstrong NJ, Athanasiu L, Axelsson T, Beecham AH, Beiser A, Bernard M, Blanton SH, Bohlken MM, Boks MP, Bralten J, Brickman AM, Carmichael O, Chakravarty MM, Chen Q, Ching CRK, Chouraki V, Cuellar-Partida G, Crivello F, Den Braber A, Doan NT, Ehrlich S, Giddaluru S, Goldman AL, Gottesman RF, Grimm O, Griswold ME, Guadalupe T, Gutman BA, Hass J, Haukvik UK, Hoehn D, Holmes AJ, Hoogman M, Janowitz D, Jia T, Jorgensen KN, Karbalai N, Kasperaviciute D, Kim S, Klein M, Kraemer B, Lee PH, Liewald DCM, Lopez LM, Luciano M, Macare C, Marquand AF, Matarin M, Mather KA, Mattheisen M, McKay DR, Milaneschi Y, Munoz Maniega S, Nho K, Nugent AC, Nyquist P, Loohuis LMO, Oosterlaan J, Papmeyer M, Pirpamer L, Putz B, Ramasamy A, Richards JS, Risacher SL, Roiz-Santianez R, Rommelse N, Ropele S, Rose EJ, Royle NA, Rundek T, Samann PG, Saremi A, Satizabal CL, Schmaal L, Schork AJ, Shen L, Shin J, Shumskaya E, Smith AV, Sprooten E, Strike LT, Teumer A, Tordesillas-Gutierrez D, Toro R, Trabzuni D, Trompet S, Vaidya D, Van der Grond J, Van der Lee SJ, Van der Meer D, Van Donkelaar MMJ, Van Eijk KR, Van Erp TGM, Van Rooij D, Walton E, Westlye LT, Whelan CD, Windham BG, Winkler AM, Wittfeld K, Woldehawariat G, Wolf C, Wolfers T, Yanek LR, Yang J, Zijdenbos A, Zwiers MP, Agartz I, Almasy L, Ames D, Amouyel P, Andreassen OA, Arepalli S, Assareh AA, Barral S, Bastin ME, Becker DM, Becker JT, Bennett DA, Blangero J, van Bokhoven H, Boomsma DI, Brodaty H, Brouwer RM, Brunner HG, Buckner RL, Buitelaar JK, Bulayeva KB, Cahn W, Calhoun VD, Cannon DM, Cavalleri GL, Cheng CY, Cichon S, Cookson MR, Corvin A, Crespo-Facorro B, Curran JE, Czisch M, Dale AM, Davies GE, De Craen AJM, De Geus EJC, De Jager PL, De Zubicaray GI, Deary IJ, Debette S, DeCarli C, Delanty N, Depondt C, DeStefano A, Dillman A, Djurovic S, Donohoe G, Drevets WC, Duggirala R, Dyer TD, Enzinger C, Erk S, Espeseth T, Fedko IO, Fernandez G, Ferrucci L, Fisher SE, Fleischman DA, Ford I, Fornage M, Foroud TM, Fox PT, Francks C, Fukunaga M, Gibbs JR, Glahn DC, Gollub RL, Goring HHH, Green RC, Gruber O, Gudnason V, Guelfi S, Haberg AK, Hansell NK, Hardy J, Hartman CA, Hashimoto R, Hegenscheid K, Heinz A, Le Hellard S, Hernandez DG, Heslenfeld DJ, Ho BC, Hoekstra PJ, Hoffmann W, Hofman A, Holsboer F, Homuth G, Hosten N, Hottenga JJ, Huentelman M, Hulshoff Pol HE, Ikeda M, Jack CR, Jr, Jenkinson M, Johnson R, Jonsson EG, Jukema JW, Kahn RS, Kanai R, Kloszewska I, Knopman DS, Kochunov P, Kwok JB, Lawrie SM, Lemaitre H, Liu X, Longo DL, Lopez OL, Lovestone S, Martinez O, Martinot JL, Mattay VS, McDonald C, McIntosh AM, McMahon FJ, McMahon KL, Mecocci P, Melle I, Meyer-Lindenberg A, Mohnke S, Montgomery GW, Morris DW, Mosley TH, Muhleisen TW, Muller-Myhsok B, Nalls MA, Nauck M, Nichols TE, Niessen WJ, Nothen MM, Nyberg L, Ohi K, Olvera RL, Ophoff RA, Pandolfo M, Paus T, Pausova Z, Penninx B, Pike GB, Potkin SG, Psaty BM, Reppermund S, Rietschel M, Roffman JL, Romanczuk-Seiferth N, Rotter JI, Ryten M, Sacco RL, Sachdev PS, Saykin AJ, Schmidt R, Schmidt H, Schofield PR, Sigursson S, Simmons A, Singleton A, Sisodiya SM, Smith C, Smoller JW, Soininen H, Steen VM, Stott DJ, Sussmann JE, Thalamuthu A, Toga AW, Traynor BJ, Troncoso J, Tsolaki M, Tzourio C, Uitterlinden AG, Hernandez MCV, Van der Brug M, van der Lugt A, van der Wee NJA, Van Haren NEM, van ’t Ent D, Van Tol MJ, Vardarajan BN, Vellas B, Veltman DJ, Volzke H, Walter H, Wardlaw JM, Wassink TH, Weale ME, Weinberger DR, Weiner MW, Wen W, Westman E, White T, Wong TY, Wright CB, Zielke RH, Zonderman AB, Martin NG, Van Duijn CM, Wright MJ, Longstreth WT, Schumann G, Grabe HJ, Franke B, Launer LJ, Medland SE, Seshadri S, Thompson PM, Ikram MA. Novel genetic loci associated with hippocampal volume. Nat Commun. 2017;8:13624. doi: 10.1038/ncomms13624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoffbuhr KC, Moses LM, Jerdonek MA, Naidu S, Hoffman EP. Associations between MeCP2 mutations, X-chromosome inactivation, and phenotype. Ment Retard Dev Disabil Res Rev. 2002;8:99–105. doi: 10.1002/mrdd.10026. [DOI] [PubMed] [Google Scholar]
- Huang H, Shu N, Mishra V, Jeon T, Chalak L, Wang ZJ, Rollins N, Gong G, Cheng H, Peng Y, Dong Q, He Y. Development of human brain structural networks through infancy and childhood. Cereb Cortex. 2015;25:1389–1404. doi: 10.1093/cercor/bht335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang H, Zhang J, Wakana S, Zhang W, Ren T, Richards LJ, Yarowsky P, Donohue P, Graham E, van Zijl PC, Mori S. White and gray matter development in human fetal, newborn and pediatric brains. Neuroimage. 2006;33:27–38. doi: 10.1016/j.neuroimage.2006.06.009. [DOI] [PubMed] [Google Scholar]
- Insel TR. Rethinking schizophrenia. Nature. 2010;468:187–193. doi: 10.1038/nature09552. [DOI] [PubMed] [Google Scholar]
- Jacobson SW, Carr LG, Croxford J, Sokol RJ, Li TK, Jacobson JL. Protective effects of the alcohol dehydrogenase-ADH1B allele in children exposed to alcohol during pregnancy. J Pediatr. 2006;148:30–37. doi: 10.1016/j.jpeds.2005.08.023. [DOI] [PubMed] [Google Scholar]
- Kang HJ, Kawasawa YI, Cheng F, Zhu Y, Xu X, Li M, Sousa AM, Pletikos M, Meyer KA, Sedmak G, Guennel T, Shin Y, Johnson MB, Krsnik Z, Mayer S, Fertuzinhos S, Umlauf S, Lisgo SN, Vortmeyer A, Weinberger DR, Mane S, Hyde TM, Huttner A, Reimers M, Kleinman JE, Sestan N. Spatio-temporal transcriptome of the human brain. Nature. 2011;478:483–489. doi: 10.1038/nature10523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karlsen TH, Melum E, Franke A. The utility of genome-wide association studies in hepatology. Hepatology. 2010;51:1833–1842. doi: 10.1002/hep.23564. [DOI] [PubMed] [Google Scholar]
- King E, Campbell A, Belger A, Grewen K. Prenatal Nicotine Exposure Disrupts Infant Neural Markers of Orienting. Nicotine Tob Res. 2017 doi: 10.1093/ntr/ntx177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kinney HC, Ottoson CK, White WF. Three-dimensional distribution of 3H-naloxone binding to opiate receptors in the human fetal and infant brainstem. J Comp Neurol. 1990;291:55–78. doi: 10.1002/cne.902910106. [DOI] [PubMed] [Google Scholar]
- Knickmeyer RC, Wang J, Zhu H, Geng X, Woolson S, Hamer RM, Konneker T, Lin W, Styner M, Gilmore JH. Common variants in psychiatric risk genes predict brain structure at birth. Cereb Cortex. 2014;24:1230–1246. doi: 10.1093/cercor/bhs401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Knothe C, Doehring A, Ultsch A, Lotsch J. Methadone induces hypermethylation of human DNA. Epigenomics. 2016;8:167–179. doi: 10.2217/epi.15.78. [DOI] [PubMed] [Google Scholar]
- Krishnan ML, Van Steenwinckel J, Schang AL, Yan J, Arnadottir J, Le Charpentier T, Csaba Z, Dournaud P, Cipriani S, Auvynet C, Titomanlio L, Pansiot J, Ball G, Boardman JP, Walley AJ, Saxena A, Mirza G, Fleiss B, Edwards AD, Petretto E, Gressens P. Integrative genomics of microglia implicates DLG4 (PSD95) in the white matter development of preterm infants. Nat Commun. 2017;8:428. doi: 10.1038/s41467-017-00422-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krishnan ML, Wang Z, Silver M, Boardman JP, Ball G, Counsell SJ, Walley AJ, Montana G, Edwards AD. Possible relationship between common genetic variation and white matter development in a pilot study of preterm infants. Brain Behav. 2016;6:e00434. doi: 10.1002/brb3.434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee PH, Perlis RH, Jung JY, Byrne EM, Rueckert E, Siburian R, Haddad S, Mayerfeld CE, Heath AC, Pergadia ML, Madden PA, Boomsma DI, Penninx BW, Sklar P, Martin NG, Wray NR, Purcell SM, Smoller JW. Multi-locus genome-wide association analysis supports the role of glutamatergic synaptic transmission in the etiology of major depressive disorder. Transl Psychiatry. 2012;2:e184. doi: 10.1038/tp.2012.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leech SL, Richardson GA, Goldschmidt L, Day NL. Prenatal substance exposure: effects on attention and impulsivity of 6-year-olds. Neurotoxicol Teratol. 1999;21:109–118. doi: 10.1016/s0892-0362(98)00042-7. [DOI] [PubMed] [Google Scholar]
- Lenroot RK, Giedd JN. The changing impact of genes and environment on brain development during childhood and adolescence: initial findings from a neuroimaging study of pediatric twins. Dev Psychopathol. 2008;20:1161–1175. doi: 10.1017/S0954579408000552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lester BM, Padbury JF. Third pathophysiology of prenatal cocaine exposure. Dev Neurosci. 2009;31:23–35. doi: 10.1159/000207491. [DOI] [PubMed] [Google Scholar]
- Lewis JD, Evans AC, Pruett JR, Jr, Botteron KN, McKinstry RC, Zwaigenbaum L, Estes AM, Collins DL, Kostopoulos P, Gerig G, Dager SR, Paterson S, Schultz RT, Styner MA, Hazlett HC, Piven J, Infant Brain Imaging Study, N. The Emergence of Network Inefficiencies in Infants With Autism Spectrum Disorder. Biol Psychiatry. 2017;82:176–185. doi: 10.1016/j.biopsych.2017.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li G, Wang L, Shi F, Lyall AE, Ahn M, Peng Z, Zhu H, Lin W, Gilmore JH, Shen D. Cortical thickness and surface area in neonates at high risk for schizophrenia. Brain Struct Funct. 2016;221:447–461. doi: 10.1007/s00429-014-0917-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lussier AA, Stepien KA, Weinberg J, Kobor MS. Prenatal alcohol exposure alters gene expression in the rat brain: Experimental design and bioinformatic analysis of microarray data. Data Brief. 2015;4:239–252. doi: 10.1016/j.dib.2015.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mars B, Collishaw S, Hammerton G, Rice F, Harold GT, Smith D, Bevan Jones R, Sellers R, Potter R, Craddock N, Thapar AK, Heron J, Thapar A. Longitudinal symptom course in adults with recurrent depression: Impact on impairment and risk of psychopathology in offspring. Journal of affective disorders. 2015;182:32–38. doi: 10.1016/j.jad.2015.04.018. [DOI] [PubMed] [Google Scholar]
- Masten AS, Cicchetti D. Developmental cascades. Dev Psychopathol. 2010;22:491–495. doi: 10.1017/S0954579410000222. [DOI] [PubMed] [Google Scholar]
- Matijasevich A, Murray J, Cooper PJ, Anselmi L, Barros AJD, Barros FC, Santos IS. Trajectories of maternal depression and offspring psychopathology at 6 years: 2004 Pelotas cohort study. Journal of affective disorders. 2015;174:424–431. doi: 10.1016/j.jad.2014.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Michael N, Erfurth A, Ohrmann P, Arolt V, Heindel W, Pfleiderer B. Neurotrophic effects of electroconvulsive therapy: a proton magnetic resonance study of the left amygdalar region in patients with treatment-resistant depression. Neuropsychopharmacology. 2003;28:720–725. doi: 10.1038/sj.npp.1300085. [DOI] [PubMed] [Google Scholar]
- Moffitt TE, Caspi A, Newman DL, Silva PA. Behavioral Observations at Age 3 Years Predict Adult Psychiatric Disorders: Longitudinal Evidence From a Birth Cohort. Archives of General Psychiatry. 1996;53:1033–1039. doi: 10.1001/archpsyc.1996.01830110071009. [DOI] [PubMed] [Google Scholar]
- Nakano T, Watanabe H, Homae F, Taga G. Prefrontal cortical involvement in young infants’ analysis of novelty. Cereb Cortex. 2009;19:455–463. doi: 10.1093/cercor/bhn096. [DOI] [PubMed] [Google Scholar]
- Nestler EJ, Alreja M, Aghajanian GK. Molecular and cellular mechanisms of opiate action: studies in the rat locus coeruleus. Brain Res Bull. 1994;35:521–528. doi: 10.1016/0361-9230(94)90166-x. [DOI] [PubMed] [Google Scholar]
- Nestler EJ, Alreja M, Aghajanian GK. Molecular control of locus coeruleus neurotransmission. Biol Psychiatry. 1999;46:1131–1139. doi: 10.1016/s0006-3223(99)00158-4. [DOI] [PubMed] [Google Scholar]
- Nowakowski R, Hayes N. General principles of NCS development. Blackwell Publishers; 2002. [Google Scholar]
- O’Donnell KJ, Meaney MJ. Fetal Origins of Mental Health: The Developmental Origins of Health and Disease Hypothesis. Am J Psychiatry. 2017;174:319–328. doi: 10.1176/appi.ajp.2016.16020138. [DOI] [PubMed] [Google Scholar]
- Oertel BG, Doehring A, Roskam B, Kettner M, Hackmann N, Ferreiros N, Schmidt PH, Lotsch J. Genetic-epigenetic interaction modulates mu-opioid receptor regulation. Hum Mol Genet. 2012;21:4751–4760. doi: 10.1093/hmg/dds314. [DOI] [PubMed] [Google Scholar]
- Pawlby S, Hay DF, Sharp D, Waters CS, O’Keane V. Antenatal depression predicts depression in adolescent offspring: Prospective longitudinal community-based study. Journal of affective disorders. 2009;113:236–243. doi: 10.1016/j.jad.2008.05.018. [DOI] [PubMed] [Google Scholar]
- Pearson RM, Evans J, Kounali D, Lewis G, Heron J, Ramchandani PG, O’Connor TG, Stein A. Maternal depression during pregnancy and the postnatal period: risks and possible mechanisms for offspring depression at age 18 years. JAMA psychiatry. 2013;70:1312–1319. doi: 10.1001/jamapsychiatry.2013.2163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qiu A, Anh TT, Li Y, Chen H, Rifkin-Graboi A, Broekman BF, Kwek K, Saw SM, Chong YS, Gluckman PD, Fortier MV, Meaney MJ. Prenatal maternal depression alters amygdala functional connectivity in 6-month-old infants. Transl Psychiatry. 2015a;5:e508. doi: 10.1038/tp.2015.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qiu A, Rifkin-Graboi A, Chen H, Chong YS, Kwek K, Gluckman PD, Fortier MV, Meaney MJ. Maternal anxiety and infants’ hippocampal development: timing matters. Transl Psychiatry. 2013;3:e306. doi: 10.1038/tp.2013.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qiu A, Shen M, Buss C, Chong YS, Kwek K, Saw SM, Gluckman PD, Wadhwa PD, Entringer S, Styner M, Karnani N, Heim CM, O’Donnell KJ, Holbrook JD, Fortier MV, Meaney MJ, the, G.s.g Effects of Antenatal Maternal Depressive Symptoms and Socio-Economic Status on Neonatal Brain Development are Modulated by Genetic Risk. Cereb Cortex. 2017;27:3080–3092. doi: 10.1093/cercor/bhx065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qiu A, Tuan TA, Ong ML, Li Y, Chen H, Rifkin-Graboi A, Broekman BF, Kwek K, Saw SM, Chong YS, Gluckman PD, Fortier MV, Holbrook JD, Meaney MJ. COMT haplotypes modulate associations of antenatal maternal anxiety and neonatal cortical morphology. Am J Psychiatry. 2015b;172:163–172. doi: 10.1176/appi.ajp.2014.14030313. [DOI] [PubMed] [Google Scholar]
- Raschle N, Zuk J, Ortiz-Mantilla S, Sliva DD, Franceschi A, Grant PE, Benasich AA, Gaab N. Pediatric neuroimaging in early childhood and infancy: challenges and practical guidelines. Ann N Y Acad Sci. 2012;1252:43–50. doi: 10.1111/j.1749-6632.2012.06457.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Riancho JA. Genome-wide association studies (GWAS) in complex diseases: advantages and limitations. Reumatol Clin. 2012;8:56–57. doi: 10.1016/j.reuma.2011.07.005. [DOI] [PubMed] [Google Scholar]
- Rifkin-Graboi A, Bai J, Chen H, Hameed WB, Sim LW, Tint MT, Leutscher-Broekman B, Chong YS, Gluckman PD, Fortier MV, Meaney MJ, Qiu A. Prenatal maternal depression associates with microstructure of right amygdala in neonates at birth. Biol Psychiatry. 2013;74:837–844. doi: 10.1016/j.biopsych.2013.06.019. [DOI] [PubMed] [Google Scholar]
- Rifkin-Graboi A, Meaney MJ, Chen H, Bai J, Hameed WB, Tint MT, Broekman BF, Chong YS, Gluckman PD, Fortier MV, Qiu A. Antenatal maternal anxiety predicts variations in neural structures implicated in anxiety disorders in newborns. J Am Acad Child Adolesc Psychiatry. 2015;54:313–321 e312. doi: 10.1016/j.jaac.2015.01.013. [DOI] [PubMed] [Google Scholar]
- Ross EJ, Graham DL, Money KM, Stanwood GD. Developmental consequences of fetal exposure to drugs: what we know and what we still must learn. Neuropsychopharmacology. 2015;40:61–87. doi: 10.1038/npp.2014.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salzwedel AP, Grewen KM, Goldman BD, Gao W. Thalamocortical functional connectivity and behavioral disruptions in neonates with prenatal cocaine exposure. Neurotoxicol Teratol. 2016;56:16–25. doi: 10.1016/j.ntt.2016.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salzwedel AP, Grewen KM, Vachet C, Gerig G, Lin W, Gao W. Prenatal drug exposure affects neonatal brain functional connectivity. J Neurosci. 2015;35:5860–5869. doi: 10.1523/JNEUROSCI.4333-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanchez ES, Bigbee JW, Fobbs W, Robinson SE, Sato-Bigbee C. Opioid addiction and pregnancy: perinatal exposure to buprenorphine affects myelination in the developing brain. Glia. 2008;56:1017–1027. doi: 10.1002/glia.20675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schou J, Prockop LD, Dahlstrom G, Rohde C. Penetration of delta-9-tetrahydrocannabinol and 11-OH-delta-9-tetrahydrocannabinol through the blood-brain barrier. Acta Pharmacol Toxicol (Copenh) 1977;41:33–38. doi: 10.1111/j.1600-0773.1977.tb02120.x. [DOI] [PubMed] [Google Scholar]
- Selley DE, Nestler EJ, Breivogel CS, Childers SR. Opioid receptor-coupled G-proteins in rat locus coeruleus membranes: decrease in activity after chronic morphine treatment. Brain Res. 1997;746:10–18. doi: 10.1016/s0006-8993(96)01125-0. [DOI] [PubMed] [Google Scholar]
- Shen MD, Kim SH, McKinstry RC, Gu H, Hazlett HC, Nordahl CW, Emerson RW, Shaw D, Elison JT, Swanson MR, Fonov VS, Gerig G, Dager SR, Botteron KN, Paterson S, Schultz RT, Evans AC, Estes AM, Zwaigenbaum L, Styner MA, Amaral DG, Piven J, Infant Brain Imaging Study, N., Infant Brain Imaging Study Network, T.I.B.I.S.N.i.a.N.I.o.H.-f.A.C.o.E.p., consists of a consortium of eight universities in the United, S., Canada. Piven J, Hazlett HC, Chappell C, Dager S, Estes A, Shaw D, Botteron K, McKinstry R, Constantino J, Pruett J, Schultz R, Zwaigenbaum L, Elison J, Evans AC, Collins DL, Pike GB, Fonov V, Kostopoulos P, Das S, Gerig G, Styner M, Gu H. Increased Extra-axial Cerebrospinal Fluid in High-Risk Infants Who Later Develop Autism. Biol Psychiatry. 2017;82:186–193. doi: 10.1016/j.biopsych.2017.02.1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi F, Yap PT, Gao W, Lin W, Gilmore JH, Shen D. Altered structural connectivity in neonates at genetic risk for schizophrenia: A combined study using morphological and white matter networks. Neuroimage. 2012;62:1622–1633. doi: 10.1016/j.neuroimage.2012.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Skolnick P, Popik P, Trullas R. Glutamate-based antidepressants: 20 years on. Trends in pharmacological sciences. 2009;30:563–569. doi: 10.1016/j.tips.2009.09.002. [DOI] [PubMed] [Google Scholar]
- Smyser CD, Inder TE, Shimony JS, Hill JE, Degnan AJ, Snyder AZ, Neil JJ. Longitudinal analysis of neural network development in preterm infants. Cereb Cortex. 2010;20:2852–2862. doi: 10.1093/cercor/bhq035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stein JL, Medland SE, Vasquez AA, Hibar DP, Senstad RE, Winkler AM, Toro R, Appel K, Bartecek R, Bergmann O, Bernard M, Brown AA, Cannon DM, Chakravarty MM, Christoforou A, Domin M, Grimm O, Hollinshead M, Holmes AJ, Homuth G, Hottenga JJ, Langan C, Lopez LM, Hansell NK, Hwang KS, Kim S, Laje G, Lee PH, Liu X, Loth E, Lourdusamy A, Mattingsdal M, Mohnke S, Maniega SM, Nho K, Nugent AC, O’Brien C, Papmeyer M, Putz B, Ramasamy A, Rasmussen J, Rijpkema M, Risacher SL, Roddey JC, Rose EJ, Ryten M, Shen L, Sprooten E, Strengman E, Teumer A, Trabzuni D, Turner J, van Eijk K, van Erp TG, van Tol MJ, Wittfeld K, Wolf C, Woudstra S, Aleman A, Alhusaini S, Almasy L, Binder EB, Brohawn DG, Cantor RM, Carless MA, Corvin A, Czisch M, Curran JE, Davies G, de Almeida MA, Delanty N, Depondt C, Duggirala R, Dyer TD, Erk S, Fagerness J, Fox PT, Freimer NB, Gill M, Goring HH, Hagler DJ, Hoehn D, Holsboer F, Hoogman M, Hosten N, Jahanshad N, Johnson MP, Kasperaviciute D, Kent JW, Jr, Kochunov P, Lancaster JL, Lawrie SM, Liewald DC, Mandl R, Matarin M, Mattheisen M, Meisenzahl E, Melle I, Moses EK, Muhleisen TW, Nauck M, Nothen MM, Olvera RL, Pandolfo M, Pike GB, Puls R, Reinvang I, Renteria ME, Rietschel M, Roffman JL, Royle NA, Rujescu D, Savitz J, Schnack HG, Schnell K, Seiferth N, Smith C, Steen VM, Valdes Hernandez MC, Van den Heuvel M, van der Wee NJ, Van Haren NE, Veltman JA, Volzke H, Walker R, Westlye LT, Whelan CD, Agartz I, Boomsma DI, Cavalleri GL, Dale AM, Djurovic S, Drevets WC, Hagoort P, Hall J, Heinz A, Jack CR, Jr, Foroud TM, Le Hellard S, Macciardi F, Montgomery GW, Poline JB, Porteous DJ, Sisodiya SM, Starr JM, Sussmann J, Toga AW, Veltman DJ, Walter H, Weiner MW, Alzheimer’s Disease Neuroimaging, I., Consortium, E., Consortium, I., Saguenay Youth Study, G. Bis JC, Ikram MA, Smith AV, Gudnason V, Tzourio C, Vernooij MW, Launer LJ, DeCarli C, Seshadri S, Cohorts for, H., Aging Research in Genomic Epidemiology, C. Andreassen OA, Apostolova LG, Bastin ME, Blangero J, Brunner HG, Buckner RL, Cichon S, Coppola G, de Zubicaray GI, Deary IJ, Donohoe G, de Geus EJ, Espeseth T, Fernandez G, Glahn DC, Grabe HJ, Hardy J, Hulshoff Pol HE, Jenkinson M, Kahn RS, McDonald C, McIntosh AM, McMahon FJ, McMahon KL, Meyer-Lindenberg A, Morris DW, Muller-Myhsok B, Nichols TE, Ophoff RA, Paus T, Pausova Z, Penninx BW, Potkin SG, Samann PG, Saykin AJ, Schumann G, Smoller JW, Wardlaw JM, Weale ME, Martin NG, Franke B, Wright MJ, Thompson PM, Enhancing Neuro Imaging Genetics through Meta-Analysis, C. Identification of common variants associated with human hippocampal and intracranial volumes. Nat Genet. 2012;44:552–561. doi: 10.1038/ng.2250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Streissguth AP, Dehaene P. Fetal alcohol syndrome in twins of alcoholic mothers: concordance of diagnosis and IQ. Am J Med Genet. 1993;47:857–861. doi: 10.1002/ajmg.1320470612. [DOI] [PubMed] [Google Scholar]
- Swanson JD, Wadhwa PM. Developmental origins of child mental health disorders. J Child Psychol Psychiatry. 2008;49:1009–1019. doi: 10.1111/j.1469-7610.2008.02014.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tau GZ, Peterson BS. Normal development of brain circuits. Neuropsychopharmacology. 2010;35:147–168. doi: 10.1038/npp.2009.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thompson BL, Levitt P, Stanwood GD. Prenatal exposure to drugs: effects on brain development and implications for policy and education. Nat Rev Neurosci. 2009;10:303–312. doi: 10.1038/nrn2598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thompson PM, Cannon TD, Narr KL, van Erp T, Poutanen VP, Huttunen M, Lonnqvist J, Standertskjold-Nordenstam CG, Kaprio J, Khaledy M, Dail R, Zoumalan CI, Toga AW. Genetic influences on brain structure. Nat Neurosci. 2001;4:1253–1258. doi: 10.1038/nn758. [DOI] [PubMed] [Google Scholar]
- Van Bockstaele EJ, Reyes BA, Valentino RJ. The locus coeruleus: A key nucleus where stress and opioids intersect to mediate vulnerability to opiate abuse. Brain Res. 2010;1314:162–174. doi: 10.1016/j.brainres.2009.09.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Bockstaele EJ, Valentino RJ. Neuropeptide regulation of the locus coeruleus and opiate-induced plasticity of stress responses. Adv Pharmacol. 2013;68:405–420. doi: 10.1016/B978-0-12-411512-5.00019-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van den Bergh BR, Mulder EJ, Mennes M, Glover V. Antenatal maternal anxiety and stress and the neurobehavioural development of the fetus and child: links and possible mechanisms. A review. Neurosci Biobehav Rev. 2005;29:237–258. doi: 10.1016/j.neubiorev.2004.10.007. [DOI] [PubMed] [Google Scholar]
- Vestal-Laborde AA, Eschenroeder AC, Bigbee JW, Robinson SE, Sato-Bigbee C. The opioid system and brain development: effects of methadone on the oligodendrocyte lineage and the early stages of myelination. Dev Neurosci. 2014;36:409–421. doi: 10.1159/000365074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Videman M, Tokariev A, Stjerna S, Roivainen R, Gaily E, Vanhatalo S. Effects of prenatal antiepileptic drug exposure on newborn brain activity. Epilepsia. 2016;57:252–262. doi: 10.1111/epi.13281. [DOI] [PubMed] [Google Scholar]
- Walhovd KB, Moe V, Slinning K, Due-Tonnessen P, Bjornerud A, Dale AM, van der Kouwe A, Quinn BT, Kosofsky B, Greve D, Fischl B. Volumetric cerebral characteristics of children exposed to opiates and other substances in utero. Neuroimage. 2007;36:1331–1344. doi: 10.1016/j.neuroimage.2007.03.070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walhovd KB, Westlye LT, Moe V, Slinning K, Due-Tonnessen P, Bjornerud A, van der Kouwe A, Dale AM, Fjell AM. White matter characteristics and cognition in prenatally opiate- and polysubstance-exposed children: a diffusion tensor imaging study. AJNR Am J Neuroradiol. 2010;31:894–900. doi: 10.3174/ajnr.A1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walsh T, McClellan JM, McCarthy SE, Addington AM, Pierce SB, Cooper GM, Nord AS, Kusenda M, Malhotra D, Bhandari A, Stray SM, Rippey CF, Roccanova P, Makarov V, Lakshmi B, Findling RL, Sikich L, Stromberg T, Merriman B, Gogtay N, Butler P, Eckstrand K, Noory L, Gochman P, Long R, Chen Z, Davis S, Baker C, Eichler EE, Meltzer PS, Nelson SF, Singleton AB, Lee MK, Rapoport JL, King MC, Sebat J. Rare structural variants disrupt multiple genes in neurodevelopmental pathways in schizophrenia. Science. 2008;320:539–543. doi: 10.1126/science.1155174. [DOI] [PubMed] [Google Scholar]
- Warner TD, Behnke M, Eyler FD, Padgett K, Leonard C, Hou W, Garvan CW, Schmalfuss IM, Blackband SJ. Diffusion tensor imaging of frontal white matter and executive functioning in cocaine-exposed children. Pediatrics. 2006;118:2014–2024. doi: 10.1542/peds.2006-0003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Warren KR, Li TK. Genetic polymorphisms: impact on the risk of fetal alcohol spectrum disorders. Birth Defects Res A Clin Mol Teratol. 2005;73:195–203. doi: 10.1002/bdra.20125. [DOI] [PubMed] [Google Scholar]
- Waters CS, Hay DF, Simmonds JR, van Goozen SHM. Antenatal depression and children’s developmental outcomes: potential mechanisms and treatment options. European child & adolescent psychiatry. 2014;23:957. doi: 10.1007/s00787-014-0582-3. [DOI] [PubMed] [Google Scholar]
- Wen DJ, Poh JS, Ni SN, Chong YS, Chen H, Kwek K, Shek LP, Gluckman PD, Fortier MV, Meaney MJ, Qiu A. Influences of prenatal and postnatal maternal depression on amygdala volume and microstructure in young children. Transl Psychiatry. 2017a;7:e1103. doi: 10.1038/tp.2017.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wen DJ, Soe NN, Sim LW, Sanmugam S, Kwek K, Chong YS, Gluckman PD, Meaney MJ, Rifkin-Graboi A, Qiu A. Infant frontal EEG asymmetry in relation with postnatal maternal depression and parenting behavior. Transl Psychiatry. 2017b;7:e1057. doi: 10.1038/tp.2017.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilcox T, Haslup JA, Boas DA. Dissociation of processing of featural and spatiotemporal information in the infant cortex. Neuroimage. 2010;53:1256–1263. doi: 10.1016/j.neuroimage.2010.06.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilcox T, Stubbs J, Hirshkowitz A, Boas DA. Object processing and functional organization of the infant cortex. Neuroimage. 2012;62:1883–1840. doi: 10.1016/j.neuroimage.2012.05.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wolff JJ, Gerig G, Lewis JD, Soda T, Styner MA, Vachet C, Botteron KN, Elison JT, Dager SR, Estes AM, Hazlett HC, Schultz RT, Zwaigenbaum L, Piven J, Network, I. Altered corpus callosum morphology associated with autism over the first 2 years of life. Brain. 2015;138:2046–2058. doi: 10.1093/brain/awv118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xia K, Zhang J, Ahn M, Jha S, Crowley JJ, Szatkiewicz J, Li T, Zou F, Zhu H, Hibar D, Thompson P, Consortium, E. Sullivan PF, Styner M, Gilmore JH, Knickmeyer RC. Genome-wide association analysis identifies common variants influencing infant brain volumes. Transl Psychiatry. 2017;7:e1188. doi: 10.1038/tp.2017.159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yehuda R, Flory JD, Southwick S, Charney DS. Developing an agenda for translational studies of resilience and vulnerability following trauma exposure. Ann N Y Acad Sci. 2006;1071:379–396. doi: 10.1196/annals.1364.028. [DOI] [PubMed] [Google Scholar]