Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Apr 1.
Published in final edited form as: Child Adolesc Psychiatr Clin N Am. 2012 Feb 17;21(2):237–259. doi: 10.1016/j.chc.2012.01.004

Developmental Risk I: Depression and the Developing Brain

John M Weir a, Arthurine Zakama b, Uma Rao a,b,c
PMCID: PMC3338920  NIHMSID: NIHMS352100  PMID: 22537725

SYNOPSIS

This article discusses recent findings on the neurobiology of pediatric depression as well as the interplay between genetic and environmental factors in determining the risk for the disorder. Utilizing data from both animal and human studies, the authors focus on the evolving understanding of the developmental neurobiology of emotional regulation, cognitive function and social behavior as it applies to the risk and clinical course of depression. Treatment implications and directions for future research are also discussed.

Keywords: Adolescent, Depression, Brain, Development, Neurobiology

INTRODUCTION

In the past three decades, public health recognition of depression in children and adolescents has increased significantly. The prevalence of pediatric depression is on the rise14, and depression during this developmental period is associated with significant impairment in multiple social domains58. Furthermore, there is evidence that early depressive episodes persist and/or recur into adult life along with ongoing psychosocial difficulties8,9. A growing body of research has been identifying the neurobiological and psychological correlates8,10,11. In addition, recent studies have begun to identify specific genetic and experiential risk factors1215. The aim of this article is to describe recent findings on the neurobiology of pediatric depression as well as the interplay between genetic and environmental factors in determining the risk for the disorder. In particular, utilizing data from both animal and human studies, we will focus on the evolving understanding of the developmental neurobiology of emotional regulation, cognitive function and social behavior as it applies to the risk and clinical course of depression. Treatment implications and directions for future research also will be discussed.

DELOPMENTAL INFLUENCES ON THE VULNERABILTY TO DEPRESSION

The risk for depression increases markedly during the transition from childhood to adolescence1. Adolescence is a crucial developmental stage marked by a confluence of physical, biological, psychological and social challenges1619. There are significant physical maturational changes (e.g., the onset of puberty), social-cognitive advances (e.g., ability for more abstract thinking and generalizations across situations and time), interpersonal transitions (e.g., changes in social roles in family and peer relationships), and social-contextual changes (e.g., school transitions). Although these maturational transitions offer tremendous opportunities for youth, because the developing brain regions underlying emotional, cognitive and behavioral systems mature at different rates, and because these systems are under the control of both common and independent biological processes, this developmental period also is marked by heightened vulnerability16,18,2022. The normative developmental transitions associated with adolescence might serve as sensitive periods for the activation of specific processes involved in the onset, persistence and recurrence of depressive episodes2325.

THE DEVELOPMENTAL NEUROBIOLOGY OF ADOLESCENCE

Adolescence is perhaps the greatest time of neural change and maturation since infancy26,27. Simultaneously, this period of brain maturation is marked by improvements in the ability to understand social and emotional cues, as well as an increase in the responsiveness to and importance of peer and other interpersonal relationships2831. Also, there is a gradual increase in the capacity for cognitive control and executive function, including abstract thought, organization, decision-making and planning, and response-inhibition16,30,32,33. Until recently, there was limited research on the neurobiological changes that accompany the emotional and cognitive changes that occur during adolescence. The application of magnetic resonance imaging (MRI) techniques has enabled researchers to examine the specific areas and circuits within the brain that are involved in the development of emotional and cognitive abilities31,3436.

Although there is a minimal increase in brain size after early-school-age years, remodeling of grey and white matter occurs throughout adolescence and into early adulthood34,37. In the grey matter, these changes are non-linear and region-specific. The grey matter changes take the form of increased myelination of different cortical connections and/or synaptic pruning, with a net reduction in volume 26,3740. There is a simultaneous linear increase in white matter density associated with increases in the diameter and myelination of the axons forming the fiber tracts alongside increased neural size and proliferation of glia27,3942.

Myelination increases the speed of neural transmission27,43. Synaptic pruning is the process by which excess connections (synapses) between neurons are removed. Synapse elimination is believed to reduce the immature pattern of processing44. The elimination of this immature pattern of processing is adaptive in that in its immature state it requires greater metabolic activity and the recruitment of a wider array of structures44. Additionally, pruning appears to increase the efficiency of cognitive processing through the creation of dedicated neural networks36. For instance, synaptic overproduction followed by selective pruning allows for maximum efficiency in associative memory functions45,46. Disturbances in these developmental patterns can adversely affect behavioral, emotional and cognitive control21,22.

Neural Circuits

Prominent developmental transformations are seen in prefrontal cortex (PFC) and limbic brain regions of adolescents across a variety of species16,18. The above-described emotional and cognitive processes and social behavior (i.e., self-regulation) appear to depend on the maturation of PFC and limbic system interconnectivity4751. The limbic system consists of diverse neural structures, including the cingulate cortex, amygdala and hippocampus, and it regulates emotional experience and motivational learning48,49,5155. Furthermore, maturation of connections between the PFC, basal ganglia and cerebellum also appear to be crucial for the development of higher cognitive functions56.

The PFC mediates the highest cognitive capacities, including reasoning, planning and behavioral control57,58. This relatively large and complex associative brain region has been shown to develop along with other higher-order association regions as children mature from adolescence into adulthood26,58. Structural neuroimaging studies using growth mapping techniques suggest that the prefrontal cortex matures more slowly than other regions of the brain26,59, and that its development parallels the improvements in cognitive control and behavioral inhibition that emerge during the adolescent transition into adulthood60. Frontal lobe maturation, particularly thinning of the cortical gray matter, has been associated with better performance on verbal memory tests in children aged 7–16 years61, and PFC volume in healthy adolescents has been associated with greater ability to inhibit behavioral responses62. Using diffusion tensor imaging (DTI) technology, changes in white matter microstructure have been studied. These studies indicated that anisotropy, a measure that reflects myelin-related restriction of water diffusion across axons, was significantly lower in the frontal white matter in pediatric samples than in adults, suggesting less myelination63,64.

Functional MRI (fMRI) studies indicate that core regions of the neural circuitry underlying cognitive control are on-line early in development32. However, age-related changes in localized processes across the brain, and in establishing long-range connections that support top-down (cortical-subcortical) modulation of behavior and more effective neural processing for optimal mature executive function, have been demonstrated32,6570. With respect to affect regulation, adolescents seem to show a greater magnitude of amygdala activation in response to facial expressions of emotion compared to children and adults7173. The exaggerated amygdala activation in response to emotional cues in adolescents might be related to their intense and variable emotional responses7476. In contrast to the exaggerated amygdala responses to emotional cues, preliminary evidence indicates that the PFC is under-recruited in adolescents compared to adults73,77.

Although some neuroscientists describe cognition and emotion as separable processes implemented by different regions of the brain, such as the amygdala for emotion and the PFC for cognition, functional interactions between the amygdala and PFC mediate emotional influences on cognitive processes and vice versa. These mental processes are inextricably linked and represented in dynamic neural networks composed of interconnected prefrontal and limbic brain structures48,49,51. During adolescence, social relationships take on a new importance and adolescents become adept at reading social and emotional cues, and modulating their affective responses28,78. Evidence suggests that children have difficulty managing interference from competing distractions and the level of difficulty seems to be correlated with the immaturity of posterior and frontal association cortices70. As children mature, they show an increased ability to attend to incoming information and control their behavior in a goal-directed manner60,68,77,79. This development seems to emerge in conjunction with a progressive “frontalization” of functional activity associated with inhibitory processing32,66,70.

Neurotransmitter Systems

In addition to connective and structural changes in the central nervous system, adolescents undergo dramatic alterations in virtually all neurotransmitter systems, including innervation patterns, neurotransmitter levels and signaling mechanisms18,80,81. Developmental studies have shown that neurotransmitter systems generally follow a trajectory of overproduction and pruning, such that changes in expression typically peak during late childhood/early adolescence and are then reduced to reach adult levels. This pattern of overproduction and regressive elimination are believed to fine-tune the brain for efficiency but it also represents a state of vulnerability to exogenous influences18,22,80.

Cholinergic neurons projecting from the basal forebrain innervate the cerebral cortex during critical periods of neural development82. Acetylcholine stimulation may help to promote a favorable environment for neuronal maturation and the refinement of cortical connectivity. Acetylcholine also is likely to play a critical role in neural plasticity. Nicotinic acetylcholine receptors (nAChRs) appear early during development and are expressed throughout the nervous system. They not only exist on neuronal cell bodies and dendrites but also are located on axon terminals and are involved in multiple neurotransmitter release, including acetylcholine, dopamine, 5-HT, gamma-aminobutyric acid (GABA), glutamate and norepinephrine83. Muscarinic receptors gradually appear in the postnatal cortex and may be more dependent on the presence of nAChRs84. The cholinergic receptors are expressed at high levels in the developing cortex but then decline progressively to a significant extent during adolescence, with the cholinergic innervation of the PFC reaching mature levels18. The timing of cholinergic cortical innervation is of primary importance for the normal development of cognitive functions as nAChRs are involved in attention, learning and memory85.

The mesostriatal and mesocorticolimbic dopamine pathways are involved in processing natural rewards and reward-directed behavior86,87. The mesostriatal-mesocorticolimbic dopamine system includes reciprocal dopamine projections from the ventral tegmental area in the midbrain into the ventral striatum, the limbic structures (amygdala in particular), and the orbitofrontal cortex88. The temporal relationships among exposure to rewards, dopamine neuronal firing activity and extracellular dopamine concentrations suggest that ventral striatal dopamine release is involved in forming associations between salient contextual stimuli and internal rewarding events86,89.

Adolescence is associated with substantial development of the dopaminergic system. There is an increase in dopaminergic input to the PFC indexed by an increase in the density of dopaminergic fibers and transporters18, which is partially offset by developmental decline in dopamine synthesis and turnover after early adolescence80. Dopamine receptors are overproduced in early adolescence, followed by pruning that is more evident in subcortical than prefrontal regions18,90. The net effect of increased dopaminergic projection to the PFC, which shows less pronounced receptor pruning, is a shift in the relative balance between subcortical and cortical dopaminergic systems, with dominance of the mesocortical dopaminergic system. The development of the dopamine system during adolescence is likely to have an influence on the pruning of PFC neurons18. Dopaminergic input to the PFC, then, likely contributes to the coupling of salient cortico-cortical connections, and concomitantly to the pruning of connections that do not have significant salience, thereby influencing the important adolescent neurodevelopmental process of prefrontal pruning and myelination24.

Serotonin (5-HT) is involved in neural plasticity, a process through which modification of the functional properties of neurons and their networks occurs based on experience91. It has been hypothesized that neural plasticity occurs through a reversal of neuronal maturation that reinstates neuronal functions lost during development92. Such a dynamic process may involve a number of mechanisms, including neurite outgrowth, synaptogenesis, neurogenesis and cell survival during brain development and even in adulthood91. There is evidence of significant 5- HT synaptic pruning in the basal forebrain around puberty in rats93, and 5-HT1A receptor binding appears to decrease most dramatically in humans during adolescence94.

Serotonin inhibits and opposes dopamine activity, particularly in relation to dopamine’s role in aggressive and impulsive behaviors9597. By puberty, dopamine input to the PFC is up to 3 times greater than serotonin input98, and PFC concentrations of dopamine precursor are much greater than those of 5-HT precursor in pubertal rhesus monkeys99. The relative imbalance in the dopamine-serotonin activity during adolescence might explain the enhanced sensitivity to appetitive (rewarding) situations, resulting in a higher prevalence of risky behaviors18,100,101.

Maturational changes during adolescence also have been detected in glutaminergic and GABAergic systems. The behavioral effects of agonists for a specific glutamate receptor, the Nmethyl- D-aspartate (NMDA) receptor, appear to peak late in the pre-adolescent period in rats102, and this coincides with greater NMDA agonist sensitivity103. Glutaminergic inputs to the PFC appear to decrease during adolescence18. GABA opposes the modulating excitatory effects of glutamate104. GABA receptors achieve maturity in adolescence105, and GABAergic input to the PFC appears to decrease strongly through adolescence in humans18.

In summary, there are significant developmental changes in the neural circuits involved in emotional and cognitive regulation from childhood through adolescence. The following sections will discuss how these maturational changes might be associated with increased vulnerability to depression during adolescence.

THE DEVELOPMENTAL NEUROBIOLOGY OF DEPRESSIVE DISORDER: THEORETICAL MODELS

Utilizing developmental, integrative and neuroscience frameworks, several theoretical models have been proposed for the increased vulnerability to depression during adolescence. Some of these models are described.

The Social Information Processing Model

Nelson and colleagues106 have proposed that developmental changes in social behavior during adolescence are correlated with the maturation of a brain system referred to as the social information processing network. This network consists of three specific nodes: the detection node, the affective node, and the cognitive-regulatory node. They hypothesized that these nodes develop along different trajectories, such that the development of the affective node, approximately equivalent to the subcortical limbic system, outpaces maturation of the cortically-based cognitive-regulatory node. The mismatch is proposed to create a vulnerability in which strong emotional responses to social stimuli are not tempered by the yet-to-mature regulatory mechanisms.

The Triadic Model

Ernst and Korelitz21 put forth a triadic model comprised of three primary systems, namely the affective system (which includes the amygdala), the reward system (which includes the ventral striatum), and the cognitive/response inhibition system (which includes the PFC). Each system/node has its own developmental trajectory, which creates a state of flux during adolescence. Final behavioral outcomes are likely to depend on the dominant node of a given stage or could result from a weakened node that fails to perform regulatory functions. In its simplified form, the triadic model explains exaggerated reactivity to a number of emotional stimuli7476, changes in reward sensitivity107,108, and the significant lag in cortical control and cognitive development.

The Dysregulated Positive Affect Model

A third model, proposed by Forbes and Dahl109, focuses on the relationship between adolescent depression and the development of the reward system. They conceptualized depression as a reduction in positive affectivity (a factor that indexes active engagement with the environment). The approach system is a motivational system whose function is the pursuit of reward, and it is posited to include brain structures (e.g., the nucleus accumbens) that mediate processing of reward information. From this view, depression is associated with deficits in the approach system. They posited a link between the development of neural systems underlying reward processing, which may become vulnerable to dysregulation as a consequence of remodeling during adolescence, and a predisposition to depression, particularly in vulnerable youth with a temperamental characteristic of low positive affect.

Integration of the Theoretical Models

An integration of the various models suggests that the increased vulnerability to depression and other psychiatric disorders during adolescence may be due to an imbalance between the relative structural and functional maturity of brain systems critical to emotional and incentive-based behavior (subcortical regions such as the amygdala and ventral striatum) compared to brain systems mediating cognitive and impulse control (e.g., PFC), suggesting that the PFC exerts less regulatory control over subcortical regions relative to adults16,20,21,101,106.

This framework provides a heuristic model for explaining the neurodevelopmental basis for the affective and behavioral changes observed in adolescence. By demonstrating how the development of regulatory mechanisms lag behind development of affective brain systems, the model seems particularly appropriate for explaining the increased rates of dysregulated behaviors, especially antisocial behaviors, that emerge during adolescence but decline during adulthood when regulatory brain systems have reached adult levels of maturity110,111. However, it is not able to explain the increased rates of depression that start in adolescence but persist through adulthood9, by which time, presumably, the regulatory mechanisms whose delayed development putatively gave rise to affective dysregulation, have matured26,40. The theory also appears to hold that affective and motivational systems are primarily comprised of subcortical structures, whereas the regulatory systems are cortical. Recently, the concept that affect and affective regulation (or affect and cognition) can even be said to exist as separable processes has been questioned112115.

In contrast to the above-described models that implicated the delayed development of the PFC compared to limbic areas as being responsible for the increased vulnerability to depression in adolescence, Davey and colleagues24 proposed that the maturation of the PFC itself might be responsible for the development and maintenance of depression. According to this model, there is a cost to the PFC’s ability to make decisions in complex social environments that take into account the consequences of decisions into the future, resulting in a heightened vulnerability to depression when anticipated future rewards are not attained.

As described above, there is substantial remodeling and maturation of the dopaminergic reward system and PFC during adolescence, that coincides with the adolescent entering the complex world of adult peer and romantic relationships, where the rewards that can be obtained (such as group affiliation, romantic love, social status) are abstract and temporally distant from the proximal context. Development of the PFC makes it possible to pursue such complex and distal rewards, which are, however, tenuous and more readily frustrated than the more immediate rewards. Davey and Colleagues24 hypothesized that when these distant rewards are unattainable, they suppress the reward system. When such suppression is extensive and occurs for an extended period of time, it manifests as depressive disorder.

The functional significance of the dopaminergic system’s more extensive integration with PFC during adolescence is that the nature of the represented rewards becomes more sophisticated. The net result is the ability of adolescents to be motivated by, and to respond to, rewards that are more distal and complex86,89. Serotonin interacts with the dopaminergic system to further shape reward function116,117, possibly by reducing impulsive over-responding to proximal affective stimuli in favor of maintaining affective engagement with the long-term goals96,118.

Davey and colleagues24 proposed that the initial episodes of clinical depression during adolescence often result from the frustration, or omission, of a highly anticipated social reward(s). Abstract social rewards have a greater salience and are associated with an active state of arousal119,120. When an anticipated reward is omitted, it has the effect of transiently suppressing the neural reward system86. Omission of rewards that are extended in their representation into the more distant future will cause a correspondingly prolonged suppression of the reward system, resulting in depression.

THE DEVELOPMENTAL NEUROBIOLOGY OF DEPRESSIVE DISORDER: EMPIRICAL DATA

Structural Brain Changes

Hippocampus

The hippocampus has been a focal area of research in both animal and human studies because depression is recognized as a stress-sensitive illness and the hippocampus is highly sensitive to stress, particularly during the early developmental period121124. The hippocampus also is involved in mood regulation and cognitive function125. In animal models, extreme or chronic psychosocial stress was associated with dendritic atrophy of hippocampal pyramidal neurons and impaired neurogenesis in the dentate gyrus126128. In a recent investigation, adult female cynomolgus macaque monkeys which exhibited spontaneously occurring depressive behavior manifested reduced volume compared with non-depressed controls specifically in the anterior portion of the hippocampus 129. This finding is notable in that this region of the hippocampus has been implicated in emotional functioning. In a developmental study, subjecting infant monkeys to early-life stress lead to reductions in glucocorticoid and mineralocorticoid receptors in the hippocampus during adolescence (compared to non-stressed controls130).

Human studies, including both pediatric and adult samples, also reported a reduction in hippocampal volume in association with depression121,131. In a recent study, reduced hippocampal volume was observed in healthy adolescents at high familial risk for depression, particularly in those who experienced high levels of adversity in childhood132. Among youth who experienced high levels of adversity, reduced hippocampal volume partly accounted for the increased vulnerability to depression during longitudinal follow-up132. In another investigation, adult patients with depression showed a greater decline in grey matter density of the hippocampus than controls after 3 years, particularly in those who failed to remit from the index depressive episode133. Although morphological changes in the hippocampus have been associated with depression, not all studies replicated these findings. The variability in findings might be attributed to sample size, developmental stage of the sample, number of depressive episodes, duration of illness, family history of depression, history of early-life adversity, comorbid symptoms and methodology of the morphometric analysis131,134.

Amygdala

The amygdala is of importance to depression research due to its posited role in stress responses, as well as emotional and mood processes. In human adult studies, there was significant variability in amygdala changes associated with depression135. The variability across studies was accounted for by medication status such that studies that included only medicated individuals showed increased amygdala volume, whereas studies with only unmedicated persons showed a decrease in amygdala volume135. In a pediatric sample of medication-naïve patients with depression, an increased ratio of the amygdala to hippocampal volume was observed compared to age- and gender-matched controls, but this difference was accounted for by the severity of associated anxiety symptoms136. In a separate investigation, depressed youngsters had significant reductions of left and right amygdala volumes compared with healthy subjects. No significant correlations were found between amygdala volumes and depressive symptom severity, age at onset of illness, or illness duration.

Frontal Lobes

In adult human studies, depressed patients showed large volume reductions in frontal regions, especially in the anterior cingulate and orbitofrontal cortex, and sub-genual region of the PFC137,138. Gender, medication status, stage of illness and family history appear to affect the nature of the findings in a region-specific manner. In a pediatric sample, patients with non-familial depression had significantly increased left PFC volume compared to patients with familial depression as well as with healthy controls139. Left PFC volume correlated with severity of depression in familial but not in non-familial patients with depression139. Taken together, decreased left PFC volume in familial depression in youth and adults might result from degeneration of the left PFC139141, whereas larger left PFC volume in pediatric patients with non-familial depression might be due to developmental alterations in PFC maturation.

Striatum

Several studies in depressed adults reported grey matter deficits in the striatum, especially in the caudate nucleus137,142. Reduced caudate volume also was observed in adolescents with depression (Rao et al., unpublished data).

Summary of Structural Brain Changes

Morphometric changes have been observed in a number of brain regions in association with depressive disorder, particularly those involving corticostriatal and corticolimbic networks, in adults143,144. Limited studies in pediatric samples showed similar patterns145,146. Several DTI studies reported reduced white matter integrity (fractional anisotropy) in adult patients with depression, particularly in the frontal and temporal regions147150. Microstructural white matter abnormalities also were detected during the firstepisode of depression in young adult patients151 as well as in depressed adolescents152. In a pilot study, we reported alterations in white matter tracts in healthy adolescents at high familial risk for depression, suggesting that it might be a vulnerability marker for depression153. Post- mortem studies in animals and adult humans revealed alterations in glial cells in these networks154156. Glial cells not only protect neurons through the production of myelin, but they are also dynamic partners participating in brain metabolism and communication between neurons156.

Functional Brain Changes

Consistent with structural brain changes associated with depressive disorder, functional imaging (fMRI) studies in adults also implicated impaired corticostriatal and corticolimbic circuits51,143,157,158. Most of this research has focused on resting state data159,160, or the processing of negative or positive emotional stimuli160,161. A summary of these data indicate that patients with depression show increased neural activity in response to negative cues and diminished neural activity in response to positive stimuli in emotion-related brain circuits (e.g., amygdala and ventral striatum). Some of these abnormalities in the processing of emotional information persist after symptom remission and they have also been found in healthy individuals who are at heightened risk for the development of mood disorders. Limited data in pediatric populations also reported similar deficits in these neural networks although the direction of change (i.e., increased versus decreased response) has not been consistent across pediatric studies or in comparison with adult data146,162167.

Biochemical studies utilizing magnetic resonance spectroscopy (MRS) reported alterations in N-acetyl aspartate, glutamate/glutamine/GABA, creatinine/phosphocreatinine, choline and myoinositol concentrations in specific regions of the corticostriatal and corticolimbic networks in adult patients with depression160,168170. Modest amount of research in pediatric samples is consistent with the findings in adults, suggesting developmental continuities146,170172.

In summary, data from functional neuroimaging studies in youngsters and adults indicate alterations in corticolimbic and corticostriatal circuits, similar to structural brain changes.

Summary of Neuroimaging Findings in Pediatric Depression

Although a growing body of research in pediatric depression has identified structural and functional brain changes, it has raised more questions than answers. This is, in great part, due to the modest sample sizes with cross-sectional designs. For instance, it is not clear how the maturational changes across child, adolescent and adult development relate to the vulnerability and maintenance of depression. More information is needed on which neural changes are specific to depression and how family history, severity of illness, symptom patterns and comorbid conditions influence the findings131. Also, it is not known whether the neural changes are pre-existing and increase vulnerability to the disorder or if they are a consequence of the illness132,153,165. Further, it is not known with any certainty if the observed brain changes are temporary, state-like conditions which resolve without any sequalae, are temporary but still place an individual on a delayed trajectory towards normal development, or whether any disruption to the normal maturational process during this period permanently and deleteriously affects the neurobiological systems. The last of these possibilities may well be the most likely explanation based on data regarding the increased risk for recurrent depressive episodes173,174. The effect of disease course on the neurobiological substrate also has not been studied133. The utility of these neural markers in the diagnosis, treatment and prognosis of the disorder should be established, as well as neurobiological changes in response to treatment175,176. In the final section of this article, future research directions will be discussed to address these issues.

INDIVIDUAL DIFFERENCES IN DEVELOPMENT AND VULNERABILITY TO DEPRESSION

Although adolescence is associated with dramatic maturational changes in multiple domains and this developmental period is marked by vulnerability to depression, it is important to recognize that the behavioral and neurobiological responses are highly subject to individual differences and only a small subset develops the disorder. Such individual differences may take the form of heritable characteristics such as stable personality traits, differences in neurotransmitter profiles and biologically-governed changes in hormones or other effects of puberty. Environmental factors (e.g., the social context, such as one’s social status among peers) also make a contribution to individual differences. Depression itself has a significant heritable component177, but stress interacts with the genetic diathesis to determine the clinical manifestation of the disorder178180. In the following section, the moderating influence of genetic and environmental factors on the relationship between neurobiological markers and vulnerability to depressive illness will be described.

Gene-Environment Interactions in Determining the Risk for Depression

Research indicates that genetic factors account for anywhere between 24%–58% of the variance in depression177. The relative contribution of genetics to depression vulnerability seems to vary with age. Some studies reported that the genetic influence on depression appears to increase with age, such that depression in adolescents appears to be influenced more by genes than is depression in children181,182. Contrary to this finding, other investigators found that the influence of genes decreases as children grow into adolescence183,184. The genetic risk also may be moderated by gender. Eley and Stevenson185 found that genetic factors made a greater contribution to depression in males but not females. Silberg and colleagues186 reported the opposite finding, with genes playing a greater role in females as they matured into adolescence, but not with males. Clearly, more research is needed before any meaningful conclusions can be made with regard to the relationships among genetic factors, development, gender and depression vulnerability.

A growing body of research has indicated that the genetic vulnerability is modified by adverse environmental conditions, particularly during early development, to increase the risk for depression178,187. For instance, in a longitudinal study of a birth cohort, Caspi and colleagues188 found that individuals with the short allele of the serotonin transporter gene-linked polymorphic region (5-HTTLPR) were at elevated risk for developing a depressive episode in adult life only if they experienced severe maltreatment in childhood. On the other hand, another investigation showed that the presence of positive social support to be protective against depression in children with 5-HTTLPR short allele and a prior maltreatment history189. However, not all studies found a relationship between 5-HTTLPR polymorphism in determining the risk for depression190. In addition to methodological differences across studies, factors such as stress sensitivity might determine the contribution of environmental factors on vulnerability to depression. For instance, Wichers and colleagues180 found that greater stress sensitivity required less environmental influence for the clinical manifestation of depression.

Another candidate gene that has received significant attention in depression research is the corticotropin-releasing hormone type 1 receptor (CRHR1) because it mediates the hormonal responses to stress178,191. In a large community study of adults, a single nucleoside polymorphism (rs110402) of the CRHR1 gene moderated the effects of childhood abuse on depressive symptoms in adult life in males only; females with childhood maltreatment were at elevated risk for depressive symptoms regardless of the genetic alleles191. Additionally, the presence of the rare allele (the A allele in the case of rs110402) was protective against depression in males exposed to childhood abuse.

Genetic variants of the Val66Met polymorphism of brain-derived neurotrophic factor (BDNF) gene also have been studies in relation to gene-environment interactions on depression risk192. In such studies, childhood adversity seems to have a greater impact on depressive symptoms in Met allele carriers of the BDNF gene than in the Val/Val group. BDNF is important for neural plasticity193. In animal models, depressive states were associated with reduced BDNF levels in the brain, and central administration of BDNF has been shown to reverse such depressive states. In an investigation of children with and without maltreatment, a three-way interaction was observed among BDNF genotype, 5-HTTLPR and maltreatment history in predicting depression194. Children with the Met allele of the BDNF gene and two short alleles of 5-HTTLPR had the highest depression scores, but the vulnerability associated with these two genotypes was only evident in maltreated children. However, social support was protective against depression in the vulnerable group.

Taken together, these data support gene-environment interactions in the etiology of depression across the lifespan.

Genetic and Environmental Influences on Neurobiological Responses and Vulnerability to Depression

There is a growing interest in understanding genetic influences on the variability in brain structure and function195197. For example, genetic variants of the 5-HTTLPR and BDNF polymorphisms were associated with amygdala, hippocampal and prefrontal volumes195,197,198. Moreover, individuals carrying the 5HTTLPR short (risk) allele or BDNF Met allele had smaller hippocampal and/or amygdala volumes when they had a history of childhood maltreatment compared with those who only had one risk factor (environmental or genetic). Independent of genetic risk, childhood stress predicted additional hippocampal white matter alterations195,198. Although depressed patients had reduced PFC volume, individuals carrying the non-risk (long) allele and who also experienced high levels of childhood stress had larger PFC volumes, suggesting a protective effect198. Structural brain changes due to stress represent part of the mechanism by which the illness risk and outcome might be genetically-mediated.

Genetic factors also influence neurobiological responses. In a meta-analysis, there was an association between the magnitude of amygdala activation in response to emotional cues and 5-HTTLPR196. To our knowledge, there are no data on the gene-environment interactions in brain responses in relation to depressive disorder. In the study described above, the A allele of CRHR1 gene was associated with a less robust hormonal response in men, possibly serving as a potential mediator of protection against depression191.

Summary of Findings on the Associations among Genes, Neurobiology, Environment and Depression

Genetic polymorphisms modulate brain structure and function. The structural and functional brain changes might serve as intermediate phenotypes in determining the risk for depressive illness. Environmental insults can further exacerbate the neurobiological alterations in at-risk individuals and magnify the risk for the disorder. In a similar vein, enriched/supportive environment can ameliorate the risk in genetically-vulnerable persons. A better understanding of the gene-environment interactions, and the mechanisms (e.g., neurobiological changes) through which risk and resilience for the disorder occur, will be helpful in developing more specific and effective preventive and treatment interventions.

Treatment Implications for Depression

While there is still much to learn in terms of understanding the neurobiology of depressive illness across the lifespan in general and pediatric depression in particular, the available data offer potential options for intervention. Given that depression is associated with reduced hippocampal volume and possibly other structural brain changes, interventions that can potentially reverse these changes could be helpful. Potential therapies such as antidepressant agents, electroconvulsive therapy and exercise that have been shown to promote neurogenesis ought to be considered199201. Lithium has been found to reverse the glial cell changes202. Anti-glucocorticoid agents and CRH antagonists can be helpful in reducing the toxic effects of hypercortisolemia203. Psychotherapeutic interventions also can induce brain changes204.

Future Directions for Research

Perhaps the most obvious implication from the current chapter is the need for more prospective longitudinal research in youngsters. Investigators should identify clear structure/function hypotheses that utilize, but go beyond, the current diagnostic system in order to translate findings from clinical neuroscience research into a new classification system based on pathophysiology and etiological processes205. Such models also may be able to explain the high rates of comorbidity between depression and other psychiatric conditions. Studies of depressed children and adolescents have a number of advantages compared with studies of adults; confounding factors such as repeated episodes, long duration of illness, multiple medications and co-occurring medical problems are minimized. It is also important to include high-risk youth who have not yet developed a depressive episode in order to identify pre-morbid factors. Another focus of future research should center on evaluating the stability of biological abnormalities. Given that adolescence is a high risk period for the onset of depression, in a relatively short follow-up period, it is possible to identify both pre-existing and “scar” markers of the illness.

It is important to obtain functional neuroimaging measures before and after pharmacological or behavioral interventions. Results from such studies will shed light on the mechanisms through which treatments work and may provide new intervention targets for drug development206,207. In addition to identifying potential mediators of different treatments, it is important for neuroimaging studies to identify baseline predictors of treatment outcome (i.e., the moderators). The identification of mediators and moderators of treatment would help move the field towards more “personalized” care208. Exciting times lie ahead in our efforts to tackle the mechanisms involved in pediatric depression and its treatment response.

Acknowledgments

This work was supported, in part, by the following grants from the National Institutes of Health (DA17805, MH68391, RR003032 and RR026140) and from the Endowed Chair in Brain and Behavior Research at Meharry Medical College.

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.

The authors have no financial conflicts to disclose.

REFERENCES

  • 1.Kessler RC, Avenevoli S, Ries Merikangas K. Mood disorders in children and adolescents: an epidemiologic perspective. Biological psychiatry. 2001 Jun 15;49(12):1002–1014. doi: 10.1016/s0006-3223(01)01129-5. [DOI] [PubMed] [Google Scholar]
  • 2.Kovacs M, Gatsonis C. Secular trends in age at onset of major depressive disorder in a clinical sample of children. Journal of psychiatric research. 1994 May-Jun;28(3):319–329. doi: 10.1016/0022-3956(94)90014-0. [DOI] [PubMed] [Google Scholar]
  • 3.Lewinsohn PM, Hops H, Roberts RE, Seeley JR, Andrews JA. Adolescent psychopathology: I. Prevalence and incidence of depression and other DSM-III-R disorders in high school students. Journal of abnormal psychology. 1993 Feb;102(1):133–144. doi: 10.1037//0021-843x.102.1.133. [DOI] [PubMed] [Google Scholar]
  • 4.Ryan ND, Williamson DE, Iyengar S, et al. A secular increase in child and adolescent onset affective disorder. Journal of the American Academy of Child and Adolescent Psychiatry. 1992 Jul;31(4):600–605. doi: 10.1097/00004583-199207000-00004. [DOI] [PubMed] [Google Scholar]
  • 5.Puig-Antich J, Lukens E, Davies M, Goetz D, Brennan-Quattrock J, Todak G. Psychosocial functioning in prepubertal major depressive disorders. I Interpersonal relationships during the depressive episode. Archives of general psychiatry. 1985 May;42(5):500–507. doi: 10.1001/archpsyc.1985.01790280082008. [DOI] [PubMed] [Google Scholar]
  • 6.Puig-Antich J, Lukens E, Davies M, Goetz D, Brennan-Quattrock J, Todak G. Psychosocial functioning in prepubertal major depressive disorders. II Interpersonal relationships after sustained recovery from affective episode. Archives of general psychiatry. 1985 May;42(5):511–517. doi: 10.1001/archpsyc.1985.01790280093010. [DOI] [PubMed] [Google Scholar]
  • 7.Puig-Antich J, Kaufman J, Ryan ND, et al. The psychosocial functioning and family environment of depressed adolescents. Journal of the American Academy of Child and Adolescent Psychiatry. 1993 Mar;32(2):244–253. doi: 10.1097/00004583-199303000-00003. [DOI] [PubMed] [Google Scholar]
  • 8.Rao U, Chen LA. Characteristics, correlates, and outcomes of childhood and adolescent depressive disorders. Dialogues in clinical neuroscience. 2009;11(1):45–62. doi: 10.31887/DCNS.2009.11.1/urao. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Rao U. Links between depression and substance abuse in adolescents: neurobiological mechanisms. American journal of preventive medicine. 2006 Dec;31(6 Suppl 1):S161–S174. doi: 10.1016/j.amepre.2006.07.002. [DOI] [PubMed] [Google Scholar]
  • 10.Hankin BL, Oppenheimer C, Jenness J, Barrocas A, Shapero BG, Goldband J. Developmental origins of cognitive vulnerabilities to depression: review of processes contributing to stability and change across time. Journal of clinical psychology. 2009 Dec;65(12):1327–1338. doi: 10.1002/jclp.20625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Zalsman G, Oquendo MA, Greenhill L, et al. Neurobiology of depression in children and adolescents. Child and adolescent psychiatric clinics of North America. 2006 Oct;15(4):843–868. vii–viii. doi: 10.1016/j.chc.2006.05.010. [DOI] [PubMed] [Google Scholar]
  • 12.Franic S, Middeldorp CM, Dolan CV, Ligthart L, Boomsma DI. Childhood and adolescent anxiety and depression: beyond heritability. Journal of the American Academy of Child and Adolescent Psychiatry. 2010 Aug;49(8):820–829. doi: 10.1016/j.jaac.2010.05.013. [DOI] [PubMed] [Google Scholar]
  • 13.Rice F. The genetics of depression in childhood and adolescence. Current psychiatry reports. 2009 Apr;11(2):167–173. doi: 10.1007/s11920-009-0026-9. [DOI] [PubMed] [Google Scholar]
  • 14.Rutter M. Gene-environment interplay. Depression and anxiety. 2010;27(1):1–4. doi: 10.1002/da.20641. [DOI] [PubMed] [Google Scholar]
  • 15.Schlossberg K, Massler A, Zalsman G. Environmental risk factors for psychopathology. The Israel journal of psychiatry and related sciences. 2010;47(2):139–143. [PubMed] [Google Scholar]
  • 16.Somerville LH, Jones RM, Casey BJ. A time of change: behavioral and neural correlates of adolescent sensitivity to appetitive and aversive environmental cues. Brain and cognition. 2010 Feb;72(1):124–133. doi: 10.1016/j.bandc.2009.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sisk CL, Foster DL. The neural basis of puberty and adolescence. Nature neuroscience. 2004 Oct;7(10):1040–1047. doi: 10.1038/nn1326. [DOI] [PubMed] [Google Scholar]
  • 18.Spear L. Neurobehavioral changes in adolescence. Current Directions in Psychological Science. 2000;9(4):111–114. [Google Scholar]
  • 19.Blakemore SJ, Burnett S, Dahl RE. The role of puberty in the developing adolescent brain. Human brain mapping. 2010 Jun;31(6):926–933. doi: 10.1002/hbm.21052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Brenhouse HC, Andersen SL. Developmental trajectories during adolescence in males and females: A cross-species understanding of underlying brain changes. Neuroscience and biobehavioral reviews. 2011 May 12; doi: 10.1016/j.neubiorev.2011.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ernst M, Korelitz KE. Cerebral maturation in adolescence: behavioral vulnerability. L'Encephale. 2009 Dec;35(Suppl 6):S182–S189. doi: 10.1016/S0013-7006(09)73469-4. [DOI] [PubMed] [Google Scholar]
  • 22.Paus T, Keshavan M, Giedd JN. Why do many psychiatric disorders emerge during adolescence? Nature reviews. Neuroscience. 2008 Dec;9(12):947–957. doi: 10.1038/nrn2513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Andersen SL, Teicher MH. Stress, sensitive periods and maturational events in adolescent depression. Trends in neurosciences. 2008 Apr;31(4):183–191. doi: 10.1016/j.tins.2008.01.004. [DOI] [PubMed] [Google Scholar]
  • 24.Davey CG, Yucel M, Allen NB. The emergence of depression in adolescence: development of the prefrontal cortex and the representation of reward. Neuroscience and biobehavioral reviews. 2008;32(1):1–19. doi: 10.1016/j.neubiorev.2007.04.016. [DOI] [PubMed] [Google Scholar]
  • 25.Rudolph KD, Hammen C, Daley SE. Mood disorders. In: Wolfe DA, Mash EJ, editors. Behavioral and emotional disorders in adolescents : nature, assessment, and treatment. New York: Guilford Press; 2006. pp. 300–342. [Google Scholar]
  • 26.Gogtay N, Giedd JN, Lusk L, et al. Dynamic mapping of human cortical development during childhood through early adulthood. Proceedings of the National Academy of Sciences of the United States of America. 2004 May 25;101(21):8174–8179. doi: 10.1073/pnas.0402680101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Paus T. Growth of white matter in the adolescent brain: myelin or axon? Brain and cognition. 2010 Feb;72(1):26–35. doi: 10.1016/j.bandc.2009.06.002. [DOI] [PubMed] [Google Scholar]
  • 28.Herba C, Phillips M. Annotation: Development of facial expression recognition from childhood to adolescence: behavioural and neurological perspectives. Journal of child psychology and psychiatry, and allied disciplines. 2004 Oct;45(7):1185–1198. doi: 10.1111/j.1469-7610.2004.00316.x. [DOI] [PubMed] [Google Scholar]
  • 29.Rosso IM, Young AD, Femia LA, Yurgelun-Todd DA. Cognitive and emotional components of frontal lobe functioning in childhood and adolescence. Annals of the New York Academy of Sciences. 2004 Jun;1021:355–362. doi: 10.1196/annals.1308.045. [DOI] [PubMed] [Google Scholar]
  • 30.Yurgelun-Todd D. Emotional and cognitive changes during adolescence. Current opinion in neurobiology. 2007 Apr;17(2):251–257. doi: 10.1016/j.conb.2007.03.009. [DOI] [PubMed] [Google Scholar]
  • 31.Burnett S, Sebastian C, Cohen Kadosh K, Blakemore SJ. The social brain in adolescence: Evidence from functional magnetic resonance imaging and behavioural studies. Neuroscience and biobehavioral reviews. 2011 Aug;35(8):1654–1664. doi: 10.1016/j.neubiorev.2010.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Luna B, Padmanabhan A, O'Hearn K. What has fMRI told us about the development of cognitive control through adolescence? Brain and cognition. 2010 Feb;72(1):101–113. doi: 10.1016/j.bandc.2009.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Paus T. Mapping brain maturation and cognitive development during adolescence. Trends in cognitive sciences. 2005 Feb;9(2):60–68. doi: 10.1016/j.tics.2004.12.008. [DOI] [PubMed] [Google Scholar]
  • 34.Durston S, Hulshoff Pol HE, Casey BJ, Giedd JN, Buitelaar JK, van Engeland H. Anatomical MRI of the developing human brain: what have we learned? Journal of the American Academy of Child and Adolescent Psychiatry. 2001 Sep;40(9):1012–1020. doi: 10.1097/00004583-200109000-00009. [DOI] [PubMed] [Google Scholar]
  • 35.Chugani HT. Biological basis of emotions: brain systems and brain development. Pediatrics. 1998 Nov;102(5 Suppl E):1225–1229. [PubMed] [Google Scholar]
  • 36.Luna B, Sweeney JA. The emergence of collaborative brain function: FMRI studies of the development of response inhibition. Annals of the New York Academy of Sciences. 2004 Jun;1021:296–309. doi: 10.1196/annals.1308.035. [DOI] [PubMed] [Google Scholar]
  • 37.Wilke M, Krageloh-Mann I, Holland SK. Global and local development of gray and white matter volume in normal children and adolescents. Experimental brain research. Experimentelle Hirnforschung. Experimentation cerebrale. 2007 Apr;178(3):296–307. doi: 10.1007/s00221-006-0732-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Sowell ER, Thompson PM, Holmes CJ, Jernigan TL, Toga AW. In vivo evidence for post-adolescent brain maturation in frontal and striatal regions. Nature neuroscience. 1999 Oct;2(10):859–861. doi: 10.1038/13154. [DOI] [PubMed] [Google Scholar]
  • 39.Giorgio A, Watkins KE, Chadwick M, et al. Longitudinal changes in grey and white matter during adolescence. NeuroImage. 2010 Jan 1;49(1):94–103. doi: 10.1016/j.neuroimage.2009.08.003. [DOI] [PubMed] [Google Scholar]
  • 40.Giedd JN, Blumenthal J, Jeffries NO, et al. Brain development during childhood and adolescence: a longitudinal MRI study. Nature neuroscience. 1999 Oct;2(10):861–863. doi: 10.1038/13158. [DOI] [PubMed] [Google Scholar]
  • 41.Schmithorst VJ, Yuan W. White matter development during adolescence as shown by diffusion MRI. Brain and cognition. 2010 Feb;72(1):16–25. doi: 10.1016/j.bandc.2009.06.005. [DOI] [PubMed] [Google Scholar]
  • 42.Asato MR, Terwilliger R, Woo J, Luna B. White matter development in adolescence: a DTI study. Cereb Cortex. 2010 Sep;20(9):2122–2131. doi: 10.1093/cercor/bhp282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yurgelun-Todd DA, Killgore WD, Young AD. Sex differences in cerebral tissue volume and cognitive performance during adolescence. Psychological reports. 2002 Dec;91(3 Pt 1):743–757. doi: 10.2466/pr0.2002.91.3.743. [DOI] [PubMed] [Google Scholar]
  • 44.Durston S, Casey BJ. What have we learned about cognitive development from neuroimaging? Neuropsychologia. 2006;44(11):2149–2157. doi: 10.1016/j.neuropsychologia.2005.10.010. [DOI] [PubMed] [Google Scholar]
  • 45.Chechik G, Meilijson I, Ruppin E. Synaptic pruning in development: a computational account. Neural computation. 1998 Oct 1;10(7):1759–1777. doi: 10.1162/089976698300017124. [DOI] [PubMed] [Google Scholar]
  • 46.Mimura K, Kimoto T, Okada M. Synapse efficiency diverges due to synaptic pruning following overgrowth. Physical review. E, Statistical, nonlinear, and soft matter physics. 2003 Sep;68(3 Pt 1) doi: 10.1103/PhysRevE.68.031910. 031910. [DOI] [PubMed] [Google Scholar]
  • 47.Disner SG, Beevers CG, Haigh EA, Beck AT. Neural mechanisms of the cognitive model of depression. Nature reviews. Neuroscience. 2011 Jul 6; doi: 10.1038/nrn3027. [DOI] [PubMed] [Google Scholar]
  • 48.Heatherton TF, Wagner DD. Cognitive neuroscience of self-regulation failure. Trends in cognitive sciences. 2011 Mar;15(3):132–139. doi: 10.1016/j.tics.2010.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Price JL, Drevets WC. Neurocircuitry of mood disorders. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 2010 Jan;35(1):192–216. doi: 10.1038/npp.2009.104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Liston C, Watts R, Tottenham N, et al. Frontostriatal microstructure modulates efficient recruitment of cognitive control. Cereb Cortex. 2006 Apr;16(4):553–560. doi: 10.1093/cercor/bhj003. [DOI] [PubMed] [Google Scholar]
  • 51.Mayberg HS. Modulating dysfunctional limbic-cortical circuits in depression: towards development of brain-based algorithms for diagnosis and optimised treatment. British medical bulletin. 2003;65:193–207. doi: 10.1093/bmb/65.1.193. [DOI] [PubMed] [Google Scholar]
  • 52.Bellani M, Baiano M, Brambilla P. Brain anatomy of major depression II. Focus on amygdala. Epidemiology and psychiatric science. 2011 Mar;20(1):33–36. doi: 10.1017/s2045796011000096. [DOI] [PubMed] [Google Scholar]
  • 53.Hamani C, Mayberg H, Stone S, Laxton A, Haber S, Lozano AM. The subcallosal cingulate gyrus in the context of major depression. Biological psychiatry. 2011 Feb 15;69(4):301–308. doi: 10.1016/j.biopsych.2010.09.034. [DOI] [PubMed] [Google Scholar]
  • 54.Hariri AR, Bookheimer SY, Mazziotta JC. Modulating emotional responses: effects of a neocortical network on the limbic system. Neuroreport. 2000 Jan 17;11(1):43–48. doi: 10.1097/00001756-200001170-00009. [DOI] [PubMed] [Google Scholar]
  • 55.Rogers MA, Bradshaw JL, Pantelis C, Phillips JG. Frontostriatal deficits in unipolar major depression. Brain research bulletin. 1998 Nov 1;47(4):297–310. doi: 10.1016/s0361-9230(98)00126-9. [DOI] [PubMed] [Google Scholar]
  • 56.Heyder K, Suchan B, Daum I. Cortico-subcortical contributions to executive control. Acta psychologica. 2004 Feb-Mar;115(2–3):271–289. doi: 10.1016/j.actpsy.2003.12.010. [DOI] [PubMed] [Google Scholar]
  • 57.Baxter MG. Introduction to the special section on "translational models of prefrontal cortical function". Behavioral neuroscience. 2011 Jun;125(3):279–281. doi: 10.1037/a0023841. [DOI] [PubMed] [Google Scholar]
  • 58.Fuster JM. Frontal lobe and cognitive development. Journal of neurocytology. 2002 Mar-Jun;31(3–5):373–385. doi: 10.1023/a:1024190429920. [DOI] [PubMed] [Google Scholar]
  • 59.Sowell ER, Thompson PM, Leonard CM, Welcome SE, Kan E, Toga AW. Longitudinal mapping of cortical thickness and brain growth in normal children. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2004 Sep 22;24(38):8223–8231. doi: 10.1523/JNEUROSCI.1798-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Casey BJ, Tottenham N, Liston C, Durston S. Imaging the developing brain: what have we learned about cognitive development? Trends in cognitive sciences. 2005 Mar;9(3):104–110. doi: 10.1016/j.tics.2005.01.011. [DOI] [PubMed] [Google Scholar]
  • 61.Sowell ER, Thompson PM, Rex D, et al. Mapping sulcal pattern asymmetry and local cortical surface gray matter distribution in vivo: maturation in perisylvian cortices. Cereb Cortex. 2002 Jan;12(1):17–26. doi: 10.1093/cercor/12.1.17. [DOI] [PubMed] [Google Scholar]
  • 62.Casey BJ, Castellanos FX, Giedd JN, et al. Implication of right frontostriatal circuitry in response inhibition and attention-deficit/hyperactivity disorder. Journal of the American Academy of Child and Adolescent Psychiatry. 1997 Mar;36(3):374–383. doi: 10.1097/00004583-199703000-00016. [DOI] [PubMed] [Google Scholar]
  • 63.Lebel C, Walker L, Leemans A, Phillips L, Beaulieu C. Microstructural maturation of the human brain from childhood to adulthood. NeuroImage. 2008 Apr 15;40(3):1044–1055. doi: 10.1016/j.neuroimage.2007.12.053. [DOI] [PubMed] [Google Scholar]
  • 64.Klingberg T, Vaidya CJ, Gabrieli JD, Moseley ME, Hedehus M. Myelination and organization of the frontal white matter in children: a diffusion tensor MRI study. Neuroreport. 1999 Sep 9;10(13):2817–2821. doi: 10.1097/00001756-199909090-00022. [DOI] [PubMed] [Google Scholar]
  • 65.Casey BJ, Cohen JD, Jezzard P, et al. Activation of prefrontal cortex in children during a nonspatial working memory task with functional MRI. NeuroImage. 1995 Sep;2(3):221–229. doi: 10.1006/nimg.1995.1029. [DOI] [PubMed] [Google Scholar]
  • 66.Rubia K, Overmeyer S, Taylor E, et al. Functional frontalisation with age: mapping neurodevelopmental trajectories with fMRI. Neuroscience and biobehavioral reviews. 2000 Jan;24(1):13–19. doi: 10.1016/s0149-7634(99)00055-x. [DOI] [PubMed] [Google Scholar]
  • 67.Schlaggar BL, Brown TT, Lugar HM, Visscher KM, Miezin FM, Petersen SE. Functional neuroanatomical differences between adults and school-age children in the processing of single words. Science. 2002 May 24;296(5572):1476–1479. doi: 10.1126/science.1069464. [DOI] [PubMed] [Google Scholar]
  • 68.Tamm L, Menon V, Reiss AL. Maturation of brain function associated with response inhibition. Journal of the American Academy of Child and Adolescent Psychiatry. 2002 Oct;41(10):1231–1238. doi: 10.1097/00004583-200210000-00013. [DOI] [PubMed] [Google Scholar]
  • 69.Gaillard WD, Hertz-Pannier L, Mott SH, Barnett AS, LeBihan D, Theodore WH. Functional anatomy of cognitive development: fMRI of verbal fluency in children and adults. Neurology. 2000 Jan 11;54(1):180–185. doi: 10.1212/wnl.54.1.180. [DOI] [PubMed] [Google Scholar]
  • 70.Durston S, Davidson MC, Tottenham N, et al. A shift from diffuse to focal cortical activity with development. Developmental science. 2006 Jan;9(1):1–8. doi: 10.1111/j.1467-7687.2005.00454.x. [DOI] [PubMed] [Google Scholar]
  • 71.Somerville LH, Fani N, McClure-Tone EB. Behavioral and neural representation of emotional facial expressions across the lifespan. Developmental neuropsychology. 2011 May;36(4):408–428. [Google Scholar]
  • 72.Guyer AE, Monk CS, McClure-Tone EB, et al. A developmental examination of amygdala response to facial expressions. Journal of cognitive neuroscience. 2008 Sep;20(9):1565–1582. doi: 10.1162/jocn.2008.20114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Hare TA, Tottenham N, Galvan A, Voss HU, Glover GH, Casey BJ. Biological substrates of emotional reactivity and regulation in adolescence during an emotional go-nogo task. Biological psychiatry. 2008 May 15;63(10):927–934. doi: 10.1016/j.biopsych.2008.03.015015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Arnett JJ. Adolescent storm and stress, reconsidered. The American psychologist. 1999 May;54(5):317–326. doi: 10.1037//0003-066x.54.5.317. [DOI] [PubMed] [Google Scholar]
  • 75.Buchanan CM, Eccles JS, Becker JB. Are adolescents the victims of raging hormones: evidence for activational effects of hormones on moods and behavior at adolescence. Psychological bulletin. 1992 Jan;111(1):62–107. doi: 10.1037/0033-2909.111.1.62. [DOI] [PubMed] [Google Scholar]
  • 76.Larson RW, Moneta G, Richards MH, Wilson S. Continuity, stability, and change in daily emotional experience across adolescence. Child development. 2002 Jul-Aug;73(4):1151–1165. doi: 10.1111/1467-8624.00464. [DOI] [PubMed] [Google Scholar]
  • 77.Monk CS, McClure EB, Nelson EE, et al. Adolescent immaturity in attention-related brain engagement to emotional facial expressions. NeuroImage. 2003 Sep;20(1):420–428. doi: 10.1016/s1053-8119(03)00355-0. [DOI] [PubMed] [Google Scholar]
  • 78.Baird AA, Gruber SA, Fein DA, et al. Functional magnetic resonance imaging of facial affect recognition in children and adolescents. Journal of the American Academy of Child and Adolescent Psychiatry. 1999 Feb;38(2):195–199. doi: 10.1097/00004583-199902000-00019. [DOI] [PubMed] [Google Scholar]
  • 79.Luna B, Garver KE, Urban TA, Lazar NA, Sweeney JA. Maturation of cognitive processes from late childhood to adulthood. Child development. 2004 Sep-Oct;75(5):1357–1372. doi: 10.1111/j.1467-8624.2004.00745.x. [DOI] [PubMed] [Google Scholar]
  • 80.Andersen SL. Trajectories of brain development: point of vulnerability or window of opportunity? Neuroscience and biobehavioral reviews. 2003 Jan-Mar;27(1–2):3–18. doi: 10.1016/s0149-7634(03)00005-8. [DOI] [PubMed] [Google Scholar]
  • 81.Daws LC, Gould GG. Ontogeny and regulation of the serotonin transporter: providing insights into human disorders. Pharmacology & therapeutics. 2011 Jul;131(1):61–79. doi: 10.1016/j.pharmthera.2011.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Bruel-Jungerman E, Lucassen PJ, Francis F. Cholinergic influences on cortical development and adult neurogenesis. Behavioural brain research. 2011 Aug 10;221(2):379–388. doi: 10.1016/j.bbr.2011.01.021. [DOI] [PubMed] [Google Scholar]
  • 83.Dani JA. Overview of nicotinic receptors and their roles in the central nervous system. Biological psychiatry. 2001 Feb 1;49(3):166–174. doi: 10.1016/s0006-3223(00)01011-8. [DOI] [PubMed] [Google Scholar]
  • 84.Aubert I, Cecyre D, Gauthier S, Quirion R. Comparative ontogenic profile of cholinergic markers, including nicotinic and muscarinic receptors, in the rat brain. The Journal of comparative neurology. 1996 May 20;369(1):31–55. doi: 10.1002/(SICI)1096-9861(19960520)369:1<31::AID-CNE3>3.0.CO;2-L. [DOI] [PubMed] [Google Scholar]
  • 85.Berger-Sweeney J. The cholinergic basal forebrain system during development and its influence on cognitive processes: important questions and potential answers. Neuroscience and biobehavioral reviews. 2003 Sep;27(4):401–411. doi: 10.1016/s0149-7634(03)00070-8. [DOI] [PubMed] [Google Scholar]
  • 86.Schultz W. Dopamine signals for reward value and risk: basic and recent data. Behavioral and brain functions : BBF. 2010;6:24. doi: 10.1186/1744-9081-6-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Berridge KC, Robinson TE. Parsing reward. Trends in neurosciences. 2003 Sep;26(9):507–513. doi: 10.1016/S0166-2236(03)00233-9. [DOI] [PubMed] [Google Scholar]
  • 88.Ongur D, Price JL. The organization of networks within the orbital and medial prefrontal cortex of rats, monkeys and humans. Cereb Cortex. 2000 Mar;10(3):206–219. doi: 10.1093/cercor/10.3.206. [DOI] [PubMed] [Google Scholar]
  • 89.Spanagel R, Weiss F. The dopamine hypothesis of reward: past and current status. Trends in neurosciences. 1999 Nov;22(11):521–527. doi: 10.1016/s0166-2236(99)01447-2. [DOI] [PubMed] [Google Scholar]
  • 90.Andersen SL, Thompson AT, Rutstein M, Hostetter JC, Teicher MH. Dopamine receptor pruning in prefrontal cortex during the periadolescent period in rats. Synapse. 2000 Aug;37(2):167–169. doi: 10.1002/1098-2396(200008)37:2<167::AID-SYN11>3.0.CO;2-B. [DOI] [PubMed] [Google Scholar]
  • 91.Mattson MP, Maudsley S, Martin B. BDNF and 5-HT: a dynamic duo in age-related neuronal plasticity and neurodegenerative disorders. Trends in neurosciences. 2004 Oct;27(10):589–594. doi: 10.1016/j.tins.2004.08.001. [DOI] [PubMed] [Google Scholar]
  • 92.Kobayashi K, Ikeda Y, Sakai A, et al. Reversal of hippocampal neuronal maturation by serotonergic antidepressants. Proceedings of the National Academy of Sciences of the United States of America. 2010 May 4;107(18):8434–8439. doi: 10.1073/pnas.0912690107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Dinopoulos A, Dori I, Parnavelas JG. The serotonin innervation of the basal forebrain shows a transient phase during development. Brain research. Developmental brain research. 1997 Mar 17;99(1):38–52. doi: 10.1016/s0165-3806(96)00198-8. [DOI] [PubMed] [Google Scholar]
  • 94.Dillon KA, Gross-Isseroff R, Israeli M, Biegon A. Autoradiographic analysis of serotonin 5-HT1A receptor binding in the human brain postmortem: effects of age and alcohol. Brain research. 1991 Jul 19;554(1–2):56–64. doi: 10.1016/0006-8993(91)90171-q. [DOI] [PubMed] [Google Scholar]
  • 95.Goveas JS, Csernansky JG, Coccaro EF. Platelet serotonin content correlates inversely with life history of aggression in personality-disordered subjects. Psychiatry research. 2004 Apr 15;126(1):23–32. doi: 10.1016/j.psychres.2004.01.006. [DOI] [PubMed] [Google Scholar]
  • 96.Katz LD. Dopamine and serotonin: integrating current affective engagement with longer-term goals. Behavioral and Brain Sciences. 1999;22(3):527. [Google Scholar]
  • 97.van der Vegt BJ, Lieuwes N, Cremers TI, de Boer SF, Koolhaas JM. Cerebrospinal fluid monoamine and metabolite concentrations and aggression in rats. Hormones and behavior. 2003 Sep;44(3):199–208. doi: 10.1016/s0018-506x(03)00132-6. [DOI] [PubMed] [Google Scholar]
  • 98.Lambe EK, Krimer LS, Goldman-Rakic PS. Differential postnatal development of catecholamine and serotonin inputs to identified neurons in prefrontal cortex of rhesus monkey. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2000 Dec 1;20(23):8780–8787. doi: 10.1523/JNEUROSCI.20-23-08780.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Goldman-Rakic PS, Brown RM. Postnatal development of monoamine content and synthesis in the cerebral cortex of rhesus monkeys. Brain research. 1982 Jul;256(3):339–349. doi: 10.1016/0165-3806(82)90146-8. [DOI] [PubMed] [Google Scholar]
  • 100.Zeeb FD, Robbins TW, Winstanley CA. Serotonergic and dopaminergic modulation of gambling behavior as assessed using a novel rat gambling task. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 2009 Sep;34(10):2329–2343. doi: 10.1038/npp.2009.62. [DOI] [PubMed] [Google Scholar]
  • 101.Steinberg L, Albert D, Cauffman E, Banich M, Graham S, Woolard J. Age differences in sensation seeking and impulsivity as indexed by behavior and self-report: evidence for a dual systems model. Developmental psychology. 2008 Nov;44(6):1764–1778. doi: 10.1037/a0012955. [DOI] [PubMed] [Google Scholar]
  • 102.Frantz KJ, Van Hartesveldt C. The locomotor effects of quinpirole in rats depend on age and gender. Pharmacology, biochemistry, and behavior. 1999 Dec;64(4):821–826. doi: 10.1016/s0091-3057(99)00162-8. [DOI] [PubMed] [Google Scholar]
  • 103.Subramaniam S, McGonigle P. Regional profile of developmental changes in the sensitivity of the N-methyl-D-aspartate receptor to polyamines. Journal of neurochemistry. 1994 Apr;62(4):1408–1415. doi: 10.1046/j.1471-4159.1994.62041408.x. [DOI] [PubMed] [Google Scholar]
  • 104.Johnson BA. Recent advances in the development of treatments for alcohol and cocaine dependence: focus on topiramate and other modulators of GABA or glutamate function. CNS drugs. 2005;19(10):873–896. doi: 10.2165/00023210-200519100-00005. [DOI] [PubMed] [Google Scholar]
  • 105.Nurse S, Lacaille JC. Late maturation of GABA(B) synaptic transmission in area CA1 of the rat hippocampus. Neuropharmacology. 1999 Nov;38(11):1733–1742. doi: 10.1016/s0028-3908(99)00122-7. [DOI] [PubMed] [Google Scholar]
  • 106.Nelson EE, Leibenluft E, McClure EB, Pine DS. The social re-orientation of adolescence: a neuroscience perspective on the process and its relation to psychopathology. Psychological medicine. 2005 Feb;35(2):163–174. doi: 10.1017/s0033291704003915. [DOI] [PubMed] [Google Scholar]
  • 107.Ernst M, Paulus MP. Neurobiology of decision making: a selective review from a neurocognitive and clinical perspective. Biological psychiatry. 2005 Oct 15;58(8):597–604. doi: 10.1016/j.biopsych.2005.06.004. [DOI] [PubMed] [Google Scholar]
  • 108.Galvan A, Hare TA, Parra CE, et al. Earlier development of the accumbens relative to orbitofrontal cortex might underlie risk-taking behavior in adolescents. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2006 Jun 21;26(25):6885–6892. doi: 10.1523/JNEUROSCI.1062-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Forbes EE, Dahl RE. Neural systems of positive affect: relevance to understanding child and adolescent depression? Dev Psychopathol. 2005 Summer;17(3):827–850. doi: 10.1017/S095457940505039X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Moffitt TE. Adolescence-limited and life-course-persistent antisocial behavior: a developmental taxonomy. Psychological review. 1993 Oct;100(4):674–701. [PubMed] [Google Scholar]
  • 111.Rutter M, Kim-Cohen J, Maughan B. Continuities and discontinuities in psychopathology between childhood and adult life. Journal of child psychology and psychiatry, and allied disciplines. 2006 Mar-Apr;47(3–4):276–295. doi: 10.1111/j.1469-7610.2006.01614.x. [DOI] [PubMed] [Google Scholar]
  • 112.Campos JJ, Frankel CB, Camras L. On the nature of emotion regulation. Child development. 2004 Mar-Apr;75(2):377–394. doi: 10.1111/j.1467-8624.2004.00681.x. [DOI] [PubMed] [Google Scholar]
  • 113.Phillips ML, Drevets WC, Rauch SL, Lane R. Neurobiology of emotion perception I: The neural basis of normal emotion perception. Biological psychiatry. 2003 Sep 1;54(5):504–514. doi: 10.1016/s0006-3223(03)00168-9. [DOI] [PubMed] [Google Scholar]
  • 114.Salzman CD, Fusi S. Emotion, cognition, and mental state representation in amygdala and prefrontal cortex. Annual review of neuroscience. 2010;33:173–202. doi: 10.1146/annurev.neuro.051508.135256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Wager TD, Davidson ML, Hughes BL, Lindquist MA, Ochsner KN. Prefrontal-subcortical pathways mediating successful emotion regulation. Neuron. 2008 Sep 25;59(6):1037–1050. doi: 10.1016/j.neuron.2008.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Benloucif S, Galloway MP. Facilitation of dopamine release in vivo by serotonin agonists: studies with microdialysis. European journal of pharmacology. 1991 Jul 23;200(1):1–8. doi: 10.1016/0014-2999(91)90658-d. [DOI] [PubMed] [Google Scholar]
  • 117.Di Mascio M, Di Giovanni G, Di Matteo V, Prisco S, Esposito E. Selective serotonin reuptake inhibitors reduce the spontaneous activity of dopaminergic neurons in the ventral tegmental area. Brain research bulletin. 1998 Aug;46(6):547–554. doi: 10.1016/s0361-9230(98)00054-9. [DOI] [PubMed] [Google Scholar]
  • 118.Spoont MR. Modulatory role of serotonin in neural information processing: implications for human psychopathology. Psychological bulletin. 1992 Sep;112(2):330–350. doi: 10.1037/0033-2909.112.2.330. [DOI] [PubMed] [Google Scholar]
  • 119.Panksepp J. Affective neuroscience of the emotional BrainMind: evolutionary perspectives and implications for understanding depression. Dialogues in clinical neuroscience. 2010;12(4):533–545. doi: 10.31887/DCNS.2010.12.4/jpanksepp. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Bechara A, Damasio AR. The somatic marker hypothesis: a neural theory of economic decision. Games and Economic Behavior. 2005;52:336–372. [Google Scholar]
  • 121.MacQueen G, Frodl T. The hippocampus in major depression: evidence for the convergence of the bench and bedside in psychiatric research? Molecular psychiatry. 2011 Mar;16(3):252–264. doi: 10.1038/mp.2010.80. [DOI] [PubMed] [Google Scholar]
  • 122.Sapolsky RM. Stress and plasticity in the limbic system. Neurochemical research. 2003 Nov;28(11):1735–1742. doi: 10.1023/a:1026021307833. [DOI] [PubMed] [Google Scholar]
  • 123.Spinelli S, Chefer S, Suomi SJ, Higley JD, Barr CS, Stein E. Early-life stress induces long-term morphologic changes in primate brain. Archives of general psychiatry. 2009 Jun;66(6):658–665. doi: 10.1001/archgenpsychiatry.2009.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.McEwen BS. Stress and hippocampal plasticity. Annual review of neuroscience. 1999;22:105–122. doi: 10.1146/annurev.neuro.22.1.105. [DOI] [PubMed] [Google Scholar]
  • 125.Campbell S, Macqueen G. The role of the hippocampus in the pathophysiology of major depression. Journal of psychiatry & neuroscience : JPN. 2004 Nov;29(6):417–426. [PMC free article] [PubMed] [Google Scholar]
  • 126.Samuels BA, Hen R. Neurogenesis and affective disorders. The European journal of neuroscience. 2011 Mar;33(6):1152–1159. doi: 10.1111/j.1460-9568.2011.07614.x. [DOI] [PubMed] [Google Scholar]
  • 127.Thomas RM, Hotsenpiller G, Peterson DA. Acute psychosocial stress reduces cell survival in adult hippocampal neurogenesis without altering proliferation. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2007 Mar 14;27(11):2734–2743. doi: 10.1523/JNEUROSCI.3849-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Fuchs E, Flugge G. Chronic social stress: effects on limbic brain structures. Physiology & behavior. 2003 Aug;79(3):417–427. doi: 10.1016/s0031-9384(03)00161-6. [DOI] [PubMed] [Google Scholar]
  • 129.Willard SL, Friedman DP, Henkel CK, Shively CA. Anterior hippocampal volume is reduced in behaviorally depressed female cynomolgus macaques. Psychoneuroendocrinology. 2009 Nov;34(10):1469–1475. doi: 10.1016/j.psyneuen.2009.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Arabadzisz D, Diaz-Heijtz R, Knuesel I, et al. Primate early life stress leads to long-term mild hippocampal decreases in corticosteroid receptor expression. Biological psychiatry. 2010 Jun 1;67(11):1106–1109. doi: 10.1016/j.biopsych.2009.12.016. [DOI] [PubMed] [Google Scholar]
  • 131.McKinnon MC, Yucel K, Nazarov A, MacQueen GM. A meta-analysis examining clinical predictors of hippocampal volume in patients with major depressive disorder. Journal of psychiatry & neuroscience : JPN. 2009 Jan;34(1):41–54. [PMC free article] [PubMed] [Google Scholar]
  • 132.Rao U, Chen LA, Bidesi AS, Shad MU, Thomas MA, Hammen CL. Hippocampal changes associated with early-life adversity and vulnerability to depression. Biological psychiatry. 2010 Feb 15;67(4):357–364. doi: 10.1016/j.biopsych.2009.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Frodl TS, Koutsouleris N, Bottlender R, et al. Depression-related variation in brain morphology over 3 years: effects of stress? Archives of general psychiatry. 2008 Oct;65(10):1156–1165. doi: 10.1001/archpsyc.65.10.1156. [DOI] [PubMed] [Google Scholar]
  • 134.Campbell S, Marriott M, Nahmias C, MacQueen GM. Lower hippocampal volume in patients suffering from depression: a meta-analysis. The American journal of psychiatry. 2004 Apr;161(4):598–607. doi: 10.1176/appi.ajp.161.4.598. [DOI] [PubMed] [Google Scholar]
  • 135.Hamilton JP, Siemer M, Gotlib IH. Amygdala volume in major depressive disorder: a meta-analysis of magnetic resonance imaging studies. Molecular psychiatry. 2008 Nov;13(11):993–1000. doi: 10.1038/mp.2008.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.MacMillan S, Szeszko PR, Moore GJ, et al. Increased amygdala: hippocampal volume ratios associated with severity of anxiety in pediatric major depression. Journal of child and adolescent psychopharmacology. 2003 Spring;13(1):65–73. doi: 10.1089/104454603321666207. [DOI] [PubMed] [Google Scholar]
  • 137.Koolschijn PC, van Haren NE, Lensvelt-Mulders GJ, Hulshoff Pol HE, Kahn RS. Brain volume abnormalities in major depressive disorder: a meta-analysis of magnetic resonance imaging studies. Human brain mapping. 2009 Nov;30(11):3719–3735. doi: 10.1002/hbm.20801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Lorenzetti V, Allen NB, Fornito A, Yucel M. Structural brain abnormalities in major depressive disorder: a selective review of recent MRI studies. Journal of affective disorders. 2009 Sep;117(1–2):1–17. doi: 10.1016/j.jad.2008.11.021. [DOI] [PubMed] [Google Scholar]
  • 139.Nolan CL, Moore GJ, Madden R, et al. Prefrontal cortical volume in childhood-onset major depression: preliminary findings. Archives of general psychiatry. 2002 Feb;59(2):173–179. doi: 10.1001/archpsyc.59.2.173. [DOI] [PubMed] [Google Scholar]
  • 140.Botteron KN, Raichle ME, Drevets WC, Heath AC, Todd RD. Volumetric reduction in left subgenual prefrontal cortex in early onset depression. Biological psychiatry. 2002 Feb 15;51(4):342–344. doi: 10.1016/s0006-3223(01)01280-x. [DOI] [PubMed] [Google Scholar]
  • 141.Drevets WC. Neuroimaging studies of mood disorders. Biological psychiatry. 2000 Oct 15;48(8):813–829. doi: 10.1016/s0006-3223(00)01020-9. [DOI] [PubMed] [Google Scholar]
  • 142.Kim MJ, Hamilton JP, Gotlib IH. Reduced caudate gray matter volume in women with major depressive disorder. Psychiatry research. 2008 Nov 30;164(2):114–122. doi: 10.1016/j.pscychresns.2007.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Drevets WC, Price JL, Furey ML. Brain structural and functional abnormalities in mood disorders: implications for neurocircuitry models of depression. Brain structure & function. 2008 Sep;213(1–2):93–118. doi: 10.1007/s00429-008-0189-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Peng J, Liu J, Nie B, et al. Cerebral and cerebellar gray matter reduction in first-episode patients with major depressive disorder: A voxel-based morphometry study. European journal of radiology. 2010 May 11; doi: 10.1016/j.ejrad.2010.04.006. [DOI] [PubMed] [Google Scholar]
  • 145.Pine DS. Brain development and the onset of mood disorders. Seminars in clinical neuropsychiatry. 2002 Oct;7(4):223–233. doi: 10.1053/scnp.2002.35218. [DOI] [PubMed] [Google Scholar]
  • 146.Rosenberg DR, Macmaster FP, Mirza Y, et al. Reduced anterior cingulate glutamate in pediatric major depression: a magnetic resonance spectroscopy study. Biological psychiatry. 2005 Nov 1;58(9):700–704. doi: 10.1016/j.biopsych.2005.05.007. [DOI] [PubMed] [Google Scholar]
  • 147.Kieseppa T, Eerola M, Mantyla R, et al. Major depressive disorder and white matter abnormalities: a diffusion tensor imaging study with tract-based spatial statistics. Journal of affective disorders. 2010 Jan;120(1–3):240–244. doi: 10.1016/j.jad.2009.04.023. [DOI] [PubMed] [Google Scholar]
  • 148.Maller JJ, Thomson RH, Lewis PM, Rose SE, Pannek K, Fitzgerald PB. Traumatic brain injury, major depression, and diffusion tensor imaging: making connections. Brain research reviews. 2010 Sep;64(1):213–240. doi: 10.1016/j.brainresrev.2010.04.003. [DOI] [PubMed] [Google Scholar]
  • 149.Sexton CE, Mackay CE, Ebmeier KP. A systematic review of diffusion tensor imaging studies in affective disorders. Biological psychiatry. 2009 Nov 1;66(9):814–823. doi: 10.1016/j.biopsych.2009.05.024. [DOI] [PubMed] [Google Scholar]
  • 150.Shimony JS, Sheline YI, D'Angelo G, et al. Diffuse microstructural abnormalities of normal-appearing white matter in late life depression: a diffusion tensor imaging study. Biological psychiatry. 2009 Aug 1;66(3):245–252. doi: 10.1016/j.biopsych.2009.02.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Ma N, Li L, Shu N, et al. White matter abnormalities in first-episode, treatment-naive young adults with major depressive disorder. The American journal of psychiatry. 2007 May;164(5):823–826. doi: 10.1176/ajp.2007.164.5.823. [DOI] [PubMed] [Google Scholar]
  • 152.Cullen KR, Klimes-Dougan B, Muetzel R, et al. Altered white matter microstructure in adolescents with major depression: a preliminary study. Journal of the American Academy of Child and Adolescent Psychiatry. 2010 Feb;49(2):173–183. doi: 10.1097/00004583-201002000-00011. e171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Huang H, Fan X, Williamson DE, Rao U. White matter changes in healthy adolescents at familial risk for unipolar depression: a diffusion tensor imaging study. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 2011 Feb;36(3):684–691. doi: 10.1038/npp.2010.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Hamidi M, Drevets WC, Price JL. Glial reduction in amygdala in major depressive disorder is due to oligodendrocytes. Biological psychiatry. 2004 Mar 15;55(6):563–569. doi: 10.1016/j.biopsych.2003.11.006. [DOI] [PubMed] [Google Scholar]
  • 155.Leventopoulos M, Ruedi-Bettschen D, Knuesel I, Feldon J, Pryce CR, Opacka-Juffry J. Long-term effects of early life deprivation on brain glia in Fischer rats. Brain research. 2007 Apr 20;1142:119–126. doi: 10.1016/j.brainres.2007.01.039. [DOI] [PubMed] [Google Scholar]
  • 156.Rajkowska G, Miguel-Hidalgo JJ. Gliogenesis and glial pathology in depression. CNS & neurological disorders drug targets. 2007 Jun;6(3):219–233. doi: 10.2174/187152707780619326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Pizzagalli DA, Holmes AJ, Dillon DG, et al. Reduced caudate and nucleus accumbens response to rewards in unmedicated individuals with major depressive disorder. The American journal of psychiatry. 2009 Jun;166(6):702–710. doi: 10.1176/appi.ajp.2008.08081201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Rigucci S, Serafini G, Pompili M, Kotzalidis GD, Tatarelli R. Anatomical and functional correlates in major depressive disorder: the contribution of neuroimaging studies. The world journal of biological psychiatry : the official journal of the World Federation of Societies of Biological Psychiatry. 2010 Mar;11(2 Pt 2):165–180. doi: 10.1080/15622970903131571. [DOI] [PubMed] [Google Scholar]
  • 159.Greicius MD, Flores BH, Menon V, et al. Resting-state functional connectivity in major depression: abnormally increased contributions from subgenual cingulate cortex and thalamus. Biological psychiatry. 2007 Sep 1;62(5):429–437. doi: 10.1016/j.biopsych.2006.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Hasler G, Northoff G. Discovering imaging endophenotypes for major depression. Molecular psychiatry. 2011 Jun;16(6):604–619. doi: 10.1038/mp.2011.23. [DOI] [PubMed] [Google Scholar]
  • 161.Leppanen JM. Emotional information processing in mood disorders: a review of behavioral and neuroimaging findings. Current opinion in psychiatry. 2006 Jan;19(1):34–39. doi: 10.1097/01.yco.0000191500.46411.00. [DOI] [PubMed] [Google Scholar]
  • 162.Davey CG, Allen NB, Harrison BJ, Yucel M. Increased amygdala response to positive social feedback in young people with major depressive disorder. Biological psychiatry. 2011 Apr 15;69(8):734–741. doi: 10.1016/j.biopsych.2010.12.004. [DOI] [PubMed] [Google Scholar]
  • 163.Forbes EE, Christopher May J, Siegle GJ, et al. Reward-related decision-making in pediatric major depressive disorder: an fMRI study. Journal of child psychology and psychiatry, and allied disciplines. 2006 Oct;47(10):1031–1040. doi: 10.1111/j.1469-7610.2006.01673.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Forbes EE, Hariri AR, Martin SL, et al. Altered striatal activation predicting real-world positive affect in adolescent major depressive disorder. The American journal of psychiatry. 2009 Jan;166(1):64–73. doi: 10.1176/appi.ajp.2008.07081336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Gotlib IH, Hamilton JP, Cooney RE, Singh MK, Henry ML, Joormann J. Neural processing of reward and loss in girls at risk for major depression. Archives of general psychiatry. 2010 Apr;67(4):380–387. doi: 10.1001/archgenpsychiatry.2010.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Roberson-Nay R, McClure EB, Monk CS, et al. Increased amygdala activity during successful memory encoding in adolescent major depressive disorder: An FMRI study. Biological psychiatry. 2006 Nov 1;60(9):966–973. doi: 10.1016/j.biopsych.2006.02.018. [DOI] [PubMed] [Google Scholar]
  • 167.Shad MU, Bidesi AP, Chen LA, Ernst M, Rao U. Neurobiology of decision making in depressed adolescents: a functional magnetic resonance imaging study. Journal of the American Academy of Child and Adolescent Psychiatry. 2011 Jun;50(6):612–621. doi: 10.1016/j.jaac.2011.03.011. e612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Konarski JZ, McIntyre RS, Soczynska JK, Kennedy SH. Neuroimaging approaches in mood disorders: technique and clinical implications. Annals of clinical psychiatry : official journal of the American Academy of Clinical Psychiatrists. 2007 Oct-Dec;19(4):265–277. doi: 10.1080/10401230701653435. [DOI] [PubMed] [Google Scholar]
  • 169.Milne A, MacQueen GM, Yucel K, Soreni N, Hall GB. Hippocampal metabolic abnormalities at first onset and with recurrent episodes of a major depressive disorder: a proton magnetic resonance spectroscopy study. NeuroImage. 2009 Aug 1;47(1):36–41. doi: 10.1016/j.neuroimage.2009.03.031. [DOI] [PubMed] [Google Scholar]
  • 170.Yildiz-Yesiloglu A, Ankerst DP. Review of 1H magnetic resonance spectroscopy findings in major depressive disorder: a meta-analysis. Psychiatry research. 2006 Jun 30;147(1):1–25. doi: 10.1016/j.pscychresns.2005.12.004. [DOI] [PubMed] [Google Scholar]
  • 171.Kondo DG, Hellem TL, Sung YH, et al. Review: magnetic resonance spectroscopy studies of pediatric major depressive disorder. Depression research and treatment. 2011;2011 doi: 10.1155/2011/650450. 650450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Olvera RL, Caetano SC, Stanley JA, et al. Reduced medial prefrontal N-acetyl-aspartate levels in pediatric major depressive disorder: a multi-voxel in vivo(1)H spectroscopy study. Psychiatry research. 2010 Nov 30;184(2):71–76. doi: 10.1016/j.pscychresns.2010.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Kessler RC, Walters EE. Epidemiology of DSM-III-R major depression and minor depression among adolescents and young adults in the National Comorbidity Survey. Depression and anxiety. 1998;7(1):3–14. doi: 10.1002/(sici)1520-6394(1998)7:1<3::aid-da2>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
  • 174.Birmaher B, Arbelaez C, Brent D. Course and outcome of child and adolescent major depressive disorder. Child and adolescent psychiatric clinics of North America. 2002 Jul;11(3):619–637. doi: 10.1016/s1056-4993(02)00011-1. x. [DOI] [PubMed] [Google Scholar]
  • 175.Gerber AJ, Peterson BS. Applied brain imaging. Journal of the American Academy of Child and Adolescent Psychiatry. 2008 Mar;47(3):239. doi: 10.1097/CHI.0b013e3181635e00. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Pavuluri MN, Sweeney JA. Integrating functional brain neuroimaging and developmental cognitive neuroscience in child psychiatry research. Journal of the American Academy of Child and Adolescent Psychiatry. 2008 Nov;47(11):1273–1288. doi: 10.1097/CHI.0b013e318185d2d1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Uhl GR, Grow RW. The burden of complex genetics in brain disorders. Archives of general psychiatry. 2004 Mar;61(3):223–229. doi: 10.1001/archpsyc.61.3.223. [DOI] [PubMed] [Google Scholar]
  • 178.Nugent NR, Tyrka AR, Carpenter LL, Price LH. Gene-environment interactions: early life stress and risk for depressive and anxiety disorders. Psychopharmacology. 2011 Mar;214(1):175–196. doi: 10.1007/s00213-010-2151-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Silberg JL, Maes H, Eaves LJ. Genetic and environmental influences on the transmission of parental depression to children's depression and conduct disturbance: an extended Children of Twins study. Journal of child psychology and psychiatry, and allied disciplines. 2010 Jun;51(6):734–744. doi: 10.1111/j.1469-7610.2010.02205.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Wichers M, Geschwind N, Jacobs N, et al. Transition from stress sensitivity to a depressive state: longitudinal twin study. The British journal of psychiatry : the journal of mental science. 2009 Dec;195(6):498–503. doi: 10.1192/bjp.bp.108.056853. [DOI] [PubMed] [Google Scholar]
  • 181.Rice F, Harold GT, Thapar A. Assessing the effects of age, sex and shared environment on the genetic aetiology of depression in childhood and adolescence. Journal of child psychology and psychiatry, and allied disciplines. 2002 Nov;43(8):1039–1051. doi: 10.1111/1469-7610.00231. [DOI] [PubMed] [Google Scholar]
  • 182.Scourfield J, Rice F, Thapar A, Harold GT, Martin N, McGuffin P. Depressive symptoms in children and adolescents: changing aetiological influences with development. Journal of child psychology and psychiatry, and allied disciplines. 2003 Oct;44(7):968–976. doi: 10.1111/1469-7610.00181. [DOI] [PubMed] [Google Scholar]
  • 183.Gjone H, Stevenson J, Sundet JM, Eilertsen DE. Changes in heritability across increasing levels of behavior problems in young twins. Behavior genetics. 1996 Jul;26(4):419–426. doi: 10.1007/BF02359486. [DOI] [PubMed] [Google Scholar]
  • 184.O'Connor TG, Neiderhiser JM, Reiss D, Hetherington EM, Plomin R. Genetic contributions to continuity, change, and co-occurrence of antisocial and depressive symptoms in adolescence. Journal of child psychology and psychiatry, and allied disciplines. 1998 Mar;39(3):323–336. [PubMed] [Google Scholar]
  • 185.Eley TC, Stevenson J. Exploring the covariation between anxiety and depression symptoms: a genetic analysis of the effects of age and sex. Journal of child psychology and psychiatry, and allied disciplines. 1999 Nov;40(8):1273–1282. [PubMed] [Google Scholar]
  • 186.Silberg J, Pickles A, Rutter M, et al. The influence of genetic factors and life stress on depression among adolescent girls. Archives of general psychiatry. 1999 Mar;56(3):225–232. doi: 10.1001/archpsyc.56.3.225. [DOI] [PubMed] [Google Scholar]
  • 187.Karg K, Burmeister M, Shedden K, Sen S. The serotonin transporter promoter variant (5-HTTLPR), stress, and depression meta-analysis revisited: evidence of genetic moderation. Archives of general psychiatry. 2011 May;68(5):444–454. doi: 10.1001/archgenpsychiatry.2010.189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Caspi A, Sugden K, Moffitt TE, et al. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science. 2003 Jul 18;301(5631):386–389. doi: 10.1126/science.1083968. [DOI] [PubMed] [Google Scholar]
  • 189.Kaufman J, Yang BZ, Douglas-Palumberi H, et al. Social supports and serotonin transporter gene moderate depression in maltreated children. Proceedings of the National Academy of Sciences of the United States of America. 2004 Dec 7;101(49):17316–17321. doi: 10.1073/pnas.0404376101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Risch N, Herrell R, Lehner T, et al. Interaction between the serotonin transporter gene (5-HTTLPR), stressful life events, and risk of depression: a meta-analysis. JAMA : the journal of the American Medical Association. 2009 Jun 17;301(23):2462–2471. doi: 10.1001/jama.2009.878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Heim C, Bradley B, Mletzko TC, et al. Effect of Childhood Trauma on Adult Depression and Neuroendocrine Function: Sex-Specific Moderation by CRH Receptor 1 Gene. Frontiers in behavioral neuroscience. 2009;3:41. doi: 10.3389/neuro.08.041.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Aguilera M, Arias B, Wichers M, et al. Early adversity and 5-HTT/BDNF genes: new evidence of gene-environment interactions on depressive symptoms in a general population. Psychological medicine. 2009 Sep;39(9):1425–1432. doi: 10.1017/S0033291709005248. [DOI] [PubMed] [Google Scholar]
  • 193.Castren E, Rantamaki T. Role of brain-derived neurotrophic factor in the aetiology of depression: implications for pharmacological treatment. CNS drugs. 2010 Jan 1;24(1):1–7. doi: 10.2165/11530010-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 194.Kaufman J, Yang BZ, Douglas-Palumberi H, et al. Brain-derived neurotrophic factor-5-HTTLPR gene interactions and environmental modifiers of depression in children. Biological psychiatry. 2006 Apr 15;59(8):673–680. doi: 10.1016/j.biopsych.2005.10.026. [DOI] [PubMed] [Google Scholar]
  • 195.Gatt JM, Nemeroff CB, Dobson-Stone C, et al. Interactions between BDNF Val66Met polymorphism and early life stress predict brain and arousal pathways to syndromal depression and anxiety. Molecular psychiatry. 2009 Jul;14(7):681–695. doi: 10.1038/mp.2008.143. [DOI] [PubMed] [Google Scholar]
  • 196.Munafo MR, Brown SM, Hariri AR. Serotonin transporter (5-HTTLPR) genotype and amygdala activation: a meta-analysis. Biological psychiatry. 2008 May 1;63(9):852–857. doi: 10.1016/j.biopsych.2007.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Frodl T, Moller HJ, Meisenzahl E. Neuroimaging genetics: new perspectives in research on major depression? Acta psychiatrica Scandinavica. 2008 Nov;118(5):363–372. doi: 10.1111/j.1600-0447.2008.01225.x. [DOI] [PubMed] [Google Scholar]
  • 198.Frodl T, Reinhold E, Koutsouleris N, et al. Childhood stress, serotonin transporter gene and brain structures in major depression. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology. 2010 May;35(6):1383–1390. doi: 10.1038/npp.2010.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Duman RS, Monteggia LM. A neurotrophic model for stress-related mood disorders. Biological psychiatry. 2006 Jun 15;59(12):1116–1127. doi: 10.1016/j.biopsych.2006.02.013. [DOI] [PubMed] [Google Scholar]
  • 200.Griffin EW, Mulally S, Foley C, Warmington SA, O'Mara SM, Kelly AM. Aerobic exercise improves hippocampal function and increases BDNF in the serum of young adult males. Physiology & behavior. 2011 Jun 23; doi: 10.1016/j.physbeh.2011.06.005. [DOI] [PubMed] [Google Scholar]
  • 201.Lucassen PJ, Meerlo P, Naylor AS, et al. Regulation of adult neurogenesis by stress, sleep disruption, exercise and inflammation: Implications for depression and antidepressant action. European neuropsychopharmacology : the journal of the European College of Neuropsychopharmacology. 2010 Jan;20(1):1–17. doi: 10.1016/j.euroneuro.2009.08.003. [DOI] [PubMed] [Google Scholar]
  • 202.Bowley MP, Drevets WC, Ongur D, Price JL. Low glial numbers in the amygdala in major depressive disorder. Biological psychiatry. 2002 Sep 1;52(5):404–412. doi: 10.1016/s0006-3223(02)01404-x. [DOI] [PubMed] [Google Scholar]
  • 203.Holsboer F, Ising M. Central CRH system in depression and anxiety--evidence from clinical studies with CRH1 receptor antagonists. European journal of pharmacology. 2008 Apr 7;583(2–3):350–357. doi: 10.1016/j.ejphar.2007.12.032. [DOI] [PubMed] [Google Scholar]
  • 204.Linden DE. Brain imaging and psychotherapy: methodological considerations and practical implications. European archives of psychiatry and clinical neuroscience. 2008 Nov;258(Suppl 5):71–75. doi: 10.1007/s00406-008-5023-1. [DOI] [PubMed] [Google Scholar]
  • 205.Kupfer DJ, First MB, Regier DA. A Research Agenda for DSM-V. 2002 [Google Scholar]
  • 206.Leibenluft E. Skating to where the puck will be: the importance of neuroimaging literacy in child psychiatry. Journal of the American Academy of Child and Adolescent Psychiatry. 2008 Nov;47(11):1213–1216. doi: 10.1097/CHI.0b013e318185dab8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Mason GF, Krystal JH. MR spectroscopy: its potential role for drug development for the treatment of psychiatric diseases. NMR in biomedicine. 2006 Oct;19(6):690–701. doi: 10.1002/nbm.1080. [DOI] [PubMed] [Google Scholar]
  • 208.Kraemer HC, Wilson GT, Fairburn CG, Agras WS. Mediators and moderators of treatment effects in randomized clinical trials. Archives of general psychiatry. 2002 Oct;59(10):877–883. doi: 10.1001/archpsyc.59.10.877. [DOI] [PubMed] [Google Scholar]

RESOURCES