Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Dec 5.
Published in final edited form as: Physiol Behav. 2012 May 27;107(5):623–640. doi: 10.1016/j.physbeh.2012.05.014

Have studies of the developmental regulation of behavioral phenotypes revealed the mechanisms of gene-environment interactions?

F Scott Hall 1, Maria T G Perona 1
PMCID: PMC3447116  NIHMSID: NIHMS381065  PMID: 22643448

Abstract

This review addresses the recent convergence of our long-standing knowledge of the regulation of behavioral phenotypes by developmental experience with recent advances in our understanding of mechanisms regulating gene expression. This review supports a particular perspective on the developmental regulation of behavioral phenotypes: That the role of common developmental experiences (e.g. maternal interactions, peer interactions, exposure to a complex environment, etc.) is to fit individuals to the circumstances of their lives within bounds determined by long-standing (evolutionary) mechanisms that have shaped responses to critical and fundamental types of experience via those aspects of gene structure that regulate gene expression. The phenotype of a given species is not absolute for a given genotype but rather variable within bounds that are determined by mechanisms regulated by experience (e.g. epigenetic mechanisms). This phenotypic variation is not necessarily random, or evenly distributed along a continuum of description or measurement, but often highly disjointed, producing distinct, even opposing, phenotypes. The potentiality for these varying phenotypes is itself the product of evolution, the potential for alternative phenotypes itself conveying evolutionary advantage. Examples of such phenotypic variation, resulting from environmental or experiential influences, have a long history of study in neurobiology, and a number of these will be discussed in this review: neurodevelopmental experiences that produce phenotypic variation in visual perception, cognitive function, and emotional behavior. Although other examples will be discussed, particular emphasis will be made on the role of social behavior on neurodevelopment and phenotypic determination. It will be argued that an important purpose of some aspects of social behavior is regulation of neurobehavioral phenotypes by experience via genetic regulatory mechanisms.

Keywords: Environmental enrichment, social isolation, maternal deprivation, gene-environment interaction, epigenetic mechanisms

Introduction

Over the last 50 years there has been accumulating evidence that particular experiences, often at particular critical periods during development, produce permanent alterations in behavioral phenotypes – and thus account for a substantial proportion of the variability in these phenotypes. Although all species have variability (the building blocks of evolution), substantial variability was found to be the result of differences in experience of particular types, at particular ages, affecting particular behaviors [1]. This review will begin by summarizing some of the main findings from investigations that have sought to purposefully manipulate aspects of early experience in order to determine the relationship between that experience and variability in later behavioral phenotypes, as well as the underlying biological mechanisms that produce this variation. This is an extensive literature, so this review is intended to be neither exhaustive for any specific experimental manipulation, or to completely summarize the types of altered experiences that produce changes in neurobehavioral phenotypes; rather, the examples taken are intended to illustrate the extent of variation in neurobehavioral phenotypes that can be produced by altered experience, as well as the depth of understanding of the processes involved that can, with the advent of modern genomics and epigenetics, be used to elaborate precise genetic and epigenetic mechanisms controlling what are usually termed “gene-environment” interactions.

Effects of early experience on visual system development

Hubel and Wiesel were awarded the Nobel Prize in 1981 for their work on visual perception. A very important aspect of this work was that visual input to the cerebral cortex is a critical regulator of visual system development and, in particular, cellular survival [2]. Grossly put, their findings have often been described as “use it, or lose it”, a statement that is to the point in some respects, but underestimates the subtleties of their findings, as they identified a number of limitations of the principle: not all brain structures were affected in the same manner and the effects of sensory input on visual system development were highly dependent on when that visual input occurred, thus identifying specific “critical periods” for visual system development. Their classical experiments on monocular deprivation [3] demonstrated that deprivation of input from one eye led to reduced responses in primary visual cortical neurons in response to input from that eye and a shift in the proportion of cells responding only to the non-deprived eye, e.g. a shift in ocular dominance [4]. Importantly, deprivation of input did not lead to reductions in the numbers of cells responsive to the deprived eye in the lateral geniculate nucleus [5], suggesting that only some levels of the visual system were regulated by early life visual experience. The same changes in visual cortex function were observed whether light input was completely occluded or only partially occluded by a translucent device that eliminated form vision with far less impact on total light input [4]. Importantly, this indicates that it is not deprivation of all visual input that is important to the development of the ocular dominance columns in striate cortex, but specifically visual input of aspects of form, to which these cells were responsive.

Thus, exposure to visual stimuli during development determines overall sensitivity to particular types of visual stimuli in the mature animal, i.e. the visual behavioral phenotype. Importantly, deprivation of visual input only produced these changes in ocular dominance if they occurred during a certain critical developmental periods [5], between 1 and 3 months of age in cats, and were not observed when similar experiments were done in adult animals. Studies in very young animals, done just as the eyes opened, indicated that the effects of deprivation influenced the survival of cells or cellular connections [2]. The discovery at about the same time of neurotrophic factors which were necessary for neuronal cell survival [6] provided a potential explanation for such phenomena. The release of neurotrophins consequent to cellular activity may provide an explanation for certain classes of experience-dependent neurodevelopmental processes in which the release of trophic factors occurs in the same synapses that are activated by the experience, subsequently affecting the function of those synapses and the survival of those cells. However, other types of experience-dependent neurodevelopmental processes involve more distributed regulation, across many aspects of behavioral function, involving diverse brain regions. Although it is certainly likely that trophic factors are involved in these processes as well, such mechanisms alone cannot account for them, and other signaling mechanisms are likely to be involved, including endocrine signals.

Environmental Enrichment

One way to think of the studies on visual system development is that experience with specific stimuli can affect the development of those parts of the cerebral cortex that respond to those stimuli. At about the same time that these findings were reported Rosenzwieg and colleagues demonstrated that overall environmental complexity (e.g. enrichment, termed by them “environmental complexity and training” or ECT) has very broad effects on the cerebral cortex and behavior dependent on the cerebral cortex [7]. As was the case for differences in visual experience, environmental enrichment produces changes in brain anatomy and chemistry (summarized initially in [8]). This “enrichment” consisted of housing from weaning of groups of rats in large cages with platforms for food and water and ample room for climbing on the wire sides. A small wooden maze and a set of “toys” (varying in shape and color) were presented on a rotating basis which the rats could manipulate, and interact with; indeed many of the toys were large enough to climb on, including a “swing” and a “ladder” (see Fig. 1 in Krech et al, 1960). Additionally, the rats were exposed to a variety of mazes during this enrichment period as well. Thus, environmental enrichment in these studies consisted of multiple components that separately might affect neurological and behavioral development, including exposure to complex visual and tactile cues, motor learning experiences associated with climbing and manipulating objects, spatial learning and hypothesis testing associated with maze training, exposure to learning under motivated circumstances and exposure to the physical cues and species typical interactions provided by other rats. From the outset it was recognized that each of these aspects of the enrichment experience might have separate effects on behavior and brain function. This was done to maximize the possibility of producing detectable changes in brain chemistry and function. It was recognized from the outset that the effects might be attributable to only some of the components of the ECT procedure; indeed, this was incorporated in the original experimental design with two control groups: an isolated condition (IC), which eliminates all components of the ECT procedure, including social interaction, and a social condition (SC) which eliminates all components of the ECT procedure except social interaction. The SC procedure is often found to produce effects that are intermediate between the ECT and IC groups, but at times the SC and ECT groups are not different, suggesting that the effects are purely the result of deprivation of social interaction.

Early findings with environmental enrichment demonstrated enhancement of learning in ECT rats, although often the comparisons were only to IC rats and limited to specific types of procedures. For instance, although there is no difference between ECT and IC rats in acquisition of a visual discrimination, IC rats are substantially impaired in acquisition of subsequent reversals of that discrimination [7]. These changes in learning are accompanied by dramatic changes in brain function. Gross brain weight, particularly of the cerebral cortex, is greater in ECT rats [911], as well as cortical thickness [1214], numbers of glia [13], size of neuronal perikarya [14], length of postsynaptic thickenings [15] and the density of dendritic spines in occipital cortical pyramidal neurons [16]. The large increase in glial numbers (but not neurons) accounts for the increased cortical volume but actually leads to reduced synaptic counts per unit area under some conditions [15]. Diamond et al. (1975) also had a social condition, and the SC rats were similar to the ECT rats indicating that the main effects were due to impoverishment (or social isolation) rather than enrichment, in this case. Surprisingly this increase in synaptic size is accompanied by an overall decrease in the number of synapses of about 33%, but a shift in the frequency distribution towards larger synapses (52% on average) in ECT rats compared to IC rats, resulting in an overall increase of total synaptic area of about 40% [17]. A series of detailed anatomical studies by Greenough and colleagues [1822] on occipital cortex morphology in EC, SC and IC rats specified these differences in more detail. Reduced neuronal density is found in EC rats, but along with a greater number of synapses per neuron [21], increased length of synaptic contact zones [18], increased dendritic length and branching [22], and increased glial, capillary and mitochondrial measures indicative of metabolic adjustments [19, 23]. As for the more gross anatomical changes mentioned above, SC rats are intermediate to EC and IC rats for some measures [18, 21], but not others [19, 23], for which EC rats are different from both SC and IC rats. Gross anatomical changes are also accompanied neurochemical changes including increases in acetylcholinesterase activity [9, 24]. Subsequent studies have also identified differences in serotonin, dopamine, glutamate, and other neurotransmitters systems [2530], which is not surprising given the widespread morphological changes to neuronal structure and distribution produced by environmental enrichment identified in these and subsequent studies (see [31]) for a complete review of this literature). Indeed, environmental enrichment reduces apoptosis and increases neurogenesis [32].

One of the most fundamental questions about the effects of environmental enrichment (and all other neurodevelopmental manipulations) involves the question of critical periods. One of the earliest such studies [24] found that environmental enrichment of both young and adult rats increases the weight of cortical subregions as well as total acetylcholinesterase activity. Indeed, for some measures the effects in adults actually appear to be more pronounced, but other measures indicate that although enrichment has effects in both young and adult animals, including measures of cortical thickness across diverse brain regions [33], the basis of these enrichment induced changes differs in young and adult rats in a variety of ways. In that study the effects of social housing conditions were most prominent at earlier ages and environmental enrichment at later ages, and affected somewhat different brain regions. Importantly, many of the changes in cortical weight resulting from enrichment can be observed in individually housed/enriched rats [34]. By contrast, differences in the length of postsynaptic thickenings in occipital cortex appear to be largely due to social isolation rather than environmental enrichment per se, e.g. SC rats are similar to ECT rats, and both are different from IC rats [15]. Thus, there appear to be somewhat separable effects of alterations in the physical environment and the social environment in studies of environmental enrichment.

Another way to address the question of critical periods is to determine whether later enrichment can reverse the effects of impoverishment: If the effects of enrichment/impoverishment have a critical period, then they should not be reversed by later enrichment. Some of the first studies found that later enrichment does reverse some of the effects of early impoverishment [35, 36], but these first findings were limited to the most basic measures of gross weight of cortical tissue and total acetylcholinesterase activity. In an experiment examining the persistence of the effects of environmental enrichment after subsequent environmental impoverishment, the persistence of the effects of enrichment were determined by both the duration of the initial ECT experience as well as the duration of the subsequent IC experience [37]. When the initial experience incorporates the entire period of juvenile development, up to 80 days, the effects of ECT on both gross brain measures and neurochemical measures are highly persistent. Persistence of the effects of enrichment is observed for behavioral consequences as well [38].

These changes in cortical structure and chemistry are associated with improved learning in a variety of tasks [30, 3943]. However, many of these comparisons have been between EC and IC rats, so at least some of these differences may be attributed to isolation-induced differences, discussed in more detail in a later section. Some learning differences in EC rats are associated with increases in REM sleep [40]. Importantly, this study used both SC and IC comparison groups; the IC group was impaired at initial learning compared to both EC and SC groups, while EC was superior to both groups in memory recall. Since EC groups had substantially more REM sleep than either SC or IC groups this was taken to be the basis of this effect. However, effects on memory alone do not account for enhanced learning in EC rats, as they also adopt more effective learning strategies than IC rats [41]. The extent to which deprivation of social interaction plays a role the differences observed between EC and IC rats is a complex question. Conspecifics no doubt constitute a major component of environmental complexity, contributing in some ways as general environmental stimuli (e.g. as complex visual, olfactory and tactile stimuli) and also in a specific interactive, experiential manner. Thus under some circumstances IC rats perform more poorly in learning paradigms compared to both EC and SC rats, while under other, more demanding circumstances EC rats perform superiorly to both SC and IC rats [44]. One approach to isolating these effects is to examine the effects of environmental complexity under equivalent social conditions; enrichment has been found to affect exploratory behavior [45], and improve learning [46] in isolated rats. Even the mildly enriching procedure of placing a gerbil, otherwise reared in isolation, in a different environment each day for 30 day increases dopamine fiber density in the prefrontal cortex and basolateral amygdala [47].

Studies have suggested that environmental enrichment affects dopaminergic systems based on behavioral, neurochemical and pharmacological data, including both reduced [4853] and increased responses to psychostimulants [5355]. Although the data is not entirely consistent, it would appear that self-administration is reduced while acute locomotor effects are increased. As part of this series of studies increased impulsivity was observed in IC rats, and opposite effects of amphetamine in EC and IC rats on impulsive choice [56], and faster extinction in EC rats and greater reinstatement in IC rats [51]. In a test of Pavlovian conditioned approach to examine incentive salience, EC rats were found to be “goal-trackers”, while IC rats were “sign-trackers” [57]. However, these studies have generally compared EC and IC conditions, and therefore cannot separate the consequences of environmental enrichment from isolation rearing, an important consideration given the profound effects that isolation rearing alone has on these behaivors. Some studies have included an SC group. EC rats had increased responses to both the locomotor stimulant and reinforcing effects of amphetamine compared to both IC and SC rats [58], but in a later study IC rats had increased self-administration compared to both EC and SC rats [50]. The protective effects of “enrichment” were also associated with the social component of enrichment, rather than the non-social component, in a study of cocaine escalation [59]. Schrijver et al. [43] specifically set out to dissociate the effects of social deprivation and environmental enrichment on cognitive function by examining the enrichment in both isolation-reared and socially reared rats. Those authors examined behavior dependent on hippocampal neocortical pathways (spatial memory formation) and behavior dependent on prefrontal corticostriatal pathways (reversal learning). Isolation rearing impaired reversal learning, but did not impair spatial learning and memory; while environmental enrichment improved spatial learning, but had no effect on reversal learning. However, some of these effects are not neurodevelopmental in their basis, as recent studies have shown that environmental enrichment after initial drug treatments in adults eliminated cocaine conditioned place preference, sensitization and reduced cue or drug induced reinstatement [6062] (see also [31]) for a complete discussion of this work). Another factor that may make the social and non-social aspects of enrichment especially hard to separate is that environmental enrichment has been shown to increase social interaction and social play in adolescent rats in comparison to socially housed rats housed in a non-enriched environment [63, 64].

The effects of social deprivation on emotional development in primates

While the first studies of environmental enrichment, which included a social component to enrichment, focused primarily on the effects of enrichment on cognitive function and consequent changes in brain structure, other studies specifically examined the effects of social deprivation on emotional development. The seminal studies of Harlow found that prolonged periods of social deprivation in primates produces profound impairments of emotional and social behavior [6567]. Quite interestingly given the effects of environmental enrichment on learning in rats discussed above, and despite rather extreme behavioral disruptions in socially deprived monkeys, cognitive deficits are limited in deprived monkeys which are so abnormal in many other respects. Indeed, social deprivation of primates, and rodents as well, leads to such abnormal behavior that social deprivation models have been suggested to model a variety of pathological conditions: the self-clutching and crouching behavior exhibited by socially deprived rhesus monkeys upon their initial release from these conditions has been compared to autistic children [67]; isolation housing of adult animals has been suggested to model phobia and anxiety [68]; isolation-rearing of rats has been suggested to model schizophrenia [1, 69]; repeated maternal deprivation in rodents has been suggested to model depression [70]; while both maternal deprivation and isolation rearing produce phenotypes that predispose individuals to addiction, although perhaps via different mechanisms [71].

One of the most important questions in considering deprivation of social experience as models of these disorders, and the potential underlying mechanisms, is the extent to which deprivation of specific types of social interaction induce specific changes in subsequent behavior. The key to this question is the nature of the deprivation experience itself, and how that is related to the behavioral regulators of the natural development of the organism across the lifespan. Harlow initiated his studies from a developmental perspective by describing the development of multiple affectional systems in rhesus monkeys (Macaca mulatta): mother-infant, infant-mother, peer, heterosexual and paternal [72]. Each of these is defined by the nature of the interactors (e.g. mother, infant, peer, etc.) and is related to distinct developmental stages. Furthermore, the development of each of these affectional systems is also subdivided into distinct stages based upon the type of social interactions between the actors. Although most famous for his studies of contact comfort with “cloth mothers”, Harlow described the effects of social deprivation under a variety conditions: differing durations, occurring at different ages, involving different types of social behavior and consequently affecting the development of systems related to these different affectional systems. Infant monkeys raised apart from their mothers would prefer a cuddly cloth surrogate mother without food over a wire surrogate mother that provided food [65, 66]. This is not to say that provision of nutrition was not important. Infant monkeys would prefer a lactating cloth surrogate over a non-lactating cloth surrogate, but contact comfort determined the infant’s preference for physical contact more than nutrition. Indeed, the cloth surrogate was a sufficiently adequate maternal replacement that deprived infant monkeys, in the presence of the surrogate, would exhibit somewhat normal exploration of inanimate and animate objects. This pattern of exploration using the cloth surrogate as a base is similar to that observed in normal infants with their mothers, as was the immediate return and contact clinging to the surrogate mother when frightened. Other sensory aspects of a surrogate mother are also preferred, including movement (e.g. rocking), warmth and texture [73, 74]. Infant monkeys respond quite similarly to removal of actual mothers and cloth-surrogate mothers at 6 months of age [75]. Presence of the attachment object in a novel circumstance, or in the presence of a stranger, evokes contact cling, while absence of the attachment object evokes agitation, vocalization and fearful responses. However, one should not conclude from these findings that the surrogate mother was a complete replacement for the real thing. Indeed, in considering the degree of normalcy engendered by the surrogate mother Harlow pointed out that one of the main roles of mothers is to shape the developing behavior of the infant monkey [76] – an essential point when considering the critical aspects of experience that might mediate the effects of maternal deprivation. The maternal role includes the security and protection roles previously described that are so necessary for initial interactions with the physical and social environment that lead to the transition away from infant behaviors like excess clinging and non-nutritive sucking. This process is more often initiated by the mother than the infant and leads the infant towards age-appropriate social behaviors with peers and adults. One immediate role of the mother as infants begin to interact with peers is to regulate play behavior, particularly levels of aggression. These functions of maternal experience cannot be replaced by an inanimate surrogate and indicate how these experiences regulate subsequent behavioral competency. Importantly, one can also infer interactions between the consequences of maternal behavior directed towards the infant at this stage and later social development. Another important role of the mother is to train her infant to comprehend gestural and vocal communication. Infant monkeys deprived of the experiences necessary to regulate emotional reactions and communicate with other monkeys are asocial and subject over time to increasing ostracism and violence from other young monkeys. Finally, it should also be noted that different maternal attributes, some captured by the inanimate maternal surrogate and some not, affect the development of different behavioral and psychological attributes in infants. This diversity of neuro-regulatory factors increases as other types of social interactions are considered.

Studies of responses of infant rhesus monkeys to separation from their mothers [7779] described a sequence of reactions beginning with agitation (characterized by frantic activity, climbing, screeching and crying), followed by a period of despair (characterized by greatly reduced activity) and finally proceeding to a period of “grief and mourning”. Confirming the central role of the mother as an emotional base from which subsequent exploration of the world originates, when infant monkeys were allowed a period of access to other infant monkeys during a period of maternal separation peer-peer interactions were decreased [7779]. In those same studies, when maternally deprived monkeys were reunited with their mothers aspects of infant-mother/mother-infant interaction increased for a short time (e.g. cradling, embracing, clinging, and ventral contact). During this period peer-peer contact was also increased, over pre-separation levels, but was also of a somewhat different quality, more aggressive than prior to the separation. Nonetheless, surrogate-reared rhesus monkeys, when subsequently allowed access to peers, initially exhibited abnormal behavior, but their behavior gradually normalized to a large extent over the first year of life [80]. Play was an important part of this process of normalization subsequent to maternal deprivation. This did not happen when monkeys were subsequently isolated from their peers [81]. These experiments emphasize the interacting and complex nature of life events occurring during different developmental stages.

Extended periods of early deprivation that deprived young monkeys of both maternal and peer social interactions, termed “total social isolation” [82, 83], has been consistently associated with a variety of types of aberrant behavior. These include compulsive oral behavior, repetitive stereotypical behavior, decreased exploration of non-social stimuli, aggression, self-injurious behavior and an inability to form normal social and sexual attachments later in life. The severity of these symptoms increases with the duration of the deprivation and total social isolates were described as “depressed” [84], the longer the period of deprivation the greater the deficits. Monkeys isolated for 6 months show profound changes in behavior in a novel playroom when placed with control monkeys, with almost no behavior directed at other monkeys (contact play or threat), and minimal activity play (parallel play). In this circumstance the effects of long term deprivation are very consistent over long periods of subsequent testing, manifested by asociality of the isolated monkeys. Over time, in that experiment, control monkeys exhibited more and more aggression towards the isolates, to the point where testing had to be terminated.

Although extremely illuminating as to the degree of behavioral disturbance that can be engendered by social deprivation, the combination of altered experience in these experimental designs makes it difficult to isolate causal factors. Although maternal deprivation has very profound effects by itself, Harlow noted of the role of early social experience that “play is the variable of primary importance in the development of normal social, sexual and maternal functions” [74]. Furthermore, play itself is by no means a unitary construct, there being a number of distinct types of play in primates that appear to have quite different developmental roles [85, 86]. The consequences of deprivation of these experiences early in life are highly persistent; later in life (10 yrs), rhesus monkeys isolated for the first 9 months of life exhibited self-injurious behavior (e.g. self-biting, slapping, or head-banging) in response to a stressor or conditioned aversive stimulus [87].

When deprived rhesus monkeys later became mothers themselves (note that this was accomplished with some difficulty as they were highly incompetent sexually) they were extremely abusive and neglectful to their infants [8890]. These studies of what were termed “motherless mothers” raise the very interesting possibility that deviant behavior patterns may be passed from generation to generation because maternal deprivation leads to aberrant maternal behavior that leads to abnormal, impaired or insufficient maternal behavior in the next generation. Indeed, when offspring of motherless mothers were studied they were found to be deficient in social play and sexual behavior, but more aggressive [89], the same attributes that characterized their mothers. Indeed, this might be considered to be a “purer” experiment of the consequences of decreased maternal interactions because it was not imposed by the experimenters, but rather a natural consequence of the behavioral tendencies of the mothers. Furthermore, common allelic variants may also produce variation in maternal behavior [91] that might be similarly expected to affect behavioral development.

Two fundamentally important issues regarding these effects involve the persistence and reversibility of the consequences of these early experiences. Although effects of total social isolation are observed after 3 months, they are largely reversible, but this is not the case for longer periods of deprivation [82]. However, even in more severe cases resulting from extended isolation for 6 or 12 months, aberrant social behavior could be rehabilitated, to a certain extent, by subsequent exposure to peers, but only younger peers [83, 92]. Most of the outcomes explored in these primate models involved emotional and social behaviors. Another important issue is the degree of specificity resulting from deprivation of specific social experiences. Importantly, although producing substantial disturbances in emotional and social function, the 3 month isolation paradigm produces no differences in learning [9395], in stark contrast with environmental enrichment paradigms. Social deprivation in primates may produce some of same changes as isolation rearing in rodents. Thus, it appears that maternal deprivation in monkeys reduces self-administration of psychostimulants [96], although self-administration of drugs in primates appears to be affected by a variety of manipulations affecting interacting factors, including social dominance, which seem to be mediated, at least in part, by changes in D2 receptor function [97].

The behavioral consequences of social deprivation in rhesus monkeys indicate the potential involvement of several brain systems, but particularly those involved in regulation of responses to aversive stimuli, stress and other aspects of emotional behavior. The mechanisms underlying these changes have been better elaborated in studies of maternal deprivation in rodents, particularly as they contrast with the effects of post-weaning peer deprivation in rodents.

Maternal deprivation in rodents

Although the consequences of maternal deprivation are often discussed in terms of “stress” or “adversity”, and certainly do affect stress responsiveness, the effects of maternal deprivation are highly dependent upon the developmental state of the pup. The effects of maternal deprivation differ over the first three weeks of life prior to weaning depending in part upon the age of the pup when the deprivation occurs [1, 98]. Indeed, different intervening factors mediate the consequences of deprivation at different ages. At earlier ages these are largely passive (e.g. warmth, nutrition, physical contact) while at later ages these become much more active (e.g. active contact, non-nutritive suckling, maternal licking). Hofer has described these factors as “hidden regulators” [98], a concept which could certainly be applied to other types of environmental and experiential effects. These include changes in gross physiological parameters such as changes in heart rate and respiration during separation from the dam between 7 and 16 days postnatal [99103], as well as behavioral changes, including the levels of locomotion, rearing, digging, self-grooming and ultrasonic vocalizations (USV) during this period [103]. The behavioral effects depend in part on whether the isolation occurs in the nest or a novel environment [103], indicating that different sensory aspects of the experience may mediate different physiological and behavioral consequences of separation. For instance, although cardiovascular effects could be reversed by feeding, the behavioral activation that was observed during this period could not [104], while the presence of a non-lactating foster mother during deprivation would reverse the behavioral activation but not the cardiac changes [105, 106]. Thus, a different and distinct component of pup-dam interaction appears to regulate each consequence of maternal deprivation. Supporting this conclusion, another consequence of maternal deprivation, sleep disturbance, was regulated by a completely different factor – milk composition and rhythmicity of milk delivery [107].

Many of the changes that occur in rodents that are maternally deprived very early in life involve short-term responses to loss of the sole source of nutrition. Thus mechanisms controlling metabolism and growth are down-regulated immediately after an extended period of maternal deprivation, including ornithine decarboxylase [108], growth hormone [109] and heart rate [110], although the brain may be partially protected from some of these changes [108]. Suppression of growth hormone release can be reversed by administration of growth hormone-releasing factor [111]; interestingly, these effects are dependent on 5-HT2A/2C signaling. Some long-term effects of maternal deprivation are observed, including reductions in weight and feeding and changes in food preferences [112].

Many of the neurodevelopmental regulators of neonatal development involve active interaction between the dam and pup, which become more important as the pup develops increased behavioral capability and complexity. An important response to separation from the dam is USV, which elicits attention from the dam in the nest or pup retrieval if the pup is outside of the nest. When the mother forages away from the nest, her extended absence elicits USV from the pups after a period of time [113]; thus, the duration of USV is normally regulated by the time required for her foraging, and thus variably dependent upon environmental conditions, such as the density of food resources. The production of USV in pups is determined by a number of specific sensory aspects of isolation, including temperature. USV is minimal at thermoneutrality, but increases with slight deviations with temperature [114]. Although the suppressive effect of littermates during isolation is at least partially due to effects on thermoregulation, the ability of littermates alone to suppress USV increases with age, while the effects of temperature wane, indicating that other factors are present. In pups less than a week old maintenance of temperature alone is sufficient to suppress USV [115], while in 5 to13 day old pups tactile and olfactory cues are necessary to reduce USV [116], and in pups over 14 days of age the presence of a sibling or dam is necessary to suppress USV [113]. At this age passive tactile, olfactory and thermal cues are sufficient to suppress USV, e.g. an anesthetized pup or an anesthetized dam [117, 118].

Hypothalamic-pituitary-adrenal (HPA) axis activity, and changes in HPA axis sensitivity, are important in these effects, but are certainly not the only mechanisms involved. For instance, there is evidence of endogenous opioid release during maternal deprivation [119], the greatest opioid release being observed in the presence of nest bedding, which partially suppresses USV. This may indicate that stimuli associated with the dam come to produce opioid release – the comparison here to contact comfort is perhaps not unwarranted. This reduction in USV could be reversed by naltrexone, which has no effect in the absence of nest bedding, and may indicate that the involvement of endogenous opioids is an adaptive response to familiar or dam-associated stimuli. Under unfamiliar conditions other behavioral changes are observed, including locomotor activity, USV and hyperalgesia [120123]. These could also be considered to be adaptive responses, the behavior which is most adaptive varying with the duration and chronicity of deprivation. Importantly, vocal interactions may be reciprocal, as recent studies in octodon degus have demonstrated effects of maternal vocalizations on the behavior of maternally deprived pups [124, 125].

Much work on maternal deprivation has examined the role of maternal interactions in the development of HPA axis reactivity. USV is emitted during the first hour after separation, but then subsides, followed by increased signs of stress including plasma corticosterone increases [126]. Although adrenocortical responses to some stimuli are reduced or absent during the first two weeks of life [127], the so-called “stress hyporesponsive period”, extended periods (2 hr or more) of maternal deprivation can increase corticosterone levels from postnatal day 5 onward [126], responses becoming more robust thereafter [128, 129]. Maternally deprived pups exhibit HPA responses to stressors at ages which non-deprived pups are not yet responsive [130, 131]. In contrast to some of the other effects of maternal deprivation, the corticosterone response to maternal deprivation could not be reduced by milk [132] or the presence of siblings [133], but was reduced by the presence of the dam [132]. Returning a pup to the dam after a stressor reduces stress responses [134], but importantly the ability of a dam to inhibit pup stress responses is quickly lost after 20 days of age [133, 135].

There is a critical period for the effects of maternal deprivation on the HPA axis reactivity of about 6 to 15 days postnatal, with reduced effected observed earlier (4–6 days postnatal), and later (20 days postnatal) [112, 129131, 136141]. Enhanced responses to stressors are associated with reduced glucocorticoid negative feedback [129, 139, 142] and enhanced sensitivity to adrenocorticotropic hormone (ACTH) [128, 134]. Maternal deprivation accelerates HPA axis development [143], but it also appears that different aspects of pup-dam interactions regulate different aspects of these neuroendocrine changes [144], which also include changes in corticotrophin releasing factor (CRF) [137, 138, 145], and norepinephrine [146] function.

Either a single long episode or repeated shorter episodes of maternal deprivation induce permanent changes neural and neuroendocrine function; the duration, age of the pups and chronicity of the experience influence these permanent outcomes. As for more acute effects of maternal deprivation many, though not all, of the effects described above are reversed by anogenital stroking during deprivation [138, 147]. Interestingly, although dexamethasone treatment blocked the effects of maternal deprivation on ACTH and corticosterone, many of the central consequences (e.g. reduced hippocampal glucocorticoid receptor (GR) mRNA, elevated paraventricular nucleus (PVN) cFos mRNA and reduced PVN CRF mRNA) were unaffected, but could be reversed by stroking or stroking plus feeding [147]. This indicates that the aspects of the deprivation experience that affect these functions are more than just elevations in HPA axis activity, so, not surprisingly, the nature of the deprivation environment is a critical aspect of any maternal deprivation procedure [148].

Deeper understanding of the underlying mechanisms of maternal deprivation has come from comparisons to mice that received very short periods of deprivation (5–15 minutes), termed “handling”, in addition to unhandled controls. Many of the long-term effects of handling have been observed to be in direct opposition to the effects of extended periods of maternal deprivation: although handling accelerates the development of stress response early in life [149], it produces permanent reductions in HPA axis responsivity. The critical factor here has been determined to be increases maternal attention when the pup is returned to the dam [150, 151]. Thus, when assessed as adults, handled pups have decreased basal corticosterone, ACTH and CRF levels [152, 153], reduced stress-activated CRF, ACTH and corticosterone responses [154156], and reduced behavioral indices of emotional responses to stressors [157]. These changes occur in part because of increases in glucocorticoid negative feedback in the hippocampus [155, 158, 159]. Indeed, differences in stress responses may account for some apparent effects of neonatal handling on learning, such as impaired aversive conditioning [160], particularly since some other types of learning unaffected [161] are enhanced [160].

In contrast to the effects of handling, extended maternal deprivation enhances activity of the HPA axis and this enhancement persists into adulthood. Increased stress responsivity [162167] in maternally deprived animals is mediated in large part by decreased negative feedback mediated by hippocampal glucocorticoid receptors [163, 168, 169], but is also associated with increased activity at other levels of the HPA axis [112, 153, 163, 165, 167, 170, 171]. Changes in hippocampal feedback to the HPA axis are part of broader constellation of changes in hippocampal circuitry and function which may involve brain derived neurotrophic factor (BDNF) and glutamate function [172]. However, although maternally deprived rats exhibit some learning deficits in adulthood, these seem to be associated with stress hyper-responsiveness and can be reversed by glucocorticoid receptor or adrenergic receptor antagonism [167].

Maternally deprived rodents are anxious in adulthood [112, 162164, 166, 173175], exhibit increased depressive-like behavior [167, 175, 176] and increased ethanol consumption [162, 177, 178], which correlates with stressor evoked corticosterone release [162]. Consistent with hypotheses that these types of experience relate to the etiology of depression both increased anxiety and increased ethanol consumption can reversed by antidepressant treatments [162, 177]. Perhaps also consistent this possibility are reductions in responses to drug rewards and other responses associated with mesolimbic dopamine function [179181], consistent with reduced dopamine turnover in the striatum [182]. However, not all responses to reinforcers or stimulants are reduced [181, 183186], and dopamine responses to amphetamine are actually increased in the nucleus accumbens [187]. Levels of dopamine D1 and D2 receptors are unaltered in most brain regions in maternally deprived or handled rats compared to unhandled controls, including the cortex and striatum [178]. Thus, although some maternal deprivation procedures seem to produce consistent effects on responses to drugs of abuse, the responses across paradigms are variable suggesting that procedural factors, particularly those that alter the nature of the deprivation experience, may account for what appear to be conflicting results, as suggested previously [98], and as discussed recently [188]. Indeed, those authors have found that lengthy maternal deprivation produces a leftward shift in the dose response curve for cocaine self-administration, while brief maternal separation substantially reduces self-administration, compared to non-handled controls [189]. In many of the experiments described above pups were separated from the dam in groups, and repeatedly throughout the neonatal period. By contrast, a procedure that involves isolation from both the dam and littermates at early postnatal ages, and consequently may be more stressful, increases cocaine self-administration [190, 191]. However, this procedure does not affect other behavioral responses to cocaine [190], and increases ventral striatal dopamine release in response to cocaine [192]. Artificial rearing, which also combines maternal and sibling isolation increases incentive salience in a conditioned approach task, increases sucrose consumption and the conditioned place preference produced by morphine [193, 194]. Other authors have suggested that maternal deprivation produces differences in responses to cocaine that are observable in adolescence, but not adulthood [195].

Maternal deprivation can also produce impairments in prepulse inhibition of acoustic startle (PPI) [196], but does not always do so, depending on strain and other factors [197199], e.g. genetic background, and subsequent experience. Maternal deprivation can even reverse PPI deficits induced by subsequent isolation rearing [200]. Notably, the effects of maternal deprivation are affected by subsequent stress, perhaps due to the sensitized stress mechanisms in these animals [201], effects that include altered expression of serotonin transporter and tryptophan hydroxylase 2 mRNA in the dorsal raphe [202, 203]. Although the effects of maternal deprivation on PPI are equivocal, it facilitates latent inhibition (LI) [204], while, by contrast, post-weaning social isolation and prenatal stress do not affect LI but do impair PPI [204, 205]. Furthermore, Lehmann et al. (2000) found that maternal deprivation normalizes the impairments of PPI produced by prenatal stress, while prenatal stress normalizes the effects of maternal deprivation on LI. Obviously, equating the consequences of these experiences solely to stress is a gross oversimplification.

It might be supposed that long-term changes in HPA axis function are simply a response to elevated HPA activity produced by the deprivation experience; however, the changes in glucocorticoid receptor feedback that occur in the hippocampus are dependent upon both thyroid hormones and serotonin [206208]. Serotonin has been implicated in the effects of maternal deprivation in rodents [209] and primates [210]. When assessed in adults maternal deprivation decreases hippocampal serotonin levels [182], elevates hippocampal 5-HT1A and 5-HT1B receptor mRNA levels [211], elevates 5-HT1A and serotonin transporter levels in several brain regions [212] and produces other alterations in serotonergic function [211, 213]. Although, neonatal handling induces serotonin release, this only occurs in regions that later demonstrate decreases in both glucocorticoid and serotonin 5-HT2 receptor binding [209]. Supporting the contention that maternal deprivation may constitute an animal model of depression, chronic desipramine reverses some of the physiological consequences of maternal deprivation, as well as increased in hippocampal 5-HT1B levels, but interestingly, not the changes in hippocampal glucocorticoid receptors [211].

Many of the effects of maternal deprivation are thought to be the result of altered maternal interactions, although some authors have argued that the effects of maternal deprivation on maternal care and changes HPA function can be dissociated [214]. Those authors have proposed that two aspects of maternal deprivation paradigms, alterations in maternal care and stress, determine stress phenotypes in these paradigms [215]. Nonetheless, rat strains that exhibit differences in maternal behavior exhibit effects similar to enforced maternal deprivation, as cross-fostering experiments have demonstrated in Fisher and Lewis rats [216]. Pups of either strain raised by Fisher dams were more active in a novel environment and more responsive to the locomotor stimulant effects of d-amphetamine, and this was related to increased levels of arched-back nursing in Fisher dams. Similarly, maternal care is negatively correlated with ethanol and cocaine preference in adulthood [217], although interestingly in this study both maternal deprivation groups actually had greater maternal care than controls, consistent with Macri et al. (2004), who showed temporal changes in the pattern of maternal care. Natural variation in maternal care produces identical phenotypes to those observed after maternal deprivation and maternal handling [218], and paradigms that alter foraging requirements produce alterations in maternal care [219]. Furthermore, as was previously demonstrated for primates, rats that had experienced maternal deprivation as pups show deficits in maternal behavior in adulthood [220, 221]. Not surprisingly, complete maternal and sibling deprivation (artificial rearing; AR) produces profound impairments in social learning that require olfactory cues about conspecifics [161]. Artificial rearing also impairs maternal behavior when AR rats have offspring, an effect that can be reversed by simulating “licking” during the AR experience [222], as can some of the other sequelae of AR [223]. Other aspects of the deprivation experience also contribute to these behavioral changes, including exposure to maternal odors and social stimulation from littermates [224], auditory stimulation from the mother [124] and prolactin in milk [225].

Deprivation of social play in rodents

Many studies of environmental enrichment in rodents confound the sequelae of social deprivation and reduced environmental complexity, although some studies have shown separate contributions of each to the sequelae of environmental enrichment paradigms. Although social isolation paradigms can also be confounded with non-social environmental effects (see [226]), such paradigms more specifically assess the consequences of deprivation of social contact in rodents under standard (e.g. non-enriched) housing conditions. The most common procedure involves permanent isolation from weaning (isolation-rearing). Prior to weaning, at about 21 days of age, rodent behavior is directly largely towards the dam, while after weaning rodent behavior is directed primarily at same-sex conspecifics [227]. The most characteristic type of social interaction at this age is rough-and-tumble play which is replaced by adult types of social interactions by about 50 days of age. Adolescent animals are highly motivated to engage in this type of play. When otherwise deprived of contact with other adolescent animals, subjects allowed a period of time to interact with another adolescent animal will engage largely in this behavior [228], which is highly motivated [229]. Indeed, socially isolated weanling rats will even choose social interaction over food when food-deprived. Short play experiences can mitigate the effects of isolation [230]. Isolated animals will exhibit preferences for places paired with social interactions, effects that are greater in adolescents than adults [231]. The fact that play itself is rewarding has substantial importance for understanding the neurodevelopmental sequelae of play and conversely, deprivation of play. In this regard, it is interesting to note that males, which exhibit more pronounced play behavior, may be more susceptible to the effects of isolation-rearing [232]. However, it must be noted that aspects of non-physical contact also contribute to the effects of isolation rearing [233].

Since many consequences of isolation rearing are thought to be due to deprivation of play experience, it is interesting to note that the consequences of some other procedures that produce similar effects may also be associated with changes in social play. For instance, the effects of prenatal stress and environmental enrichment on HPA axis function may be related to their effects on social play in adolescent rats [63], and rhesus monkeys raised with a surrogate played less with age-mates than mother-raised monkeys [67], raising the possibility that some of the effects observed in those experiments are the result of deprivation of social play. These examples illustrate the difficulty of separating causality in neurodevelopment.

Play likely sub-serves multiple purposes, including the development of dominance hierarchies, which develop in early adolescence when rough-and-tumble play dominates their social interactions. Rough-and-tumble play involves mock-fighting, or wrestling, among similarly aged young mice, with winners that chase or pin the losers, that run away or get pinned. One of the interesting aspects of this process is that the winners and losers become more consistent over time until they assume distinct phenotypes, distinct enough that we categorize them with those names, dominant and subordinate. Serotonin plays a role in this developmental process. Serotonin lesions enhance both dominant and subordinate responses [234], perhaps accelerating this process towards more adult type behavior, while fluoxetine treatment does the opposite [235].

When adolescent animals are isolated at weaning they do not go through this process; thus, the immediate question would be, in the absence of this normal process of socialization, what phenotype results? One hallmark of isolation rearing is increased weight gain [236], which might be associated with dominance, as dominant rats typically weigh more than subordinate animals. Another prototypical effect of isolation rearing that could be associated with a dominant phenotype is enhanced aggression [237], although it must be noted that isolation in adulthood (isolation housing) increases aggression as well (e.g. [238]). However, the behavior of isolation-reared rats is different from normal dominant individuals and distinctly aberrant in several respects from animals reared under social conditions, even if isolated as adults [239]. One of the first findings in isolation-reared rats was muricide [240], which is rarely seen in non-isolated rats. Aggression is also observed in isolated mice [237, 241] under conditions that do not normally evoke aggression in socially housed mice. One of the results of the development of dominant and subordinate traits is reduction of intraspecies aggression, indeed this is one of the reasons for the development of these traits. Isolation rearing impairs the development of these behaviors and the ability to recognize social signals, although this is certainly a process that begins with imprinting and social interaction, including maternal interaction and imprinting, at an early period (see discussion by [242]).

Isolation rearing has been suggested to alter the trajectory of the development of adult-like behavioral flexibility and exploratory behavior [243]. Thus, most behavioral consequences of isolation rearing are not simply interpretable in terms of a dominant-like phenotype - but appear to involve increased responsiveness to a variety of environmental stimuli. The most consistently replicated consequence of isolation rearing that has been observed is locomotor hyperactivity in a novel environment [48, 244247]. Novelty is an essential component, and isolation-reared rats have increased exploratory tendencies [248], or perhaps reduced habituation as their locomotor behavior is characterized by a shift towards more repetitive, straight, distance-covering movements characteristic of initial exploratory responses [249]. Based on a simple comparison between isolation rearing induced hyperactivity and stimulant-induced locomotion, isolation rearing was hypothesized to produce a hyperdopaminergic state, which has been confirmed by a variety of findings. Pharmacological studies have consistently found increased responses to both direct and indirect dopamine agonists in a variety of circumstances [48, 50, 237, 250260]. In some cases isolation rearing has been shown to alter responses to amphetamine while maternal deprivation does not [204]. Isolation-reared rats exhibit a variety of behavioral characteristics associated with enhanced dopaminergic function [244, 250, 261, 262]. Based on this evidence for dopaminergic hyperactivity, the effect of isolation rearing on PPI was examined [246, 263]. Isolation-reared rats have impaired PPI, similar to that observed in schizophrenia or after the administration of dopaminergic agonists. Enhanced dopamine function in the nucleus accumbens is the critical mediator of these effects as 6-OHDA lesions of the nucleus accumbens ameliorate PPI deficits in isolation reared rats [264]. Furthermore, these deficits in PPI can be reversed by drugs with established or suspected antipsychotic efficacy [263, 265268]. Although latent inhibition was not found to be affected by isolation rearing from weaning [246], isolation rearing beginning at postnatal day 28 may do so [269]. Behavioral findings indicative of enhanced dopamine function are supported by in vivo microdialysis studies demonstrating increased reactivity of ventral and dorsal striatal dopamine systems to indirect dopamine agonists [246, 257, 270, 271] and conditioned stimuli [272]. There are strain differences in these effects [271], notably Sprague-Dawley rats are less sensitive to the effects of isolation rearing, that are also observed for differences in tissue dopamine levels [273]. Importantly, the effects of isolation rearing on PPI are permanent, and not reversed by later social housing [274] nor are they observed in animals isolated as adults [246].

One way to consider some of the changes discussed above are that they represent a generally enhanced responsiveness to certain environmental stimuli, perhaps over others, which would make sense from an evolutionary sense since such animals, more than those in social groups, would have to respond to all threats and identify all resources necessary to survive. In this light, it is not surprising that isolation-reared animals show enhanced responses to other classes of stimuli. Isolation-reared rats exhibit enhanced anxiety-like responses in most standard rodent anxiety models [275282] and are more emotionally reactive [283]. Similarly, although isolation-reared rats exhibit a preference for a novel environment, this is dependent on the aversiveness of the environment [284], and similar effects are observed in anxiety tests [285, 286], and account for apparent learning deficits in Stone’s Maze [287]. Although there is evidence that long-term isolation is stressful [275, 288], unlike maternal deprivation, examination of basal HPA axis function in isolation-reared animals has produced inconsistent results [275, 289, 290] and HPA axis activation does not appear to be different in most studies ([291293] but see [294]). Although less well characterized than changes in serotonergic and dopaminergic systems, there may also be changes in gabaergic mechanisms and neuroactive steroids (for review see [295]) as well as cannabinoid systems, which may be relevant to these effects [296]. Isolation-rearing may affect responses to other chronic stressors however [297, 298]. At least some effects of isolation-rearing on anxiety, and perhaps associated impairments in learning, may be more related to acute housing state, as these effects are at least partially reversible by later social/enriched housing, and isolation of social/enriched reared animals have the opposite effects [299]. Isolation-rearing is also associated with depressive-like behavior [282], while environmental enrichment has opposite effects [300303], and some of these effects are reversed by antidepressant treatments. The effects of isolation on anxiety and depressive phenotypes may be more related to acute housing state as both the behavioral effects and associated reductions in hypothalamic α-melanocyte stimulating hormone were reversed by resocialization [282].

Just as the consequences of isolation-rearing contrast sharply with those of maternal deprivation, they also contrast sharply with those of environmental enrichment. Although in some circumstances isolation-rearing has been found to produce impairments in learning [7, 41, 287, 304308], many of these effects appear to be the result of other consequences of isolation-rearing that indirectly affect learning, such as increased anxiety, and indeed increased learning is observed in some paradigms [309, 310]. Some of these learning impairments may reflect more specific changes in cognition that result from isolation-rearing rather than the apparently broad cognitive enhancing effects produced by environmental enrichment. For instance, isolation-reared rats are not impaired on acquisition of either a spatial or a non-spatial visual discrimination, nor reversal learning using an intra-dimensional shift, but are substantially impaired at switching selective attention from previously relevant perceptual dimensions to previously irrelevant perceptual dimensions, that is between spatial and non-spatial visual cues [311]. Selective impairments of extra-dimensional set shifting are produced by 6-OHDA lesions of prefrontal cortex [312], which produce elevations in striatal dopamine similar to those observed in isolation-rearing. Reversal learning is impaired by isolation rearing, and these deficits are ameliorated by clozapine [313]. Several aspects of behavioral choice appear to be affected by isolation rearing. Isolation-reared rats also do not inhibit responding during extinction, or when sated prior to testing, like socially reared rats [308, 314316]. Motor impulsivity in a go/no-go task is not affected by isolation rearing, but cognitive impulsivity in a delayed reinforcement task is reduced [317]. Similarly, Dalley and colleagues (2002) found increased perseverative behavior in the 5-choice serial reaction time task in isolation-reared rats; there were no differences in impulsive behavior, except after amphetamine administration which increased impulsive behavior in socially reared rats, but not in isolation-reared rats.

As mentioned above, the effects of isolation rearing on prefrontal cortex function may be quite different from other dopamine terminal areas. Tissue levels of dopamine metabolites are reduced in the prefrontal cortex [318, 319] as are dopamine fiber densities in isolation-reared gerbils (meriones unguiculatus) [320] consistent with the original suggestion by Blanc et al. (1980) that reductions in prefrontal cortex dopamine function is associated with increases in striatal dopaminergic function. Jones et al., (1992) also found changes in dopamine laterality in the prefrontal cortex of isolation-reared rats, which suggests alterations in mesocorticolimbic inputs which seems to be confirmed by electrophysiological results [321]. An in vivo microdialysis study in mice found that basal prefrontal DA levels are higher in isolation-reared subjects, but DA release stimulated by K+ or a 5-HT1A agonist is reduced [237]. Another study failed to find basal differences in the prefrontal cortex, but isolation-reared increased the ability of atypical antipsychotics to increase prefrontal dopamine release [322], effects suggested to be mediated by 5-HT1A receptors. There is also a reduction in calbindin-D28k expression and dendritic atrophy in prefronto-cortical pyramidal cells [323] indicative of broader changes in prefrontal function. Antipsychotics, including clozapine and olanzapine, can increase dopamine release in the prefrontal cortex and this effect is enhanced in isolation-reared rats [324]. However, this may involve their affinity for serotonin receptors, which have also been implicated in the effects of isolation rearing.

By contrast to studies demonstrating increased presynaptic dopamine function in isolation-reared rats, postsynaptic function, particularly dopamine D2 receptor function, has often been found to be reduced. Pharmacological responses to directly acting postsynaptic agonists and antagonists are reduced [252, 270, 325, 326], as are D2 mRNA levels in throughout the striatum [327], D2 binding in the striatum [328] and cAMP responses to D2 stimulation in the nucleus accumbens [271], despite an increased proportion of high-affinity D2 receptors [329]. These postsynaptic changes are thought to be compensatory changes produces by elevated dopamine release. Hyperdopaminergic function in isolation-reared rats has been suggested to constitute and animal model of schizophrenia [1], and likely involves altered interactions with the prefrontal cortex, amygdala and hippocampus. Indeed basal c-fos levels are increased in the hippocampus, but decreased in the amygdala [330], an effect that parallels a shift in sensitivity aversive conditioning to contextual and discrete stimuli in trace conditioning [331]. Levels of the presynaptic protein CDCrel-1, which has been associated with schizophrenia [332], are reduced in the striatum of isolation-reared rats, but increased in the hippocampus [333], as are synaptophysin levels and the high correlation between synaptophysin and CDCrel-1 levels observed in socially reared rats is reduced in isolation-reared rats. Levels of n-acetylaspartate, reductions of which are also observed in schizophrenia [334], are also reduced in the temporal cortex of isolated rats [335].

While certain behavioral differences in isolation-reared rats are consistent with elevated mesolimbic dopamine function, others are more consistent with reduced serotonin function. Post-mortem studies have found evidence for reduced serotonin turnover in isolation-reared rats [270, 336, 337], reductions in serotonin synthesis [338, 339], and reduced tissue levels of serotonin [303, 340]. Jaffe et al. (1993) also found reduced hippocampal serotonin release ex vivo in hippocampal tissue slices. In vivo microdialysis experiments have also demonstrated reduced serotonin release in the hippocampus and prefrontal cortex [341343]. These results are consistent with reduced immunohistochemical staining for serotonergic terminals in the hippocampus [344], although other brain regions may have increased serotonin levels [345], at least in gerbils. Changes in presynaptic serotonin function, like other neurochemical changes in isolation-reared rats, may be highly regionally dependent however. Aversive stimuli and exposure to conditioned stimuli produce increases in serotonin release in the nucleus accumbens that are not observed in socially reared rats [346]. This would seem to indicate that changes in serotonin function are not simply a matter of globally reduced serotonin function, but reflect a shift in the types of responses affected by serotonin in different brain areas, perhaps also mediated by different serotonin receptor subtypes. Consistent with this interpretation, changes in postsynaptic serotonin receptor function are also observed in isolation-reared rats, including increased density of the 5-HT2A receptor, reductions in the 5-HT1A receptor [337, 347] and 5-HT1A mediated dopamine release in the prefrontal cortex [348] and increased sensitivity to some postsynaptic serotonin agonists [349, 350]. Dorsal raphe 5-HT1A receptor function is enhanced [351].

An important consideration for the effects of isolation rearing, as for other neurodevelopmental experiences, is the extent to which these effects are linked to specific developmental periods in which the isolation occurs, which has been demonstrated for a number of the behavioral consequences discussed above [243, 352]. There may be differential effects of isolation on prepubertal and postpubertal adolescent development on some of these behavioral effects [353]. With regard to the specificity of the effects of isolation rearing, it is interesting to note that some other early manipulations, such as stress during the post-weaning period [354], reduce social play and social investigation, which may actually underlie some of the effects of “stress” during this period. By contrast daily handling reverses some of the effects of isolation-rearing [355], but not others [271].

The best approach to specifying the neurodevelopmental window for isolation-rearing effects would be to isolate the subjects for different periods of time during adolescence and then rehouse them socially to determine if the effects are permanent. Although this has not been done very extensively, there are some illuminating examples. An early study of this type failed to find a “developmental window” for the effects of isolation-rearing on PPI, with only complete post-weaning isolation being effective [356]. However, isolating adolescent rats from postnatal days 30 to 35, the peak period of social play, produced immediate sex-dependent behavioral changes at postnatal day 36, associated with reductions in synaptic markers in the prefrontal cortex [357]. Assessment in adulthood found that this experience produces long-lasted reductions in synaptic markers in the infralimbic prefrontal cortex and cingulate gyrus [358]. In a procedure in which rats were isolated for 3 weeks at weaning, and then rehoused socially for 2 weeks, persistent effects of isolation on anxiety and fear-like behavior were observed [359], effects that were associated with CRF receptor alterations in the dorsal raphe that prolonged CRF induced serotonin release in the nucleus accumbens [360, 361]. A 2 week period of isolation produced increases in the number of vasoactive intestinal peptide immunoreactive neurons in the medial prefrontal cortex, along with reductions in the arborizations of pyramidal neurons, which were not reversed by resocialization [362].

The mechanisms of gene-environment interactions

The previous sections have briefly summarized the long history of accumulating evidence that several specific types of experience, and in particular social interactions and learning experiences that have circumscribed developmental windows, resulting in critical periods for neurobehavioral developmental. As suggested by Hofer [98] these experiences act as neurodevelopmental regulators that produce alternative behavioral and psychological phenotypes. It would appear that the range of these possibilities is genetically predetermined, based on the interaction of experience with mechanisms controlling gene expression; that is, for all of these experiences, activation of sets of genes by transcription factors presumably produce permanent epigenetic reprogramming of the organism. As discussed below, the genes that are affected and the epigenetic mechanisms involved are just starting to be elucidated. Furthermore, although some of these experiences are of the more mundane “use it or lose it” type, many of these experiences influence neurobehavioral development much more broadly. Indeed, this may in fact be a raison d’être for the existence of many of these behaviors, to act as neurodevelopmental regulators, as it is just this flexibility of phenotype, from generation to generation, in the face of changing environmental demands, that explains the evolutionary benefit that they convey. Furthermore, such mechanisms may be of critical importance for understanding those mechanisms that explain the evolution of certain characteristics of species that produce only a few offspring in each generation (e.g. mammals, and in particular primates), relative to those mechanisms that explain the evolution of characteristics of species that produce large numbers of offspring in each generation (e.g. insects); given this major difference in rate of population increase the solutions to problems that exert evolutionary pressures may be quite different. With current advancements in genomics and epigenetics it should be possible to identify the set of epigenetic programs that control each of these neurodevelopmental processes. This final section discusses the first small steps in that direction.

Epigenetic Mechanisms

The original definition of “epigenetic” mechanisms involved inherited differences in DNA structure (from one organism to its offspring or within a cell lineage) that did not involve changes in DNA sequence; such changes include alterations in CpG methylation, chromatin structure and transcriptional activity (among others, see [363]), as well as the recent recognition of the effects of microRNA on gene expression [364]. In part because of the understanding of some of the mechanisms involved the term “epigenetics” has come to have a broader meaning, of “the combination of mechanisms that confers long-term programming to genes leading to a change in gene function without a change in gene sequence” [365]. An important aspect of the machinery that produces changes in methylation and chromatin restructuring is that many of the proteins involved are relatively non-specific in their actions, but are recruited to particular gene locations by transcription factors [366]. Because DNA methylation alters the covalent structure of the DNA [367], this makes this mechanism particularly interesting as regards long-term changes in gene regulation resulting from differential experience early in life. Thus, it has recently been suggested that “programmed changes in DNA methylation have evolved to serve as an interface between the dynamic environment and the static genome” [368], that much of the inter-individual variation in many phenotypes that is not accounted for by sequence variation may be the result of these epigenetic mechanisms [369], but that under certain circumstances this can be maladaptive [370, 371]. Nonetheless, in many cases authors continue to emphasize stress as the primary mediator of these effects (e.g [372, 373], even when substantial data suggests that many of these effects may be mediated by other aspects of the situations involved – it is not the absence of the experience that does the regulation, but rather the experience. In any case for many experiences, such as maternal deprivation, it remains to be seen what portion of the permanent consequences of deprivation are due to stress, and what are due to alterations in other types of social and non-social interactions with the dam. However, the description of regulation of epigenetic markers during development by Szyf and colleagues, particularly in response to social stimuli, as a “system-wide adaptation of the DNA methylation pattern to an anticipated environment” should apply to many types of “neurodevelopmental regulators” as was described by Hofer [98].

Environmental Enrichment

Superficially it might be supposed that the mechanisms of environmental enrichment result solely from a simple perspective of “use it or lose it”. That is, simply as a matter of conservation of resources that each animal preserves systems that are active and eliminates systems that are inactive in order to maximize efficiency of energy use. Indeed, at about the time the effects of environmental enrichment were being first described, a mechanism that preserves active neuronal connections was discovered: neurotrophic factors [374]. Such a mechanism explains the survival of active cells and differences in synaptic contacts. However, such mechanisms do not necessarily explain the broader consequences of environmental enrichment. One of the areas of the cerebral cortex that is most consistently affected, and which has the greatest magnitude of response to environmental enrichment, is the visual cortical area of the occipital cortex. However, environmental enrichment induces increases in the weight of the occipital cortex even in blinded rats [375]. Thus, visual input alone does not mediate the changes observed in this region.

Surprisingly few studies have addressed changes in gene expression resulting from environmental enrichment. Consistent with the role of neurotrophins in shaping cell survival environmental enrichment induces earlier expression of brain derived neurotrophic factor (BDNF) and glutamic acid decarboxylase (GAD) 65/67, which has been associated with accelerated visual system development [376]. Environmental enrichment also induces the transcription factor NGFI-A [377], which is a transcription factor induced by NGF [378], as well as the transcription factors Arc, c-fos, and zif-268 in the cerebral cortex and hippocampus [379, 380], and ΔFosB in the striatum [381]. A study using a transgenic strain that coupled activation of fos to a reporter gene found that a single exposure to enrichment induced fos in diverse brain regions [382], although this is no doubt a transient signal and only the first in a series of events leading to permanent epigenetic changes. In the nucleus accumbens the phosphorylated (transcriptionally active) form of the transcription factor cyclic adenosine monophosphate response element binding protein (pCREB) is reduced in EC rats compared to IC rats, which was linked to reduced cocaine self-administration and other behavioral effects of enrichment [53]. Those authors also showed that some of the apparent effects of enrichment could be reproduced by administration of a short hairpin RNA via a viral vector that down-regulated pCREB. This, like many other studies of environmental enrichment did not dissociate the effects of enrichment and social deprivation, so it is uncertain which neurodevelopmental regulator mediates these effects. However, it seems likely that a large proportion of the effects are due to social isolation, particularly given the similarity of the effects on cocaine administration to those of Howes et al. [257] in isolation-reared rats.

Even with the limited data currently available it would appear that the affects of environmental enrichment involve diverse transcription factors that activate diverse classes of genes. So far, the genes and transcription factors that have been analyzed were based on a priori assumptions about their importance, so it remains to be seen what a more comprehensive and unbiased genome-wide approach would find. Potential epigenetic mechanisms involved in the effects of environmental enrichment are just starting to be considered. Environmental enrichment has been shown to increase histone acetylation and methylation in the hippocampus and cerebral cortex, which was associated with improved learning and memory in a neurodegenerative model [383]. Early, but not later environmental enrichment also ameliorated the effects of a transgenic Rett Syndrome model involving MeCP2, a gene involved in epigenetic modifications [384, 385].

Isolation-rearing

The permanent behavioral changes that results from isolation-rearing have been linked to particular neurotransmitter systems and particular brain regions. Although no comprehensive analysis of transcription factors has been performed, some of these neurochemical and neuroanatomical changes have been linked to particular transcription factors such as c-fos. Thus, cocaine-induced FOS immunoreactivity in the amygdala, nucleus accumbens and striatum is greatly enhanced by isolation rearing [257], while stress-induced reactivity is enhanced in specific subregions of the frontal cortex, dorsal hippocampus, periaqueductal gray and amygdala [343]. No comprehensive examination of gene expression changes after isolation rearing has been conducted but one recent study found reduced expression of numerous serotonin receptor subtypes in the prefrontal cortex of isolation reared mice, including 5-HT1A, 5-HT1B, 5-HT2A, 5-HT2C, 5-HT3A, 5-HT6 and 5-HT7, while more restricted reductions were observed in the hypothalamus and midbrain, and no differences except for up-regulation of 5-HT6 gene expression were found in the hippocampus [386]. The full meaning of these changes remain to be seen, but they are exceptionally striking, as well as consistent with the results of a variety of other approaches that have examined serotonin function in isolation reared rodents. In isolated prairie voles (microtus ochrogaster) isolation-rearing induces anxiety, as it does in other rodents species, which is associated with increased mRNA expression for vasopressin, oxytocin, corticotrophin releasing factor and tyrosine hydroxylase in the paraventricular nucleus of the hypothalamus [387].

Neither more comprehensive gene expression analyses nor examination of epigenetic mechanisms have been applied to isolation rearing models.

Maternal Deprivation

The epigenetic mechanisms underlying experiential regulation of behavioral phenotypes has been most examined with regards to maternal deprivation. As discussed earlier, the hippocampal feedback to the HPA axis is regulated during development by maternal behavior, leading to alterations in the duration of HPA axis activity in response to stressors. Although some of these developmental consequences of maternal deprivation may be related directly to glucocorticoid receptor activity, broader changes are also suggested by the involvement of neurotrophic factors, such as the transient increase in hippocampal expression of nerve growth factor (NGF) mRNA after 45 minutes of deprivation [388], and transcription factors cFOS and NGFI-B in the in PVN [137]. Some gene expression changes are only observed after longer or repeated periods of deprivation, including changes in CRF mRNA in the periventricular nucleus [137, 138], CRF2 receptor mRNA in the ventromedial nucleus [389] and arginine vasopressin in the PVN [373]. In adult rats that were maternally deprived long-lasting changes in BDNF are observed in the hippocampus, striatum and prefrontal cortex [390], and both BDNF and glial derived neurotrophic factor (GDNF) levels are reduced in adult mice that were maternally deprived [391]. Longer-term changes may go in the opposite direction from acute changes as increases in hypothalamic CRF mRNA levels are observed long term [153, 163, 165, 171]. Many of these gene expression changes are relatively permanent, including reduced glucocorticoid receptor mRNA, reduced hippocampal BDNF and NMDA receptor subunit mRNA, and increased arginine vasopressin mRNA [163, 172, 373]. These broader changes in gene expression may also be driven by changes in endocrine function. Repeated early (postnatal day 1 to 10) maternal deprivation increases pre-opiomelanocortin mRNA in the pituitary [373], which appeared to be independent of changes in glucocorticoid receptor expression.

Thus, maternal deprivation, like other experiences, produces a sequence of gene expression changes. The initial changes are short-lasting, but if they are sufficiently strong, prolonged or repeated, they induce induction of other genes that are permanently affected. It would then appear that the key to understanding the consequences of maternal deprivation would be the mechanisms by which such experiences activate transcription factors that produce long-term changes in gene expression, and the subsequent consequences on cell function and morphology. Indeed, a series of studies has shown that levels of maternal behavior (specifically related to the amount of maternal licking and grooming) mediate the pattern of behavioral effects associated with maternal deprivation by altering DNA methylation patterns, histone acetylation and binding of NGFI-A to the glucocorticoid receptor gene promotor [392, 393], and presumably other genes as well. Maternal deprivation produces a number of behavioral changes, so it is not surprising that epigenetic changes associated with maternal care affect the transcription of several hundred genes [394], including others that may be affected by other transcription factors, such as estrogen receptor alpha [395, 396], and clusters of protocadherin genes involved in synaptogenesis [397]. Changes in glucocorticoid receptor expression in the hippocampus have been specifically linked to a sequence of cell signaling events that include thyroid-hormone dependent increases in serotonin 5-HT7 receptors, increased activation of cAMP-dependent protein kinase A, induction NGFI-A and binding of both NGFI-A and CBP to the glucocorticoid receptor promotor [207, 398400]. Differences in arginine vasopressin gene expression after repeated maternal deprivation have been specifically associated with reduced methylation of a promoter region of the AVP gene involved in the binding of methyl CpG binding protein 2 (MeCP2), in part mediated by CAMKII activity dependent phosphorylation of MeCP2 [373]. Interestingly, MeCP2 also affects the transcription of BDNF [401]. Importantly these methylation patterns, consequent changes in glucocorticoid receptor expression and behavior could be reversed pharmacologically by treatment with histone deacetylase inhibitors or methionine in adulthood [392, 394, 402], raising the possibility that even relatively permanent consequences of epigenetic modifications of gene expression may be open to pharmacological intervention. Finally, some allelic variation association with neurodevelopmental phenotypes may act by modifying epigenetic modifications. For instance, serotonin gene polymorphisms may exert their effects by altering neurodevelopmental regulation of gene expression by early life experience, as has been suggested by recent studies in rats [403], primates [404], and humans [405].

Summary and Conclusions

This review has reviewed several, but certainly not all, circumstances in which specific types of experience, in particular social experience, during defined critical periods in life, produce permanent changes in behavior associated with alterations in brain chemistry, anatomy and function. This is far from a complete list of such experiences, but includes some of the most well characterized ones, although certainly each of these could be further divided into component aspects that may have independent mechanisms. The seminal studies of Harlow and colleagues in primates, and the studies of Meaney and colleagues that elaborated the effects of maternal behavior on HPA axis function in rodents, have now lead to an elegant series of studies by Meaney, Szyf and colleagues that have elaborated the precise mechanism underlying the epigenetic mechanism underlying alterations in the HPA axis by maternal interaction. Indeed, Meaney has suggested that these mechanisms are adaptive, and that evolution may have shaped the genome to produce intergenerational transmission of traits via the epigenome to better fit organisms to the circumstances of their lives (the situations of offspring are likely to be similar to that of their parents) [406]. The review here of the effects of alterations in early life experiences certainly supports this view, even without an understanding of the epigenome. These models, and now their underlying epigenetic basis, have been linked to psychiatric outcomes, e.g. early life adversity (child abuse) has been linked to DNA methylation patterns in rRNA [407] and the glucocorticoid receptor gene in the hippocampus [408]. All the same, even if some of the ultimate consequences are deleterious, the initial neurodevelopmentally determined behavioral outcomes are likely to be highly adaptive. Further elaboration of the epigenetic basis of neurodevelopmental mechanisms will lead toward an understanding of this fundamental issue surrounding the effects of early life experience and the importance of gene-environment interactions in determining behavioral and psychological characteristics.

Highlights for Review.

  • Early experience, including visual experience, environmental enrichment, maternal and adolescent peer interactions produce permanent changes in behavior

  • The effects of early experience a mediated by particular regulators (e.g. maternal licking and grooming or play)

  • The effects of different types of experience are distinct, but interactive

  • Changes in brain function, chemistry and anatomy accompany this distinct behavioral changes

  • These changes in brain function are mediated by epigenetic programs

Acknowledgements

This work was supported by funding from the Intramural Research Program of the National Institute on Drug Abuse (USA).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Hall FS. Social deprivation of neonatal, adolescent, and adult rats has distinct neurochemical and behavioral consequences. Crit Rev Neurobiol. 1998;12:129–162. doi: 10.1615/critrevneurobiol.v12.i1-2.50. [DOI] [PubMed] [Google Scholar]
  • 2.Hubel DH, Wiesel TN. Receptive Fields Of Cells In Striate Cortex Of Very Young, Visually Inexperienced Kittens. J Neurophysiol. 1963;26:994–1002. doi: 10.1152/jn.1963.26.6.994. [DOI] [PubMed] [Google Scholar]
  • 3.Hubel DH, Wiesel TN. Effects Of Monocular Deprivation In Kittens. Naunyn Schmiedebergs Arch Exp Pathol Pharmakol. 1964;248:492–497. doi: 10.1007/BF00348878. [DOI] [PubMed] [Google Scholar]
  • 4.Wiesel TN, Hubel DH. Single-Cell Responses In Striate Cortex Of Kittens Deprived Of Vision In One Eye. J Neurophysiol. 1963;26:1003–1017. doi: 10.1152/jn.1963.26.6.1003. [DOI] [PubMed] [Google Scholar]
  • 5.Wiesel TN, Hubel DH. Effects Of Visual Deprivation On Morphology And Physiology Of Cells In The Cats Lateral Geniculate Body. J Neurophysiol. 1963;26:978–993. doi: 10.1152/jn.1963.26.6.978. [DOI] [PubMed] [Google Scholar]
  • 6.Levi-Montalcini R, Angeletti PU. Essential role of the nerve growth factor in the survival and maintenance of dissociated sensory and sympathetic embryonic nerve cells in vitro. Dev Biol. 1963;7:653–659. doi: 10.1016/0012-1606(63)90149-0. [DOI] [PubMed] [Google Scholar]
  • 7.Krech D, Rosenzweig MR, Bennett EL. Relations between chemistry and problem-solving among rats raised in enriched and impoverished environments. J Comp Physiol Psychol. 1962;55:801–807. doi: 10.1037/h0044220. [DOI] [PubMed] [Google Scholar]
  • 8.Bennett EL, Diamond MC, Krech D, Rosenzweig MR. Chemical And Anatomical Plasticity Brain. Science. 1964;146:610–619. doi: 10.1126/science.146.3644.610. [DOI] [PubMed] [Google Scholar]
  • 9.Rosenzweig MR, Krech D, Bennett EL, Diamond MC. Effects of environmental complexity and training on brain chemistry and anatomy: a replication and extension. J Comp Physiol Psychol. 1962;55:429–437. doi: 10.1037/h0041137. [DOI] [PubMed] [Google Scholar]
  • 10.Zolman JF, Morimoto H. Cerebral changes related to duration of environmental complexity and locomotor activity. J Comp Physiol Psychol. 1965;60:382–387. doi: 10.1037/h0022547. [DOI] [PubMed] [Google Scholar]
  • 11.Bennett EL, Rosenzweig MR, Diamond MC. Rat brain: effects of environmental enrichment on wet and dry weights. Science. 1969;163:825–826. doi: 10.1126/science.163.3869.825. [DOI] [PubMed] [Google Scholar]
  • 12.Diamond MC, Krech D, Rosenzweig MR. The Effects Of An Enriched Environment On The Histology Of The Rat Cerebral Cortex. J Comp Neurol. 1964;123:111–120. doi: 10.1002/cne.901230110. [DOI] [PubMed] [Google Scholar]
  • 13.Diamond MC, Law F, Rhodes H, Lindner B, Rosenzweig MR, Krech D, et al. Increases in cortical depth and glia numbers in rats subjected to enriched environment. J Comp Neurol. 1966;128:117–126. doi: 10.1002/cne.901280110. [DOI] [PubMed] [Google Scholar]
  • 14.Diamond MC, Lindner B, Raymond A. Extensive Cortical Depth Measurements and Neuron Size Increases in the Cortex of Environmentally Enrished Rats. J Comp Neurol. 1967;131:357–364. [Google Scholar]
  • 15.Diamond MC, Lindner B, Johnson R, Bennett EL, Rosenzweig MR. Differences in occipital cortical synapses from environmentally enriched, impoverished, and standard colony rats. J Neurosci Res. 1975;1:109–119. doi: 10.1002/jnr.490010203. [DOI] [PubMed] [Google Scholar]
  • 16.Globus A, Rosenzweig MR, Bennett EL, Diamond MC. Effects of differential experience on dendritic spine counts in rat cerebral cortex. J Comp Physiol Psychol. 1973;82:175–181. doi: 10.1037/h0033910. [DOI] [PubMed] [Google Scholar]
  • 17.Mollgaard K, Diamond MC, Bennett EL, Rosenzweig MR, Lindner B. Quantitative synaptic changes with differential experience in rat brain. Int J Neurosci. 1971;2:113–127. doi: 10.3109/00207457109148764. [DOI] [PubMed] [Google Scholar]
  • 18.Sirevaag AM, Greenough WT. Differential rearing effects on rat visual cortex synapses. II. Synaptic morphometry. Brain Res. 1985;351:215–226. doi: 10.1016/0165-3806(85)90193-2. [DOI] [PubMed] [Google Scholar]
  • 19.Sirevaag AM, Greenough WT. Differential rearing effects on rat visual cortex synapses. III. Neuronal and glial nuclei, boutons, dendrites, and capillaries. Brain Res. 1987;424:320–332. doi: 10.1016/0006-8993(87)91477-6. [DOI] [PubMed] [Google Scholar]
  • 20.Sirevaag AM, Greenough WT. A multivariate statistical summary of synaptic plasticity measures in rats exposed to complex, social and individual environments. Brain Res. 1988;441:386–392. doi: 10.1016/0006-8993(88)91420-5. [DOI] [PubMed] [Google Scholar]
  • 21.Turner AM, Greenough WT. Differential rearing effects on rat visual cortex synapses. I. Synaptic and neuronal density and synapses per neuron. Brain Res. 1985;329:195–203. doi: 10.1016/0006-8993(85)90525-6. [DOI] [PubMed] [Google Scholar]
  • 22.Wallace CS, Kilman VL, Withers GS, Greenough WT. Increases in dendritic length in occipital cortex after 4 days of differential housing in weanling rats. Behav Neural Biol. 1992;58:64–68. doi: 10.1016/0163-1047(92)90937-y. [DOI] [PubMed] [Google Scholar]
  • 23.Sirevaag AM, Black JE, Shafron D, Greenough WT. Direct evidence that complex experience increases capillary branching and surface area in visual cortex of young rats. Brain Res. 1988;471:299–304. doi: 10.1016/0165-3806(88)90107-1. [DOI] [PubMed] [Google Scholar]
  • 24.Rosenzweig MR, Bennett EL, Krech D. Cerebral Effects Of Environmental Complexity And Training Among Adult Rats. J Comp Physiol Psychol. 1964;57:438–439. doi: 10.1037/h0046387. [DOI] [PubMed] [Google Scholar]
  • 25.Lehmann K, Teuchert-Noodt G, Dawirs RR. Postnatal rearing conditions influence ontogeny of adult dopamine transporter (DAT) immunoreactivity of the striatum in gerbils. J Neural Transm. 2002;109:1129–1137. doi: 10.1007/s00702-002-0765-9. [DOI] [PubMed] [Google Scholar]
  • 26.Lehmann K, Lesting J, Polascheck D, Teuchert-Noodt G. Serotonin fibre densities in, subcortical areas: differential effects of isolated rearing and methamphetamine. Dev Brain Res. 2003;147:143–152. doi: 10.1016/s0165-3806(03)00130-5. [DOI] [PubMed] [Google Scholar]
  • 27.Zhu J, Green T, Bardo MT, Dwoskin LP. Environmental enrichment enhances sensitization to GBR 12935-induced activity and decreases dopamine transporter function in the medial prefrontal cortex. Behavioural Brain Research. 2004;148:107–117. doi: 10.1016/s0166-4328(03)00190-6. [DOI] [PubMed] [Google Scholar]
  • 28.Zhu J, Apparsundaram S, Bardo MT, Dwoskin LP. Environmental enrichment decreases cell surface expression of the dopamine transporter in rat medial prefrontal cortex. J Neurochem. 2005;93:1434–1443. doi: 10.1111/j.1471-4159.2005.03130.x. [DOI] [PubMed] [Google Scholar]
  • 29.Lesting J, Neddens J, Busche A, Teuchert-Noodt G. Hemisphere-specific effects on serotonin but not dopamine innervation in the nucleus accumbens of gerbils caused by isolated rearing and a single early methamphetamine challenge. Brain Research. 2005;1035:168–176. doi: 10.1016/j.brainres.2004.12.005. [DOI] [PubMed] [Google Scholar]
  • 30.Melendez RI, Gregory ML, Bardo MT, Kalivas PW. Impoverished rearing environment alters metabotrophic glutamate receptor expression and function in the prefrontal cortex. Neuropsychopharmacology. 2004;29:1980–1987. doi: 10.1038/sj.npp.1300507. [DOI] [PubMed] [Google Scholar]
  • 31.Solinas M, Thiriet N, Chauvet C, Jaber M. Environmental Enrichment and Drug Addiction. Behav Pharmacol. 2010;21:583-. doi: 10.1016/j.pneurobio.2010.08.002. [DOI] [PubMed] [Google Scholar]
  • 32.Young D, Lawlor PA, Leone P, Dragunow M, During MJ. Environmental enrichment inhibits spontaneous apoptosis, prevents seizures and is neuroprotective. Nat Med. 1999;5:448–453. doi: 10.1038/7449. [DOI] [PubMed] [Google Scholar]
  • 33.Diamond MC, Rosenzweig MR, Bennett EL, Lindner B, Lyon L. Effects of environmental enrichment and impoverishment on rat cerebral cortex. J Neurobiol. 1972;3:47–64. doi: 10.1002/neu.480030105. [DOI] [PubMed] [Google Scholar]
  • 34.Rosenzweig MR, Bennett EL. Cerebral changes in rats exposed individually to an enriched environment. J Comp Physiol Psychol. 1972;80:304–313. doi: 10.1037/h0032978. [DOI] [PubMed] [Google Scholar]
  • 35.Rosenzweig MR, Krech D, Bennett EL, Zolman JF. Variation in environmental complexity and brain measures. J Comp Physiol Psychol. 1962;55:1092–1095. doi: 10.1037/h0042758. [DOI] [PubMed] [Google Scholar]
  • 36.Rosenzweig MR, Bennett EL, Diamond MC. Effects of differential environments on brain anatomy and brain chemistry. Proc Annu Meet Am Psychopathol Assoc. 1967;56:45–56. [PubMed] [Google Scholar]
  • 37.Bennett EL, Rosenzweig MR, Diamond MC, Morimoto H, Hebert M. Effects of successive environments on brain measures. Physiol Behav. 1974;12:621–631. doi: 10.1016/0031-9384(74)90212-1. [DOI] [PubMed] [Google Scholar]
  • 38.Amaral OB, Vargas RS, Hansel G, Izquierdo I, Souza DO. Duration of environmental enrichment influences the magnitude and persistence of its behavioral effects on mice. Physiol Behav. 2008;93:388–394. doi: 10.1016/j.physbeh.2007.09.009. [DOI] [PubMed] [Google Scholar]
  • 39.Freeman BJ, Ray OS. Strain, sex, and environment effects on appetitively and aversively motivated learning tasks. Dev Psychobiol. 1972;5:101–109. doi: 10.1002/dev.420050203. [DOI] [PubMed] [Google Scholar]
  • 40.Gutwein BM, Fishbein W. Paradoxical sleep and memory (I): Selective alterations following enriched and impoverished environmental rearing. Brain Res Bull. 1980;5:9–12. doi: 10.1016/0361-9230(80)90276-2. [DOI] [PubMed] [Google Scholar]
  • 41.Juraska JM, Henderson C, Muller J. Differential rearing experience, gender, and radial maze performance. Dev Psychobiol. 1984;17:209–215. doi: 10.1002/dev.420170302. [DOI] [PubMed] [Google Scholar]
  • 42.Bourgeon S, Xerri C, Coq JO. Abilities in tactile discrimination of textures in adult rats exposed to enriched or impoverished environments. Behav Brain Res. 2004;153:217–231. doi: 10.1016/j.bbr.2003.12.002. [DOI] [PubMed] [Google Scholar]
  • 43.Schrijver NC, Pallier PN, Brown VJ, Wurbel H. Double dissociation of social and environmental stimulation on spatial learning and reversal learning in rats. Behav Brain Res. 2004;152:307–314. doi: 10.1016/j.bbr.2003.10.016. [DOI] [PubMed] [Google Scholar]
  • 44.Greenough WT, Wood WE, Madden TC. Possible memory storage differences among mice reared in environments varying in complexity. Behav Biol. 1972;7:717–722. doi: 10.1016/s0091-6773(72)80078-6. [DOI] [PubMed] [Google Scholar]
  • 45.Widman DR, Rosellini RA. Restricted daily exposure to environmental enrichment increases the diversity of exploration. Physiol Behav. 1990;47:57–62. doi: 10.1016/0031-9384(90)90042-3. [DOI] [PubMed] [Google Scholar]
  • 46.Brillaud E, Morillion D, de Seze R. Modest environmental enrichment: effect on a radial maze validation and well being of rats. Brain Res. 2005;1054:174–182. doi: 10.1016/j.brainres.2005.06.069. [DOI] [PubMed] [Google Scholar]
  • 47.Lehmann K, Grund T, Bagorda A, Bagorda F, Grafen K, Winter Y, et al. Developmental effects on dopamine projections and hippocampal cell proliferation in the rodent model of postweaning social and physical deprivation can be triggered by brief changes of environmental context. Behav Brain Res. 2009;205:26–31. doi: 10.1016/j.bbr.2009.07.016. [DOI] [PubMed] [Google Scholar]
  • 48.Smith JK, Neill JC, Costall B. Post-weaning housing conditions influence the behavioural effects of cocaine and d-amphetamine. Psychopharmacology (Berl) 1997;131:23–33. doi: 10.1007/s002130050261. [DOI] [PubMed] [Google Scholar]
  • 49.Bardo MT, Bowling SL, Rowlett JK, Manderscheid P, Buxton ST, Dwoskin LP. Environmental enrichment attenuates locomotor sensitization, but not in vitro dopamine release, induced by amphetamine. Pharmacol Biochem Behav. 1995;51:397–405. doi: 10.1016/0091-3057(94)00413-d. [DOI] [PubMed] [Google Scholar]
  • 50.Bardo MT, Klebaur JE, Valone JM, Deaton C. Environmental enrichment decreases intravenous self-administration of amphetamine in female and male rats. Psychopharmacology (Berl) 2001;155:278–284. doi: 10.1007/s002130100720. [DOI] [PubMed] [Google Scholar]
  • 51.Stairs DJ, Klein ED, Bardo MT. Effects of environmental enrichment on extinction and reinstatement of amphetamine self-administration and sucrose-maintained responding. Behav Pharmacol. 2006;17:597–604. doi: 10.1097/01.fbp.0000236271.72300.0e. [DOI] [PubMed] [Google Scholar]
  • 52.Rahman S, Bardo MT. Environmental enrichment increases amphetamine-induced glutamate neurotransmission in the nucleus accumbens: a neurochemical study. Brain Res. 2008;1197:40–46. doi: 10.1016/j.brainres.2007.12.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Green TA, Alibhai IN, Roybal CN, Winstanley CA, Theobald DE, Birnbaum SG, et al. Environmental enrichment produces a behavioral phenotype mediated by low cyclic adenosine monophosphate response element binding (CREB) activity in the nucleus accumbens. Biol Psychiatry. 2010;67:28–35. doi: 10.1016/j.biopsych.2009.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Bardo MT, Valone JM, Robinet PM, Shaw WB, Dwoskin LP. Environmental enrichment enhances the stimulant effect of intravenous amphetamine: Seach for a cellular mechanism in the nucleus accumbens. Psychobiology. 1999;27:292–299. [Google Scholar]
  • 55.Bowling SL, Rowlett JK, Bardo MT. The effect of environmental enrichment on amphetamine-stimulated locomotor activity, dopamine synthesis and dopamine release. Neuropharmacology. 1993;32:885–893. doi: 10.1016/0028-3908(93)90144-r. [DOI] [PubMed] [Google Scholar]
  • 56.Perry JL, Stairs DJ, Bardo MT. Impulsive choice and environmental enrichment: effects of d-amphetamine and methylphenidate. Behav Brain Res. 2008;193:48–54. doi: 10.1016/j.bbr.2008.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Beckmann JS, Bardo MT. Environmental enrichment reduces attribution of incentive salience to a food-associated stimulus. Behav Brain Res. 2012;226:331–334. doi: 10.1016/j.bbr.2011.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bowling SL, Bardo MT. Locomotor and rewarding effects of amphetamine in enriched, social, and isolate reared rats. Pharmacol Biochem Behav. 1994;48:459–464. doi: 10.1016/0091-3057(94)90553-3. [DOI] [PubMed] [Google Scholar]
  • 59.Gipson CD, Beckmann JS, El-Maraghi S, Marusich JA, Bardo MT. Effect of environmental enrichment on escalation of cocaine self-administration in rats. Psychopharmacology (Berl) 2011;214:557–566. doi: 10.1007/s00213-010-2060-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Solinas M, Chauvet C, Thiriet N, El Rawas R, Jaber M. Reversal of cocaine addiction by environmental enrichment. Proc Natl Acad Sci U S A. 2008;105:17145–17150. doi: 10.1073/pnas.0806889105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Thiel KJ, Pentkowski NS, Peartree NA, Painter MR, Neisewander JL. Environmental living conditions introduced during forced abstinence alter cocaine-seeking behavior and Fos protein expression. Neuroscience. 2010;171:1187–1196. doi: 10.1016/j.neuroscience.2010.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Thiel KJ, Engelhardt B, Hood LE, Peartree NA, Neisewander JL. The interactive effects of environmental enrichment and extinction interventions in attenuating cue-elicited cocaine-seeking behavior in rats. Pharmacol Biochem Behav. 2011;97:595–602. doi: 10.1016/j.pbb.2010.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Morley-Fletcher S, Rea M, Maccari S, Laviola G. Environmental enrichment during adolescence reverses the effects of prenatal stress on play behaviour and HPA axis reactivity in rats. Eur J Neurosci. 2003;18:3367–3374. doi: 10.1111/j.1460-9568.2003.03070.x. [DOI] [PubMed] [Google Scholar]
  • 64.Laviola G, Rea M, Morley-Fletcher S, Di Carlo S, Bacosi A, De Simone R, et al. Beneficial effects of enriched environment on adolescent rats from stressed pregnancies. Eur J Neurosci. 2004;20:1655–1664. doi: 10.1111/j.1460-9568.2004.03597.x. [DOI] [PubMed] [Google Scholar]
  • 65.Harlow HF. The nature of love. Am Psychol. 1958;13:673–685. [Google Scholar]
  • 66.Harlow HF, Zimmermann RR. Affectional responses in the infant monkey; orphaned baby monkeys develop a strong and persistent attachment to inanimate surrogate mothers. Science. 1959;130:421–432. doi: 10.1126/science.130.3373.421. [DOI] [PubMed] [Google Scholar]
  • 67.Harlow HF, Harlow M. Learning to love. Am Sci. 1966;54:244–272. [PubMed] [Google Scholar]
  • 68.Garattini S, Valzelli L. Is the isolated animal a possible model for phobia and anxiety? Prog. Neuropsychopharmacol. 1981;5:159–165. doi: 10.1016/0364-7722(81)90065-5. [DOI] [PubMed] [Google Scholar]
  • 69.Fone KCF, Porkess MV. Behavioural and neurochemical effects of post-weaning social isolation in rodents - Relevance to developmental neuropsychiatric disorders. Neurosci Biobehav R. 2008;32:1087–1102. doi: 10.1016/j.neubiorev.2008.03.003. [DOI] [PubMed] [Google Scholar]
  • 70.Matthews K, Robbins TW. Early experience as a determinant of adult behavioural responses to reward: the effects of repeated maternal separation in the rat. Neurosci Biobehav Rev. 2003;27:45–55. doi: 10.1016/s0149-7634(03)00008-3. [DOI] [PubMed] [Google Scholar]
  • 71.Andersen SL, Teicher MH. Desperately driven and no brakes: developmental stress exposure and subsequent risk for substance abuse. Neurosci Biobehav Rev. 2009;33:516–524. doi: 10.1016/j.neubiorev.2008.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Harlow HF, Harlow MK. The affectional systems. In: Schrier AM, Harlow HF, Stollnitz F, editors. Behavior of nonhuman primates. New York: Academic Press; 1965. pp. 287–334. [Google Scholar]
  • 73.Furchner CS, Harlow HF. Preference for various surrogate surfaces among infant rhesus monkeys. Psychonomic Sci. 1969;17:279–280. [Google Scholar]
  • 74.Harlow HF, Suomi SJ. Nature of love--simplified. Am Psychol. 1970;25:161–168. doi: 10.1037/h0029383. [DOI] [PubMed] [Google Scholar]
  • 75.Meyer JS, Novak MA, Bowman RE, Harlow HF. Behavioral and hormonal effects of attachment object separation in surrogate-peer-reared and mother-reared infant rhesus monkeys. Dev Psychobiol. 1975;8:425–435. doi: 10.1002/dev.420080507. [DOI] [PubMed] [Google Scholar]
  • 76.Harlow HF, Harlow MK, Suomi SJ. From thought to therapy: lessons from a primate laboratory. Am Sci. 1971;59:538–549. [PubMed] [Google Scholar]
  • 77.Seay B, Hansen E, Harlow HF. Mother-infant separation in monkeys. J Child Psychol Psychiatry. 1962;3:123–132. doi: 10.1111/j.1469-7610.1962.tb02047.x. [DOI] [PubMed] [Google Scholar]
  • 78.Seay B, Harlow HF. Maternal separation in the rhesus monkey. J Nerv Ment Dis. 1965;140:434–441. doi: 10.1097/00005053-196506000-00006. [DOI] [PubMed] [Google Scholar]
  • 79.Suomi SJ, Collins ML, Harlow HF, Ruppenthal GC. Effects of maternal and peer separations on young monkeys. J Child Psychol Psychiatry. 1976;17:101–112. doi: 10.1111/j.1469-7610.1976.tb00382.x. [DOI] [PubMed] [Google Scholar]
  • 80.Suomi SJ, Harlow HF. Monkeys at play. Nat Hist. 1971;80:72–76. [Google Scholar]
  • 81.Suomi SJ, Harlow HF. Effects of differential removal from group on social development of Rhesus monkeys. J Child Psychol Psychiatry. 1975;16:149–164. doi: 10.1111/j.1469-7610.1975.tb01264.x. [DOI] [PubMed] [Google Scholar]
  • 82.Harlow HF, Dodsworth RO, Harlow MK. Total social isolation in monkeys. Proc Natl Acad Sci U S A. 1965;54:90–97. doi: 10.1073/pnas.54.1.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Harlow HF, Suomi SJ. Social recovery by isolation-reared monkeys. Proc Natl Acad Sci U S A. 1971;68:1534–1538. doi: 10.1073/pnas.68.7.1534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.McKinney WT, Jr, Suomi SJ, Harlow HF. Depression in primates. Am J Psychiatry. 1971;127:1313–1320. doi: 10.1176/ajp.127.10.1313. [DOI] [PubMed] [Google Scholar]
  • 85.Mears CE, Harlow HF. Play: early and eternal. Proc Natl Acad Sci U S A. 1975;72:1878–1882. doi: 10.1073/pnas.72.5.1878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Harlow HF, Lauersdorf HE. Sex differences in passion and play. Perspect Biol Med. 1974;17:348–360. doi: 10.1353/pbm.1974.0054. [DOI] [PubMed] [Google Scholar]
  • 87.Gluck JP, Otto MW, Beauchamp AJ. Respondent conditioning of self-injurious behavior in early socially deprived rhesus monkeys (Macaca mulatta) J Abnorm Psychol. 1985;94:222–226. doi: 10.1037//0021-843x.94.2.222. [DOI] [PubMed] [Google Scholar]
  • 88.Seay B, Alexander BK, Harlow HF. Maternal Behavior Of Socially Deprived Rhesus Monkeys. J Abnorm Psychol. 1964;69:345–354. doi: 10.1037/h0040539. [DOI] [PubMed] [Google Scholar]
  • 89.Arling GL, Harlow HF. Effects of social deprivation on maternal behavior of rhesus monkeys. J Comp Physiol Psychol. 1967;64:371–377. doi: 10.1037/h0025221. [DOI] [PubMed] [Google Scholar]
  • 90.Ruppenthal GC, Arling GL, Harlow HF, Sackett GP, Suomi SJ. A 10-year perspective of motherless-mother monkey behavior. J Abnorm Psychol. 1976;85:341–349. doi: 10.1037//0021-843x.85.4.341. [DOI] [PubMed] [Google Scholar]
  • 91.Higham JP, Barr CS, Hoffman CL, Mandalaywala TM, Parker KJ, Maestripieri D. Mu-opioid receptor (OPRM1) variation, oxytocin levels and maternal attachment in free-ranging rhesus macaques Macaca mulatta. Behav Neurosci. 2011;125:131–136. doi: 10.1037/a0022695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Suomi SJ, Harlow HF, McKinney WT., Jr Monkey psychiatrists. Am J Psychiatry. 1972;128:927–932. doi: 10.1176/ajp.128.8.927. [DOI] [PubMed] [Google Scholar]
  • 93.Gluck JP. Successive acquisition and extinction of barpressing: The effects of differential rearing in monkeys. Madison: University of Wisconsin; 1970. [Google Scholar]
  • 94.Griffin GA, Harlow HF. Effects of three months of total social deprivation on social adjustment and learning in the rhesus monkey. Child Dev. 1966;37:533–547. [PubMed] [Google Scholar]
  • 95.Harlow HF, Schiltz MA, Harlow MK. Second International Congress of Primatology. Basel: Kargen; 1969. Effects of social isolation on the learning performance of rhesus monkeys. [Google Scholar]
  • 96.Corcoran SBE, Howell LL. Impact of early life stress on the reinforcing and behavioral-stimulant effects of psychostimulants in rhesus monkeys. Behav Pharmacol. 2010;21:69–76. doi: 10.1097/FBP.0b013e3283359f53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Nader MA, Czoty PW. PET imaging of dopamine D2 receptors in monkey models of cocaine abuse: Genetic predisposition versus environmental modulation. Am J Psychiat. 2005;162:1473–1482. doi: 10.1176/appi.ajp.162.8.1473. [DOI] [PubMed] [Google Scholar]
  • 98.Hofer MA. Early relationships as regulators of infant physiology and behavior. Acta Paediatr Suppl. 1994;397:9–18. doi: 10.1111/j.1651-2227.1994.tb13260.x. [DOI] [PubMed] [Google Scholar]
  • 99.Richardson R, Siegel MA, Campbell BA. Effect of maternal presence on the fear response to an unfamiliar environment as measured by heart rate in rats as a function of age. Dev Psychobiol. 1988;21:613–633. doi: 10.1002/dev.420210702. [DOI] [PubMed] [Google Scholar]
  • 100.Hofer MA, Reiser MF. The development of cardiac rate regulation in preweanling rats. Psychosom Med. 1969;31:372–388. doi: 10.1097/00006842-196909000-00003. [DOI] [PubMed] [Google Scholar]
  • 101.Hofer MA. Physiological responses of infant rats to separation from their mothers. Science. 1970;168:871–873. doi: 10.1126/science.168.3933.871. [DOI] [PubMed] [Google Scholar]
  • 102.Hofer MA. The effects of brief maternal separations on behavior and heart rate of two week old rat pups. Physiol Behav. 1973;10:423–427. doi: 10.1016/0031-9384(73)90200-x. [DOI] [PubMed] [Google Scholar]
  • 103.Hofer MA, Shair HN. Isolation distress in two-week-old rats: influence of home cage, social companions, and prior experience with littermates. Dev Psychobiol. 1987;20:465–476. doi: 10.1002/dev.420200410. [DOI] [PubMed] [Google Scholar]
  • 104.Hofer MA. The role of nutrition in the physiological and behavioral effects of early maternal separation on infant rats. Psychosom Med. 1973;35:350–359. doi: 10.1097/00006842-197307000-00009. [DOI] [PubMed] [Google Scholar]
  • 105.Hofer MA. Maternal separation affects infant rats' behavior. Behav Biol. 1973;9:629–633. doi: 10.1016/s0091-6773(73)80057-4. [DOI] [PubMed] [Google Scholar]
  • 106.Hofer MA, Weiner H. Development and mechanisms of cardiorespiratory responses to maternal deprivation in rat pups. Psychosom Med. 1971;33:353–362. doi: 10.1097/00006842-197107000-00005. [DOI] [PubMed] [Google Scholar]
  • 107.Hofer MA, Shair H. Control of sleep-wake states in the infant rat by features of the mother-infant relationship. Dev Psychobiol. 1982;15:229–243. doi: 10.1002/dev.420150307. [DOI] [PubMed] [Google Scholar]
  • 108.Lau C, Cameron AM, Antolick LL, Stanton ME. Repeated maternal separation in the neonatal rat: cellular mechanisms contributing to brain growth sparing. J Dev Physiol. 1992;17:265–276. [PubMed] [Google Scholar]
  • 109.Kuhn CM, Butler SR, Schanberg SM. Selective depression of serum growth hormone during maternal deprivation in rat pups. Science. 1978;201:1034–1036. doi: 10.1126/science.684424. [DOI] [PubMed] [Google Scholar]
  • 110.Hofer MA. Early stages in the organization of cardiovascular control. Proc Soc Exp Biol Med. 1984;175:147–157. doi: 10.3181/00379727-175-41780. [DOI] [PubMed] [Google Scholar]
  • 111.Katz LM, Nathan L, Kuhn CM, Schanberg SM. Inhibition of GH in maternal separation may be mediated through altered serotonergic activity at 5-HT2A and 5-HT2C receptors. Psychoneuroendocrinology. 1996;21:219–235. doi: 10.1016/0306-4530(95)00043-7. [DOI] [PubMed] [Google Scholar]
  • 112.Penke Z, Felszeghy K, Fernette B, Sage D, Nyakas C, Burlet A. Postnatal maternal deprivation produces long-lasting modifications of the stress response, feeding and stress-related behaviour in the rat. Eur J Neurosci. 2001;14:747–755. doi: 10.1046/j.0953-816x.2001.01691.x. [DOI] [PubMed] [Google Scholar]
  • 113.Hofer MA, Shair H. Ultrasonic vocalization during social interaction and isolation in 2-week-old rats. Dev Psychobiol. 1978;11:495–504. doi: 10.1002/dev.420110513. [DOI] [PubMed] [Google Scholar]
  • 114.Blumberg MS, Efimova IV, Alberts JR. Ultrasonic vocalizations by rat pups: the primary importance of ambient temperature and the thermal significance of contact comfort. Dev Psychobiol. 1992;25:229–250. doi: 10.1002/dev.420250402. [DOI] [PubMed] [Google Scholar]
  • 115.Allin JT, Banks EM. Effects of temperature on ultrasound production by infant albino rats. Dev Psychobiol. 1971;4:149–156. doi: 10.1002/dev.420040206. [DOI] [PubMed] [Google Scholar]
  • 116.Oswalt GL, Meier GW. Olfactory, thermal, and tactual influences on infantile ultrasonic vocalization in rats. Dev Psychobiol. 1975;8:129–135. doi: 10.1002/dev.420080205. [DOI] [PubMed] [Google Scholar]
  • 117.Hofer MA, Shair H. Ultrasonic vocalization during social interaction and isolation in 2-weeek-old rats. Dev Psychobiol. 1978;11:495–504. doi: 10.1002/dev.420110513. [DOI] [PubMed] [Google Scholar]
  • 118.Hofer MA, Shair H. Sensory processes in the control of isolation-induced ultrasonic vocalization by 2-week-old rats. J Comp Physiol Psychol. 1980;94:271–279. doi: 10.1037/h0077665. [DOI] [PubMed] [Google Scholar]
  • 119.Shoemaker WJ, Kehoe P. Effect of isolation conditions on brain regional enkephalin and beta-endorphin levels and vocalizations in 10-day-old rat pups. Behav Neurosci. 1995;109:117–122. doi: 10.1037//0735-7044.109.1.117. [DOI] [PubMed] [Google Scholar]
  • 120.Kehoe P, Blass EM. Opioid-mediation of separation distress in 10-day-old rats: reversal of stress with maternal stimuli. Dev Psychobiol. 1986;19:385–398. doi: 10.1002/dev.420190410. [DOI] [PubMed] [Google Scholar]
  • 121.Blass EM, Shide DJ, Zaw-Mon C, Sorrentino J. Mother as shield: differential effects of contact and nursing on pain responsivity in infant rats--evidence for nonopioid mediation. Behav Neurosci. 1995;109:342–353. doi: 10.1037//0735-7044.109.2.342. [DOI] [PubMed] [Google Scholar]
  • 122.Winslow JT, Insel TR. Endogenous opioids: do they modulate the rat pup's response to social isolation? Behav Neurosci. 1991;105:253–263. doi: 10.1037//0735-7044.105.2.253. [DOI] [PubMed] [Google Scholar]
  • 123.Kehoe P, Shoemaker W. Opioid-dependent behaviors in infant rats: effects of prenatal exposure to ethanol. Pharmacol Biochem Behav. 1991;39:389–394. doi: 10.1016/0091-3057(91)90197-a. [DOI] [PubMed] [Google Scholar]
  • 124.Ziabreva I, Schnabel R, Poeggel G, Braun K. Mother's voice "buffers" separation-induced receptor changes in the prefrontal cortex of octodon degus. Neuroscience. 2003;119:433–441. doi: 10.1016/s0306-4522(03)00123-4. [DOI] [PubMed] [Google Scholar]
  • 125.Braun K, Kremz P, Wetzel W, Wagner T, Poeggel G. Influence of parental deprivation on the behavioral development in Octodon degus: modulation by maternal vocalizations. Dev Psychobiol. 2003;42:237–245. doi: 10.1002/dev.10096. [DOI] [PubMed] [Google Scholar]
  • 126.Kuhn CM, Pauk J, Schanberg SM. Endocrine responses to mother-infant separation in developing rats. Dev Psychobiol. 1990;23:395–410. doi: 10.1002/dev.420230503. [DOI] [PubMed] [Google Scholar]
  • 127.Sapolsky RM, Meaney MJ. Maturation of the adrenocortical stress response: neuroendocrine control mechanisms and the stress hyporesponsive period. Brain Res. 1986;396:64–76. doi: 10.1016/s0006-8993(86)80190-1. [DOI] [PubMed] [Google Scholar]
  • 128.Stanton ME, Gutierrez YR, Levine S. Maternal deprivation potentiates pituitary-adrenal stress responses in infant rats. Behav Neurosci. 1988;102:692–700. doi: 10.1037//0735-7044.102.5.692. [DOI] [PubMed] [Google Scholar]
  • 129.Viau V, Sharma S, Meaney MJ. Changes in plasma adrenocorticotropin, corticosterone, corticosteroid-binding globulin, and hippocampal glucocorticoid receptor occupancy/translocation in rat pups in response to stress. J Neuroendocrinol. 1996;8:1–8. doi: 10.1111/j.1365-2826.1996.tb00680.x. [DOI] [PubMed] [Google Scholar]
  • 130.Suchecki D, Nelson DY, Van Oers H, Levine S. Activation and inhibition of the hypothalamic-pituitary-adrenal axis of the neonatal rat: effects of maternal deprivation. Psychoneuroendocrinology. 1995;20:169–182. doi: 10.1016/0306-4530(94)00051-b. [DOI] [PubMed] [Google Scholar]
  • 131.Avishai-Eliner S, Yi SJ, Newth CJ, Baram TZ. Effects of maternal and sibling deprivation on basal and stress induced hypothalamic-pituitary-adrenal components in the infant rat. Neurosci Lett. 1995;192:49–52. doi: 10.1016/0304-3940(95)11606-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Stanton ME, Wallstrom J, Levine S. Maternal contact inhibits pituitary-adrenal stress responses in preweanling rats. Dev Psychobiol. 1987;20:131–145. doi: 10.1002/dev.420200204. [DOI] [PubMed] [Google Scholar]
  • 133.Stanton ME, Levine S. Inhibition of infant glucocorticoid stress response: specific role of maternal cues. Dev Psychobiol. 1990;23:411–426. doi: 10.1002/dev.420230504. [DOI] [PubMed] [Google Scholar]
  • 134.Rosenfeld P, Gutierrez YA, Martin AM, Mallett HA, Alleva E, Levine S. Maternal regulation of the adrenocortical response in preweanling rats. Physiol Behav. 1991;50:661–671. doi: 10.1016/0031-9384(91)90001-5. [DOI] [PubMed] [Google Scholar]
  • 135.Stanton ME, Levine S. Maternal modulation of infant glucocorticoid stress response: role of age and maternal deprivation. Psychobiology. 1988;16:223–228. [Google Scholar]
  • 136.van Oers HJ, de Kloet ER, Levine S. Early vs. late maternal deprivation differentially alters the endocrine and hypothalamic responses to stress. Brain Res Dev Brain Res. 1998;111:245–252. doi: 10.1016/s0165-3806(98)00143-6. [DOI] [PubMed] [Google Scholar]
  • 137.Smith MA, Kim SY, van Oers HJ, Levine S. Maternal deprivation and stress induce immediate early genes in the infant rat brain. Endocrinology. 1997;138:4622–4628. doi: 10.1210/endo.138.11.5529. [DOI] [PubMed] [Google Scholar]
  • 138.Schmidt MV, Oitzl MS, Levine S, de Kloet ER. The HPA system during the postnatal development of CD1 mice and the effects of maternal deprivation. Brain Res Dev Brain Res. 2002;139:39–49. doi: 10.1016/s0165-3806(02)00519-9. [DOI] [PubMed] [Google Scholar]
  • 139.Vazquez DM, Van Oers H, Levine S, Akil H. Regulation of glucocorticoid and mineralocorticoid receptor mRNAs in the hippocampus of the maternally deprived infant rat. Brain Res. 1996;731:79–90. doi: 10.1016/0006-8993(96)00465-9. [DOI] [PubMed] [Google Scholar]
  • 140.Frisone DF, Frye CA, Zimmerberg B. Social isolation stress during the third week of life has age-dependent effects on spatial learning in rats. Behav Brain Res. 2002;128:153–160. doi: 10.1016/s0166-4328(01)00315-1. [DOI] [PubMed] [Google Scholar]
  • 141.Walker CD. Chemical sympathectomy and maternal separation affect neonatal stress responses and adrenal sensitivity to ACTH. Am J Physiol. 1995;268:R1281–R1288. doi: 10.1152/ajpregu.1995.268.5.R1281. [DOI] [PubMed] [Google Scholar]
  • 142.van Oers HJ, de Kloet ER, Li C, Levine S. The ontogeny of glucocorticoid negative feedback: influence of maternal deprivation. Endocrinology. 1998;139:2838–2846. doi: 10.1210/endo.139.6.6037. [DOI] [PubMed] [Google Scholar]
  • 143.Vazquez DM. Stress and the developing limbic-hypothalamic-pituitary-adrenal axis. Psychoneuroendocrinology. 1998;23:663–700. doi: 10.1016/s0306-4530(98)00029-8. [DOI] [PubMed] [Google Scholar]
  • 144.Levine S. Primary social relationships influence the development of the hypothalamic--pituitary--adrenal axis in the rat. Physiol Behav. 2001;73:255–260. doi: 10.1016/s0031-9384(01)00496-6. [DOI] [PubMed] [Google Scholar]
  • 145.Dent GW, Smith MA, Levine S. Rapid induction of corticotropin-releasing hormone gene transcription in the paraventricular nucleus of the developing rat. Endocrinology. 2000;141:1593–1598. doi: 10.1210/endo.141.5.7455. [DOI] [PubMed] [Google Scholar]
  • 146.Dent GW, Smith MA, Levine S. Stress-induced alterations in locus coeruleus gene expression during ontogeny. Brain Res Dev Brain Res. 2001;127:23–30. doi: 10.1016/s0165-3806(01)00108-0. [DOI] [PubMed] [Google Scholar]
  • 147.van Oers HJ, de Kloet ER, Whelan T, Levine S. Maternal deprivation effect on the infant's neural stress markers is reversed by tactile stimulation and feeding but not by suppressing corticosterone. J Neurosci. 1998;18:10171–10179. doi: 10.1523/JNEUROSCI.18-23-10171.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Zimmerberg B, Shartrand AM. Temperature-dependent effects of maternal separation on growth, activity, and amphetamine sensitivity in the rat. Dev Psychobiol. 1992;25:213–226. doi: 10.1002/dev.420250306. [DOI] [PubMed] [Google Scholar]
  • 149.Levine S, Lewis GW. Critical period for effects of infantile experience on maturation of stress response. Science. 1959;129:42–43. doi: 10.1126/science.129.3340.42. [DOI] [PubMed] [Google Scholar]
  • 150.Francis D, Diorio J, LaPlante P, Weaver S, Seckl JR, Meaney MJ. The role of early environmental events in regulating neuroendocrine development. Moms, pups, stress, and glucocorticoid receptors. Ann N Y Acad Sci. 1996;794:136–152. doi: 10.1111/j.1749-6632.1996.tb32517.x. [DOI] [PubMed] [Google Scholar]
  • 151.de Kloet ER, Rots NY, Cools AR. Brain-corticosteroid hormone dialogue: slow and persistent. Cell Mol Neurobiol. 1996;16:345–356. doi: 10.1007/BF02088100. [DOI] [PubMed] [Google Scholar]
  • 152.Meaney MJ, Aitken DH, Sharma S, Viau V. Basal ACTH, corticosterone and corticosterone-binding globulin levels over the diurnal cycle, and age-related changes in hippocampal type I and type II corticosteroid receptor binding capacity in young and aged, handled and nonhandled rats. Neuroendocrinology. 1992;55:204–213. doi: 10.1159/000126116. [DOI] [PubMed] [Google Scholar]
  • 153.Plotsky PM, Meaney MJ. Early, postnatal experience alters hypothalamic corticotropin-releasing factor (CRF) mRNA, median eminence CRF content and stress-induced release in adult rats. Brain Res Mol Brain Res. 1993;18:195–200. doi: 10.1016/0169-328x(93)90189-v. [DOI] [PubMed] [Google Scholar]
  • 154.Levine S. Plasma-free corticosteroid response to electric shock in rats stimulated in infancy. Science. 1962;135:795–796. doi: 10.1126/science.135.3506.795-a. [DOI] [PubMed] [Google Scholar]
  • 155.Meaney MJ, Aitken DH, Viau V, Sharma S, Sarrieau A. Neonatal handling alters adrenocortical negative feedback sensitivity and hippocampal type II glucocorticoid receptor binding in the rat. Neuroendocrinology. 1989;50:597–604. doi: 10.1159/000125287. [DOI] [PubMed] [Google Scholar]
  • 156.Zarrow MX, Campbell PS, Denenberg VH. Handling in infancy: increased levels of the hypothalamic corticotropin releasing factor (CRF) following exposure to a novel situation. Proc Soc Exp Biol Med. 1972;141:356–358. doi: 10.3181/00379727-141-36776. [DOI] [PubMed] [Google Scholar]
  • 157.Levine S, Haltmeyer GC, Karas GG, Denenberg VH. Physiological and behavioral effects of infantile stimulation. Physiol Behav. 1967;2:55. [Google Scholar]
  • 158.Meaney MJ, Aitken DH, Sharma S, Viau V, Sarrieau A. Postnatal handling increases hippocampal glucocorticoid receptors and enhances adrenocrotical negative-feedback efficacy in the rat. Neuroendocrinology. 1989;50:597. doi: 10.1159/000125287. [DOI] [PubMed] [Google Scholar]
  • 159.Meaney MJ, Aitken DH, Bodnoff SR, Iny LJ, Tatarewicz JE, Sapolsky RM. Early postnatal handling alters glucocorticoid receptor concentrations in selected brain regions. Behav Neurosci. 1985;99:765–770. doi: 10.1037//0735-7044.99.4.765. [DOI] [PubMed] [Google Scholar]
  • 160.Kosten TA, Karanian DA, Yeh J, Haile CN, Kim JJ, Kehoe P, et al. Memory impairments and hippocampal modifications in adult rats with neonatal isolation stress experience. Neurobiology of Learning and Memory. 2007;88:167–176. doi: 10.1016/j.nlm.2007.03.011. [DOI] [PubMed] [Google Scholar]
  • 161.Levy F, Melo AI, Galef G, Madden M, Fleming AS. Complete maternal deprivation affects social but not spatial learning in adult rats. Developmental Psychobiology. 2003;43:177–191. doi: 10.1002/dev.10131. [DOI] [PubMed] [Google Scholar]
  • 162.Huot RL, Thrivikraman KV, Meaney MJ, Plotsky PM. Development of adult ethanol preference and anxiety as a consequence of neonatal maternal separation in Long Evans rats and reversal with antidepressant treatment. Psychopharmacology (Berl) 2001;158:366–373. doi: 10.1007/s002130100701. [DOI] [PubMed] [Google Scholar]
  • 163.Francis DD, Diorio J, Plotsky PM, Meaney MJ. Environmental enrichment reverses the effects of maternal separation on stress reactivity. J Neurosci. 2002;22:7840–7843. doi: 10.1523/JNEUROSCI.22-18-07840.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Kalinichev M, Easterling KW, Plotsky PM, Holtzman SG. Long-lasting changes in stress-induced corticosterone response and anxiety-like behaviors as a consequence of neonatal maternal separation in Long-Evans rats. Pharmacol Biochem Behav. 2002;73:131–140. doi: 10.1016/s0091-3057(02)00781-5. [DOI] [PubMed] [Google Scholar]
  • 165.Ladd CO, Owens MJ, Nemeroff CB. Persistent changes in corticotropin-releasing factor neuronal systems induced by maternal deprivation. Endocrinology. 1996;137:1212–1218. doi: 10.1210/endo.137.4.8625891. [DOI] [PubMed] [Google Scholar]
  • 166.Wigger A, Neumann ID. Periodic maternal deprivation induces gender-dependent alterations in behavioral and neuroendocrine responses to emotional stress in adult rats. Physiol Behav. 1999;66:293–302. doi: 10.1016/s0031-9384(98)00300-x. [DOI] [PubMed] [Google Scholar]
  • 167.Aisa B, Tordera R, Lasheras B, Del Rio J, Ramirez MJ. Cognitive impairment associated to HPA axis hyperactivity after maternal separation in rats. Psychoneuroendocrinology. 2007;32:256–266. doi: 10.1016/j.psyneuen.2006.12.013. [DOI] [PubMed] [Google Scholar]
  • 168.Meaney MJ, Diorio J, Francis D, Widdowson J, LaPlante P, Caldji C, et al. Early environmental regulation of forebrain glucocorticoid receptor gene expression: implications for adrenocortical responses to stress. Dev Neurosci. 1996;18:49–72. doi: 10.1159/000111395. [DOI] [PubMed] [Google Scholar]
  • 169.Sutanto W, Rosenfeld P, de Kloet ER, Levine S. Long-term effects of neonatal maternal deprivation and ACTH on hippocampal mineralocorticoid and glucocorticoid receptors. Brain Res Dev Brain Res. 1996;92:156–163. doi: 10.1016/0165-3806(95)00213-8. [DOI] [PubMed] [Google Scholar]
  • 170.Rots NY, de Jong J, Workel JO, Levine S, Cools AR, De Kloet ER. Neonatal maternally deprived rats have as adults elevated basal pituitary-adrenal activity and enhanced susceptibility to apomorphine. J Neuroendocrinol. 1996;8:501–506. doi: 10.1046/j.1365-2826.1996.04843.x. [DOI] [PubMed] [Google Scholar]
  • 171.Husum H, Mathe AA. Early life stress changes concentrations of neuropeptide Y and corticotropin-releasing hormone in adult rat brain. Lithium treatment modifies these changes. Neuropsychopharmacology. 2002;27:756–764. doi: 10.1016/S0893-133X(02)00363-9. [DOI] [PubMed] [Google Scholar]
  • 172.Roceri M, Hendriks W, Racagni G, Ellenbroek BA, Riva MA. Early maternal deprivation reduces the expression of BDNF and NMDA receptor subunits in rat hippocampus. Mol Psychiatry. 2002;7:609–616. doi: 10.1038/sj.mp.4001036. [DOI] [PubMed] [Google Scholar]
  • 173.Maciag CM, Dent G, Gilligan P, He L, Dowling K, Ko T, et al. Effects of a non-peptide CRF antagonist (DMP696) on the behavioral and endocrine sequelae of maternal separation. Neuropsychopharmacology. 2002;26:574–582. doi: 10.1016/S0893-133X(01)00398-0. [DOI] [PubMed] [Google Scholar]
  • 174.Ogawa T, Mikuni M, Kuroda Y, Muneoka K, Mori KJ, Takahashi K. Periodic maternal deprivation alters stress response in adult offspring: potentiates the negative feedback regulation of restraint stress-induced adrenocortical response and reduces the frequencies of open field-induced behaviors. Pharmacol Biochem Behav. 1994;49:961–967. doi: 10.1016/0091-3057(94)90250-x. [DOI] [PubMed] [Google Scholar]
  • 175.Ryu V, Yoo SB, Kang DW, Lee JH, Jahng JW. Post-weaning isolation promotes food intake and body weight gain in rats that experienced neonatal maternal separation. Brain Research. 2009;1295:127–134. doi: 10.1016/j.brainres.2009.08.006. [DOI] [PubMed] [Google Scholar]
  • 176.Mourlon V, Baudin A, Blanc O, Lauber A, Giros B, Naudon L, et al. Maternal deprivation induces depressive-like behaviours only in female rats. Behavioural Brain Research. 2010;213:278–287. doi: 10.1016/j.bbr.2010.05.017. [DOI] [PubMed] [Google Scholar]
  • 177.Kalinichev M, Easterling KW, Holtzman SG. Early neonatal experience of Long-Evans rats results in long-lasting changes in reactivity to a novel environment and morphine-induced sensitization and tolerance. Neuropsychopharmacology. 2002;27:518–533. doi: 10.1016/S0893-133X(02)00326-3. [DOI] [PubMed] [Google Scholar]
  • 178.Ploj K, Roman E, Nylander I. Long-term effects of maternal separation on ethanol intake and brain opioid and dopamine receptors in male wistar rats. Neuroscience. 2003;121:787–799. doi: 10.1016/s0306-4522(03)00499-8. [DOI] [PubMed] [Google Scholar]
  • 179.Matthews K, Robbins TW, Everitt BJ, Caine SB. Repeated neonatal maternal separation alters intravenous cocaine self-administration in adult rats. Psychopharmacology (Berl) 1999;141:123–134. doi: 10.1007/s002130050816. [DOI] [PubMed] [Google Scholar]
  • 180.Matthews K, Hall FS, Wilkinson LS, Robbins TW. Retarded acquisition and reduced expression of conditioned locomotor activity in adult rats following repeated early maternal separation: effects of prefeeding, d-amphetamine, dopamine antagonists and clonidine. Psychopharmacology (Berl) 1996;126:75–84. doi: 10.1007/BF02246414. [DOI] [PubMed] [Google Scholar]
  • 181.Matthews K, Wilkinson LS, Robbins TW. Repeated maternal separation of preweanling rats attenuates behavioral responses to primary and conditioned incentives in adulthood. Physiol Behav. 1996;59:99–107. doi: 10.1016/0031-9384(95)02069-1. [DOI] [PubMed] [Google Scholar]
  • 182.Matthews K, Dalley JW, Matthews C, Tsai TH, Robbins TW. Periodic maternal separation of neonatal rats produces region- and gender-specific effects on biogenic amine content in postmortem adult brain. Synapse. 2001;40:1–10. doi: 10.1002/1098-2396(200104)40:1<1::AID-SYN1020>3.0.CO;2-E. [DOI] [PubMed] [Google Scholar]
  • 183.Shalev U, Kafkafi N. Repeated maternal separation does not alter sucrose-reinforced and open-field behaviors. Pharmacol Biochem Behav. 2002;73:115–122. doi: 10.1016/s0091-3057(02)00756-6. [DOI] [PubMed] [Google Scholar]
  • 184.Planeta CS, Marin MT. Effect of cocaine on periadolescent rats with or without early maternal separation. Brazilian Journal of Medical and Biological Research. 2002;35:1367–1371. doi: 10.1590/s0100-879x2002001100015. [DOI] [PubMed] [Google Scholar]
  • 185.Faure J, Stein DJ, Daniels W. Maternal separation fails to render animals more susceptible to methamphetamine-induced conditioned place preference. Metab Brain Dis. 2009;24:541–559. doi: 10.1007/s11011-009-9158-1. [DOI] [PubMed] [Google Scholar]
  • 186.Hensleigh E, Smedley L, Pritchard LM. Sex, But Not Repeated Maternal Separation During the First Postnatal Week, Influences Novel Object Exploration and Amphetamine Sensitivity. Developmental Psychobiology. 2011;53:132–140. doi: 10.1002/dev.20499. [DOI] [PubMed] [Google Scholar]
  • 187.Hall FS, Wilkinson LS, Humby T, Robbins TW. Maternal deprivation of neonatal rats produces enduring changes in dopamine function. Synapse. 1999;32:37–43. doi: 10.1002/(SICI)1098-2396(199904)32:1<37::AID-SYN5>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
  • 188.Moffett MC, Vicentic A, Kozel M, Plotsky P, Francis DD, Kuhar MJ. Maternal separation alters drug intake patterns in adulthood in rats. Biochem Pharmacol. 2007;73:321–330. doi: 10.1016/j.bcp.2006.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Moffett MC, Harley J, Francis D, Sanghani SP, Davis WI, Kuhar MJ. Maternal separation and handling affects cocaine self-administration in both the treated pups as adults and the dams. J Pharmacol Exp Ther. 2006;317:1210–1218. doi: 10.1124/jpet.106.101139. [DOI] [PubMed] [Google Scholar]
  • 190.Kosten TA, Sanchez H, Zhang XY, Kehoe P. Neonatal isolation enhances acquisition of cocaine self-administration and food responding in female rats. Behavioural Brain Research. 2004;151:137–149. doi: 10.1016/j.bbr.2003.08.010. [DOI] [PubMed] [Google Scholar]
  • 191.Kosten TA, Zhang XY, Kehoe P. Heightened cocaine and food self-administration in female rats with neonatal isolation experience. Neuropsychopharmacology. 2006;31:70–76. doi: 10.1038/sj.npp.1300779. [DOI] [PubMed] [Google Scholar]
  • 192.Kosten TA, Zhang XY, Kehoe P. Neurochemical and behavioral responses to cocaine in adult male rats with neonatal isolation experience. J Pharmacol Exp Ther. 2005;314:661–667. doi: 10.1124/jpet.104.081216. [DOI] [PubMed] [Google Scholar]
  • 193.Lomanowska AM, Rana SA, McCutcheon D, Parker LA, Wainwright PE. Artificial rearing alters the response of rats to natural and drug-mediated rewards. Developmental Psychobiology. 2006;48:301–314. doi: 10.1002/dev.20139. [DOI] [PubMed] [Google Scholar]
  • 194.Lomanowska AM, Lovic V, Rankine MJ, Mooney SJ, Robinson TE, Kraemer GW. Inadequate early social experience increases the incentive salience of reward-related cues in adulthood. Behavioural Brain Research. 2011;220:91–99. doi: 10.1016/j.bbr.2011.01.033. [DOI] [PubMed] [Google Scholar]
  • 195.Marin MT, Planeta CS. Matemal separation affects cocaine-induced locomotion and response to novelty in adolescent, but not in adult rats. Brain Research. 2004;1013:83–90. doi: 10.1016/j.brainres.2004.04.003. [DOI] [PubMed] [Google Scholar]
  • 196.Ellenbroek BA, van den Kroonenberg PTJM, Cools AR. The effects of an early stressful life event on sensorimotor gating in adult rats. Schizophrenia Research. 1998;30:251–260. doi: 10.1016/s0920-9964(97)00149-7. [DOI] [PubMed] [Google Scholar]
  • 197.Pryce CR, Bettschen D, Bahr NI, Feldon J. Comparison of the effects of infant handling, isolation, and nonhandling on acoustic startle, prepulse inhibition, locomotion, and HPA activity in the adult rat. Behavioral Neuroscience. 2001;115:71–83. doi: 10.1037/0735-7044.115.1.71. [DOI] [PubMed] [Google Scholar]
  • 198.Groenink L, Bijlsma EY, van Bogaert MJV, Oosting RS, Olivier B. Serotonin(1A) receptor deletion does not interact with maternal separation-induced increases in startle reactivity and prepulse inhibition deficits. Psychopharmacology. 2011;214:353–365. doi: 10.1007/s00213-010-1998-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Lehmann J, Pryce CR, Feldon J. Lack of effect of an early stressful life event on sensorimotor gating in adult rats. Schizophrenia Research. 2000;41:365–371. doi: 10.1016/s0920-9964(99)00080-8. [DOI] [PubMed] [Google Scholar]
  • 200.Ellenbroek BA, Cools AR. Early maternal deprivation and prepulse inhibition - The role of the postdeprivation environment. Pharmacol Biochem Behav. 2002;73:177–184. doi: 10.1016/s0091-3057(02)00794-3. [DOI] [PubMed] [Google Scholar]
  • 201.Choy KHC, van den Buuse M. Attenuated disruption of prepulse inhibition by dopaminergic stimulation after maternal deprivation and adolescent corticosterone treatment in rats. Eur Neuropsychopharm. 2008;18:1–13. doi: 10.1016/j.euroneuro.2007.03.011. [DOI] [PubMed] [Google Scholar]
  • 202.Gardner KL, Hale MW, Lightman SL, Plotsky PM, Lowry CA. Adverse early life experience and social stress during adulthood interact to increase serotonin transporter mRNA expression. Brain Research. 2009;1305:47–63. doi: 10.1016/j.brainres.2009.09.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Gardner KL, Hale MW, Oldfield S, Lightman SL, Plotsky PM, Lowry CA. ADVERSE EXPERIENCE DURING EARLY LIFE AND ADULTHOOD INTERACT TO ELEVATE tph2 mRNA EXPRESSION IN SEROTONERGIC NEURONS WITHIN THE DORSAL RAPHE NUCLEUS. Neuroscience. 2009;163:991–1001. doi: 10.1016/j.neuroscience.2009.07.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Weiss IC, Domeney AM, Moreau JL, Russig H, Feldon J. Dissociation between the effects of pre-weaning and/or post-weaning social isolation on prepulse inhibition and latent inhibition in adult Sprague-Dawley rats. Behavioural Brain Research. 2001;121:207–218. doi: 10.1016/s0166-4328(01)00166-8. [DOI] [PubMed] [Google Scholar]
  • 205.Lehmann J, Stohr T, Feldon J. Long-term effects of prenatal stress experiences and postnatal maternal separation on emotionality and attentional processes. Behav Brain Res. 2000;107:133–144. doi: 10.1016/s0166-4328(99)00122-9. [DOI] [PubMed] [Google Scholar]
  • 206.Meaney MJ, Diorio J, Francis D, LaRocque S, O'Donnell D, Smythe JW, et al. Environmental regulation of the development of glucocorticoid receptor systems in the rat forebrain. The role of serotonin. Ann N Y Acad Sci. 1994;746:260–273. doi: 10.1111/j.1749-6632.1994.tb39243.x. discussion 74, 89–93. [DOI] [PubMed] [Google Scholar]
  • 207.Meaney MJ, Aitken DH, Sapolsky RM. Thyroid hormones influence the development of hippocampal glucocorticoid receptors in the rat: a mechanism for the effects of postnatal handling on the development of the adrenocortical stress response. Neuroendocrinology. 1987;45:278–283. doi: 10.1159/000124741. [DOI] [PubMed] [Google Scholar]
  • 208.Mitchell JB, Iny LJ, Meaney MJ. The role of serotonin in the development and environmental regulation of type II corticosteroid receptor binding in rat hippocampus. Brain Res Dev Brain Res. 1990;55:231–235. doi: 10.1016/0165-3806(90)90204-c. [DOI] [PubMed] [Google Scholar]
  • 209.Smythe JW, Rowe WB, Meaney MJ. Neonatal handling alters serotonin (5-HT) turnover and 5-HT2 receptor binding in selected brain regions: relationship to the handling effect on glucocorticoid receptor expression. Brain Res Dev Brain Res. 1994;80:183–189. doi: 10.1016/0165-3806(94)90103-1. [DOI] [PubMed] [Google Scholar]
  • 210.Clarke AS, Hedeker DR, Ebert MH, Schmidt DE, McKinney WT, Kraemer GW. Rearing experience and biogenic amine activity in infant rhesus monkeys. Biol Psychiatry. 1996;40:338–352. doi: 10.1016/0006-3223(95)00663-X. [DOI] [PubMed] [Google Scholar]
  • 211.Stewart CA, Petrie RX, Balfour DJ, Matthews K, Reid IC. Enhanced evoked responses after early adversity and repeated platform exposure: the neurobiology of vulnerability? Biol Psychiatry. 2004;55:868–870. doi: 10.1016/j.biopsych.2003.12.009. [DOI] [PubMed] [Google Scholar]
  • 212.Vicentic A, Francis D, Moffett M, Lakatos A, Rogge G, Hubert GW, et al. Maternal separation alters serotonergic transporter densities and serotonergic 1A receptors in rat brain. Neuroscience. 2006;140:355–365. doi: 10.1016/j.neuroscience.2006.02.008. [DOI] [PubMed] [Google Scholar]
  • 213.Vazquez DM, Lopez JF, Van Hoers H, Watson SJ, Levine S. Maternal deprivation regulates serotonin 1A and 2A receptors in the infant rat. Brain Res. 2000;855:76–82. doi: 10.1016/s0006-8993(99)02307-0. [DOI] [PubMed] [Google Scholar]
  • 214.Macri S, Mason GJ, Wurbel H. Dissociation in the effects of neonatal maternal separations on maternal care and the offspring's HPA and fear responses in rats. Eur J Neurosci. 2004;20:1017–1024. doi: 10.1111/j.1460-9568.2004.03541.x. [DOI] [PubMed] [Google Scholar]
  • 215.Macri S, Wurbel H. Developmental plasticity of HPA and fear responses in rats: a critical review of the maternal mediation hypothesis. Horm Behav. 2006;50:667–680. doi: 10.1016/j.yhbeh.2006.06.015. [DOI] [PubMed] [Google Scholar]
  • 216.Wood GK, Marcotte ER, Quirion R, Srivastava LK. Strain differences in the behavioural outcome of neonatal ventral hippocampal lesions are determined by the postnatal environment and not genetic factors. Eur J Neurosci. 2001;14:1030–1034. doi: 10.1046/j.0953-816x.2001.01716.x. [DOI] [PubMed] [Google Scholar]
  • 217.Francis DD, Kuhar MJ. Frequency of maternal licking and grooming correlates negatively with vulnerability to cocaine and alcohol use in rats. Pharmacol Biochem Behav. 2008;90:497–500. doi: 10.1016/j.pbb.2008.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Liu D, Diorio J, Tannenbaum B, Caldji C, Francis D, Freedman A, et al. Maternal care, hippocampal glucocorticoid receptors, and hypothalamic-pituitary-adrenal responses to stress. Science. 1997;277:1659–1662. doi: 10.1126/science.277.5332.1659. [DOI] [PubMed] [Google Scholar]
  • 219.Macri S, Wurbel H. Effects of variation in postnatal maternal environment on maternal behaviour and fear responses in rats. Anim Behav. 2007;73:171–184. [Google Scholar]
  • 220.Lovic V, Gonzalez A, Fleming AS. Maternally separated rats show deficits in maternal care in adulthood. Dev Psychobiol. 2001;39:19–33. doi: 10.1002/dev.1024. [DOI] [PubMed] [Google Scholar]
  • 221.Rees SL, Fleming AS. How early maternal separation and juvenile experience with pups affect maternal behavior and emotionality in adult postpartum rats. Animal Learning & Behavior. 2001;29:221–233. [Google Scholar]
  • 222.Gonzalez A, Lovic V, Ward GR, Wainwright PE, Fleming AS. Intergenerational effects of complete maternal deprivation and replacement stimulation on maternal behavior and emotionality in female rats. Dev Psychobiol. 2001;38:11–32. doi: 10.1002/1098-2302(2001)38:1<11::aid-dev2>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
  • 223.Lovic V, Fleming AS, Fletcher PJ. Early life tactile stimulation changes adult rat responsiveness to amphetamine. Pharmacol Biochem Behav. 2006;84:497–503. doi: 10.1016/j.pbb.2006.06.013. [DOI] [PubMed] [Google Scholar]
  • 224.Melo AI, Lovic V, Gonzalez A, Madden M, Sinopoli K, Fleming AS. Maternal and littermate deprivation disrupts maternal behavior and social-learning of food preference in adulthood: Tactile stimulation, nest odor, and social rearing prevent these effects. Developmental Psychobiology. 2006;48:209–219. doi: 10.1002/dev.20130. [DOI] [PubMed] [Google Scholar]
  • 225.Melo AI, Perez-Ledezma M, Clapp C, Arnold E, Rivera JC, Fleming AS. Effects of prolactin deficiency during the early postnatal period on the development of maternal behavior in female rats: Mother's milk makes the difference. Horm Behav. 2009;56:281–291. doi: 10.1016/j.yhbeh.2009.06.005. [DOI] [PubMed] [Google Scholar]
  • 226.Kim JW, Kirkpatrick B. Social isolation in animal models of relevance to neuropsychiatric disorders. Biol. Psychiat. 1996;40:918–922. doi: 10.1016/0006-3223(95)00546-3. [DOI] [PubMed] [Google Scholar]
  • 227.Panksepp J, Beatty WW. Social deprivation and play in rats. Behav. Neural Biol. 1980;30:197–206. doi: 10.1016/s0163-1047(80)91077-8. [DOI] [PubMed] [Google Scholar]
  • 228.Einon DF, Morgan MJ, Kibbler CC. Brief periods of socialization and later behavior in the rat. Dev. Psychobiol. 1978;11:213–225. doi: 10.1002/dev.420110305. [DOI] [PubMed] [Google Scholar]
  • 229.Ikemoto S, Panksepp J. The effects of early social isolation on the motivation for social play in juvenile rats. Dev. Psychobiol. 1992;25:261–274. doi: 10.1002/dev.420250404. [DOI] [PubMed] [Google Scholar]
  • 230.Colonnello V, Iacobucci P, Anderson MP, Panksepp J. Brief Periods of Positive Peer Interactions Mitigate the Effects of Total Social Isolation in Young Octodon degus. Developmental Psychobiology. 2011;53:280–290. doi: 10.1002/dev.20520. [DOI] [PubMed] [Google Scholar]
  • 231.Douglas LA, Varlinskaya EI, Spear LP. Rewarding properties of social interactions in adolescent and adult male and female rats: Impact of social versus isolate housing of subjects and partners. Dev. Psychobiol. 2004;45:153–162. doi: 10.1002/dev.20025. [DOI] [PubMed] [Google Scholar]
  • 232.Pietropaolo S, Singer P, Feldon J, Yee BK. The postweaning social isolation in C57BL/6 mice: preferential vulnerability in the male sex. Psychopharmacology. 2008;197:613–628. doi: 10.1007/s00213-008-1081-3. [DOI] [PubMed] [Google Scholar]
  • 233.Pietropaolo S, Feldon J, Yee BK. Nonphysical contact between cagemates alleviates the social isolation syndrome in C57BL/6 male mice. Behavioral Neuroscience. 2008;122:505–515. doi: 10.1037/0735-7044.122.3.505. [DOI] [PubMed] [Google Scholar]
  • 234.Knutson B, Panksepp J. Effects of serotonin depletion on the play of juvenile rats. Ann N Y Acad Sci. 1997;807:475–477. doi: 10.1111/j.1749-6632.1997.tb51942.x. [DOI] [PubMed] [Google Scholar]
  • 235.Knutson B, Panksepp J, Pruitt D. Effects of fluoxetine on play dominance in juvenile rats. Aggress Behav. 1996;22:297–307. [Google Scholar]
  • 236.Sahakian BJ, Burdess C, Luckhurst H, Trayhurn P. Hyperactivity and obesity: the interaction of social isolation and cafeteria feeding. Physiol Behav. 1982;28:117–124. doi: 10.1016/0031-9384(82)90112-3. [DOI] [PubMed] [Google Scholar]
  • 237.Matsuda T, Sakaue M, Ago Y, Sakamoto Y, Koyama Y, Baba A. Functional alteration of brain dopaminergic system in isolated aggressive mice. Nihon Shinkei Seishin Yakurigaku Zasshi. 2001;21:71–76. [PubMed] [Google Scholar]
  • 238.Malkesman O, Maayan R, Weizman A, Weller A. Aggressive behavior and HPA axis hormones after social isolation in adult rats of two different genetic animal models for depression. Behavioural Brain Research. 2006;175:408–414. doi: 10.1016/j.bbr.2006.09.017. [DOI] [PubMed] [Google Scholar]
  • 239.Wolffgramm J, Heyne A. Social-Behavior, Dominance, and Social Deprivation of Rats Determine Drug Choice. Pharmacol Biochem Behav. 1991;38:389–399. doi: 10.1016/0091-3057(91)90297-f. [DOI] [PubMed] [Google Scholar]
  • 240.Valzelli L, Bernasconi S. Psychoactive Drug Effect on Behavioral-Changes Induced by Prolonged Socio-Environmental Deprivation in Rats. Psychol Med. 1976;6:271–276. doi: 10.1017/s0033291700013829. [DOI] [PubMed] [Google Scholar]
  • 241.Koike H, Ibi D, Mizoguchi H, Nagai T, Nitta A, Takuma K, et al. Behavioral abnormality and pharmacologic response in social isolation-reared mice. Behavioural Brain Research. 2009;202:114–121. doi: 10.1016/j.bbr.2009.03.028. [DOI] [PubMed] [Google Scholar]
  • 242.Insel TR, Fernald RD. How the brain processes social information: Searching for the social brain. Annu Rev Neurosci. 2004;27:697–722. doi: 10.1146/annurev.neuro.27.070203.144148. [DOI] [PubMed] [Google Scholar]
  • 243.Arakawa H. Interaction between isolation rearing and social development on exploratory behavior in male rats. Behav Process. 2005;70:223–234. doi: 10.1016/j.beproc.2005.07.002. [DOI] [PubMed] [Google Scholar]
  • 244.Jones GH, Robbins TW, Marsden CA. Isolation-rearing retards the acquisition of schedule-induced polydipsia in rats. Physiol Behav. 1989;45:71–77. doi: 10.1016/0031-9384(89)90167-4. [DOI] [PubMed] [Google Scholar]
  • 245.Einon DF, Morgan MJ. Early isolation produces enduring hyperactivity in the rat, but no effect upon spontaneous alternation. Q. J. Exp. Psychol. 1978;30:151–156. doi: 10.1080/14640747808400663. [DOI] [PubMed] [Google Scholar]
  • 246.Wilkinson LS, Killcross SS, Humby T, Hall FS, Geyer MA, Robbins TW. Social isolation in the rat produces developmentally specific deficits in prepulse inhibition of the acoustic startle response without disrupting latent inhibition. Neuropsychopharmacology. 1994;10:61–72. doi: 10.1038/npp.1994.8. [DOI] [PubMed] [Google Scholar]
  • 247.Gentsch C, Lichtsteiner M, Kraeuchi K, Feer H. Different reaction patterns in individually and socially reared rats during exposures to novel environments. Behav Brain Res. 1982;4:45–54. doi: 10.1016/0166-4328(82)90163-2. [DOI] [PubMed] [Google Scholar]
  • 248.Sahakian BJ, Robbins TW, Iversen SD. The effects of isolation rearing on exploration in the rat. Anim. Learn. Behav. 1977;5:193. [Google Scholar]
  • 249.Paulus MP, Bakshi VP, Geyer MA. Isolation rearing affects sequential organization of motor behavior in post-pubertal but not pre-pubertal Lister and Sprague-Dawley rats. Behav Brain Res. 1998;94:271–280. doi: 10.1016/s0166-4328(97)00158-7. [DOI] [PubMed] [Google Scholar]
  • 250.Jones GH, Marsden CA, Robbins TW. Increased sensitivity to amphetamine and reward-related stimuli following social isolation in rats: possible disruption of dopamine-dependent mechanisms of the nucleus accumbens. Psychopharmacology (Berl) 1990;102:364–372. doi: 10.1007/BF02244105. [DOI] [PubMed] [Google Scholar]
  • 251.Einon DF, Sahakian BJ. Environmentally induced differences in susceptibility of rats to CNS stimulants and CNS depressants: evidence against a unitary explanation. Psychopharmacology (Berl) 1979;61:299–307. doi: 10.2196/40080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Phillips GD, Howes SR, Whitelaw RB, Wilkinson LS, Robbins TW, Everitt BJ. Isolation rearing enhances the locomotor response to cocaine and a novel environment, but impairs the intravenous self-administration of cocaine. Psychopharmacology (Berl) 1994;115:407–418. doi: 10.1007/BF02245084. [DOI] [PubMed] [Google Scholar]
  • 253.Sahakian BJ, Robbins TW, Morgan MJ, Iversen SD. The effects of psychomotor stimulants on stereotypy and locomotor activity in socially-deprived and control rats. Brain Res. 1975;84:195–205. doi: 10.1016/0006-8993(75)90975-0. [DOI] [PubMed] [Google Scholar]
  • 254.Chitkara B, Durcan MJ, Campbell IC. Apomorphine-induced stereotypy: function of age and rearing environment. Pharmacol Biochem Behav. 1984;21:671–673. doi: 10.1016/s0091-3057(84)80056-8. [DOI] [PubMed] [Google Scholar]
  • 255.Weiss IC, Domeney AM, Heidbreder CA, Moreau JL, Feldon J. Early social isolation, but not maternal separation, affects behavioral sensitization to amphetamine in male and female adult rats. Pharmacol Biochem Behav. 2001;70:397–409. doi: 10.1016/s0091-3057(01)00626-8. [DOI] [PubMed] [Google Scholar]
  • 256.Green TA, Gehrke BJ, Bardo MT. Environmental enrichment decreases intravenous amphetamine self-administration in rats: dose-response functions for fixed- and progressive-ratio schedules. Psychopharmacology (Berl) 2002;162:373–378. doi: 10.1007/s00213-002-1134-y. [DOI] [PubMed] [Google Scholar]
  • 257.Howes SR, Dalley JW, Morrison CH, Robbins TW, Everitt BJ. Leftward shift in the acquisition of cocaine self-administration in isolation-reared rats: relationship to extracellular levels of dopamine, serotonin and glutamate in the nucleus accumbens and amygdala-striatal FOS expression. Psychopharmacology (Berl) 2000;151:55–63. doi: 10.1007/s002130000451. [DOI] [PubMed] [Google Scholar]
  • 258.Boyle AE, Gill K, Smith BR, Amit Z. Differential effects of an early housing manipulation on cocaine-induced activity and self-administration in laboratory rats. Pharmacol Biochem Behav. 1991;39:269–274. doi: 10.1016/0091-3057(91)90178-5. [DOI] [PubMed] [Google Scholar]
  • 259.Schenk S, Lacelle G, Gorman K, Amit Z. Cocaine self-administration in rats influenced by environmental conditions: implications for the etiology of drug abuse. Neurosci Lett. 1987;81:227–231. doi: 10.1016/0304-3940(87)91003-2. [DOI] [PubMed] [Google Scholar]
  • 260.Yajie D, Lin K, Baoming L, Lan M. Enhanced cocaine self-administration in adult rats with adolescent isolation experience. Pharmacol Biochem Behav. 2005;82:673–677. doi: 10.1016/j.pbb.2005.11.007. [DOI] [PubMed] [Google Scholar]
  • 261.Hall FS, Humby T, Wilkinson LS, Robbins TW. The effects of isolation-rearing on sucrose consumption in rats. Physiol Behav. 1997;62:291–297. doi: 10.1016/s0031-9384(97)00116-9. [DOI] [PubMed] [Google Scholar]
  • 262.Morgan M, Einon D. Incentive motivation and behavioral inhibition in socially-isolated rats. Physiol Behav. 1975;15:405–409. doi: 10.1016/0031-9384(75)90205-x. [DOI] [PubMed] [Google Scholar]
  • 263.Geyer MA, Wilkinson LS, Humby T, Robbins TW. Isolation rearing of rats produces a deficit in prepulse inhibition of acoustic startle similar to that in schizophrenia. Biol Psychiatry. 1993;34:361–372. doi: 10.1016/0006-3223(93)90180-l. [DOI] [PubMed] [Google Scholar]
  • 264.Powell SB, Geyer MA, Preece MA, Pitcher LK, Reynolds GP, Swerdlow NR. Dopamine depletion of the nucleus accumbens reverses isolation-induced deficits in prepulse inhibition in rats. Neuroscience. 2003;119:233–240. doi: 10.1016/s0306-4522(03)00122-2. [DOI] [PubMed] [Google Scholar]
  • 265.Geyer MA, Swerdlow NR, Lehmann-Masten V, Teschendorf HJ, Traut M, Gross G. Effects of LU-111995 in three models of disrupted prepulse inhibition in rats. J Pharmacol Exp Ther. 1999;290:716–724. [PubMed] [Google Scholar]
  • 266.Varty GB, Higgins GA. Examination of drug-induced and isolation-induced disruptions of prepulse inhibition as models to screen antipsychotic drugs. Psychopharmacology (Berl) 1995;122:15–26. doi: 10.1007/BF02246437. [DOI] [PubMed] [Google Scholar]
  • 267.Cilia J, Reavill C, Hagan JJ, Jones DN. Long-term evaluation of isolation-rearing induced prepulse inhibition deficits in rats. Psychopharmacology (Berl) 2001;156:327–337. doi: 10.1007/s002130100786. [DOI] [PubMed] [Google Scholar]
  • 268.Bakshi VP, Swerdlow NR, Braff DL, Geyer MA. Reversal of isolation rearing-induced deficits in prepulse inhibition by seroquel and olanzapine. Biol. Psychiat. 1998;43:436–445. doi: 10.1016/s0006-3223(97)00246-1. [DOI] [PubMed] [Google Scholar]
  • 269.Shao F, Jin J, Meng QX, Liu M, Xie X, Lin WJ, et al. Pubertal isolation alters latent inhibition and DA in nucleus accumbens of adult rats. Physiology & Behavior. 2009;98:251–257. doi: 10.1016/j.physbeh.2009.05.021. [DOI] [PubMed] [Google Scholar]
  • 270.Jones GH, Hernandez TD, Kendall DA, Marsden CA, Robbins TW. Dopaminergic and serotonergic function following isolation rearing in rats: study of behavioural responses and postmortem and in vivo neurochemistry. Pharmacol Biochem Behav. 1992;43:17–35. doi: 10.1016/0091-3057(92)90635-s. [DOI] [PubMed] [Google Scholar]
  • 271.Hall FS, Wilkinson LS, Humby T, Inglis W, Kendall DA, Marsden CA, et al. Isolation rearing in rats: pre- and postsynaptic changes in striatal dopaminergic systems. Pharmacol Biochem Behav. 1998;59:859–872. doi: 10.1016/s0091-3057(97)00510-8. [DOI] [PubMed] [Google Scholar]
  • 272.Fulford AJ, Marsden CA. Effect of isolation-rearing on conditioned dopamine release in vivo in the nucleus accumbens of the rat. J Neurochem. 1998;70:384–390. doi: 10.1046/j.1471-4159.1998.70010384.x. [DOI] [PubMed] [Google Scholar]
  • 273.Leng A, Feldon J, Ferger B. Long-term social isolation and medial prefrontal cortex: dopaminergic and cholinergic neurotransmission. Pharmacol Biochem Behav. 2004;77:371–379. doi: 10.1016/j.pbb.2003.11.011. [DOI] [PubMed] [Google Scholar]
  • 274.Cilia J, Reavill C, Hagan JJ, Jones DNC. Long-term evaluation of isolation-rearing induced prepulse inhibition deficits in rats. Psychopharmacology. 2001;156:327–337. doi: 10.1007/s002130100786. [DOI] [PubMed] [Google Scholar]
  • 275.Gamallo A, Villanua A, Trancho G, Fraile A. Stress adaptation and adrenal activity in isolated and crowded rats. Physiol Behav. 1986;36:217–221. doi: 10.1016/0031-9384(86)90006-5. [DOI] [PubMed] [Google Scholar]
  • 276.Parker V, Morinan A. The socially isolated rat as a model for anxiety. Neuropharmacology. 1986;25:663. [Google Scholar]
  • 277.Da Silva NL, Ferreira VM, Carobrez Ade P, Morato GS. Individual housing from rearing modifies the performance of young rats on the elevated plus-maze apparatus. Physiol Behav. 1996;60:1391–1396. doi: 10.1016/s0031-9384(96)00254-5. [DOI] [PubMed] [Google Scholar]
  • 278.Molina-Hernandez M, Tellez-Alcantara P, Perez-Garcia J. Isolation rearing induced fear-like behavior without affecting learning abilities of Wistar rats. Prog Neuropsychopharmacol Biol Psychiatry. 2001;25:1111–1123. doi: 10.1016/s0278-5846(01)00168-3. [DOI] [PubMed] [Google Scholar]
  • 279.Hol T, Van den Berg CL, Van Ree JM, Spruijt BM. Isolation during the play period in infancy decreases adult social interactions in rats. Behav Brain Res. 1999;100:91–97. doi: 10.1016/s0166-4328(98)00116-8. [DOI] [PubMed] [Google Scholar]
  • 280.Morinan A, Parker V. Are socialy isolated rats anxious? Br J Pharmacol. 1985;86:460P. [Google Scholar]
  • 281.Einon D, Tye NC. Chlordiazepoxide and isolation induced timidity in rats. Psychopharmacologia. 1975;44:83–85. doi: 10.1007/BF00421188. [DOI] [PubMed] [Google Scholar]
  • 282.Kokare DM, Dandekar MP, Singru PS, Gupta GL, Subhedar NK. Involvement of alpha- MSH in the social isolation induced anxiety- and depression-like behaviors in rat. Neuropharmacology. 2010;58:1009–1018. doi: 10.1016/j.neuropharm.2010.01.006. [DOI] [PubMed] [Google Scholar]
  • 283.Gendreau PL, Petitto JM, Gariepy JL, Lewis MH. D-1 dopamine receptor mediation of social and nonsocial emotional reactivity in mice: Effects of housing and strain difference in motor activity. Behavioral Neuroscience. 1997;111:424–434. doi: 10.1037//0735-7044.111.2.424. [DOI] [PubMed] [Google Scholar]
  • 284.Hall FS, Humby T, Wilkinson LS, Robbins TW. The effects of isolation-rearing on preference by rats for a novel environment. Physiol Behav. 1997;62:299–303. doi: 10.1016/s0031-9384(97)00117-0. [DOI] [PubMed] [Google Scholar]
  • 285.Wongwitdecha N, Marsden CA. Social isolation increases aggressive behaviour and alters the effects of diazepam in the rat social interaction test. Behav Brain Res. 1996;75:27–32. doi: 10.1016/0166-4328(96)00181-7. [DOI] [PubMed] [Google Scholar]
  • 286.Hall FS, Huang S, Fong GW, Pert A, Linnoila M. Effects of isolation-rearing on locomotion, anxiety and responses to ethanol in Fawn Hooded and Wistar rats. Psychopharmacology (Berl) 1998;139:203–209. doi: 10.1007/s002130050705. [DOI] [PubMed] [Google Scholar]
  • 287.Holson RR. Feeding neophobia: a possible explanation for the differential maze performance of rats reared in enriched or isolated environments. Physiol Behav. 1986;38:191–201. doi: 10.1016/0031-9384(86)90154-x. [DOI] [PubMed] [Google Scholar]
  • 288.Hatch AM, Wiberg GS, Zawidzka Z, Cann M, Airth JM, Grice HC. Isolation syndrome in the rat. Toxicol. Appl. Pharmacol. 1965;7:737–745. doi: 10.1016/0041-008x(65)90132-8. [DOI] [PubMed] [Google Scholar]
  • 289.Holson RR, Scallet AC, Ali SF, Turner BB. "Isolation stress" revisited: isolation-rearing effects depend on animal care methods. Physiol Behav. 1991;49:1107–1118. doi: 10.1016/0031-9384(91)90338-o. [DOI] [PubMed] [Google Scholar]
  • 290.Sanchez MM, Aguado F, Sanchez-Toscano F, Saphier D. Effects of prolonged social isolation on responses of neurons in the bed nucleus of the stria terminalis, preoptic area, and hypothalamic paraventricular nucleus to stimulation of the medial amygdala. Psychoneuroendocrinology. 1995;20:525–541. doi: 10.1016/0306-4530(94)00083-m. [DOI] [PubMed] [Google Scholar]
  • 291.Hall FS, Sundstrom JM, Lerner J, Pert A. Enhanced corticosterone release after a modified forced swim test in Fawn hooded rats is independent of rearing experience. Pharmacol Biochem Behav. 2001;69:629–634. doi: 10.1016/s0091-3057(01)00556-1. [DOI] [PubMed] [Google Scholar]
  • 292.Schrijver NC, Bahr NI, Weiss IC, Wurbel H. Dissociable effects of isolation rearing and environmental enrichment on exploration, spatial learning and HPA activity in adult rats. Pharmacol Biochem Behav. 2002;73:209–224. doi: 10.1016/s0091-3057(02)00790-6. [DOI] [PubMed] [Google Scholar]
  • 293.Holson RR, Scallet AC, Ali SF, Sullivan P, Gough B. Adrenocortical, beta-endorphin and behavioral responses to graded stressors in differentially reared rats. Physiol Behav. 1988;42:125–130. doi: 10.1016/0031-9384(88)90286-7. [DOI] [PubMed] [Google Scholar]
  • 294.Weiss IC, Pryce CR, Jongen-Relo AL, Nanz-Bahr NI, Feldon J. Effect of social isolation on stress-related behavioural and neuroendocrine state in the rat. Behavioural Brain Research. 2004;152:279–295. doi: 10.1016/j.bbr.2003.10.015. [DOI] [PubMed] [Google Scholar]
  • 295.Serra M, Sanna E, Mostallino MC, Biggio G. Social isolation stress and neuroactive steroids. Eur Neuropsychopharm. 2007;17:1–11. doi: 10.1016/j.euroneuro.2006.03.004. [DOI] [PubMed] [Google Scholar]
  • 296.Sciolino NR, Bortolato M, Eisenstein SA, Fu J, Oveisi F, Hohmann AG, et al. Social Isolation and Chronic Handling Alter Endocannabinoid Signaling and Behavioral Reactivity to Context in Adult Rats. Neuroscience. 2010;168:371–386. doi: 10.1016/j.neuroscience.2010.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.D'Aquila PS, Brain P, Willner P. Effects of chronic mild stress on performance in behavioural tests relevant to anxiety and depression. Physiol Behav. 1994;56:861–867. doi: 10.1016/0031-9384(94)90316-6. [DOI] [PubMed] [Google Scholar]
  • 298.Von Frijtag JC, Schot M, van den Bos R, Spruijt BM. Individual housing during the play period results in changed responses to and consequences of a psychosocial stress situation in rats. Dev Psychobiol. 2002;41:58–69. doi: 10.1002/dev.10057. [DOI] [PubMed] [Google Scholar]
  • 299.Hellemans KGC, Benge LC, Olmstead MC. Adolescent enrichment partially reverses the social isolation syndrome. Dev Brain Res. 2004;150:103–115. doi: 10.1016/j.devbrainres.2004.03.003. [DOI] [PubMed] [Google Scholar]
  • 300.Brenes JC, Fornaguera J. Effects of environmental enrichment and social isolation on sucrose consumption and preference: Associations with depressive-like behavior and ventral striatum dopamine. Neuroscience Letters. 2008;436:278–282. doi: 10.1016/j.neulet.2008.03.045. [DOI] [PubMed] [Google Scholar]
  • 301.Brenes JC, Rodriguez O, Fornaguera J. Differential effect of environment enrichment and social isolation on depressive-like behavior, spontaneous activity and serotonin and norepinephrine concentration in prefrontal cortex and ventral striatum. Pharmacol Biochem Behav. 2008;89:85–93. doi: 10.1016/j.pbb.2007.11.004. [DOI] [PubMed] [Google Scholar]
  • 302.Brenes JC, Fornaguera J. The effect of chronic fluoxetine on social isolation-induced changes on sucrose consumption, immobility behavior, and on serotonin and dopamine function in hippocampus and ventral striatum. Behavioural Brain Research. 2009;198:199–205. doi: 10.1016/j.bbr.2008.10.036. [DOI] [PubMed] [Google Scholar]
  • 303.Brenes JC, Padilla M, Fornaguera J. A detailed analysis of open-field habituation and behavioral and neurochemical antidepressant-like effects in postweaning enriched rats. Behavioural Brain Research. 2009;197:125–137. doi: 10.1016/j.bbr.2008.08.014. [DOI] [PubMed] [Google Scholar]
  • 304.Greenough WT, Madden TC, Fleischmann TB. Effects of isolation, daily handling, and enriched rearing on maze learning. Psychonomic Sci. 1972;27:279. [Google Scholar]
  • 305.Gardner EB, Boitano JJ, Mancino NS, D'Amico DP. Environmental enrichment and deprivation: effects on learning, memory and exploration. Physiol Behav. 1975;14:321–327. doi: 10.1016/0031-9384(75)90040-2. [DOI] [PubMed] [Google Scholar]
  • 306.Einon D. Spatial learning and response strategies in rats: age, sex, and rearing differences in performance. Q. J. Exp. Psychol. 1980;32:473. doi: 10.1080/14640748008401840. [DOI] [PubMed] [Google Scholar]
  • 307.Einon D, Morgan M, Will BE. Effects of postoperative environment on recovery from dorsal hippocampal lesions in young rats: tests of spatial memory and motor transfer. Q. J. Exp. Psychol. 1980;32:137. doi: 10.1080/00335558008248239. [DOI] [PubMed] [Google Scholar]
  • 308.Jones GH, Marsden CA, Robbins TW. Behavioural rigidity and rule-learning deficits following isolation-rearing in the rat: neurochemical correlates. Behav Brain Res. 1991;43:35–50. doi: 10.1016/s0166-4328(05)80050-6. [DOI] [PubMed] [Google Scholar]
  • 309.Harmer CJ, Phillips GD. Isolation rearing enhances acquisition in a conditioned inhibition paradigm. Physiology & Behavior. 1998;65:525–533. doi: 10.1016/s0031-9384(98)00207-8. [DOI] [PubMed] [Google Scholar]
  • 310.Harmer CJ, Phillips GD. Isolation rearing enhances the rate of acquisition of a discriminative approach task but does not affect the efficacy of a conditioned reward. Physiology & Behavior. 1998;63:177–184. doi: 10.1016/s0031-9384(97)00417-4. [DOI] [PubMed] [Google Scholar]
  • 311.Schrijver NC, Wurbel H. Early social deprivation disrupts attentional, but not affective, shifts in rats. Behav Neurosci. 2001;115:437–442. [PubMed] [Google Scholar]
  • 312.Roberts AC, De Salvia MA, Wilkinson LS, Collins P, Muir JL, Everitt BJ, et al. 6-Hydroxydopamine lesions of the prefrontal cortex in monkeys enhance performance on an analog of the Wisconsin Card Sort Test: possible interactions with subcortical dopamine. J Neurosci. 1994;14:2531–2544. doi: 10.1523/JNEUROSCI.14-05-02531.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Li NX, Wu XH, Li L. Chronic administration of clozapine alleviates reversal-learning impairment in isolation-reared rats. Behav Pharmacol. 2007;18:135–145. doi: 10.1097/FBP.0b013e3280d3ee83. [DOI] [PubMed] [Google Scholar]
  • 314.Morgan MJ, Einon D, Morris RG. Inhibition and isolation rearing in the rat: extinction and satiation. Physiol Behav. 1977;18:1–5. doi: 10.1016/0031-9384(77)90084-1. [DOI] [PubMed] [Google Scholar]
  • 315.Feldon J, Avnimelech-Gigus N, Weiner I. The effects of pre- and postweaning rearing conditions on latent inhibition and partial reinforcement extinction effect in male rats. Behav Neural Biol. 1990;53:189–204. doi: 10.1016/0163-1047(90)90407-w. [DOI] [PubMed] [Google Scholar]
  • 316.Morgan MJ, Einon D, Nicholas D. The effects of isolation rearing on behavioral inhibition in the rat. Q J Exp Psychol. 1975;27:615. [Google Scholar]
  • 317.Hellemans KGC, Nobrega JN, Olmstead MC. Early environmental experience alters baseline and ethanol-induced cognitive impulsivity: relationship to forebrain 5-HT1A receptor binding. Behavioural Brain Research. 2005;159:207–220. doi: 10.1016/j.bbr.2004.10.018. [DOI] [PubMed] [Google Scholar]
  • 318.Holson RR, Ali SF, Scallet AC. The effect of isolation rearing and stress on monoamines in forebrain nigrostriatal, mesolimbi, and mesocortical dopamine systems. Psychobiology of Posttraumatic Stress Disorder. 1988;537:512–514. [Google Scholar]
  • 319.Blanc G, Herve D, Simon H, Lisoprawski A, Glowinski J, Tassin JP. Response to stress of mesocortico-frontal dopaminergic neurones in rats after long-term isolation. Nature. 1980;284:265–267. doi: 10.1038/284265a0. [DOI] [PubMed] [Google Scholar]
  • 320.Neddens J, Brandenburg K, Teuchert-Noodt G, Dawirs RR. Differential environment alters ontogeny of dopamine innervation of the orbital prefrontal cortex in gerbils. Journal of Neuroscience Research. 2001;63:209–213. doi: 10.1002/1097-4547(20010115)63:2<209::AID-JNR1013>3.0.CO;2-E. [DOI] [PubMed] [Google Scholar]
  • 321.Peters YM, O'Donnell P. Social isolation rearing affects prefrontal cortical response to ventral tegmental area stimulation. Biol. Psychiat. 2005;57:1205–1208. doi: 10.1016/j.biopsych.2005.02.011. [DOI] [PubMed] [Google Scholar]
  • 322.Heidbreder CA, Foxton R, Cilia J, Hughes ZA, Shah AJ, Atkins A, et al. Increased responsiveness of dopamine to atypical, but not typical antipsychotics in the medial prefrontal cortex of rats reared in isolation. Psychopharmacology. 2001;156:338–351. doi: 10.1007/s002130100760. [DOI] [PubMed] [Google Scholar]
  • 323.Pascual R, Zamora-Leon P, Catalan-Ahumada M, Valero-Cabre A. Early social isolation decreases the expression of calbindin D-28k and dendritic branching in the medial prefrontal cortex of the rat. Int J Neurosci. 2007;117:465–476. doi: 10.1080/00207450600773459. [DOI] [PubMed] [Google Scholar]
  • 324.Heidbreder CA, Foxton R, Cilia J, Hughes ZA, Shah AJ, Atkins A, et al. Increased responsiveness of dopamine to atypical, but not typical antipsychotics in the medial prefrontal cortex of rats reared in isolation. Psychopharmacology (Berl) 2001;156:338–351. doi: 10.1007/s002130100760. [DOI] [PubMed] [Google Scholar]
  • 325.Sahakian BJ, Robbins TW. Isolation-rearing enhances tail pinch-induced oral behavior in rats. Physiol Behav. 1977;18:53–58. doi: 10.1016/0031-9384(77)90093-2. [DOI] [PubMed] [Google Scholar]
  • 326.Sundstrom JM, Hall FS, Stellar JR, Waugh EJ. Effects of isolation-rearing on intracranial self-stimulation reward of the lateral hypothalamus: baseline assessment and drug challenges. Life Sci. 2002;70:2799–2810. doi: 10.1016/s0024-3205(02)01509-6. [DOI] [PubMed] [Google Scholar]
  • 327.Bjornebekk A, Mathe AA, Brene S. Isolated Flinders Sensitive Line rats have decreased dopamine D2 receptor mRNA. Neuroreport. 2007;18:1039–1043. doi: 10.1097/WNR.0b013e3281668bf7. [DOI] [PubMed] [Google Scholar]
  • 328.Bean G, Lee T. Social isolation and cohabitation with haloperidol-treated partners: effect on density of striatal dopamine D2 receptors in the developing rat brain. Psychiatry Res. 1991;36:307–317. doi: 10.1016/0165-1781(91)90029-o. [DOI] [PubMed] [Google Scholar]
  • 329.King MV, Seeman P, Marsden CA, Fone KCF. Increased Dopamine D(2)(High) Receptors in Rats Reared in Social Isolation. Synapse. 2009;63:476–483. doi: 10.1002/syn.20624. [DOI] [PubMed] [Google Scholar]
  • 330.Muchimapura S, Fulford AJ, Mason R, Marsden CA. Isolation rearing in the rat disrupts the hippocampal response to stress. Neuroscience. 2002;112:697–705. doi: 10.1016/s0306-4522(02)00107-0. [DOI] [PubMed] [Google Scholar]
  • 331.Hall FS. Unpublished Ph.D. Thesis. University of Cambridge; 1994. The behavioral and neurochemical effects of social separation on the rat. [Google Scholar]
  • 332.Murphy KC, Jones LA, Owen MJ. High rates of schizophrenia in adults with velo-cardio-facial syndrome. Arch Gen Psychiatry. 1999;56:940–945. doi: 10.1001/archpsyc.56.10.940. [DOI] [PubMed] [Google Scholar]
  • 333.Barr AM, Young CE, Sawada K, Trimble WS, Phillips AG, Honer WG. Abnormalities of presynaptic protein CDCrel-1 in striatum of rats reared in social isolation: relevance to neural connectivity in schizophrenia. Eur J Neurosci. 2004;20:303–307. doi: 10.1111/j.0953-816X.2004.03457.x. [DOI] [PubMed] [Google Scholar]
  • 334.Nudmamud S, Reynolds LM, Reynolds GP. N-acetylaspartate and N-acetylaspartylglutamate deficits in superior temporal cortex in schizophrenia and bipolar disorder: A postmortem study. Biol. Psychiat. 2003;53:1138–1141. doi: 10.1016/s0006-3223(02)01742-0. [DOI] [PubMed] [Google Scholar]
  • 335.Harte M, Powell SB, Reynolds LM, Geyer MA, Reynolds GP. Reduced N-acetyl aspartate levels in the temporal cortex of rats reared in isolation. Eur Neuropsychopharm. 2004;14:S66–S67. doi: 10.1016/j.biopsych.2004.06.009. [DOI] [PubMed] [Google Scholar]
  • 336.Parker V, Morinan A. The socially-isolated rat as a model for anxiety. Neuropharmacology. 1986;25:663–664. [Google Scholar]
  • 337.Rilke O, Freier D, Jahkel M, Oehler J. Dynamic alterations of serotonergic metabolism and receptors during social isolation of low- and high-active mice. Pharmacol Biochem Behav. 1998;59:891–896. doi: 10.1016/s0091-3057(97)00509-1. [DOI] [PubMed] [Google Scholar]
  • 338.Segal DS, Knapp S, Kuczenski RT, Mandell AJ. The effects of environmental isolation on behavior and regional rat brain tyrosine hydroxylase and tryptophan hydroxylase activities. Behav Biol. 1973;8:47–53. doi: 10.1016/s0091-6773(73)80005-7. [DOI] [PubMed] [Google Scholar]
  • 339.Yanai J, Sze PY. Isolation reduces midbrain tryptophan hydroxylase activity in mice. Psychopharmacology (Berl) 1983;80:284–285. doi: 10.1007/BF00436171. [DOI] [PubMed] [Google Scholar]
  • 340.Jaffe EH, De Frias V, Ibarra C. Changes in basal and stimulated release of endogenous serotonin from different nuclei of rats subjected to two models of depression. Neurosci Lett. 1993;162:157–160. doi: 10.1016/0304-3940(93)90584-8. [DOI] [PubMed] [Google Scholar]
  • 341.Bickerdike MJ, Wright IK, Marsden CA. Social isolation attenuates rat forebrain 5-HT release induced by KCI stimulation and exposure to a novel environment. Behav Pharmacol. 1993;4:231–236. [PubMed] [Google Scholar]
  • 342.Dalley JW, Theobald DE, Pereira EA, Li PM, Robbins TW. Specific abnormalities in serotonin release in the prefrontal cortex of isolation-reared rats measured during behavioural performance of a task assessing visuospatial attention and impulsivity. Psychopharmacology (Berl) 2002;164:329–340. doi: 10.1007/s00213-002-1215-y. [DOI] [PubMed] [Google Scholar]
  • 343.Muchimapura S, Mason R, Marsden CA. Effect of isolation rearing on pre- and post-synaptic serotonergic function in the rat dorsal hippocampus. Synapse. 2003;47:209–217. doi: 10.1002/syn.10167. [DOI] [PubMed] [Google Scholar]
  • 344.Whitaker-Azmitia P, Zhou F, Hobin J, Borella A. Isolation-rearing of rats produces deficits as adults in the serotonergic innervation of hippocampus. Peptides. 2000;21:1755–1759. doi: 10.1016/s0196-9781(00)00327-2. [DOI] [PubMed] [Google Scholar]
  • 345.Neddens J, Bagorda F, Busche A, Horstmann S, Moll GH, Dawirs RR, et al. Epigenetic factors differentially influence postnatal maturation of serotonin (5-HT) innervation in cerebral cortex of gerbils: interaction of rearing conditions and early methamphetamine challenge. Dev Brain Res. 2003;146:119–130. doi: 10.1016/j.devbrainres.2003.09.016. [DOI] [PubMed] [Google Scholar]
  • 346.Fulford AJ, Marsden CA. Conditioned release of 5-hydroxytryptamine in vivo in the nucleus accumbens following isolation-rearing in the rat. Neuroscience. 1998;83:481–487. doi: 10.1016/s0306-4522(97)00423-5. [DOI] [PubMed] [Google Scholar]
  • 347.Preece MA, Dalley JW, Theobald DE, Robbins TW, Reynolds GP. Region specific changes in forebrain 5-hydroxytryptamine1A and 5-hydroxytryptamine2A receptors in isolation-reared rats: an in vitro autoradiography study. Neuroscience. 2004;123:725–732. doi: 10.1016/j.neuroscience.2003.10.008. [DOI] [PubMed] [Google Scholar]
  • 348.Ago Y, Sakaue M, Baba A, Matsuda T. Selective reduction by isolation rearing of 5-HT1A receptor-mediated dopamine release in vivo in the frontal cortex of mice. J Neurochem. 2002;83:353–359. doi: 10.1046/j.1471-4159.2002.01128.x. [DOI] [PubMed] [Google Scholar]
  • 349.Fone KC, Shalders K, Fox ZD, Arthur R, Marsden CA. Increased 5-HT2C receptor responsiveness occurs on rearing rats in social isolation. Psychopharmacology (Berl) 1996;123:346–352. doi: 10.1007/BF02246645. [DOI] [PubMed] [Google Scholar]
  • 350.Wright IK, Ismail H, Upton N, Marsden CA. Effect of isolation rearing on 5-HT agonist-induced responses in the rat. Psychopharmacology (Berl) 1991;105:259–263. doi: 10.1007/BF02244319. [DOI] [PubMed] [Google Scholar]
  • 351.Advani T, Hensler JG, Koek W. Effect of early rearing conditions on alcohol drinking and 5-HT(1A) receptor function in C57BL/6J mice. Int J Neuropsychoph. 2007;10:595–607. doi: 10.1017/S1461145706007401. [DOI] [PubMed] [Google Scholar]
  • 352.Arakawa H. The effects of age and isolation period on two phases of behavioral response to foot shock in isolation-reared rats. Developmental Psychobiology. 2002;41:15–24. doi: 10.1002/dev.10040. [DOI] [PubMed] [Google Scholar]
  • 353.Bakshi VP, Geyer MA. Ontogeny of isolation rearing-induced deficits in sensorimotor gating in rats. Physiology & Behavior. 1999;67:385–392. doi: 10.1016/s0031-9384(99)00082-7. [DOI] [PubMed] [Google Scholar]
  • 354.Klein ZA, Padow VA, Romeo RD. The Effects of Stress on Play and Home Cage Behaviors in Adolescent Male Rats. Developmental Psychobiology. 2010;52:62–70. doi: 10.1002/dev.20413. [DOI] [PubMed] [Google Scholar]
  • 355.Krebs-Thomson K, Giracello D, Solis A, Geyer MA. Post-weaning handling attenuates isolation-rearing induced disruptions of prepulse inhibition in rats. Behavioural Brain Research. 2001;120:221–224. doi: 10.1016/s0166-4328(00)00374-0. [DOI] [PubMed] [Google Scholar]
  • 356.Varty GB, Braff DL, Geyer MA. Is there a critical developmental 'window' for isolation rearing-induced changes in prepulse inhibition of the acoustic startle response? Behavioural Brain Research. 1999;100:177–183. doi: 10.1016/s0166-4328(98)00129-6. [DOI] [PubMed] [Google Scholar]
  • 357.Leussis MP, Andersen SL. Is adolescence a sensitive period for depression? Behavioral and neuroanatomical findings from a social stress model. Synapse. 2008;62:22–30. doi: 10.1002/syn.20462. [DOI] [PubMed] [Google Scholar]
  • 358.Leussis MP, Lawson K, Stone K, Andersen SL. The enduring effects of an adolescent social stressor on synaptic density, part II: Poststress reversal of synaptic loss in the cortex by adinazolam and MK-801. Synapse. 2008;62:185–192. doi: 10.1002/syn.20483. [DOI] [PubMed] [Google Scholar]
  • 359.Lukkes JL, Mokin MV, Scholl JL, Forster GL. Adult rats exposed to early-life social isolation exhibit increased anxiety and conditioned fear behavior, and altered hormonal stress responses. Horm Behav. 2009;55:248–256. doi: 10.1016/j.yhbeh.2008.10.014. [DOI] [PubMed] [Google Scholar]
  • 360.Lukkes JL, Summers CH, Scholl JL, Renner KJ, Forster GL. Early Life Social Isolation Alters Corticotropin-Releasing Factor Responses in Adult Rats. Neuroscience. 2009;158:845–855. doi: 10.1016/j.neuroscience.2008.10.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 361.Lukkes J, Vuong S, Scholl J, Oliver H, Forster G. Corticotropin-Releasing Factor Receptor Antagonism within the Dorsal Raphe Nucleus Reduces Social Anxiety-Like Behavior after Early-Life Social Isolation. Journal of Neuroscience. 2009;29:9955–9960. doi: 10.1523/JNEUROSCI.0854-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 362.Pascual R, Zamora-Leon SP, Valero-Cabre A. Effects of postweaning social isolation and re-socialization on the expression of vasoactive intestinal peptide (VIP) and dendritic development in the medial prefrontal cortex of the rat. Acta Neurobiol Exp. 2006;66:7–14. doi: 10.55782/ane-2006-1582. [DOI] [PubMed] [Google Scholar]
  • 363.Razin A. CpG methylation, chromatin structure and gene silencing - a three-way connection. Embo J. 1998;17:4905–4908. doi: 10.1093/emboj/17.17.4905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 364.Banerjee D, Slack F. Control of developmental timing by small temporal RNAs: a paradigm for RNA-mediated regulation of gene expression. Bioessays. 2002;24:119–129. doi: 10.1002/bies.10046. [DOI] [PubMed] [Google Scholar]
  • 365.McGowan PO, Meaney MJ, Szyf M. Diet and the epigenetic (re)programming of phenotypic differences in behavior. Brain Research. 2008;1237:12–24. doi: 10.1016/j.brainres.2008.07.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293:1074–1080. doi: 10.1126/science.1063127. [DOI] [PubMed] [Google Scholar]
  • 367.Razin A, Riggs AD. DNA Methylation and Gene-Function. Science. 1980;210:604–610. doi: 10.1126/science.6254144. [DOI] [PubMed] [Google Scholar]
  • 368.Szyf M. Early life, the epigenome and human health. Acta Paediatr. 2009;98:1082–1084. doi: 10.1111/j.1651-2227.2009.01382.x. [DOI] [PubMed] [Google Scholar]
  • 369.Meaney MJ, Szyf M. Maternal care as a model for experience-dependent chromatin plasticity? Trends Neurosci. 2005;28:456–463. doi: 10.1016/j.tins.2005.07.006. [DOI] [PubMed] [Google Scholar]
  • 370.Szyf M. The dynamic epigenome and its implications in toxicology. Toxicol Sci. 2007;100:7–23. doi: 10.1093/toxsci/kfm177. [DOI] [PubMed] [Google Scholar]
  • 371.Szyf M. DNA methylation, the early-life social environment and behavioral disorders. J Neurodev Disord. 2011;3:238–249. doi: 10.1007/s11689-011-9079-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Szyf M, Meaney MJP, Turecki G, Hallet M, Hertzman C, Power C, et al. Epigenetic mechanisms mediating the long-term impact on behavior of the social environment in early life. Amino Acids. 2009;37:16–17. [Google Scholar]
  • 373.Murgatroyd C, Patchev AV, Wu Y, Micale V, Bockmuhl Y, Fischer D, et al. Dynamic DNA methylation programs persistent adverse effects of early-life stress. Nat Neurosci. 2009;12:1559-U108. doi: 10.1038/nn.2436. [DOI] [PubMed] [Google Scholar]
  • 374.Levi-Montalcini R. Tissue And Nerve Growth Promoting Factors. Biological Aspects Of Specific Growth Promoting Factors. Proc R Soc Med. 1965;58:357–360. [PMC free article] [PubMed] [Google Scholar]
  • 375.Krech D, Rosenzweig MR, Bennett EL. Effects of complex environment and blindness on ratbrain. An experiment in replicate. Arch Neurol. 1963;8:402–412. doi: 10.1001/archneur.1963.00460040073007. [DOI] [PubMed] [Google Scholar]
  • 376.Cancedda L, Putignano E, Sale A, Viegi A, Berardi N, Maffei L. Acceleration of visual system development by environmental enrichment. J Neurosci. 2004;24:4840–4848. doi: 10.1523/JNEUROSCI.0845-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 377.Pinaud R, Tremere LA, Penner MR, Hess FF, Robertson HA, Currie RW. Complexity of sensory environment drives the expression of candidate-plasticity gene, nerve growth factor induced-A. Neuroscience. 2002;112:573–582. doi: 10.1016/s0306-4522(02)00094-5. [DOI] [PubMed] [Google Scholar]
  • 378.Milbrandt J. A nerve growth factor-induced gene encodes a possible transcriptional regulatory factor. Science. 1987;238:797–799. doi: 10.1126/science.3672127. [DOI] [PubMed] [Google Scholar]
  • 379.Pinaud R, Penner MR, Robertson HA, Currie RW. Upregulation of the immediate early gene arc in the brains of rats exposed to environmental enrichment: implications for molecular plasticity. Mol Brain Res. 2001;91:50–56. doi: 10.1016/s0169-328x(01)00121-8. [DOI] [PubMed] [Google Scholar]
  • 380.Pinaud R, Tremere LA, Penner MR, Hess FF, Barnes S, Robertson HA, et al. Plasticity-driven gene expression in the rat retina. Mol Brain Res. 2002;98:93–101. doi: 10.1016/s0169-328x(01)00328-x. [DOI] [PubMed] [Google Scholar]
  • 381.Solinas M, Thiriet N, El Rawas R, Lardeux V, Jaber M. Environmental Enrichment During Early Stages of Life Reduces the Behavioral, Neurochemical, and Molecular Effects of Cocaine. Neuropsychopharmacology. 2009;34:1102–1111. doi: 10.1038/npp.2008.51. [DOI] [PubMed] [Google Scholar]
  • 382.Ali AEA, Wilson YM, Murphy M. A single exposure to an enriched environment stimulates the activation of discrete neuronal populations in the brain of the fos-tau-lacZ mouse. Neurobiology of Learning and Memory. 2009;92:381–390. doi: 10.1016/j.nlm.2009.05.004. [DOI] [PubMed] [Google Scholar]
  • 383.Fischer A, Sananbenesi F, Wang XY, Dobbin M, Tsai LH. Recovery of learning and memory is associated with chromatin remodelling. Nature. 2007;447:178-U2. doi: 10.1038/nature05772. [DOI] [PubMed] [Google Scholar]
  • 384.Kondo M, Gray LJ, Pelka GJ, Christodoulou J, Tam PPL, Hannan AJ. Environmental enrichment ameliorates a motor coordination deficit in a mouse model of Rett syndrome - Mecp2 gene dosage effects and BDNF expression. European Journal of Neuroscience. 2008;27:3342–3350. doi: 10.1111/j.1460-9568.2008.06305.x. [DOI] [PubMed] [Google Scholar]
  • 385.Lonetti G, Angelucci A, Morando L, Boggio EM, Giustetto M, Pizzorusso T. Early Environmental Enrichment Moderates the Behavioral and Synaptic Phenotype of MeCP2 Null Mice. Biol. Psychiat. 2010;67:657–665. doi: 10.1016/j.biopsych.2009.12.022. [DOI] [PubMed] [Google Scholar]
  • 386.Bibancos T, Jardim DL, Aneas I, Chiavegatto S. Social isolation and expression of serotonergic neurotransmission-related genes in several brain areas of male mice. Genes Brain Behav. 2007;6:529–539. doi: 10.1111/j.1601-183X.2006.00280.x. [DOI] [PubMed] [Google Scholar]
  • 387.Pan YL, Liu Y, Young KA, Zhang ZB, Wang ZX. Post-weaning social isolation alters anxiety-related behavior and neurochemical gene expression in the brain of male prairie voles. Neuroscience Letters. 2009;454:67–71. doi: 10.1016/j.neulet.2009.02.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 388.Cirulli F, Micera A, Alleva E, Aloe L. Early maternal separation increases NGF expression in the developing rat hippocampus. Pharmacol Biochem Behav. 1998;59:853–858. doi: 10.1016/s0091-3057(97)00512-1. [DOI] [PubMed] [Google Scholar]
  • 389.Eghbal-Ahmadi M, Hatalski CG, Avishai-Eliner S, Baram TZ. Corticotropin releasing factor receptor type II (CRF2) messenger ribonucleic acid levels in the hypothalamic ventromedial nucleus of the infant rat are reduced by maternal deprivation. Endocrinology. 1997;138:5048–5051. doi: 10.1210/endo.138.11.5647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 390.Lippmann M, Bress A, Nemeroff CB, Plotsky PM, Monteggia LM. Long-term behavioural and molecular alterations associated with maternal separation in rats. European Journal of Neuroscience. 2007;25:3091–3098. doi: 10.1111/j.1460-9568.2007.05522.x. [DOI] [PubMed] [Google Scholar]
  • 391.Ognibene E, Adriani W, Caprioli A, Ghirardi O, Ali SF, Aloe L, et al. The effect of early maternal separation on brain derived neurotrophic factor and monoamine levels in adult heterozygous reeler mice. Prog Neuro-Psychoph. 2008;32:1269–1276. doi: 10.1016/j.pnpbp.2008.03.023. [DOI] [PubMed] [Google Scholar]
  • 392.Weaver ICG, Cervoni N, Champagne FA, D'Alessio AC, Sharma S, Jr, S, et al. Epigenetic programming by maternal behavior. Nat Neurosci. 2004;7:847–854. doi: 10.1038/nn1276. [DOI] [PubMed] [Google Scholar]
  • 393.Meaney MJ, Szyf M. Environmental programming of stress responses through DNA methylation: life at the interface between a dynamic environment and a fixed genome. Dialogues in Clinical Neuroscience. 2005;7:103–123. doi: 10.31887/DCNS.2005.7.2/mmeaney. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 394.Weaver ICG, Champagne FA, Brown SE, Dymov S, Sharma S, Meaney MJ, et al. Reversal of maternal programming of stress responses in adult offspring through methyl supplementation: Altering epigenetic marking later in life. Journal of Neuroscience. 2005;25:11045–11054. doi: 10.1523/JNEUROSCI.3652-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 395.Champagne FA, Weaver ICG, Diorio J, Dymov S, Szyf M, Meaney MJ. Maternal care associated with methylation of the estrogen receptor-alpha 1b promoter and estrogen receptor-alpha expression in the medial preoptic area of female offspring. Endocrinology. 2006;147:2909–2915. doi: 10.1210/en.2005-1119. [DOI] [PubMed] [Google Scholar]
  • 396.Champagne FA, Weaver ICG, Diorio J, Sharma S, Meaney MJ. Natural variations in maternal care are associated with estrogen receptor alpha expression and estrogen sensitivity in the medial preoptic area. Endocrinology. 2003;144:4720–4724. doi: 10.1210/en.2003-0564. [DOI] [PubMed] [Google Scholar]
  • 397.McGowan PO, Suderman M, Sasaki A, Huang TCT, Hallett M, Meaney MJ, et al. Broad Epigenetic Signature of Maternal Care in the Brain of Adult Rats. Plos One. 2011;6 doi: 10.1371/journal.pone.0014739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 398.Laplante P, Diorio J, Meaney MJ. Serotonin regulates hippocampal glucocorticoid receptor expression via a 5-HT7 receptor. Dev Brain Res. 2002;139:199–203. doi: 10.1016/s0165-3806(02)00550-3. [DOI] [PubMed] [Google Scholar]
  • 399.Meaney MJ, Diorio J, Francis D, Weaver S, Yau J, Chapman K, et al. Postnatal handling increases the expression of cAMP-inducible transcription factors in the rat hippocampus: The effects of thyroid hormones and serotonin. Journal of Neuroscience. 2000;20:3926–3935. doi: 10.1523/JNEUROSCI.20-10-03926.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 400.Weaver ICG, D'Alessio AC, Brown SE, Hellstrom IC, Dymov S, Sharma S, et al. The transcription factor nerve growth factor-inducible protein A mediates epigenetic programming: Altering epigenetic marks by immediate-early genes. Journal of Neuroscience. 2007;27:1756–1768. doi: 10.1523/JNEUROSCI.4164-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 401.Chen WG, Chang Q, Lin YX, Meissner A, West AE, Griffith EC, et al. Derepression of BDNF transcription involves calcium-dependent phosphorylation of MeCP2. Science. 2003;302:885–889. doi: 10.1126/science.1086446. [DOI] [PubMed] [Google Scholar]
  • 402.Weaver ICG, Meaney MJ, Szyf M. Maternal care effects on the hippocampal transcriptome and anxiety-mediated behaviors in the offspring that are reversible in adulthood. P Natl Acad Sci USA. 2006;103:3480–3485. doi: 10.1073/pnas.0507526103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 403.Belay H, Burton CL, Lovic V, Meaney MJ, Sokolowski M, Fleming AS. Early Adversity and Serotonin Transporter Genotype Interact With Hippocampal Glucocorticoid Receptor mRNA Expression, Corticosterone, and Behavior in Adult Male Rats. Behavioral Neuroscience. 2011;125:150–160. doi: 10.1037/a0022891. [DOI] [PubMed] [Google Scholar]
  • 404.Champoux M, Bennett A, Shannon C, Higley JD, Lesch KP, Suomi SJ. Serotonin transporter gene polymorphism, differential early rearing, and behavior in rhesus monkey neonates. Mol Psychiatr. 2002;7:1058–1063. doi: 10.1038/sj.mp.4001157. [DOI] [PubMed] [Google Scholar]
  • 405.Caspi A, Sugden K, Moffitt TE, Taylor A, Craig IW, Harrington H, et al. Influence of life stress on depression: Moderation by a polymorphism in the 5-HTT gene. Science. 2003;301:386–389. doi: 10.1126/science.1083968. [DOI] [PubMed] [Google Scholar]
  • 406.Meaney MJ. Maternal care, gene expression, and the transmission of individual differences in stress reactivity across generations. Annu Rev Neurosci. 2001;24:1161–1192. doi: 10.1146/annurev.neuro.24.1.1161. [DOI] [PubMed] [Google Scholar]
  • 407.McGowan PO, Sasaki A, Huang TCT, Unterberger A, Suderman M, Ernst C, et al. Promoter-Wide Hypermethylation of the Ribosomal RNA Gene Promoter in the Suicide Brain. Plos One. 2008;3 doi: 10.1371/journal.pone.0002085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 408.McGowan PO, Sasaki A, D'Alessio AC, Dymov S, Labonte B, Szyf M, et al. Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nat Neurosci. 2009;12:342–348. doi: 10.1038/nn.2270. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES