Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Aug 26.
Published in final edited form as: Ann N Y Acad Sci. 2008 Oct;1141:105–130. doi: 10.1196/annals.1441.030

Chronic Stress, Drug Use, and Vulnerability to Addiction

Rajita Sinha 1
PMCID: PMC2732004  NIHMSID: NIHMS123829  PMID: 18991954

Abstract

Stress is a well-known risk factor in the development of addiction and in addiction relapse vulnerability. A series of population-based and epidemiological studies have identified specific stressors and individual-level variables that are predictive of substance use and abuse. Preclinical research also shows that stress exposure enhances drug self-administration and reinstates drug seeking in drug-experienced animals. The deleterious effects of early life stress, child maltreatment, and accumulated adversity on alterations in the corticotropin releasing factor and hypothalamic-pituitary-adrenal axis (CRF/HPA), the extrahypothalamic CRF, the autonomic arousal, and the central noradrenergic systems are also presented. The effects of these alterations on the corticostriatal-limbic motivational, learning, and adaptation systems that include mesolimbic dopamine, glutamate, and gamma-amino-butyric acid (GABA) pathways are discussed as the underlying pathophysiology associated with stress-related risk of addiction. The effects of regular and chronic drug use on alterations in these stress and motivational systems are also reviewed, with specific attention to the impact of these adaptations on stress regulation, impulse control, and perpetuation of compulsive drug seeking and relapse susceptibility. Finally, research gaps in furthering our understanding of the association between stress and addiction are presented, with the hope that addressing these unanswered questions will significantly influence new prevention and treatment strategies to address vulnerability to addiction.

Keywords: chronic stress, early life stress, addiction risk, relapse, craving, mesolimbic dopamine

Introduction

Stress has long been known to increase vulnerability to addiction. The last decade has led to a dramatic increase in understanding the underlying mechanisms for this association. Behavioral and neurobiological correlates are being identified, and some evidence of molecular and cellular changes associated with chronic stress and addiction has been identified. Human studies have benefited from the emergence of sophisticated brain-imaging tools and the cross examination of laboratory-induced methods of stress and craving and their association to specific brain regions associated with reward and addiction risk. This paper focuses primarily on the association between stress and addiction in humans but also draws from the broader animal literature to support the proposed hypotheses. A definition of stress and its neural underpinnings is presented with specific emphasis on its effects on motivation and behavior. In the context of strong epidemiological evidence linking early-childhood and adult adversity and risk of addiction, results from basic and human research that point to putative mechanisms underlying this association are presented. A critical role is seen for prefrontal circuits involved in adaptive learning and executive function, including controlling distress and desires/impulses, in the association between stress and addiction risk. However, several questions remain unanswered in understanding stress-related addiction risk, and these are reviewed in order to inform future research. Finally, the effects of chronic drug use on stress and reward pathways particularly with respect to relapse risk are examined. Future directions in addressing stress-related relapse risk in clinical settings are also discussed.

Stress, Emotions, and Adaptive Behavior

The term “stress” refers to processes involving perception, appraisal, and response to harmful, threatening, or challenging events or stimuli.13 Stress experiences can be emotionally or physiologically challenging and activate stress responses and adaptive processes to regain homeostasis.2,46 Examples of emotional stressors include interpersonal conflict, loss of relationship, death of a close family member, and loss of a child. Common physiological stressors are hunger or food deprivation, sleep deprivation or insomnia, extreme hyper- or hypothermia, and drug withdrawal states. In addition, regular and binge use of many psychoactive drugs serve as pharmacological stressors. This kind of conceptualization allows the separate consideration of (1) internal and external events or stimuli that exert demands or load on the organism; (2) the neural processes that evaluate the demands and assess availability of adaptive resources to cope with the demands (appraisal); (3) the subjective, behavioral, and physiological activity that signal stress to the organism; (4) neuroadaptations in emotional and motivational brain systems associated with chronic stress; and (5) behavioral, cognitive, and physiological adaptation in response to stressors.

While stress is often associated with negative affect and distress, it can include “good stress” which is based on external and internal stimuli that are mild/moderately challenging but limited in duration and results in cognitive and behavioral responses that generate a sense of mastery and accomplishment, and can be perceived as pleasant and exciting.1,3,6,7 Such situations rely on adequate motivational and executive functioning to achieve goal-directed outcomes and homeostasis.3,6,8 However, the more prolonged, repeated, or chronic the stress—for example, states associated with increased intensity or persistence of distress—the greater the uncontrollability and unpredictability of the stressful situation, lower the sense of mastery or adaptability, and greater the magnitude of the stress response and risk for persistent homeostatic dysregulation.1,6,911 Thus, the dimensions of intensity, controllability, predictability, mastery, and adaptability are important in understanding the role of stress in increasing risk of maladaptive behaviors such as addiction.

The perception and appraisal of stress relies on specific aspects of the presenting external or internal stimuli, personality traits, availability of internal resources (including physiological condition of the individual), prior emotional state (including beliefs and expectancies), and specific brain regions mediating the appraisal of stimuli as distressing, and the resulting physiological, behavioral, and emotional experiences and adaptive responses. Brain regions such as the amygdala, hippocampus, insula, and orbitofrontal, medial prefrontal, and cingulate cortices are involved in the perception and appraisal of emotional and stressful stimuli, and the brain stem (locus ceruleus and related arousal regions), hypothalamus, thalamus, striatal, and limbic regions are involved in physiological and emotional responses. Together these regions contribute to the experience of distress. Physiological responses are manifested through the two major stress pathways, namely corticotropin releasing factor (CRF) released from the paraventricular nucleus (PVN) of the hypothalamus, which stimulates adrenocorticotrophin hormone from the anterior pituitary, which subsequently stimulates the secretion of cortisol/corticosterone from the adrenal glands, and the autonomic nervous system, which is coordinated via the sympathoadrenal medulary (SAM) systems.4,12

In addition, CRF has extensive influence in extrahypothalamic regions across the corticostriatal-limbic regions and plays a critical role in modulating subjective and behavioral stress responses.13 Furthermore, central catecholamines, particularly noradrenaline and dopamine, are involved in modulating brain motivational pathways (including the ventral tegmental area or VTA, nucleus accumbens [NAc], and the medial prefrontal [mPFC] regions) that are important in regulating distress, exerting cognitive and behavioral control, and negotiating behavioral and cognitive responses critical for adaptation and homeostasis.8,14,15 The hypothalamic and extrahypothalamic CRF pathways and central catechoamines target brain motivational pathways to critically affect adaptive and homeostatic processes. For example, different parts of the medial prefrontal cortex are involved in higher cognitive or executive control functions, such as controlling and inhibiting impulses, regulating distress, focusing and shifting attention, monitoring behavior, linking behaviors and consequences over time, considering alternatives before acting, and decision-making responses.16,17 Psychosocial and behavioral scientists have elegantly shown that with increasing levels of emotional and physiological stress or negative affect, there is a decrease in behavioral control and increases in impulsivity, and with increasing levels of distress, and chronicity of stress, greater the risk of maladaptive behaviors.1827 Neurobiological evidence shows that with increasing levels of stress, there is a decrease in prefrontal functioning and increased limbic-striatal level responding, which perpetuates low behavioral and cognitive control.28,29 Thus, the motivational brain pathways are key targets of brain stress chemicals and provide an important potential mechanism by which stress affects addiction vulnerability.

Stress and the Development of Addictive Behaviors

There is a substantial literature on the significant association between acute and chronic stress and the motivation to abuse addictive substances (see30 for review). Many of the major theories of addiction also identify an important role of stress in addiction processes. These range from psychological models of addiction that view drug use and abuse as a coping strategy to deal with stress, to reduce tension, to self medicate, and to decrease withdrawal-related distress,3137 to neurobiological models that propose incentive sensitization and stress allostasis concepts to explain how neuroadaptations in reward, learning, and stress pathways may enhance craving, loss of control, and compulsion, the key components in the transition from casual use of substances to the inability to stop chronic use despite adverse consequences, a key feature of addiction.3840 In this section, we review the converging lines of evidence that point to the critical role that stress plays in increasing addiction vulnerability.

Chronic Adversity and Increased Vulnerability to Drug Use

There is considerable evidence from population-based and clinical studies supporting a positive association between psychosocial adversity, negative affect, and chronic distress and addiction vulnerability. The evidence in this area can be categorized into three broad types. The first includes prospective studies demonstrating that adolescents facing high recent negative life events show increased levels of drug use and abuse.4155 Negative life events such as loss of parent, parental divorce and conflict, low parental support, physical violence and abuse, emotional abuse and neglect, isolation and deviant affiliation, and single-parent family structure have all been associated with increased risk of substance abuse.

The second type of evidence is the association between trauma and maltreatment, negative affect, chronic distress, and risk of substance abuse. Overwhelming evidence exists for an increased association between childhood sexual and physical abuse and victimization and increased drug use and abuse.5660 There is also some evidence that recent negative life events and physical and sexual abuse each exert somewhat independent risk on addiction vulnerability.58 In addition to sexual and physical abuse, negative affect and chronic distress states are predictive of addiction vulnerability. Findings indicate that negative affect, including temperamental negative emotionality, is associated with substance abuse risk.6167 Several studies have also shown a significant association between prevalence of mood and anxiety disorders, including post-traumatic stress disorder (PTSD), behavioral conduct problems and increased risk of substance use disorders.6878 As stress is significantly associated with prevalence of mood and anxiety disorders and chronic psychiatric distress,79,80 these associations raise the issue of whether psychiatric disorders conceptualized as chronic distress states may largely account for the significant association between stress and substance use disorders.

In the third type of evidence from population studies, recent research has examined lifetime exposure to stressors and the impact of cumulative adversity on addiction vulnerability after accounting for a number of control factors such as race/ethnicity, gender, socioeconomic status, prior drug abuse, prevalence of psychiatric disorders, family history of substance use, and behavioral and conduct problems.81,82 Cumulative adversity or stress was assessed using a checklist method and by counting the number of different events that were experienced in a given period during the lifespan. The effects of distal (events occurring more than 1 year prior) and proximal stress experiences (events during the most recent 1-year period), and their effects on meeting criteria for substance use disorders were also assessed. The findings indicate that the cumulative number of stressful events was significantly predictive of alcohol and drug dependence in a dose-dependent manner, even after accounting for control factors. Both distal and proximal events significantly and independently affected addiction vulnerability. Furthermore, the dose-dependent effects of cumulative stressors on risk for addiction existed for both genders and for Caucasian, African-American, and Hispanic race/ethnic groups. The types of adverse events significantly associated with addiction vulnerability were parental divorce or conflict, abandonment, forced to live apart from parents, loss of child by death or removal, unfaithfulness of significant other, loss of home to natural disaster, death of a close one, emotional abuse or neglect, sexual abuse, rape, physical abuse by parent, caretaker, family member, spouse, or significant other, victim of gun shooting or other violent acts, and observing violent victimization. These represent highly stressful and emotionally distressing events, which are typically uncontrollable and unpredictable in nature. Table 1 summarizes the types of life events, chronic stressors, maltreatment, and individual level variables associated with addiction risk.

TABLE 1.

Types of Adverse Life Events, Trauma, Chronic Stressors, and Individual-Level Variables Predictive of Addiction Risk

Loss of parent Physical neglect Negative emotionality
Parental divorce and conflict Physical abuse by parent/caretaker/family member/spouse/significant other Poor behavioral control
Isolation & abandonment Poor emotional control
Single-parent family structure Emotional abuse and neglect
Forced to live apart from parents Sexual abuse
Loss of child by death or removal Rape
Unfaithfulness of significant other
Loss of home to natural disaster
Death of significant other/close family member
Victim of gun shooting or other violent acts
Observing violent victimization

Stress Exposure Increases Initiation and Escalation of Drug Self-Administration

There is some evidence from animal studies to support the notion that acute exposure to stress increases initiation and escalation of drug use and abuse (see30,83 for reviews). For example, in animal models, social defeat stress, social isolation, tailpinch and foot-shock, restraint stress, and novelty stress are known to enhance acquisition of opiates, alcohol, and psychostimulant self-administration, with caveats relating to stressor type, genetic background of animals, and variations by drug type (see8487 for reviews). Also, although there are some negative findings, other evidence indicates that early life stress, using procedures such as neonatal isolation or maternal separation, and prolonged and repeated stressors representing chronic stress experiences, enhances self-administration of nicotine, psychostimulants, and alcohol and/or their acute behavioral effects.8893 Notably, sex plays an important role in stress-related sensitivity to the reinforcing effects of drugs and in stress enhancement of drug self-administration.9397 In humans, there is substantial evidence from prospective and longitudinal studies to support the effects of stress on drug use initiation and escalation in adolescents and young adults.24,98109 Furthermore, there are sex differences in the effects of early trauma and maltreatment on the increased risk of addiction.74,110114 Laboratory studies examining effects of stress exposure on drug use are limited to legal drugs such as alcohol and nicotine, for ethical reasons. Nonetheless, there is evidence that stress increases drinking and nicotine smoking (see83 for review), but the effects of drinking history, history of adversity, social stress, and expectancies are known to play a role in these experimental studies.

Possible Mechanisms Underlying Stress Effects on Addiction Vulnerability

As evidence using diverse approaches has accumulated in support of a significant effect of stress on risk of addiction, this section examines research on neurobiological links between stress and reward pathways activated by abusive drugs. It is well known that the reinforcing properties of drugs of abuse involve their activation of the mesolimbic dopaminergic (DA) pathways, which include dopamine neurons originating in the ventral tegmental area and extending to the ventral striatum and the prefrontal cortex (PFC).115117 This pathway is also involved in assigning salience to stimuli, in reward processing, and in learning and adaptation.14,118 Human brain imaging studies also support the role of these systems in drug reward, as psychostimulants, alcohol, opioids, and nicotine all activate the mesolimbic DA systems, in particular, the ventral and dorsal striatum, and such activity has been associated with the drug ratings of high or euphoria and craving.119126

However, stress exposure and increased levels of glucocorticoids (GC) also enhance dopamine release in the NAc.127132 Suppression of GC by adrenalectomy reduces extracellular levels of dopamine under basal conditions and in responses to stress and psychostimulants.131,133 However, chronic GC inhibits DA synthesis and turnover in the NAc,134 suggesting that alterations in the hypothalamic-pituitary-adrenal (HPA) axis and glucocorticoids can significantly affect DA transmission. There is also evidence that, like drugs of abuse, stress and concomitant increases in CRF and glucocorticoids enhance glutamate activity in the VTA, which in turn enhances activity of dopaminergic neurons.135138 Human brain imaging studies have further shown that stress-related increases in cortisol are associated with dopamine accumulation in the ventral striatum,125,139 and some evidence also reveals that amphetamine-induced increases in cortisol are associated with both dopamine binding in the ventral striatum and with ratings of amphetamine-induced euphoria.140 Given that both stress and drugs of abuse activate the mesolimbic pathways, it is not surprising that each results in synaptic adaptations in VTA dopamine neurons and in morphological changes in the medial prefrontal cortex.87,136,141,142

In addition to a role in reward, a growing body of human imaging studies and preclinical data indicate that the ventral striatum is also involved in aversive conditioning, in experience of aversive, pain stimuli, and in anticipation of aversive stimuli.143146 Such evidence points to a role for the mesolimbic dopamine pathways beyond reward processing, and one that more broadly involves motivation and attention to behavioral response during salient (aversive or appetitive) events.147150 Furthermore, additional regions connected to the mesolimbic DA pathways and involved in reward, learning, and adaptive and goal-directed behaviors are the amygdala, hippocampus, insula, and related corticolimbic regions.118,151 These regions, along with the mesolimbic DA pathways, play an important role in interoception, emotions and stress processing, impulse control and decision making, and in the addictive properties of drugs of abuse.29,152

Stress Mechanisms Involved in Acquisition of Drug Self-Administration

Research has also examined whether stress-related increases in acquisition of drug self-administration are mediated by corticosterone (cortisol in humans). Findings indicate that HPA-activated corticosterone release is important for acquisition of drug self-administration.131,153155 Corticosterone administration also facilitates psychomotor stimulant effects of cocaine and morphine.156 Furthermore, GC receptor antagonists injected into the VTA decrease morphine-induced locomotor activity,157 suggesting that activity of GC receptors in the VTA could mediate dopamine-dependent behavioral effects. Mice with deletion of the GR gene show a dose-dependent decrease in motivation to self-administer cocaine.158 These data suggest that HPA-related corticosterone release could at least partially mediate the dopamine increases seen after drug administration.

Although in nonhuman primates the link between cortisol, dopamine, and drug self-administration has not been reported, there is evidence that stress related to social subordination is associated with lower levels of D2 receptors and higher cocaine self-administration.159 In humans, positive emission tomography (PET) studies using [11C]raclopride indicate that acute stress exposure increases dopamine release in the ventral striatum (VS). For example, in a small-sample study, Pruessner and colleagues (2004)139 found that healthy individuals with low early-life maternal care showed greater dopamine release in the ventral striatum during an acute psychological stress task as compared to those with a history of high early-life maternal care. Furthermore, cortisol response during the stress task was correlated significantly (r = .78) to VS dopamine release. Oswald and colleagues (2005)125 also demonstrated that acute amphetamine challenge-related subjective “high” responses and concomitant increase in dopamine in the VS were each significantly associated with amphetamine-induced cortisol responses. More recently, the same group has also shown a similar significant relationship between cortisol levels and dopamine release in the VS using a psychological stress task.140 Although these data support the link between stress/cortisol and dopamine transmission, human research linking stress-induced changes in VS activity or dopamine binding and risk of addictive behavior is needed to directly establish the association between stress, mesolimbic dopamine, and addiction risk.

Early Life and Chronic Stress, Dopamine Systems, and Drug Self-Administration

There is growing evidence from basic science studies that early-life stress and chronic stress significantly affect the mesolimbic dopamine pathways and play a role in drug self-administration. Repeated and prolonged exposure to maternal separation (MS) in neonatal rats significantly alters the development of central CRF pathways.11 These animals as adults show exaggerated HPA and behavioral responses to stress.160,161 Such physiological and behavioral changes are associated with altered CRF mRNA expression in the PVN, increased CRF-like immunoreactivity in the locus ceruleus (LC), and increased CRF receptor levels in the LC and raphe nuclei.11 The adult animals also show decreased negative feedback sensitivity to glucocorticoids,162 and these changes are accompanied by decreased GC receptor expression in the hippocampus and frontal cortex.11,163 Decreased GABA receptor levels in noradrenergic cell body regions in the LC and decreased central benzodiazepine (CBZ) receptor levels in the LC and the amygdala have also been reported.164 More importantly, MS rats show significantly elevated DA responses to acute stress along with increased stress-induced behavioral sensitization and robust behavioral sensitization to psychostimulant administration.11,143,165 This cross-sensitization of stress and drugs of abuse is associated with enhanced release of DA in the NAc, lower NAc-core, and striatal DA transporter sites, and reduced D3 receptor binding sites and mRNA levels in the NAc shell.166168 In addition, chronic norepinephrine deficiency induces changes similar to sensitization that could be related to alterations in DA-signaling pathways.169,170

Early-life stress and prolonged and repeated stress also adversely affect development of the prefrontal cortex, a region that is highly dependent on environmental experiences for maturation.171 The PFC, and particularly the right PFC, plays an important role both in activating the HPA axis and autonomic responses to stress and in regulating these responses.171 For example, lesions of the ventromedial PFC result in enhanced HPA and autonomic responses to stress. High levels of glucocorticoid receptors are also found in the PFC, and chronic GC treatment results in a dramatic dendritic reorganization of PFC neurons similar to that seen in the hippocampus.172,173 Furthermore, early postnatal MS and social isolation result in abnormally high synaptic densities in the PFC and altered densities of DA and serotonin (5-HT) terminals throughout the medial PFC.174 Social defeat stress also alters feedback from the PFC and contributes to drug self-administration.84 Human studies on the neurobiological effects of child maltreatment document neuroendocrine changes as well as alterations in size and volume of prefrontal, thalamic, and cerebellar regions associated with maltreatment and with initiation of addiction.175,176 Together, the data presented in this section highlight the significance of stress effects on mesolimbic and prefrontal regions involved in stress related behavioral control.

Stress, Self-Control, and Addiction Vulnerability

High emotional stress is associated with loss of control over impulses and an inability to inhibit inappropriate behaviors and to delay gratification.20,177,178 Neurobiological data indicate that stress impairs catecholamine modulation of prefrontal circuits, which in turn impairs executive functions like working memory and self-control.17,28,179 There is also growing evidence that adolescents at risk for substance abuse who have experienced several of the stressors listed in Table 1 are more likely to show decreased emotional and behavioral control, and decreased self-control is associated with risk of substance abuse and other maladaptive behaviors.104,152,180,181 Adolescents at risk for substance abuse are known to have decreased executive functioning, low behavioral and emotional control, poor decision making, and greater levels of deviant behavior and impulsivity.24,152,182184 The corticostriatal-limbic dopamine pathways have been associated with impulsivity, decision making, and addiction risk,185,186 and as discussed in previous sections, specific regions of this pathway, such as the VTA, NAc, PFC, and amygdala, are highly susceptible to stress-related signaling and plasticity associated with early-life stress and chronic stress experiences. In a recent PET imaging study, Oswald (2007)187 examined the effects of chronic stress and impulsivity on amphetamine-induced striatal dopamine release. These findings indicated that high trait impulsivity was associated with blunted right VS dopamine release. However, these effects were modified by a significant interaction with chronic life events stress. With low to moderate stress, dopamine release was greater in low than in high impulsive subjects, but with high stress, both groups showed low DA release. These findings demonstrate the important effects of stress and impulsivity on mesolimbic dopamine transmission and highlight the fact that both factors need to be carefully considered to fully understand the role of stress and impulsivity on addiction risk.

Schematic Model of Stress Effects on Addiction

Figure 1 presents a schematic model of stress effects on addiction. It highlights cross-sensitization of stress and drug abuse on specific behavioral and neurochemical responses and indicates the common neurobiological pathways upon which both stress and drugs of abuse act. Column A lists three types of vulnerability factors: (1) developmental/individual-level factors such as frontal executive function development, negative emotionality, behavioral/self-control, impulsivity, or risk taking, and altered initial sensitivity to rewarding effects of drugs; (2) stress-related vulnerability factors such as early adverse life events, trauma and child maltreatment experiences, prolonged and chronic stress experiences; and (3) genetic influences and family history of psychopathology and addiction, which have not been discussed here but have significant interactive effects on addiction risk and in emotion and stress markers.188194 Each of these factors may influence each other to significantly affect alterations in neurobiological pathways involved in stress regulation and cognitive and behavioral control (column B). Specific synaptic changes in these pathways at molecular and cellular levels118,195 provide the basis for the mechanism by which stress and individual and genetic factors in column A interact to increase risk of maladaptive behaviors represented in column C. The model suggests that stress experiences in the presence of these vulnerability factors result in maladaptive stress and self-control responses that increase addiction risk. The specific mechanism by which the maladaptive stress responding increases this risk involves dysregulation in brain stress circuits, particularly the CRF and NE systems, and their interactions with the mesocorticolimbicstriatal dopamine pathways and its modulation by glutamate and GABA.114,196,197 Furthermore, recent evidence suggests that stress regulatory molecules, including neuropeptides such as neuropeptide (NPY) endocannabinoids, and neuroactive steroids play a role in addiction vulnerability.198203

Figure 1.

Figure 1

A schematic model of stress effects on addiction, representing the cross-sensitization of stress and drugs on behavioral and neurochemical responses, that are mediated by the stress and reward pathways. Column A lists three types of vulnerability factors: (1) developmental/individual-level factors such as frontal executive function development, negative emotionality, behavioral/self control, impulsivity or risk taking, and altered initial sensitivity to rewarding effects of drugs; (2) stress-related vulnerability factors such as early adverse life events, trauma and child maltreatment experiences, prolonged and chronic stress experiences; and (3) genetic influences and family history of psychopathology. Each of these factors influences each other to significantly affect alterations in neurobiological pathways involved in stress regulation and cognitive and behavioral control (Column B). Such changes at least partially mediate the mechanisms by which stress and individual and genetic factors in column A interact to increase risk of maladaptive behaviors represented in column C when an individual is faced with stress or challenge situations.

Drug Use and Abuse and Changes in Stress and Reward Pathways

Acute and Chronic Drug Use and Changes in Stress Responses

Acute administration of the most commonly abused drugs such as alcohol, nicotine, cocaine, amphetamines, and marijuana that activate brain reward pathways (mesocorticolimbic dopaminergic systems) also activate brain stress pathways (CRF-HPA axis and the autonomic nervous system pathways) with increases in plasma adrenocorticotropic hormone (ACTH) and corticosterone, changes in heart rate and blood pressure, and skin conductance responses.204217 On the other hand, acute exposure to opiates decreases cortisol levels in humans.218,219 Regular and chronic use of these drugs is also associated with adaptations in these systems that are specific by drug. For example, changes in heart rate and heart rate variability (HRV) are reported with regular and chronic alcohol use.220222 Sustained increases in HPA axis function in the case of psychostimulants, and tolerance to the inactivating effects of the drug in the case of morphine, nicotine, and alcohol has also been reported.223226 These direct effects of drugs of abuse on major components of the physiological stress response support their classification as pharmacological stressors.

Acute withdrawal states are associated with increases in CRF levels in CSF, plasma ACTH, cortisol, norepinephrine (NE), and epinephrine (EPI) levels.38,211,216,227231 Early abstinence is associated with high basal cortisol responses and a blunted or suppressed ACTH and cortisol response to pharmacological and psychological challenges in alcoholics and chronic smokers, while hyper-responsivity of HPA hormones in response to metyrapone has been reported in opiate and cocaine addicts.232236 Furthermore, withdrawal and abstinence from chronic alcohol is also associated with altered sympathetic and parasympathetic responses,234,237239 and altered noradrenergic responses to yohimbine challenge in early abstinence from cocaine has also been observed.240 All of the above changes highlight the significant effects of drug use and abuse on physiological stress responses.

Although acute administration of drugs increases mesolimbic dopamine,241 regular and chronic use of abusive drugs and acute withdrawal states down regulate mesolimbic dopamine pathways with decreases in basal and stimulated dopamine reported in several preclinical studies.242251 Chronic use of cocaine has also been shown to dramatically alter central noradrenergic pathways in the ventral and dorsal striatum, other areas of the fore-brain, and the ventromedial prefrontal cortex.252,253 Human brain imaging studies corroborate these preclinical data, with reduced D2 receptors and dopamine transmission in the frontal and ventral striatum regions in alcoholics and cocaine abusers during acute withdrawal and protracted withdrawal (up to 3−4 months).254256 Furthermore, blunted dopamine release in the ventral striatum and anterior caudate was associated with a preference to self-administer cocaine over receiving money in human cocaine abusers.257 These changes are similar to the effects of prolonged and repeated stressors on mesolimbic dopamine and norepinephrine deficiency noted in the previous section134,187,258 and raise the question whether chronic drug effects on extrahypothalamic CRF, noradrenergic, or glucocorticoid systems may at least partially modulate these dopamine-related changes in the corticostriatal limbic dopamine pathways.

On the other hand, acute, regular, and chronic exposure to drugs results in “sensitization” or enhanced behavioral and neurochemical response to drugs and to stress. Synaptic alterations in the VTA, NAc, and medial PFC modulated by glutamate effects on dopamine neurons and CRF and noradrenergic effects on DA and non-DA pathways contribute to behavioral sensitization of stress and drugs of abuse.210,259262 In addition, increased levels of brain derived neurotrophic factor (BDNF) in the mesolimbic dopamine regions has been associated with increases in drug seeking during abstinence from chronic drug use.263,264 Furthermore, behavioral sensitization observed with drugs of abuse and with stress are associated with synaptic changes in mesolimbic dopamine regions, particularly the VTA, NAc, and amygdala, and such changes contribute to compulsive drug seeking.118,265 Thus, there are significant physiological, neurochemical, and behavioral alterations in stress and dopaminergic pathways associated with chronic drug use, which in turn could affect craving and compulsive seeking, maintenance of drug use, and relapse risk. It is not entirely clear how long these changes persist or the extent to which there is recovery or normalization of these pathways and responses in related functional responses.

Altered Stress Responses and Craving with Chronic Drug Abuse

Clinical symptoms of irritability, anxiety, emotional distress, sleep problems, dysphoria, aggressive behaviors, and drug craving are common during early abstinence from alcohol, cocaine, opiates, nicotine, and marijuana.30,266269 A mild “negative affect” and craving state ensues postwithdrawal, associated with alterations in the stress and dopamine pathways.37,197,250,270 The severity of the these symptoms has been associated with treatment outcomes, with greater dependence and abstinence severity predictive of worse treatment outcomes.271274 Drug craving or “wanting” for drug is conceptually different from other anxiety and negative affect symptoms as it comes from “desire” or a wish for a hedonic stimulus. However, with chronic drug use, the term becomes associated with a physiological need, hunger, and strong intent to seek out the desired object, thereby representative of the more compulsive aspects of craving and drug seeking identified by addicted patients.274277 In particular, craving and compulsive seeking is strongly manifested in the context of stress exposure, drug-related cues, and drug itself and can become a potent trigger for relapse.30,274,278281 Several recent models of addiction have presented the concept that this heightened craving or wanting of drug is the behavioral manifestation of molecular and cellular changes in the stress and dopamine pathways discussed in the previous section. Indeed some support for this idea comes from laboratory and imaging studies summarized below.

In my laboratory, we have examined the effects of stress and drug-related cues on drug craving in alcoholics, cocaine-dependent individuals, and naltrexone-treated, opiate-dependent individuals in recovery. Drug craving and stress responses were assessed in treatment-engaged, abstinent, addicted individuals who were exposed to stressful and nonstressful drug-cue situations and neutral relaxing situations, using personalized guided imagery procedures as the induction method.282 Our initial findings indicated that in addicted individuals, stress imagery elicited multiple emotions of fear, sadness, and anger as compared to the stress of public speaking, which elicited increases in fear but no anger or sadness. In addition, imagery of personal stressors produced significant increases in cocaine craving, while public speaking did not.283285 Significant increases in heart rate, salivary cortisol, drug craving, and subjective anxiety were also observed with imagery exposure to stress and nonstress drug cues as compared to neutral relaxing cues in cocaine-dependent individuals.285 More recently, we have shown that stress and alcohol/drug-related stimuli similarly increase craving, anxiety, negative emotions, and physiological responses in abstinent alcoholics and in naltrexone-treated, opiate-addicted individuals.286,287 On the other hand, recently abstinent alcoholics and smokers show altered basal HPA responses and a suppressed HPA response as measured by cortisol to stress compared to their nonaddicted counterparts.288290

In a more comprehensive assessment of the biological stress response in recently abstinent cocaine-addicted individuals, we reported that brief exposure to stress and to drug cues as compared to neutral relaxing cues activated the HPA axis (with increases in ACTH, cortisol, and prolactin levels) as well as the sympthoadrenomedullary systems, as measured by plasma norepinephrine and epinephrine levels.282 Furthermore, we found little evidence of recovery or return to baseline in ACTH, NE, and EPI levels even more than 1 h after the 5-min imagery exposure. These findings were extended to directly compare abstinent cocaine-dependent individuals to a demographically matched group of healthy social drinkers, using individually calibrated personally emotional stress and drug/alcohol cue-related imagery compared to neutral imagery. Findings indicated that cocaine patients showed an enhanced sensitivity to emotional distress and physiological arousal and higher levels of drug craving to both stress and drug-cue exposure compared to controls.291 Similarly, we also compared 4-week abstinent alcoholics to matched social drinkers. The recovering alcoholics at 4 weeks abstinence showed greater levels of basal heart rate and salivary cortisol levels compared to control drinkers. Upon stress and alcohol-cue exposure, they showed persistently greater subjective distress, alcohol craving, and blood pressure responses, but a suppressed heart rate and cortisol response compared to controls.239 Interestingly, both cocaine patients and alcoholics show increased anxiety and negative emotions during drug-cue exposure, while social drinkers report lower levels of negative affect and anxiety with alcohol-cue exposure. These data provide direct evidence of high drug craving and altered hedonic responses to both stress and drug cues in addicted individuals compared to social drinkers (see Fig. 2). They also indicate that alterations in physiological stress responses are associated with high levels of stress-induced and cue-induced craving and distress states. The nature of the alterations are marked by increased emotional distress, heightened craving, altered basal responses, and blunted or suppressed physiological responses in abstinent addicted individuals compared to social drinkers.

Figure 2.

Figure 2

Mean and standard errors for peak craving and anxiety ratings during exposure to stress, drug cues, and neutral imagery conditions. (A) Peak craving is significantly higher in abstinent alcoholics and cocaine patients compared to social drinkers (P < 0.0001). (B) Peak anxiety ratings are significantly higher in abstinent alcoholics and cocaine patients compared to social drinkers (P < 0.001). (Detailed statistics provided in Fox et al.291 and Sinha et al.239)

Many studies have also examined brain regions associated with craving in addicted individuals. Exposure to drug cues known to increase craving increases activity in the amygdala and regions of the frontal cortex,292294 with gender differences in amygdala activity and frontal cortex response in cocaine-dependent individuals.295,296 Cue-induced craving for nicotine, methamphetamine, or opiates also activates regions of the prefrontal cortex, amygdala, hippocampus, insula, and VTA (see Ref. 297). As stress also increases drug craving, we examined brain activation during stress and neutral imagery in a functional magnetic resonance imaging (fMRI) study. Although healthy controls and cocaine-dependent individuals showed similar levels of distress and pulse changes during stress exposure, brain response to emotional stress in paralimbic regions such as the anterior cingulate cortex, hippocampus, and parahippocampal regions was greater in healthy controls during stress, while cocaine patients showed a striking absence of such activation.298 In contrast, cocaine patients had increased activity in the caudate and dorsal striatum region during stress that was significantly associated with stress-induced cocaine craving ratings.

Recent PET studies have also shown significant positive correlations between the dorsal striatum and drug cue–induced cocaine craving.299,300 These findings are consistent with imaging studies with alcoholic patients showing increased association between dorsal striatum regions and alcohol craving in response to presentation of alcohol-related stimuli.301,302 Using PET imaging with alcoholics and cocaine patients, research has shown a significant association between dopamine D2 receptor binding in the VS and drug craving as well as motivation for self-administration.124,303,304 On the other hand, neuropsychological and imaging studies examining prefrontal executive functions, including impulse control, decision making, and set shifting, have shown executive function deficits and hypofrontal responses in addicted individuals compared to control volunteers.305312 Together, these findings indicate that increased stress and cue-induced craving and compulsive drug-seeking states in addicted individuals are associated with greater activity in the striatum, but decreased activity in specific regions of the cingulate and prefrontal cortex and related regions involved in controlling impulses and emotions.

Stress-Induced Reinstatement of Drug Seeking and Relapse

While several efficacious behavioral and pharmacological therapies in the treatment of addiction exist, it is well known that relapse rates in addiction remain high.30,313,314 Exposure to stress, drug-related stimuli, and drugs themselves each reinstate drug-seeking behavior in animals and increase relapse susceptibility in addicted individuals.274,315317 Such data underscore the need for specific attention to the chronic relapse susceptibility as a target in addiction treatment development.

In the last decade, a substantial number of preclinical studies have shown that brain CRF, noradrenergic, and glutamatergic pathways contribute to reinstatement of drug seeking.86,316320 Neuroadaptations associated with chronic drug use include overactive brain CRF and glutamatergic pathways, altered autonomic responses, and underactive dopamine and GABA systems, and these changes may accompany the high craving states and relapse susceptibility associated with the chronic nature of addiction.118,196,197,274,313,321 Furthermore, using animal models of drug self-administration and relapse, preclinical studies have identified CRF antagonists, alpha-2-adrenergic agonists, and more recently, glutamatergic agents as important in reducing stress-induced seeking in addicted laboratory animals (see316,317,322324). These data are consistent with human findings reviewed in the previous section indicating that alterations in stress and dopaminergic pathways accompany high distress and craving states and blunted physiological and neural responses that are important in regulation of stress, craving, and impulse control.

Human research has also begun to identify markers of the stress and craving states that are predictive of relapse outcomes. To fully understand whether the increased distress and drug-craving state is predictive of relapse, we followed the inpatient treatment-engaged cocaine- and alcohol-dependent individuals in our studies described in previous sections after discharge from inpatient treatment for 90 days to assess relapse outcomes. For the cocaine group, we found that stress-induced cocaine craving in the laboratory significantly predicted time to cocaine relapse. While stress-induced ACTH and cortisol responses were not associated with time to relapse, these responses were predictive of amounts of cocaine consumed during follow-up.325 While drug cue–induced craving was not predictive of relapse in this study, there was a high correlation between stress and drug cue–induced drug craving and in stress and drug cue–induced HPA responses. These data suggest that at least in the case of cocaine dependence, stress and drug cue–induced distress states produce a similar compulsive drug-seeking state that is associated with relapse vulnerability. In alcoholics, negative mood, stress-induced alcohol craving, and blunted stress and cue-induced cortisol responses have been associated with alcohol relapse outcomes.236,326329 Nicotine-deprived smokers who were exposed to a series of stressors showed blunted ACTH, cortisol, and blood pressure responses to stress but increased nicotine withdrawal and craving scores, and these responses were predictive of nicotine relapse outcomes.289 Thus, for alcoholic and smoking samples, as in the cocaine group, it appears that the drug-craving state marked by increasing distress and compulsive motivation for drug (craving) along with poor stress regulatory responses (altered glucocorticoid feedback or increased noradrenergic arousal) results in an enhanced susceptibility to addiction relapse.

Findings from basic science and human laboratory and clinical outcome studies identify several pharmacological treatment targets to address stress-induced reinstatement of drug seeking and relapse susceptibility. Basic science data suggest CRF antagonists, alpha-2 adrenergic agonists, and glutamatergic agents could be promising in addressing stress-related relapse. Human laboratory studies are needed that will screen these agents to assess their promise with regard to intermediate markers of stress-related relapse susceptibility. Such studies would target stress- and cue-induced drug craving, craving-related anxiety, HPA measures, and heart rate or heart rate variability as well as responses in specific brain regions.297 For example, in a preliminary laboratory and clinical outcomes study, we have shown that lofexidine, an alpha-2 adrenergic agonist, significantly decreased stress-induced opiate craving and stress-induced anger ratings, while also improving opiate relapse outcomes in naltrexone-treated, opiate-dependent individuals.330 Similarly, behavioral strategies that decrease anxiety and stress-related drug craving and normalize stress responses so as to potentiate adaptive responding in high-challenge contexts would be of benefit in decreasing the effects of stress on drug seeking and relapse. For example, mindfulness based stress reduction (MBSR) is efficacious in decreasing relapse to major depression, and adaptations of these strategies could be of benefit to address relapse risk in addiction.274

Summary and Future Directions

This review focuses on the accumulating evidence from preclinical, clinical, and population studies that highly stressful situations and chronic stress increase addiction vulnerability, that is, both risk of developing addiction and risk of relapse. The types of stressors that increase addiction risk are identified in Table 1. The stressors tend to be highly emotionally, distressing events that are uncontrollable and unpredictable for both children and adults. The themes range from loss, violence, and aggression to poor support, interpersonal conflict, isolation, and trauma. There is also evidence for a dose-dependent relationship between accumulated adversity and addiction risk—the greater the number of stressors an individual is exposed to, the higher the risk of developing addiction. Work-related stressors have weaker support, but individual-level variables such as trait negative emotionality and poor self-control (possibly similar to poor executive function) appear to also contribute uniquely to addiction risk. Exposure to such stressors early in life and accumulation of stress (chronicity) result in neuroendocrine, physiological, behavioral, and subjective changes that tend to be long lasting and adversely affect development of brain systems involved in learning, motivation, and stress-related adaptive behaviors. Research that directly addresses stress-related neurobiological changes and their association with behavioral outcomes is sorely needed. Evidence to clarify the contribution of stress to alterations in mesolimbic dopamine activity and its association with drug use is also needed. Figure 1 presents a schematic model of associations that have been supported in research, as well as remaining gaps.

A review of evidence indicating the effects of drug use and abuse on stress responses and dopamine transmission is presented, along with altered emotional and motivational responses associated with craving and relapse to drug use. While substance abuse results in changes in stress and dopaminergic pathways involved in motivation, self control, and adaptive processes necessary for survival, evidence for whether such changes enhance drug seeking or craving and drug use behaviors is lacking. For example, studies on whether prior exposure to licit and illicit drugs modifies the association between stress and drug self-administration are rare. While there are specific neuroadaptations in reward and associated regions, it is also important to examine which of these changes are involved in increasing drug intake and supportive of addictive processes such as progressive loss of control, persistence of craving, and escalating drug self-administration. As stress also increases risk of mood and anxiety disorders that are highly comorbid with addiction, it is important to examine whether there are specific stress-related factors that contribute to risk for mood and anxiety disorders and addiction risk. That is, what are the resiliency factors that are protective for one set of illness but are vulnerabilities for the other. Exploration of gene–environment interactions could be particularly helpful in answering such questions.

A review of recent studies on stress-induced reinstatement to drug seeking, drug craving, and relapse susceptibility is also provided. Clinical implications include the development of new assessment procedures and markers that will be useful in identifying those who are at particular risk for stress-related relapse and testing of novel pharmacological therapies that target the link between stress and relapse risk. As shown in Figure 2, addicted individuals show enhanced sensitivity to craving and greater anxiety in stress- and drug-related situations, but whether such altered responses represent transitions due to chronic drug use or chronic stress states needs to be further examined. Research on the mechanisms by which chronic stress and drug use alter executive functions that are involved in adaptive behavioral responses is needed. Efficacious behavioral treatments focus on improving coping response. However, stress exposure and chronic distress decrease stress adaptive and coping mechanisms, and hence treatments that focus on enhancing coping may not be suitable for those with stress-related risk factors. Development of new interventions that target self-control, especially in the context of stress is needed. Systematic research on these questions will lead to a greater understanding of how stress is associated with relapse. Furthermore, such research may be significant in developing new treatment targets to reduce relapse, both in the area of medication development and in developing behavioral treatments that specifically target the effects of stress on continued drug use and relapse in addicts.

Acknowledgments

Preparation of this review was supported by grants from the National Institutes of Health, P50-DA165556, R01-AA13892, R01-DA18219, and U01-RR24925.

Footnotes

Conflicts of Interest

The author declares no conflicts of interest.

References

  • 1.Lazarus RS. Stress and Emotion: A New Synthesis. Springer Publishing Company; New York: 1999. [Google Scholar]
  • 2.Cohen S, Kessler RC, Gordon LU. Strategies for measuring stress in studies of psychiatric and physical disorders. In: Cohen S, Kessler RC, Gordon LU, editors. Measuring Stress: A Guide for Health and Social Scientists. Oxford University Press; New York: 1995. pp. 3–26. [Google Scholar]
  • 3.Levine S. Developmental determinants of sensitivity and resistance to stress. Psychoneuroendocrinology. 2005;30:939–946. doi: 10.1016/j.psyneuen.2005.03.013. [DOI] [PubMed] [Google Scholar]
  • 4.Charmandari E, Tsigos C, Chrousos G. Endocrinology of the stress response. Annu. Rev. Physiol. 2005;67:259–284. doi: 10.1146/annurev.physiol.67.040403.120816. [DOI] [PubMed] [Google Scholar]
  • 5.McEwen BS. Protective and damaging effects of stress mediators: the good and bad sides of the response to stress. Metabolism. 2002;51:2–4. doi: 10.1053/meta.2002.33183. [DOI] [PubMed] [Google Scholar]
  • 6.McEwen BS. Physiology and neurobiology of stress and adaptation: central role of the brain. Physiol. Rev. 2007;87:873–904. doi: 10.1152/physrev.00041.2006. [DOI] [PubMed] [Google Scholar]
  • 7.Selye H. The Stress of Life. McGraw-Hill; New York: 1976. [Google Scholar]
  • 8.Paulus MP. Decision-making dysfunctions in psychiatry–altered homeostatic processing? Science. 2007;318:602–606. doi: 10.1126/science.1142997. [DOI] [PubMed] [Google Scholar]
  • 9.Frankenhauser M. Psychobiological aspects of life stress. In: Levine S, Ursin H, editors. Coping and Health. Plenum Press; New York: 1980. pp. 203–223. [Google Scholar]
  • 10.Lovallo WR. Stress & Health: Biological and Psychological Interactions. Sage Publications, Inc.; Thousand Oaks, CA: 1997. [Google Scholar]
  • 11.Meaney MJ, Brake W, Gratton A. Environment regulation of the development of mesolimbic dopamine systems: A neurobiological mechanism for vulnerability to drug abuse? Psychoneuroendocrinology. 2002;27:127–138. doi: 10.1016/s0306-4530(01)00040-3. [DOI] [PubMed] [Google Scholar]
  • 12.McEwen BS. Stress and hippocampal plasticity. Annu. Rev. Neuro-sci. 1999;22:105–122. doi: 10.1146/annurev.neuro.22.1.105. [DOI] [PubMed] [Google Scholar]
  • 13.Heinrichs S. Behavioral consequences of altered corticotropin-releasing factor activation in brian: a functionalist view of affective neuroscience. In: Steckler T, Kalin NH, Reul JMHM, editors. Handbook of Stress and the Brain. Part 1: The Neurobiology of Stress. Vol. 15. Elsevier; Amsterdam: 2005. pp. 155–177. [Google Scholar]
  • 14.Berridge CW. Noradrenergic modulation of arousal. Brain Res. Rev. 2007;58(1):1–17. doi: 10.1016/j.brainresrev.2007.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Phan KL, et al. Neural substrates for voluntary suppression of negative affect: A functional magnetic resonance imaging study. Biol. Psychiatry. 2005;57:210–219. doi: 10.1016/j.biopsych.2004.10.030. [DOI] [PubMed] [Google Scholar]
  • 16.Roberts A, Robbins T, Weiskrantz L. The Prefrontal Cortex: Executive and Cognitive Functions. Oxford University Press; Oxford, UK: 1998. [Google Scholar]
  • 17.Arnsten AFT. The biology of being frazzled. Science. 1998;280:1711–1712. doi: 10.1126/science.280.5370.1711. [DOI] [PubMed] [Google Scholar]
  • 18.Mischel W. From Good Intentions to Willpower. Guilford Press; New York: 1996. [Google Scholar]
  • 19.Barkley RA. Behavioral inhibition, sustained attention, and executive functions: Constructing a unifying theory of ADHD. Psychol. Bull. 1997;121:65–94. doi: 10.1037/0033-2909.121.1.65. [DOI] [PubMed] [Google Scholar]
  • 20.Tice D, Bratslavsky E, Baumeister R. Emotional distress regulation takes precedence over impulse control: If you feel bad, do it! J. Pers. Soc. Psychol. 2001;80:53–67. [PubMed] [Google Scholar]
  • 21.Westergaard GC, et al. Physiological correlates of aggression and impulsivity in free-ranging female primates. Neuropsychopharmacology. 2003;28:1045–1055. doi: 10.1038/sj.npp.1300171. [DOI] [PubMed] [Google Scholar]
  • 22.Hayaki J, et al. Adversity among drug users: relationship to impulsivity. Drug Alcohol Depend. 2005;78:65–71. doi: 10.1016/j.drugalcdep.2004.09.002. [DOI] [PubMed] [Google Scholar]
  • 23.Greco B, Carli M. Reduced attention and increased impulsivity in mice lacking NPY Y2 receptors: relation to anxiolytic-like phenotype. Behav. Brain Res. 2006;169:325–334. doi: 10.1016/j.bbr.2006.02.002. [DOI] [PubMed] [Google Scholar]
  • 24.Fishbein DH, et al. Mediators of the stress-substance-use relationship in urban male adolescents. Prev. Sci. 2006;7:113–126. doi: 10.1007/s11121-006-0027-4. [DOI] [PubMed] [Google Scholar]
  • 25.Verdejo-Garcia A, et al. Negative emotion-driven impulsivity predicts substance dependence problems. Drug Alcohol Depend. 2007;91:213–219. doi: 10.1016/j.drugalcdep.2007.05.025. [DOI] [PubMed] [Google Scholar]
  • 26.Anestis MD, Selby EA, Joiner TE. The role of urgency in maladaptive behaviors. Behav. Res. Ther. 2007;45:3018–3029. doi: 10.1016/j.brat.2007.08.012. [DOI] [PubMed] [Google Scholar]
  • 27.Hatzinger M, et al. Hypothalamic-pituitary-adrenocortical (HPA) activity in kindergarten children: importance of gender and associations with behavioral/emotional difficulties. J. Psychiatr. Res. 2007;41:861–870. doi: 10.1016/j.jpsychires.2006.07.012. [DOI] [PubMed] [Google Scholar]
  • 28.Arnsten AFT, Goldman-Rakic PS. Noise stress impairs prefrontal cortical cognitive function in monkeys: Evidence for a hyperdopaminergic mechanism. Arch. Gen. Psychiatry. 1998;55:362–369. doi: 10.1001/archpsyc.55.4.362. [DOI] [PubMed] [Google Scholar]
  • 29.Li CS, Sinha R. Inhibitory control and emotional stress regulation: Neuroimaging evidence for frontal-limbic dysfunction in psycho-stimulant addiction. Neurosci. Biobehav. Rev. 2008;32:581–597. doi: 10.1016/j.neubiorev.2007.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Sinha R. How does stress increase risk of drug abuse and relapse? Psychopharmacology (Berl.) 2001;158:343–359. doi: 10.1007/s002130100917. [DOI] [PubMed] [Google Scholar]
  • 31.Tomkins SS. Psychological model of smoking behavior. Am. J. Public Health & the Nation's Health. 1966;56:17–20. doi: 10.2105/ajph.56.12_suppl.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Leventhal H, Cleary PD. The smoking problem: A review of the research and theory in behavioral risk modification. Psychol. Bull. 1980;88:370–405. doi: 10.1037/0033-2909.88.2.370. [DOI] [PubMed] [Google Scholar]
  • 33.Russell JA, Mehrabian A. The mediating role of emotions in alcohol use. J. Stud. Alcohol. 1975;36:1508–1536. doi: 10.15288/jsa.1975.36.1508. [DOI] [PubMed] [Google Scholar]
  • 34.Marlatt GA, Gordon JR. Relapse Prevention: Maintenance Strategies in the Treatment of Addictive Behaviors. Guilford Press; New York: 1985. [Google Scholar]
  • 35.Wills T, Shiffman S. Coping and substance abuse: A conceptual framework. In: Shiffman S, Wills T, editors. Coping and Substance Use. Academic Press; Orlando, FL: 1985. pp. 3–24. [Google Scholar]
  • 36.Khantzian EJ. The self-medication hypothesis of addictive disorders: Focus on heroin and cocaine dependence. Am. J. Psychiatry. 1985;142:1259–1264. doi: 10.1176/ajp.142.11.1259. [DOI] [PubMed] [Google Scholar]
  • 37.Baker TB, et al. Addiction motivation reformulated: An affective processing model of negative reinforcement. Psychol. Rev. 2004;111:33–51. doi: 10.1037/0033-295X.111.1.33. [DOI] [PubMed] [Google Scholar]
  • 38.Koob GF, Le Moal M. Drug abuse: Hedonic homeostatic dysregulation. Science. 1997;278:52–58. doi: 10.1126/science.278.5335.52. [DOI] [PubMed] [Google Scholar]
  • 39.Robinson TE, Berridge KC. Addiction. Annu. Rev. Psychol. 2003;54:25–53. doi: 10.1146/annurev.psych.54.101601.145237. [DOI] [PubMed] [Google Scholar]
  • 40.Hyman SE, Malenka RC. Addiction and the Brain: The Neurobiology of compulsion and its persistence. Neuroscience. 2001;2:695–703. doi: 10.1038/35094560. [DOI] [PubMed] [Google Scholar]
  • 41.Newcomb M, Harlow L. Life events and substance use among adolescents: mediating effects of perceived loss of control and meaninglessness in life. J. Pers. Soc. Psychol. 1986;51:564–577. doi: 10.1037//0022-3514.51.3.564. [DOI] [PubMed] [Google Scholar]
  • 42.Brown RI. Gambling addictions, arousal, and an affective/decision-making explanation of behavioral reversions or relapses. Int. J. Addict. 1987;22:1053–1067. doi: 10.3109/10826088709027469. [DOI] [PubMed] [Google Scholar]
  • 43.Newcomb MD, Bentler PM. Impact of adolescent drug use and social support on problems of young adults: A longitudinal study. J. Abnorm. Psychol. 1988;97:64–75. doi: 10.1037//0021-843x.97.1.64. [DOI] [PubMed] [Google Scholar]
  • 44.Chassin L, Mann LM, Sher KJ. Self-awareness theory, family history of alcoholism, and adolescent alcohol involvement. J. Abnorm. Psychol. 1998;97:206–217. doi: 10.1037//0021-843x.97.2.206. [DOI] [PubMed] [Google Scholar]
  • 45.Cooper ML, Russell M, Frone MR. Work stress and alcohol effects: a test of stress-induced drinking. J. Health Soc. Behav. 1990;31:260–276. [PubMed] [Google Scholar]
  • 46.Wills TA, Vaccaro D, McNamara G. The role of life events, family support, and competence in adolescent substance use: a test of vulnerability and protective factors. Am. J. Commun. Psychol. 1992;20:349–374. doi: 10.1007/BF00937914. [DOI] [PubMed] [Google Scholar]
  • 47.Johnson V, Pandina RJ. A longitudinal examination of the relationships among stress, coping strategies, and problems associated with alcohol use. Alcohol Clin. Exp. Res. 1993;17:696–702. doi: 10.1111/j.1530-0277.1993.tb00822.x. [DOI] [PubMed] [Google Scholar]
  • 48.Johnson V, Pandina RJ. Alcohol problems among a community sample: longitudinal influences of stress, coping, and gender. Subst. Use Misuse. 2000;35:669–686. doi: 10.3109/10826080009148416. [DOI] [PubMed] [Google Scholar]
  • 49.Turner RJ, Lloyd DA. Lifetime traumas and mental health: the significance of cumulative adversity. J. Health Soc. Behav. 1995;36:360–376. [PubMed] [Google Scholar]
  • 50.Wills TA, Cleary SD. How are social support effects mediated? A test with parental support and adolescent substance use. J. Pers. Soc. Psychol. 1996;71:937–952. doi: 10.1037//0022-3514.71.5.937. [DOI] [PubMed] [Google Scholar]
  • 51.Sher KJ, et al. The role of childhood stressors in the intergenerational transmission of alcohol use disorders. J. Stud. Alcohol. 1997;58:414–427. doi: 10.15288/jsa.1997.58.414. [DOI] [PubMed] [Google Scholar]
  • 52.Costa FM, Jessor R, Turbin MS. Transition into adolescent problem drinking: the role of psychosocial risk and protective factors. J. Stud. Alcohol. 1999;60:480–490. doi: 10.15288/jsa.1999.60.480. [DOI] [PubMed] [Google Scholar]
  • 53.Perkins HW. Stress-motivated drinking in collegiate and postcollegiate young adulthood: life course and gender patterns. J. Stud. Alcohol. 1999;60:219–227. doi: 10.15288/jsa.1999.60.219. [DOI] [PubMed] [Google Scholar]
  • 54.Burt SA, et al. Parent-child conflict and the comorbidity among childhood externalizing disorders. Arch. Gen. Psychiatry. 2003;60:505–513. doi: 10.1001/archpsyc.60.5.505. [DOI] [PubMed] [Google Scholar]
  • 55.Barrett A, Turner R. Family structure and substance use problems in adolescence and early adulthood: examining explanations for the relationship. Addiction. 2006;101:109–120. doi: 10.1111/j.1360-0443.2005.01296.x. [DOI] [PubMed] [Google Scholar]
  • 56.Dembo R, et al. The relationship between physical and sexual abuse and tobacco, alcohol, and illicit drug use among youths in a juvenile detention center. Int. J. Addict. 1988;23:351–378. doi: 10.3109/10826088809039203. [DOI] [PubMed] [Google Scholar]
  • 57.Harrison PA, Fulkerson JA, Beebe TJ. Multiple substance use among adolescent physical and sexual abuse victims. Child Abuse & Neglect. 1997;21:529–539. doi: 10.1016/s0145-2134(97)00013-6. [DOI] [PubMed] [Google Scholar]
  • 58.Clark D, Lesnick L, Hegedus A. Traumas and other adverse life events in adolescents with alcohol abuse and dependence. J. Am. Acad. Child Adolesc. Psychiatry. 1997;36:1744–1751. doi: 10.1097/00004583-199712000-00023. [DOI] [PubMed] [Google Scholar]
  • 59.Widom CS, Weiler BL, Cottler LB. Childhood victimization and drug abuse: a comparison of prospective and retrospective findings. J. Consult. Clin. Psychol. 1999;67:867–880. doi: 10.1037//0022-006x.67.6.867. [DOI] [PubMed] [Google Scholar]
  • 60.Breslau N, Davis G, Schultz L. Posttraumatic stress disorder and the incidence of nicotine, alcohol, and other drug disorders in persons who have experienced trauma. Arch. Gen. Psychiatry. 2003;60:289–294. doi: 10.1001/archpsyc.60.3.289. [DOI] [PubMed] [Google Scholar]
  • 61.Sher KJ, et al. Characteristics of children of alcoholics: Putative risk factors, substance use, and abuse and psychopathology. J. Abnorm. Psychol. 1991;100:427–448. doi: 10.1037//0021-843x.100.4.427. [DOI] [PubMed] [Google Scholar]
  • 62.Cooper ML, et al. Development and validation of a three-dimensional measure of drinking motives. Psychol. Assess. 1992;4:123–132. [Google Scholar]
  • 63.Laurent L, Catanzaro SJ, Callan MK. Stress, alcohol-related expectancies and coping preferences: A replication with adolescents of the Cooper et al. (1992) model. J. Stud. Alcohol. 1997;58:644–651. doi: 10.15288/jsa.1997.58.644. [DOI] [PubMed] [Google Scholar]
  • 64.Chen JH, et al. Gender differences in the effects of bereavement-related psychological distress on health outcomes. Psychol. Med. 1999;29:367–380. doi: 10.1017/s0033291798008137. [DOI] [PubMed] [Google Scholar]
  • 65.Stice E, Barrera M, Jr., Chassin L. Prospective differential prediction of adolescent alcohol use and problem use: examining the mechanisms of effect. J. Abnorm. Psychol. 1998;107:616–628. doi: 10.1037//0021-843x.107.4.616. [DOI] [PubMed] [Google Scholar]
  • 66.Chassin L, et al. Historical changes in cigarette smoking and smoking-related beliefs after 2 decades in a midwestern community. Health Psychol. 2003;22:347–353. doi: 10.1037/0278-6133.22.4.347. [DOI] [PubMed] [Google Scholar]
  • 67.Measelle JR, Stice E, Springer DW. A prospective test of the negative affect model of substance abuse: moderating effects of social support. Psychol. Addict. Behav. 2006;20:225–233. doi: 10.1037/0893-164X.20.3.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kandel DB, et al. Psychiatric disorders associated with substance use among children and adolescents: Findings from the Methods for the Epidemiology of Child and Adolescents Mental Disorders (MECA) Study. J. Abnorm. Child Psychol. 1997;25:121–132. doi: 10.1023/a:1025779412167. [DOI] [PubMed] [Google Scholar]
  • 69.King CA, et al. Predictors of co-morbid alcohol and substance abuse in depressed adolescents. J. Am. Acad. Child Adolesc. Psychiatry. 1996;35:743–751. doi: 10.1097/00004583-199606000-00014. [DOI] [PubMed] [Google Scholar]
  • 70.Rohde L, Roman T, Szobot C, et al. Dopamine transporter gene, response to methylphenidate and cerebral blood flow in attention-deficit/hyperactivity disorder: a pilot study. Synapse. 2003;48:87–89. doi: 10.1002/syn.10186. [DOI] [PubMed] [Google Scholar]
  • 71.Riggs PD, Whitmore EA. Substance Use Disorders and Disruptive Behavior Disorders. APA Press; Washington, DC: 1999. [Google Scholar]
  • 72.Rao U, et al. Factors associated with the development of substance use disorder in depressed adolescents. J. Am. Acad. Child Adolsc. Psychiatry. 1999;38:1109–1117. doi: 10.1097/00004583-199909000-00014. [DOI] [PubMed] [Google Scholar]
  • 73.Kessler RC, et al. The epidemiology of cooccurring addictive and mental disorders: Implications for prevention and service utilization. Am. J. Orthopsychiatry. 1996;66:17–31. doi: 10.1037/h0080151. [DOI] [PubMed] [Google Scholar]
  • 74.Sinha R, Rounsaville BJ. Sex differences in depressed substance abusers. J. Clin. Psychiatry. 2002;63:616–627. doi: 10.4088/jcp.v63n0715. [DOI] [PubMed] [Google Scholar]
  • 75.Clark DB, et al. Physical and sexual abuse, depression and alcohol use disorders in adolescents: onsets and outcomes. Drug Alcohol Depend. 2003;69:51–60. doi: 10.1016/s0376-8716(02)00254-5. [DOI] [PubMed] [Google Scholar]
  • 76.Brady KT, Sinha R. Co-occurring mental and substance use disorders: The neurobiological effects of chronic stress. Am. J. Psychiatry. 2005;162:1483–1493. doi: 10.1176/appi.ajp.162.8.1483. [DOI] [PubMed] [Google Scholar]
  • 77.Cicchetti D, Toth SL. Child maltreatment. Annu. Rev. Clin. Psychol. 2005;1:409–438. doi: 10.1146/annurev.clinpsy.1.102803.144029. [DOI] [PubMed] [Google Scholar]
  • 78.Reed PL, Anthony JC, Breslau N. Incidence of drug problems in young adults exposed to trauma and posttraumatic stress disorder: do early life experiences and predispositions matter? Arch. Gen. Psych. 2007;64:1435–1442. doi: 10.1001/archpsyc.64.12.1435. [DOI] [PubMed] [Google Scholar]
  • 79.Hammen C. Stress and depression. Annu. Rev. Clin. Psychol. 2005;1:293–319. doi: 10.1146/annurev.clinpsy.1.102803.143938. [DOI] [PubMed] [Google Scholar]
  • 80.Kessler RC. The epidemiology of dual diagnosis. Biol. Psychiatry. 2005;56:730–737. doi: 10.1016/j.biopsych.2004.06.034. [DOI] [PubMed] [Google Scholar]
  • 81.Turner RJ, Lloyd DA. Cumulative adversity and drug dependence in young adults: racial/ethnic contrasts. Addiction. 2003;98:305–315. doi: 10.1046/j.1360-0443.2003.00312.x. [DOI] [PubMed] [Google Scholar]
  • 82.Lloyd DA, Turner RJ. Cumulative life-time adversities and alcohol dependence in adolescence and young adulthood. Drug Alcohol Depend. 2008;93:217–226. doi: 10.1016/j.drugalcdep.2007.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Sinha R. Stress and drug abuse. In: Steckler NHKT, Reul JMHM, editors. Handbook of Stress and the Brain. Part 2 Stress: Integrative and Clinical Aspects. Vol. 15. Elsevier; Amsterdam: 2005. pp. 333–356. [Google Scholar]
  • 84.Miczek KA, et al. Aggression and defeat: persistent effects on cocaine self-administration and gene expression in peptidergic and aminergic mesocorticolimbic circuits. Neurosci. Biobehav. Rev. 2004;27:787–802. doi: 10.1016/j.neubiorev.2003.11.005. [DOI] [PubMed] [Google Scholar]
  • 85.Lu L, Shaham Y. The role of stress in opiate and psychostimulant addiction: evidence from animal models. In: Steckler T, Kalin N, Reul J, editors. Handbook of Stress and the Brain, Part 2 Stress: Integrative and Clinical Aspects. Vol. 15. Elsevier; San Diego, CA: 2005. pp. 315–332. [Google Scholar]
  • 86.Le AD, et al. Role of alpha-2 adrenoceptors in stress-induced reinstatement of alcohol seeking and alcohol self-administration in rats. Psychopharmacology (Berl.) 2005;179:366–373. doi: 10.1007/s00213-004-2036-y. [DOI] [PubMed] [Google Scholar]
  • 87.Cleck JN, Blendy JA. Making a bad thing worse: adverse effects of stress on drug addiction. J. Clin. Invest. 2008;118:454–461. doi: 10.1172/JCI33946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Higley JD, et al. Nonhuman primate model of alcohol abuse: Effects of early experience, personality, and stress on alcohol consumption. Proc. Natl. Acad. Sci. USA. 1991;88:7261–7265. doi: 10.1073/pnas.88.16.7261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Kosten TA, Miserendino MJD, Kehoe P. Enhanced acquisition of cocaine self-administration in adult rats with neonatal isolation stress experience. Brain Res. 2000;875:44–50. doi: 10.1016/s0006-8993(00)02595-6. [DOI] [PubMed] [Google Scholar]
  • 90.Lu L, et al. Effect of environmental stressors on opiate and psychostimulant reinforcement, reinstatement and discrimination in rats: a review. Neurosci. Biobehav. Rev. 2003;27:457–491. doi: 10.1016/s0149-7634(03)00073-3. [DOI] [PubMed] [Google Scholar]
  • 91.Moffett MC, et al. 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]
  • 92.Boyce-Rustay JM, Cameron HA, Holmes A. Chronic swim stress alters sensitivity to acute behavioral effects of ethanol in mice. Physiol. Behav. 2007;91:77–86. doi: 10.1016/j.physbeh.2007.01.024. [DOI] [PubMed] [Google Scholar]
  • 93.Park MK, et al. Age, sex and early environment contribute to individual differences in nicotine/acetaldehyde-induced behavioral and endocrine responses in rats. Pharmacol. Biochem. Behav. 2007;86:297–305. doi: 10.1016/j.pbb.2006.10.002. [DOI] [PubMed] [Google Scholar]
  • 94.Kosten TA, et al. Neonatal isolation enhances acquisition of cocaine self-administration and food responding in female rats. Behav. Brain Res. 2004;151:137–149. doi: 10.1016/j.bbr.2003.08.010. [DOI] [PubMed] [Google Scholar]
  • 95.Kosten TA, Zhang XY, Kehoe P. Heightened cocaine and food administration in female rats with neonatal isolation experience. Neuropsychopharmacology. 2006;31:70–76. doi: 10.1038/sj.npp.1300779. [DOI] [PubMed] [Google Scholar]
  • 96.Lynch W. Sex differences in vulnerability to drug self-administration. Exp. Clin. Psychopharmacol. 2006;14:34–41. doi: 10.1037/1064-1297.14.1.34. [DOI] [PubMed] [Google Scholar]
  • 97.Becker JB, et al. Stress and disease: is being female a predisposing factor? J. Neurosci. 2007;27:11851–11855. doi: 10.1523/JNEUROSCI.3565-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Tschann JM, et al. Initiation of substance use in early adolescence: the roles of pubertal timing and emotional distress. Health Psychol. 1994;13:326–333. doi: 10.1037//0278-6133.13.4.326. [DOI] [PubMed] [Google Scholar]
  • 99.Fergusson DM, Horwood LJ. Early onset cannabis use and psychosocial adjustment in young adults. Addiction. 1997;92:279–296. [PubMed] [Google Scholar]
  • 100.Simons JS, et al. Associations between alcohol use and PTSD symptoms among American Red Cross disaster relief workers responding to the 9/11/2001 attacks. Am. J. Drug Alcohol Abuse. 2005;31:285–304. [PubMed] [Google Scholar]
  • 101.Lee CM, Neighbors C, Woods BA. Marijuana motives: young adults’ reasons for using marijuana. Addict. Behav. 2007;32:1384–1394. doi: 10.1016/j.addbeh.2006.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Wills TA, et al. Contributions of positive and negative affect to adolescent substance use: Test of a bi-dimensional model in a longitudinal study. Psychol. Addict. Behav. 1999;13:327–338. [Google Scholar]
  • 103.Wills TA, et al. Coping dimensions, life stress, and adolescent substance use: a latent growth analysis. J. Abnorm. Psychol. 2001;110:309–323. doi: 10.1037//0021-843x.110.2.309. [DOI] [PubMed] [Google Scholar]
  • 104.Wills TA, et al. Behavioral and emotional self-control: relations to substance use in samples of middle and high school students. Psychol. Addict. Behav. 2006;20:265–278. doi: 10.1037/0893-164X.20.3.265. [DOI] [PubMed] [Google Scholar]
  • 105.Siqueira L, et al. The relationship of stress and coping methods to adolescent Marijuana use. Subst. Abus. 2001;22:157–166. doi: 10.1080/08897070109511455. [DOI] [PubMed] [Google Scholar]
  • 106.Butters JE. Family stressors and adolescent cannabis use: A pathway to problem use. J. Adolesc. 2002;25:645–654. doi: 10.1006/jado.2002.0514. [DOI] [PubMed] [Google Scholar]
  • 107.McGee R, et al. A longitudinal study of cannabis use and mental health from adolescence to early adulthood. Addiction. 2000;95:491–503. doi: 10.1046/j.1360-0443.2000.9544912.x. [DOI] [PubMed] [Google Scholar]
  • 108.Hayatbakhsh MR, et al. Do parents’ marital circumstances predict young adults’ DSM-IV cannabis use disorders? A prospective study. Addiction. 2006;101:1778–1786. doi: 10.1111/j.1360-0443.2006.01620.x. [DOI] [PubMed] [Google Scholar]
  • 109.Windle M, Wiesner M. Trajectories of marijuana use from adolescence to young adulthood: predictors and outcomes. Dev. Psychopathol. 2004;16:1007–1027. doi: 10.1017/s0954579404040118. [DOI] [PubMed] [Google Scholar]
  • 110.Weiss EL, Longhurst JG, Mazure CM. Childhood sexual abuse as a risk factor for depression in women: psychosocial and neurobiological correlates. Am. J. Psychiatry. 1999;156:816–828. doi: 10.1176/ajp.156.6.816. [DOI] [PubMed] [Google Scholar]
  • 111.MacMillan HL, et al. Childhood abuse and lifetime psychopathology in a community sample. Am. J. Psychiatry. 2001;158:1878–1883. doi: 10.1176/appi.ajp.158.11.1878. [DOI] [PubMed] [Google Scholar]
  • 112.Simpson T, Miller W. Concomitance between childhood sexual and physical abuse and substance use problems: A review. Clin. Psychol. Rev. 2002;22:27–77. doi: 10.1016/s0272-7358(00)00088-x. [DOI] [PubMed] [Google Scholar]
  • 113.Hyman S, et al. A gender-specific psychometric analysis of the Early Trauma Interview Short Form in cocaine dependent adults. Addict. Behav. 2004;30:847–852. doi: 10.1016/j.addbeh.2004.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Hyman SM, Garcia M, Sinha R. Gender specific associations between types of childhood maltreatment and the onset, escalation and severity of substance use in cocaine dependent adults. Am. J. Drug Alcohol Abuse. 2006;32:655–664. doi: 10.1080/10623320600919193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Di Chiara G, Imperato A. Drugs abused by humans preferentially increase synaptic dopamine concentrations in the mesolimbic system of freely moving rats. Proc. Natl. Acad. Sci. USA. 1988;85:5274–5278. doi: 10.1073/pnas.85.14.5274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Spanagel R, Weiss F. The dopamine hypothesis of reward: past and current status. Trends Neurosci. 1999;22:521–527. doi: 10.1016/s0166-2236(99)01447-2. [DOI] [PubMed] [Google Scholar]
  • 117.Pierce RC, Kumaresan V. The mesolimbic dopamine system: the final common pathway for the reinforcing effect of drugs of abuse? Neurosci. Biobehav. Rev. 2006;30:215–238. doi: 10.1016/j.neubiorev.2005.04.016. [DOI] [PubMed] [Google Scholar]
  • 118.Kauer JA, Malenka RC. Synaptic plasticity and addiction. Nat. Rev. Neurosci. 2007;8:844–858. doi: 10.1038/nrn2234. [DOI] [PubMed] [Google Scholar]
  • 119.Breiter HC, et al. Acute effects of cocaine on human brain activity and emotion. Neuron. 1997;19:591–611. doi: 10.1016/s0896-6273(00)80374-8. [DOI] [PubMed] [Google Scholar]
  • 120.Volkow N, Wang G-J, Fowler JS, et al. Reinforcing effects of psychostimulants in humans are associated with increases in brain dopamine and occupancy of D-sub-2 receptors. J. Pharm. Expt. Ther. 1999;291:409–415. [PubMed] [Google Scholar]
  • 121.Drevets W, Gautier C, Price JC, et al. Amphetamine-induced dopamine release in human ventral striatum correlates with euphoria. Biol. Psychiatry. 2001;49:81–96. doi: 10.1016/s0006-3223(00)01038-6. [DOI] [PubMed] [Google Scholar]
  • 122.Leyton M, et al. Amphetamine-induced increases in extracellular dopamine, drug wanting, and novelty seeking: a PET/[11C]raclopride study in healthy men. Neuropsychopharmacology. 2002;27:1027–1035. doi: 10.1016/S0893-133X(02)00366-4. [DOI] [PubMed] [Google Scholar]
  • 123.Brody AL, et al. Attenuation of cue-induced cigarette craving and anterior cingulate cortex activation in bupropion-treated smokers: A preliminary study. Psychiatry Res. 2004;130:269–281. doi: 10.1016/j.pscychresns.2003.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Martinez D, et al. Imaging the neurochemistry of alcohol and substance abuse. Neuroimaging Clin. N. Am. 2007;17:539–555. doi: 10.1016/j.nic.2007.07.004. [DOI] [PubMed] [Google Scholar]
  • 125.Oswald LM, et al. Relationships among ventral striatal dopamine release, cortisol secretion, and subjective responses to amphetamine. Neuropsychopharmacology. 2005;30:821–832. doi: 10.1038/sj.npp.1300667. [DOI] [PubMed] [Google Scholar]
  • 126.Yoder KK, et al. Dopamine D(2) receptor availability is associated with subjective responses to alcohol. Alcohol Clin. Exp. Res. 2005;29:965–970. doi: 10.1097/01.alc.0000171041.32716.42. [DOI] [PubMed] [Google Scholar]
  • 127.Thierry AM, et al. Selective activation of mesocortical DA system by stress. Nature. 1976;263:242–244. doi: 10.1038/263242a0. [DOI] [PubMed] [Google Scholar]
  • 128.Dunn AJ. Stress-related activation of cerebral dopaminergic systems. Ann. N. Y. Acad. Sci. 1988;537:188–205. doi: 10.1111/j.1749-6632.1988.tb42106.x. [DOI] [PubMed] [Google Scholar]
  • 129.Takahashi H, et al. Effects of nicotine and footshock stress on dopamine release in the striatum and nucleus accumbens. Brain Res. Bull. 1998;45:157–162. doi: 10.1016/s0361-9230(97)00332-8. [DOI] [PubMed] [Google Scholar]
  • 130.Kalivas PW, Duffy P. Selective activation of dopamine transmission in the shell of the nucleus accumbens by stress. Brain Res. 1995;675:325–328. doi: 10.1016/0006-8993(95)00013-g. [DOI] [PubMed] [Google Scholar]
  • 131.Piazza PV, Le Moal ML. Pathophysiological basis of vulnerability to drug abuse: role of an interaction between stress, glucocorticoids, and dopaminergic neurons. Annu. Rev. Pharmacol. Toxicol. 1996;36:359–378. doi: 10.1146/annurev.pa.36.040196.002043. [DOI] [PubMed] [Google Scholar]
  • 132.Rouge-Pont F, et al. Individual differences in stress-induced dopamine release in the nucleus accumbens are influenced by corticosterone. Eur. J. Neurosci. 1998;10:3903–3907. doi: 10.1046/j.1460-9568.1998.00438.x. [DOI] [PubMed] [Google Scholar]
  • 133.Barrot M, et al. The dopaminergic hyper-responsiveness of the shell of the nucleus accumbens is hormone-dependent. Eur. J. Neurosci. 2000;12:973–979. doi: 10.1046/j.1460-9568.2000.00996.x. [DOI] [PubMed] [Google Scholar]
  • 134.Pacak K, et al. Chronic hypercortisolemia inhibits dopamine synthesis and turnover in the nucleus accumbens: an in vivo microdialysis study. Neuroendocrinology. 2002;76:148–157. doi: 10.1159/000064522. [DOI] [PubMed] [Google Scholar]
  • 135.Overton PG, et al. Preferential occupation of mineralocorticoid receptors by corticosterone enhances glutamate-induced burst firing in rat midbrain dopaminergic neurons. Brain Res. 1996;737:146–154. doi: 10.1016/0006-8993(96)00722-6. [DOI] [PubMed] [Google Scholar]
  • 136.Saal D, et al. Drugs of abuse and stress trigger a common synaptic adaptation in dopamine neurons. Neuron. 2003;37:577–582. doi: 10.1016/s0896-6273(03)00021-7. [DOI] [PubMed] [Google Scholar]
  • 137.Ungless MA, et al. Corticotropin-releasing factor requires CRF binding protein to potentiate NMDA receptors via CRF receptor 2 in dopamine neurons. Neuron. 2003;39:401–407. doi: 10.1016/s0896-6273(03)00461-6. [DOI] [PubMed] [Google Scholar]
  • 138.Wang B, et al. Cocaine experience establishes control of midbrain glutamate and dopamine by corticotropin-releasing factor: a role in stress-induced relapse to drug seeking. J. Neurosci. 2005;25:5389–5396. doi: 10.1523/JNEUROSCI.0955-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Pruessner JC, et al. Dopamine release in response to a psychological stress in humans and its relationship to early life maternal care: a positron emission tomography study using [11C]raclopride. J. Neurosci. 2004;24:2825–2831. doi: 10.1523/JNEUROSCI.3422-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Wand GS, et al. Association of amphetamine-induced striatal dopamine release and cortisol responses to psychological stress. Neuropsychopharmacology. 2007;32:2310–2320. doi: 10.1038/sj.npp.1301373. [DOI] [PubMed] [Google Scholar]
  • 141.Robinson TE, Kolb B. Alterations in the morphology of dendrites and dendritic spines in the nucleus accumbens and prefrontal cortex following repeated treatment with amphetamine or cocaine. Eur. J. Neurosci. 1999;11:1598–1604. doi: 10.1046/j.1460-9568.1999.00576.x. [DOI] [PubMed] [Google Scholar]
  • 142.Liston C, et al. Stress-induced alterations in prefrontal cortical dendritic morphology predict selective impairments in perceptual attentional set-shifting. J. Neurosci. 2006;26:7870–7874. doi: 10.1523/JNEUROSCI.1184-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Sorg BA, Kalivas PW. Effects of cocaine and footshock stress on extracellular dopamine levels in the ventral striatum. Brain Res. 1991;559:29–36. doi: 10.1016/0006-8993(91)90283-2. [DOI] [PubMed] [Google Scholar]
  • 144.McCullough LD, Salamone JD. Anxiogenic drugs beta-CCE and FG 7142 increase extracellular dopamine levels in nucleus accumbens. Psychopharmacology (Berl.) 1992;109:379–382. doi: 10.1007/BF02245888. [DOI] [PubMed] [Google Scholar]
  • 145.Becerra L, et al. Reward circuitry activation by noxious thermal stimuli. Neuron. 2001;32:927–946. doi: 10.1016/s0896-6273(01)00533-5. [DOI] [PubMed] [Google Scholar]
  • 146.Jensen J, et al. Direct activation of the ventral striatum in anticipation of aversive stimuli. Neuron. 2003;40:1251–1257. doi: 10.1016/s0896-6273(03)00724-4. [DOI] [PubMed] [Google Scholar]
  • 147.Berridge K, Robinson TE. What is the role of dopamine in reward: Hedonic impact, reward learning, or incentive salience? Brain Res. Rev. 1998;28:309–369. doi: 10.1016/s0165-0173(98)00019-8. [DOI] [PubMed] [Google Scholar]
  • 148.Bindra D. How adaptive behavior is produced: a perceptual-motivation alternative to response reinforcement. Behav. Brain Sci. 1978;1:41–91. [Google Scholar]
  • 149.Ikemoto S, Panksepp J. The role of nucleus accumbens dopamine in motivated behavior: A unifying interpretation with special reference to reward-seeking. Brain Res. Rev. 1999;31:6–41. doi: 10.1016/s0165-0173(99)00023-5. [DOI] [PubMed] [Google Scholar]
  • 150.Salamone JD, Cousin MS, Snyder BJ. Behavioral functions of nucleus accumbens dopamine: Empirical and conceptual problems with the anhedonia hypothesis. Neurosci. Biobehav. Rev. 1997;21:341–359. doi: 10.1016/s0149-7634(96)00017-6. [DOI] [PubMed] [Google Scholar]
  • 151.Everitt BJ, Robbins TW. Neural systems of reinforcement for drug addiction: from actions to habits to compulsion. Nat. Neurosci. 2005;8:1481–1489. doi: 10.1038/nn1579. [DOI] [PubMed] [Google Scholar]
  • 152.Baler RD, Volkow ND. Drug addiction: the neurobiology of disrupted self-control. Trends Mol. Med. 2006;12:559–566. doi: 10.1016/j.molmed.2006.10.005. [DOI] [PubMed] [Google Scholar]
  • 153.Mantsch JR, Saphier D, Goeders NE. Corticosterone facilitates the acquisition of cocaine self-administration in rats: Opposite effects of the type II glucocorticoid receptor agonist dexamethasone. J. Pharmacol. Exp. Ther. 1998;287:72–80. [PubMed] [Google Scholar]
  • 154.Goeders NE. The HPA axis and cocaine reinforcement. Psychoneuroendocrinology. 2002;27:13–34. doi: 10.1016/s0306-4530(01)00034-8. [DOI] [PubMed] [Google Scholar]
  • 155.Goeders NE. Stress, motivation, and drug addiction. Curr. Dir. Psycholog. Sci. 2004;13:33–35. [Google Scholar]
  • 156.Marinelli M, et al. Corticosterone circadian secretion differentially facilitates dopamine-mediated psychomotor effect of cocaine and morphine. J. Neurosci. 1994;14:2724–2731. doi: 10.1523/JNEUROSCI.14-05-02724.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Marinelli M, et al. Dopamine-dependent responses to morphine depend on glucocorticoid receptors. Proc. Natl. Acad. Sci. USA. 1998;95:7742–7747. doi: 10.1073/pnas.95.13.7742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Deroche-Gamonet V, et al. The glucocorticoid receptor as a potential target to reduce cocaine abuse. J. Neurosci. 2003;23:4785–4790. doi: 10.1523/JNEUROSCI.23-11-04785.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Morgan D, et al. Social dominance in monkeys: Dopamine D2 receptors and cocaine self-administration. Nat. Neurosci. 2002;5:88–90. doi: 10.1038/nn798. [DOI] [PubMed] [Google Scholar]
  • 160.Plotsky PM, Meaney MJ. Early postnatal experience alters hypothalamic corticotrophon-releasing factor (CRF) mRNA, median eminence CRF content, and stress-induced release in adult rats. Mol. Brain Res. 1993;18:195–200. doi: 10.1016/0169-328x(93)90189-v. [DOI] [PubMed] [Google Scholar]
  • 161.Liu D, et al. The effects of early life events on in vivo release of norepineperine in the paraventricular nucleus of the hypothalamus and hypothalamic-pituitary-adrenal responses during stress. J. Neuroendocrinol. 2000;12:5–12. [Google Scholar]
  • 162.Ladd CO, et al. Long-term behavioral and neuroendocrine adaptations to adverse early experience. Prog. Brain Res. 2000;122:81–103. doi: 10.1016/s0079-6123(08)62132-9. [DOI] [PubMed] [Google Scholar]
  • 163.Dallman MF, et al. Regulation of the hypothalamic-pituitary-adrenal axis during stress: feedback, facilitation and feeding. Neuroscience. 1994;6:205–213. [Google Scholar]
  • 164.Caldji C, et al. The effects of early rearing environment on the development of GABAA and central benzodiazepine receptor levels and novelty-induced fearfulness in the rat. Neuropsychopharmacology. 2000;22:219–229. doi: 10.1016/S0893-133X(99)00110-4. [DOI] [PubMed] [Google Scholar]
  • 165.Robinson TE, Becker JB, Presty SK. Long-term facilitation of amphetamine-induced rotational behavior and striatal dopamine release produced by a single exposure to amphetamine: Sex differences. Brain Res. 1982;253:231–241. doi: 10.1016/0006-8993(82)90690-4. [DOI] [PubMed] [Google Scholar]
  • 166.Kalivas PW, Stewart J. Dopmaine transmission in the initiation and expression of drug- and stress-induced sensitization of motor activity. Brain Res. Rev. 1991;16:223–244. doi: 10.1016/0165-0173(91)90007-u. [DOI] [PubMed] [Google Scholar]
  • 167.Doherty MD, Gratton A. High-speed chronoamperometric measurements of mesolimbic and nigrostriatal dopamine release associated with repeated daily stress. Brain Res. 1992;586:295–302. doi: 10.1016/0006-8993(92)91639-v. [DOI] [PubMed] [Google Scholar]
  • 168.Brake WG, et al. Influence of early postnatal rearing conditions on mesococorticolimbic dopamine and behavioral responses to psychostimulants and stressors in adult rats. Eu. J. Neurosci. 2004;19:1863–1874. doi: 10.1111/j.1460-9568.2004.03286.x. [DOI] [PubMed] [Google Scholar]
  • 169.Weinshenker D, et al. Mice with chronic norepinephrine deficiency resemble amphetamine-sensitized animals. Proc. Natl. Acad. Sci. USA. 2002;99:13873–13877. doi: 10.1073/pnas.212519999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Vanderschuren LJ, Beemster P, Schoffelmeer AN. On the role of noradrenaline in psychostimulant-induced psychomotor activity and sensitization. Psychopharmacology (Berl.) 2003;169:176–185. doi: 10.1007/s00213-003-1509-8. [DOI] [PubMed] [Google Scholar]
  • 171.Gratton A, Sullivan RM. Role of prefrontal cortex in stress responsivity. In: Steckler T, Kalin NH, Reul JMHM, editors. Handbook of Stress and the Brain. Vol. 1. Elsevier; Dusseldorf: 2005. p. 838. [Google Scholar]
  • 172.Wellman CL. Dendritic reorganization in pyramidal neurons in medial prefrontal cortex after chronic corticosterone administration. J. Neurobiol. 2001;49:245–253. doi: 10.1002/neu.1079. [DOI] [PubMed] [Google Scholar]
  • 173.Sullivan RM, Gratton A. Lateralized effects of medial prefrontal cortex lesions on neuroendocrine and autonomic stress responses in rats. J. Neurosci. 1999;19:2834–2840. doi: 10.1523/JNEUROSCI.19-07-02834.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Braun K, et al. Maternal separation followed by early social deprivation affects the development of monoaminergic fiber systems in the medial prefrontal cortex of Octodon degus. Neuroscience. 2000;95:309–318. doi: 10.1016/s0306-4522(99)00420-0. [DOI] [PubMed] [Google Scholar]
  • 175.DeBellis MD. Developmental traumatology: a contributory mechanism for alcohol and substance use disorders. Psychoneuroendocrinology. 2002;27:155–170. doi: 10.1016/s0306-4530(01)00042-7. [DOI] [PubMed] [Google Scholar]
  • 176.De Bellis MD, et al. Prefrontal cortex, thalamus, and cerebellar volumes in adolescents and young adults with adolescent-onset alcohol use disorders and comorbid mental disorders. Alcohol. Clin. Exp. Res. 2005;29:1590–1600. doi: 10.1097/01.alc.0000179368.87886.76. [DOI] [PubMed] [Google Scholar]
  • 177.Mischel W, Shoda Y, Rodriguez MI. Delay of gratification in children. Science. 1989;244:933–938. doi: 10.1126/science.2658056. [DOI] [PubMed] [Google Scholar]
  • 178.Muraven M, Baumeister RF. Self-regulation and depletion of limited resources: Does self-control resemble a muscle? Psychol. Bull. 2000;126:247–259. doi: 10.1037/0033-2909.126.2.247. [DOI] [PubMed] [Google Scholar]
  • 179.Arnsten AF, Li BM. Neurobiology of executive functions: catecholamine influences on prefrontal cortical functions. Biol. Psychiatry. 2005;57:1377–1384. doi: 10.1016/j.biopsych.2004.08.019. [DOI] [PubMed] [Google Scholar]
  • 180.Wills TA, Stoolmiller M. The role of self-control in early escalation of substance use: a time-varying analysis. J. Consult. Clin. Psychol. 2002;70:986–997. doi: 10.1037//0022-006x.70.4.986. [DOI] [PubMed] [Google Scholar]
  • 181.Wills TA, et al. Self-control, symptomatology, and substance use precursors: test of a theoretical model in a community sample of 9-year-old children. Psychol. Addict. Behav. 2007;21:205–215. doi: 10.1037/0893-164X.21.2.205. [DOI] [PubMed] [Google Scholar]
  • 182.Giancola PR, et al. Executive cognitive functioning and aggressive behavior in preadolescent boys at high risk for substance abuse/dependence. J. Stud. Alcohol. 1996;57:352–359. doi: 10.15288/jsa.1996.57.352. [DOI] [PubMed] [Google Scholar]
  • 183.Giancola PR, Mezzich AC, Tarter RE. Disruptive, delinquent and aggressive behavior in female adolescents with a psychoactive substance use disorder: Relation to executive cognitive functioning. J. Stud. Alcohol. 1998;59:560–567. doi: 10.15288/jsa.1998.59.560. [DOI] [PubMed] [Google Scholar]
  • 184.Ernst M, et al. Behavioral predictors of substance-use initiation in adolescents with and without attention-deficit/hyperactivity disorder. Pediatrics. 2006;117:2030–2039. doi: 10.1542/peds.2005-0704. [DOI] [PubMed] [Google Scholar]
  • 185.Jentsch JD, Taylor JR. Sex-related differences in spatial divided attention and motor impulsivity in rats. Behav. Neurosci. 2003;117:76–83. doi: 10.1037//0735-7044.117.1.76. [DOI] [PubMed] [Google Scholar]
  • 186.Everitt B, Robbins TW. Neural systems of reinforcement for drug addiction: from actions to habits to compulsion. Nat. Neurosci. 2005;8:1481–1489. doi: 10.1038/nn1579. [DOI] [PubMed] [Google Scholar]
  • 187.Oswald LM, et al. Impulsivity and chronic stress are associated with amphetamine-induced striatal dopamine release. Neuroimage. 2007;36:153–166. doi: 10.1016/j.neuroimage.2007.01.055. [DOI] [PubMed] [Google Scholar]
  • 188.Caspi A, et al. Role of genotype in the cycle of violence in maltreated children. Science. 2002;297:851–854. doi: 10.1126/science.1072290. [DOI] [PubMed] [Google Scholar]
  • 189.Caspi A, 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]
  • 190.Kaufman J, Yang BZ, Douglas-Palumberi H, et al. Social supports and serotonin transporter gene moderate depression in maltreated children. Proc. Nat. Acad. Sci. USA. 2004;101:17316–17321. doi: 10.1073/pnas.0404376101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Kaufman J, et al. Brain-derived neurotrophic factor-5-HTTLPR gene interactions and environmental modifiers of depression in children. Biol. Psychiatry. 2006;59:673–680. doi: 10.1016/j.biopsych.2005.10.026. [DOI] [PubMed] [Google Scholar]
  • 192.Tsuang M, et al. Genetic and environmental influences on transitions in drug use. Behav. Genet. 1999;29:473–479. doi: 10.1023/a:1021635223370. [DOI] [PubMed] [Google Scholar]
  • 193.Kendler KS, Prescott CA, Neale MC. The structure of genetic and environmental risk factors for common psychiatric and substance use disorders in women and men. Arch. Gen. Psychiatry. 2003;60:929–937. doi: 10.1001/archpsyc.60.9.929. [DOI] [PubMed] [Google Scholar]
  • 194.Kreek M, et al. Genetic influences on impulsivity, risk taking, stress responsivity and vulnerability to drug abuse and addiction. Nat. Neurosci. 2005;8:1450–1457. doi: 10.1038/nn1583. [DOI] [PubMed] [Google Scholar]
  • 195.Nestler EJ. Is there a common molecular pathway for addiction? Nat. Neurosci. 2005;8:1445–1449. doi: 10.1038/nn1578. [DOI] [PubMed] [Google Scholar]
  • 196.Kalivas PW, Volkow ND. The neural basis of addiction: a pathology of motivation and choice. Am. J. Psychiatry. 2005;162:1403–1413. doi: 10.1176/appi.ajp.162.8.1403. [DOI] [PubMed] [Google Scholar]
  • 197.Koob G, Kreek MJ. Stress, dysregulation of drug reward pathways, and the transition to drug dependence. Am. J. Psychiatry. 2007;164:1149–1159. doi: 10.1176/appi.ajp.2007.05030503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Pandey SC, et al. Neuropeptide Y and alcoholism: genetic, molecular, and pharmacological evidence. Alcoholism: Clin. Exp. Res. 2003;27:149–154. doi: 10.1097/01.ALC.0000052706.21367.0E. [DOI] [PubMed] [Google Scholar]
  • 199.Gehlert D. Introduction to the reviews on neuropeptide Y. Neuropeptides. 2004;38:135–140. doi: 10.1016/j.npep.2004.07.002. [DOI] [PubMed] [Google Scholar]
  • 200.Valdez GR, Koob GF. Allostasis and dysregulation of corticotropin-releasing factor and neuropeptide Y systems: implications for the development of alcoholism. Pharmacol. Biochem. Behav. 2004;79:671–689. doi: 10.1016/j.pbb.2004.09.020. [DOI] [PubMed] [Google Scholar]
  • 201.Kathuria S, et al. Modulation of anxiety through blockade of anandamide hydrolysis. Nat. Med. 2003;9:76–81. doi: 10.1038/nm803. [DOI] [PubMed] [Google Scholar]
  • 202.DiMarzo V, Matias I. Endocannabinoid control of food intake and energy balance. Nat. Neurosci. 2005;8:585–589. doi: 10.1038/nn1457. [DOI] [PubMed] [Google Scholar]
  • 203.Di S, et al. Rapid glucocorticoid-mediated endocannabinoid release and opposing regulation of glutamate and GABA inputs to hypothalamic magnocellular neurons. Endocrinology. 2005;145:4292–4301. doi: 10.1210/en.2005-0610. [DOI] [PubMed] [Google Scholar]
  • 204.Cobb CF, Van Thiel DH. Mechanism of ethanol-induced adrenal stimulation. Alcoholism: Clin. Exp. Res. 1982;6:202–206. doi: 10.1111/j.1530-0277.1982.tb04963.x. [DOI] [PubMed] [Google Scholar]
  • 205.Cinciripini PM, et al. The effects of smoking on the mood, cardiovascular and adrenergic reactivity of heavy and light smokers in a non-stressful environment. Biol. Psychol. 1989;29:273–289. doi: 10.1016/0301-0511(89)90023-9. [DOI] [PubMed] [Google Scholar]
  • 206.Wilkins JN, et al. Nicotine from cigarette smoking increases circulating levels of cortisol, growth hormone, and prolactin in male chronic smokers. Psychopharmacology. 1982;78:305–308. doi: 10.1007/BF00433730. [DOI] [PubMed] [Google Scholar]
  • 207.Wand GS, Dobs AS. Alterations in the hypothalamic-pituitary-adrenal axis in actively drinking alcoholics. J. Clin. Endocrinol. Metab. 1991;72:1290–1295. doi: 10.1210/jcem-72-6-1290. [DOI] [PubMed] [Google Scholar]
  • 208.Baumann MH, et al. Effects of intravenous cocaine on plasma cortisol and prolactin in human cocaine abusers. Biol. Psychiatry. 1995;38:751–755. doi: 10.1016/0006-3223(95)00083-6. [DOI] [PubMed] [Google Scholar]
  • 209.Heesch CM, et al. Effects of cocaine on cortisol secretion in humans. Am. J. Med. Sci. 1995;310:61–64. doi: 10.1097/00000441-199508000-00004. [DOI] [PubMed] [Google Scholar]
  • 210.Robinson TE, Berridge KC. The neural basis of drug craving: an incentive-sensitization theory of addiction. Brain Res. Brain Res. Rev. 1993;18:247–291. doi: 10.1016/0165-0173(93)90013-p. [DOI] [PubMed] [Google Scholar]
  • 211.Mello NK, Mendelson JH. Cocaine's effects on neuroendocrine systems: clinical and preclinical studies. Pharmacol. Biochem. Behav. 1997;57:571–599. doi: 10.1016/s0091-3057(96)00433-9. [DOI] [PubMed] [Google Scholar]
  • 212.Mendelson JH, et al. Effects of low- and high-nicotine cigarette smoking on mood states and the HPA axis in men. Neuropsychopharmacology. 2005;30:1751–1763. doi: 10.1038/sj.npp.1300753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Sofuoglu M, et al. Intravenous cocaine increases plasma epinephrine and norepinephrine in humans. Pharmacol. Biochem. Behav. 2001;68:455–459. doi: 10.1016/s0091-3057(01)00482-8. [DOI] [PubMed] [Google Scholar]
  • 214.Mendelson JH, et al. Cocaine tolerance: Behavioral, cardiovascular, and neuroendocrine function in men. Neuropsychopharmacology. 1998;18:263–271. doi: 10.1016/S0893-133X(97)00146-2. [DOI] [PubMed] [Google Scholar]
  • 215.D'Souza D, et al. The psychotomimetic effects of intravenous delta-9-tetrahydrocannabinol in healthy individuals: implications for psychosis. Neuropsychopharmacology. 2004;29:1558–1572. doi: 10.1038/sj.npp.1300496. [Clinical Trial. Journal Article. Randomized Controlled Trial] [DOI] [PubMed] [Google Scholar]
  • 216.Kreek MJ, Koob GF. Drug dependence: Stress and dysregulation of brain reward pathways. Drug Alcohol Depend. 1998;51:23–47. doi: 10.1016/s0376-8716(98)00064-7. [DOI] [PubMed] [Google Scholar]
  • 217.Chen H, Fu Y, Sharp BM. Chronic nicotine self-administration augments hypothalamic-pituitary-adrenal responses to mild acute stress. Neuropsychopharmacology. 2008;33:721–730. doi: 10.1038/sj.npp.1301466. [DOI] [PubMed] [Google Scholar]
  • 218.Ho WKK, et al. Comparison of plasma hormonal levels between heroin-addicted and normal subjects. Clinica Chimica Acta. 1977;75:415–419. doi: 10.1016/0009-8981(77)90360-6. [DOI] [PubMed] [Google Scholar]
  • 219.Facchinetti F, et al. Hypothalamic-pituitary-adrenal axis of heroin addicts. Drug Alcohol Depend. 1985;15:361–366. doi: 10.1016/0376-8716(85)90014-6. [DOI] [PubMed] [Google Scholar]
  • 220.Shively CA, et al. Effects of chronic moderate alcohol consumption and novel environment on heart rate variability in primates (Macaca fascicularis). Psychopharmacology (Berl.) 2007;192:183–191. doi: 10.1007/s00213-007-0709-z. [DOI] [PubMed] [Google Scholar]
  • 221.Thayer JF, et al. Alcohol use, urinary cortisol, and heart rate variability in apparently healthy men: Evidence for impaired inhibitory control of the HPA axis in heavy drinkers. Int. J. Psychophysiol. 2006;59:244–250. doi: 10.1016/j.ijpsycho.2005.10.013. [DOI] [PubMed] [Google Scholar]
  • 222.Bar KJ, et al. Heart rate variability and sympathetic skin response in male patients suffering from acute alcohol withdrawal syndrome. Alcohol Clin. Exp. Res. 2006;30:1592–1598. doi: 10.1111/j.1530-0277.2006.00191.x. [DOI] [PubMed] [Google Scholar]
  • 223.Ignar DM, Kuhn CM. Effects of specific mu and kappa opiate tolerance and abstinence on hypothalamo-pituitary-adrenal axis secretion in the rat. J. Pharmacol. Exp. Ther. 1990;255:1287–1295. [PubMed] [Google Scholar]
  • 224.Borowsky B, Kuhn CM. Monoamine mediation of cocaine-induced hypothalamo-pituitary-adrenal activation. J. Pharmacol. Exp. Ther. 1991;256:204–210. [PubMed] [Google Scholar]
  • 225.Alcaraz C, Vargas ML, Milanes MV. Chronic naloxone-induced supersensitivity affects neither tolerance to nor physical dependence on morphine at hypothalamus-pituitary-adrenocortical axis. Neuropeptides. 1996;30:29–36. doi: 10.1016/s0143-4179(96)90051-7. [DOI] [PubMed] [Google Scholar]
  • 226.Mantsch JR, et al. Daily cocaine self-administration under long-access conditions augments restraint-induced increases in plasma corticosterone and impairs glucocorticoid receptor-mediated negative feedback in rats. Brain Res. 2007;1167:101–111. doi: 10.1016/j.brainres.2007.05.080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Adinoff B, et al. Hypothalamic-pituitary-adrenal axis functioning and cerebrospinal fluid corticotropin releasing hormone and corticotropin levels in alcoholics after recent and long-term abstinence. Arch. Gen. Psychiatry. 1990;47:325–330. doi: 10.1001/archpsyc.1990.01810160025004. [DOI] [PubMed] [Google Scholar]
  • 228.Adinoff B, et al. Disturbances of hypothalamic-pituitary-adrenal axis functioning during withdrawal in six men. Am. J. Psychiatry. 1991;148:1023–1025. doi: 10.1176/ajp.148.8.1023. [DOI] [PubMed] [Google Scholar]
  • 229.Ehrenreich H, et al. Endocrine and hemodynamic effects of stress versus systemic CRF in alcoholics during early and medium term abstinence. Alcoholism: Clin. Exp. Res. 1997;21:1285–1293. [PubMed] [Google Scholar]
  • 230.Vescovi PP, et al. Diurnal variations in plasma ACTH, cortisol and beta-endorphin levels in cocaine addicts. Hormone Res. 1992;37:221–224. doi: 10.1159/000182316. [DOI] [PubMed] [Google Scholar]
  • 231.Tsuda A, et al. Cigarette smoking and psychophysiological stress responsiveness: Effects of recent smoking and temporary abstinence. Psychopharmacology. 1996;126:226–233. doi: 10.1007/BF02246452. [DOI] [PubMed] [Google Scholar]
  • 232.Kreek MJ. Opiate and cocaine addictions: Challenge for pharmacotherapies. Pharmacol. Biochem. Behav. 1997;57:551–569. doi: 10.1016/s0091-3057(96)00440-6. [DOI] [PubMed] [Google Scholar]
  • 233.Schluger JH, et al. Altered HPA axis responsivity to metyrapone testing in methadone maintained former heroin addicts with ongoing cocaine addiction. Neuropsychopharmacology. 2001;24:568–575. doi: 10.1016/S0893-133X(00)00222-0. [DOI] [PubMed] [Google Scholar]
  • 234.Ingjaldsson JT, Laberg JC, Thayer JF. Reduced heart rate variability in chronic alcohol abuse: relationship with negative mood, chronic thought suppression, and compulsive drinking. Biol. Psychiatry. 2003;54:1427–1436. doi: 10.1016/s0006-3223(02)01926-1. [DOI] [PubMed] [Google Scholar]
  • 235.Contoreggi C, et al. Stress hormone responses to corticotropin-releasing hormone in substance abusers without severe comorbid psychiatric disease. Soc. Biol. Psychiatry. 2003;54:873–878. doi: 10.1016/s0006-3223(03)00167-7. [DOI] [PubMed] [Google Scholar]
  • 236.Adinoff B, et al. Suppression of the HPA axis stress-response: implications for relapse. Alcohol Clin. Exp. Res. 2005;29:1351–1355. doi: 10.1097/01.ALC.0000176356.97620.84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Rasmussen DD, Wilkinson CW, Raskind MA. Chronic daily ethanol and withdrawal: 6. Effects on rat sympathoadrenal activity during “abstinence.”. Alcohol. 2006;38:173–177. doi: 10.1016/j.alcohol.2006.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Rechlin T, et al. Autonomic cardiac abnormalities in alcohol-dependent patients admitted to a psychiatric department. Clin. Auton. Res. 1996;6:119–122. doi: 10.1007/BF02291234. [DOI] [PubMed] [Google Scholar]
  • 239.Sinha R, et al. Enhanced negative emotion and alcohol craving, and altered physiological responses following stress and cue exposure in alcohol dependent individuals. Neuropsychophamacol. 2008 doi: 10.1038/npp.2008.78. [Epub ahead of print 18 June: doi: 10.1038/npp.2008.78] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.McDougle CJ, et al. Noradrenergic dysregulation during discontinuation of cocaine use in addicts. Arch. Gen. Psychiatry. 1994;51:713–719. doi: 10.1001/archpsyc.1994.03950090045007. [DOI] [PubMed] [Google Scholar]
  • 241.Di Chiara G, et al. Dopamine and drug addiction: the nucleus accumbens shell connection. Neuropharmacology. 2004;47(Suppl 1):227–241. doi: 10.1016/j.neuropharm.2004.06.032. [DOI] [PubMed] [Google Scholar]
  • 242.Rossetti ZL, Hmaidan Y, Gessa GL. Marked inhibition of mesolimbic dopamine release: a common feature of ethanol, morphine, cocaine and amphetamine abstinence in rats. Eur. J. Pharmacol. 1992;221:227–234. doi: 10.1016/0014-2999(92)90706-a. [DOI] [PubMed] [Google Scholar]
  • 243.Parsons LH, Smith AD, Justice JB., Jr. Basal extracellular dopamine is decreased in the rat nucleus accumbens during abstinence from chronic cocaine. Synapse. 1991;9:60–65. doi: 10.1002/syn.890090109. [DOI] [PubMed] [Google Scholar]
  • 244.Diana M, et al. Profound decrement of mesolombic dopaminergic neuronal activity during ethanol withdrawal syndrome in rats: Electophysiological and biochemical evidence. Proc. Natl. Acad. Sci. USA. 1993;90:7966–7969. doi: 10.1073/pnas.90.17.7966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Diana M, et al. Mesolimbic dopaminergic decline after cannabinoid withdrawal. Proc. Natl. Acad. Sci. USA. 1998;95:10269–10273. doi: 10.1073/pnas.95.17.10269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Weiss F, et al. Ethanol self-administration restores withdrawal-associated deficiencies in accumbal dopamine and 5-hydroxytryptamine release in dependent rats. J. Neurosci. 1996;16:3474–3485. doi: 10.1523/JNEUROSCI.16-10-03474.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Moore RJ, et al. Effect of cocaine self-administration on striatal dopamine D1 receptors in rhesus monkeys. Synapse. 1998;28:1–9. doi: 10.1002/(SICI)1098-2396(199801)28:1<1::AID-SYN1>3.0.CO;2-G. [DOI] [PubMed] [Google Scholar]
  • 248.Zhang Y, et al. Effect of chronic “binge cocaine” on basal levels and cocaine-induced increases of dopamine in the caudate putamen and nucleus accumbens of C57BL/6J and 129/J mice. Synapse. 2003;50:191–199. doi: 10.1002/syn.10251. [DOI] [PubMed] [Google Scholar]
  • 249.Nader MA, et al. PET imaging of dopamine D2 receptors during chronic cocaine self-administration in monkeys. Nat. Neurosci. 2006;9:1050–1056. doi: 10.1038/nn1737. [DOI] [PubMed] [Google Scholar]
  • 250.Koob GF, et al. Neurobiological mechanisms in the transition from drug use to drug dependence. Neurosci. Biobehav. Rev. 2004;27:739–749. doi: 10.1016/j.neubiorev.2003.11.007. [DOI] [PubMed] [Google Scholar]
  • 251.Mateo Y, et al. Reduced dopamine terminal function and insensitivity to cocaine following cocaine binge self-administration and deprivation. Neuropsychopharmacology. 2005;30:1455–1463. doi: 10.1038/sj.npp.1300687. [DOI] [PubMed] [Google Scholar]
  • 252.Beveridge T, et al. Effects of chronic cocaine self-administration on norepinephrine transporters in the nonhuman primate brain. Psychopharmacology. 2005;180:781–788. doi: 10.1007/s00213-005-2162-1. [DOI] [PubMed] [Google Scholar]
  • 253.Porrino LJ, et al. The effects of cocaine: a shifting target over the course of addiction. Prog. Neuro-Psychopharmacol. Biol. Psychiat. 2007;31:1593–1600. doi: 10.1016/j.pnpbp.2007.08.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Volkow ND, et al. Decreased dopamine D2 receptor availability is associated with reduced frontal metabolism in cocaine abusers. Synapse. 1993;14:169–177. doi: 10.1002/syn.890140210. [DOI] [PubMed] [Google Scholar]
  • 255.Volkow ND, et al. Decreases in dopamine receptors but not in dopamine transporters in alcoholics. Alcoholism: Clin. Exp. Res. 1996;20:1594–1598. doi: 10.1111/j.1530-0277.1996.tb05936.x. [DOI] [PubMed] [Google Scholar]
  • 256.Volkow ND, et al. Decreased striatal dopaminergic responsiveness in detoxified cocaine-dependent subjects. Nature. 1997;386:830–833. doi: 10.1038/386830a0. [DOI] [PubMed] [Google Scholar]
  • 257.Martinez D, et al. Amphetamine-induced dopamine release: markedly blunted in cocaine dependence and predictive of the choice to self-administer cocaine. Am. J. Psychiatry. 2007;164:622–629. doi: 10.1176/ajp.2007.164.4.622. [DOI] [PubMed] [Google Scholar]
  • 258.Gambarana C, et al. A chronic stress that impairs reactivity in rats also decreases dopaminergic transmission in the nucleus accumbens: a microdialysis study. J. Neurochem. 1999;72:2039–2046. doi: 10.1046/j.1471-4159.1999.0722039.x. [DOI] [PubMed] [Google Scholar]
  • 259.Robinson TE, Berridge KC. The psychology and neurobiology of addiction: an incentive-sensitization view. Addiction. 2000;95(Suppl 2):S91–S117. doi: 10.1080/09652140050111681. [DOI] [PubMed] [Google Scholar]
  • 260.Nestler E, Hope B, Widnell K. Drug addiction: a model for the molecular basis of neural plasticity. Neuron. 1993;11:995–1006. doi: 10.1016/0896-6273(93)90213-b. [DOI] [PubMed] [Google Scholar]
  • 261.White F, Hu XT, Henry DJ, Zhang XF. Neurophysiological alterations in the mesocorticolimbic dopamine system during repeated cocaine administration. In: Hammer R, editor. The Neurobiology of Cocaine: Cellular and Molecular Mechanisms. CRC Press; Boca Raton, FL: 1995. pp. 95–115. [Google Scholar]
  • 262.Pierce RC, Kalivas PW. A circuitry model of the expression of behavioral sensitization to amphetamine-like stimulants. Brain Res. Rev. 1997;25:192–216. doi: 10.1016/s0165-0173(97)00021-0. [DOI] [PubMed] [Google Scholar]
  • 263.Grimm JW, Shaham Y, Hope BT. Effect of cocaine and sucrose withdrawal period on extinction behavior, cue-induced reinstatement, and protein levels of the dopamine transporter and tyrosine hydroxylase in limbic and cortical areas in rats. Behav. Pharmacol. 2002;13:379–388. doi: 10.1097/00008877-200209000-00011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Lu L, et al. Incubation of cocaine craving after withdrawal: a review of preclinical data. Neuropharmacology. 2004;47(Suppl 1):214–226. doi: 10.1016/j.neuropharm.2004.06.027. [DOI] [PubMed] [Google Scholar]
  • 265.Hyman SE, Malenka RC, Nestler EJ. Neural mechanisms of addiction: the role of reward-related learning and memory. Annu. Rev. Neurosci. 2006;29:565–598. doi: 10.1146/annurev.neuro.29.051605.113009. [DOI] [PubMed] [Google Scholar]
  • 266.Hughes JR. Tobacco withdrawal in self-quitters. J. Consult. Clin. Psychol. 1992;60:689–697. doi: 10.1037//0022-006x.60.5.689. [DOI] [PubMed] [Google Scholar]
  • 267.Kouri EM, Pope HG, Jr., Lukas SE. Changes in aggressive behavior during withdrawal from long-term marijuana use. Psychopharmacology. 1999;143:302–308. doi: 10.1007/s002130050951. [DOI] [PubMed] [Google Scholar]
  • 268.Mulvaney FD, et al. Cocaine abstinence symptomatology and treatment attrition. J. Subst. Abuse. Treat. 1999;16:129–135. doi: 10.1016/s0740-5472(98)00017-8. [DOI] [PubMed] [Google Scholar]
  • 269.Budney AJ, Hughes JR. The cannabis withdrawal syndrome. Curr. Opin. Psychiatry. 2006;19:233–238. doi: 10.1097/01.yco.0000218592.00689.e5. [DOI] [PubMed] [Google Scholar]
  • 270.Volkow N, Fowler JS. Addiction, a disease of compulsion and drive: Involvement of the orbitofrontal cortex. Cereb. Cortex. 2000;10:318–325. doi: 10.1093/cercor/10.3.318. [DOI] [PubMed] [Google Scholar]
  • 271.Baker TB, Brandon TH, Chassin L. Motivational influences on cigarette smoking. Annu. Rev. Psychol. 2004;55:463–491. doi: 10.1146/annurev.psych.55.090902.142054. [DOI] [PubMed] [Google Scholar]
  • 272.Dodge R, Sindelar J, Sinha R. The role of depressive symptoms in predicting drug abstinence in outpatient substance abuse treatment. J. Subst. Abuse Treat. 2005;28:189–196. doi: 10.1016/j.jsat.2004.12.005. [DOI] [PubMed] [Google Scholar]
  • 273.Paliwal P, Hyman SM, Sinha R. Craving predicts time to cocaine relapse: Further validation of the Now and Brief versions of the cocaine craving questionnaire. Drug Alcohol Depend. 2008;93:252–259. doi: 10.1016/j.drugalcdep.2007.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Sinha R. The role of stress in addiction relapse. Curr. Psychiatry Rep. 2007;9:388–395. doi: 10.1007/s11920-007-0050-6. [DOI] [PubMed] [Google Scholar]
  • 275.Wikler A. Recent progress in research on the neurophysiological basis morphine addiction. Am. J. Psychiatry. 1948;105:328–338. doi: 10.1176/ajp.105.5.329. [DOI] [PubMed] [Google Scholar]
  • 276.O'Brien CP, et al. Conditioning factors in drug abuse: Can they explain complusion? J. Psychopharmacol. 1998;12:15–22. doi: 10.1177/026988119801200103. [DOI] [PubMed] [Google Scholar]
  • 277.Sayette MA, et al. The measurement of drug craving. Addiction. 2000;95(Suppl 2):S189–210. doi: 10.1080/09652140050111762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Childress A, et al. Cue reactivity and cue reactivity interventions in drug dependence. NIDA Res. Monogr. 1993;137:73–95. [PubMed] [Google Scholar]
  • 279.Rohsenow DJ, et al. Cue reactivity in addictive behaviors: Theoretical and treatment implications. Int. J. Addict. 1991;25:957–993. doi: 10.3109/10826089109071030. [DOI] [PubMed] [Google Scholar]
  • 280.Foltin RW, Haney M. Conditioned effects of environmental stimuli paired with smoked cocaine in humans. Psychopharmacology. 2000;149:24–33. doi: 10.1007/s002139900340. [DOI] [PubMed] [Google Scholar]
  • 281.Stewart JA. Pathways to Relapse: Factors Controlling the Reinitiation of Drug Seeking after Abstinence. University of Nebraska Press; Lincoln: 2003. [PubMed] [Google Scholar]
  • 282.Sinha R, et al. Hypothalamic-pituitary-adrenal axis and sympatho-adreno-medullary responses during stress-induced and drug cue-induced cocaine craving states. Psychopharmacology (Berl.) 2003;170:62–72. doi: 10.1007/s00213-003-1525-8. [DOI] [PubMed] [Google Scholar]
  • 283.Sinha R, O'Malley SS. Alcohol and craving: Findings from the clinic and laboratory. Alcohol Alcohol. 1999;34:223–230. doi: 10.1093/alcalc/34.2.223. [DOI] [PubMed] [Google Scholar]
  • 284.Sinha R, Catapano D, O'Malley S. Stress-induced craving and stress response in cocaine dependent individuals. Psychopharmacology (Berl.) 1999;142:343–351. doi: 10.1007/s002130050898. [DOI] [PubMed] [Google Scholar]
  • 285.Sinha R, et al. Psychological stress, drug-related cues and cocaine craving. Psychopharmacology (Berl). 2000;152:140–148. doi: 10.1007/s002130000499. [DOI] [PubMed] [Google Scholar]
  • 286.Fox HC, et al. Stress-induced and alcohol cue-induced craving in recently abstinent alcohol dependent individuals. Alcoholism: Clin. Exp. Res. 2007;31:395–403. doi: 10.1111/j.1530-0277.2006.00320.x. [DOI] [PubMed] [Google Scholar]
  • 287.Hyman SM, et al. Stress and drug-cue-induced craving in opioid-dependent individuals in naltrexone treatment. Exp. Clin. Psychopharmacol. 2007;15:134–143. doi: 10.1037/1064-1297.15.2.134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Lovallo WR, et al. Blunted stress cortisol response in abstinent alcoholic and polysubstanceabusing men. Alcoholism: Clin. Exp. Res. 2000;24:651–658. [PubMed] [Google Scholar]
  • 289.Al'absi M, Hatsukami DK, Davis G. Attenuated adrenocorticotropic responses to Psychological stress are associated with early smoking relapse. Psychopharmacology (Berl.) 2005;181:107–117. doi: 10.1007/s00213-005-2225-3. [DOI] [PubMed] [Google Scholar]
  • 290.Badrick E, Kirschbaum C, Kumari M. The relationship between smoking status and cortisol secretion. J. Clin. Endocrinol. Metab. 2007;92:819–824. doi: 10.1210/jc.2006-2155. [DOI] [PubMed] [Google Scholar]
  • 291.Fox HC, et al. Enhanced sensitivity to stress and drug/alcohol craving in abstinent cocaine-dependent individuals compared to social drinkers. Neuropsychopharmacology. 2008;33:796–805. doi: 10.1038/sj.npp.1301470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Grant S, et al. Activation of memory circuits during cue-elicited cocaine craving. Proc. Natl. Acad. Sci. USA. 1996;93:12040–12045. doi: 10.1073/pnas.93.21.12040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Childress AR, et al. Limbic activation during cue-induced cocaine craving. Am. J. Psychiatry. 1999;156:11–18. doi: 10.1176/ajp.156.1.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Kilts C, Schweitzer JB, Quinn CK, et al. Neural activity related to drug craving in cocaine addiction. Arch. Gen. Psychiatry. 2001;58:334–341. doi: 10.1001/archpsyc.58.4.334. [DOI] [PubMed] [Google Scholar]
  • 295.Kilts CD, et al. The neural correlates of cue-induced craving in cocaine-dependent women. Am. J. Psychiatry. 2004;161:233–241. doi: 10.1176/appi.ajp.161.2.233. [DOI] [PubMed] [Google Scholar]
  • 296.Li C-S, Kosten TR, Sinha R. Sex differences in brain activation during stress imagery in abstinent cocaine users: A functional magnetic resonance imaging study. Biol. Psychiatry. 2005;57:487–494. doi: 10.1016/j.biopsych.2004.11.048. [DOI] [PubMed] [Google Scholar]
  • 297.Sinha R, Li CS. Imaging stress- and cue-induced drug and alcohol craving: association with relapse and clinical implications. Drug Alcohol Rev. 2007;26:25–31. doi: 10.1080/09595230601036960. [DOI] [PubMed] [Google Scholar]
  • 298.Sinha R, et al. Neural activity associated with stress-induced cocaine craving: A functional magnetic imaging study. Psychopharmacol. 2005;183:171–180. doi: 10.1007/s00213-005-0147-8. [DOI] [PubMed] [Google Scholar]
  • 299.Wong DF, et al. Increased occupancy of dopamine receptors in human striatum during cue-elicited cocaine craving. Neuropsychopharmacology. 2006;31:2716–2727. doi: 10.1038/sj.npp.1301194. [DOI] [PubMed] [Google Scholar]
  • 300.Volkow ND, et al. Cocaine cues and dopamine in dorsal striatum: mechanism of craving in cocaine addiction. J. Neurosci. 2006;26:6583–6588. doi: 10.1523/JNEUROSCI.1544-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Grusser S, et al. Cue-induced activation of the striatum and medial prefrontal cortex is associated with subsequent relapse in abstinent alcoholics. Psychopharmacology (Berl.) 2004;175:296–302. doi: 10.1007/s00213-004-1828-4. [DOI] [PubMed] [Google Scholar]
  • 302.Wrase J, et al. Development of alcohol-associated cues and cue-induced brain activation in alcoholics. J. Assoc. Eur. Psychiatrists. 2002;17:287–291. doi: 10.1016/s0924-9338(02)00676-4. [DOI] [PubMed] [Google Scholar]
  • 303.Heinz A, et al. Correlation between dopamine D(2) receptors in the ventral striatum and central processing of alcohol cues and craving. Am. J. Psychiatry. 2004;161:1783–1789. doi: 10.1176/appi.ajp.161.10.1783. [DOI] [PubMed] [Google Scholar]
  • 304.Martinez D, et al. Alcohol dependence is associated with blunted dopamine transmission in the ventral striatum. Biol. Psychiatry. 2005;58:779–786. doi: 10.1016/j.biopsych.2005.04.044. [DOI] [PubMed] [Google Scholar]
  • 305.Hester R, Garavan H. Executive dysfunction in cocaine addiction: evidence for discordant frontal, cingulate, and cerebellar activity. J. Neurosci. 2004;24:11017–11022. doi: 10.1523/JNEUROSCI.3321-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Kaufman J, Ross TJ, Stein EA, Garavan H. Cingulate hypoactivity in cocaine users during a GO-NOGO task as revealed by event-related functional magnetic resonance imaging. J. Neurosci. 2003;23:7839–7843. doi: 10.1523/JNEUROSCI.23-21-07839.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307.Noel X, et al. Response inhibition deficit is involved in poor decision making under risk in non-amnesic individuals with alcoholism. Neuropsychology. 2007;21:778–786. doi: 10.1037/0894-4105.21.6.778. [DOI] [PubMed] [Google Scholar]
  • 308.Ersche KD, et al. Abnormal frontal activations related to decision-making in current and former amphetamine and opiate dependent individuals. Psychopharmacology (Berl.) 2005;180:612–623. doi: 10.1007/s00213-005-2205-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Ersche KD, et al. Profile of executive and memory function associated with amphetamine and opiate dependence. Neuropsychopharmacology. 2006;31:1036–1047. doi: 10.1038/sj.npp.1300889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Ersche KD, Roiser JP, Robbins TW, Sahakian BJ. Chronic cocaine but not chronic amphetamine use is associated with perseverative responding in humans. Psychopharmacology (Berl.) 2008;197(3):421–431. doi: 10.1007/s00213-007-1051-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Paulus MP, Tapert SF, Schuckit MA. Neural activation patterns of methamphetamine-dependent subjects during decision making predict relapse. Arch. Gen. Psychiatry. 2005;62:761–768. doi: 10.1001/archpsyc.62.7.761. [DOI] [PubMed] [Google Scholar]
  • 312.Li C.-s.R., et al. Neural correlates of impulse control during stop signal inhibition in cocaine-dependent men. Neuropsychopharmacology. 2008;33:1798–1806. doi: 10.1038/sj.npp.1301568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.O'Brien CP. Anticraving medications for relapse prevention: A possible new class of psychoactive medications. Am. J. Psychiatry. 2005;162:1423–1431. doi: 10.1176/appi.ajp.162.8.1423. [DOI] [PubMed] [Google Scholar]
  • 314.Vocci F, Acri J, Elkashef A. Medication development for addictive disorders: the state of the science. Am. J. Psychiatry. 2005;162:1432–1440. doi: 10.1176/appi.ajp.162.8.1432. [DOI] [PubMed] [Google Scholar]
  • 315.Shaham Y, et al. The reinstatement model of drug relapse: history, methodology and major findings. Psychopharmacology. 2003;168:3–20. doi: 10.1007/s00213-002-1224-x. [DOI] [PubMed] [Google Scholar]
  • 316.Shaham Y, Hope BT. The role of neuroadaptations in relapse to drug seeking. Nat. Neurosci. 2005;8:1437–1439. doi: 10.1038/nn1105-1437. [DOI] [PubMed] [Google Scholar]
  • 317.Weiss F. Neurobiology of craving, conditioned reward and relapse. Curr. Opin. Pharmacol. 2005;5:9–19. doi: 10.1016/j.coph.2004.11.001. [DOI] [PubMed] [Google Scholar]
  • 318.Marinelli PW, et al. The CRF1 receptor antagonist antalarmin attenuates yohimbine-induced increases in operant alcohol self-administration and reinstatement of alcohol seeking in rats. Psychopharmacology (Berl.) 2007;195:345–355. doi: 10.1007/s00213-007-0905-x. [DOI] [PubMed] [Google Scholar]
  • 319.George O, et al. CRF-CRF1 system activation mediates withdrawal-induced increases in nicotine self-administration in nicotine-dependent rats. Proc. Natl. Acad. Sci. USA. 2007;104:17901–17902. doi: 10.1073/pnas.0707585104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320.Mantsch JR, et al. Stressor- and corticotropin releasing factor-induced reinstatement and active stress-related behavioral responses are augmented following long-access cocaine self-administration by rats. Psychopharmacology (Berl.) 2008;195:591–603. doi: 10.1007/s00213-007-0950-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321.Koob GF, Le Moal M. Plasticity of reward neurocircuitry and the ‘dark side’ of drug addiction. Nat. Neurosci. 2005;8:1442–1444. doi: 10.1038/nn1105-1442. [DOI] [PubMed] [Google Scholar]
  • 322.Lu L, et al. Systemic and central amygdala injections of the mGluR(2/3) agonist LY379268 attenuate the expression of incubation of cocaine craving. Biol. Psych. 2007;61:591–598. doi: 10.1016/j.biopsych.2006.04.011. [DOI] [PubMed] [Google Scholar]
  • 323.Zhao Y, et al. Activation of group II metabotropic glutamate receptors attenuates both stress and cue-induced ethanol-seeking and modulates c-fos expression in the hippocampus and amygdala. J. Neurosci. 2006;26:9967–9974. doi: 10.1523/JNEUROSCI.2384-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Aujla H, Martin-Fardon R, Weiss F. Rats with extended access to cocaine exhibit increased stress reactivity and sensitivity to the anxiolytic-like effects of the mGluR 2/3 agonist LY379268 during abstinence. Neuropsychopharmacology. 2007;33:1818–1826. doi: 10.1038/sj.npp.1301588. [DOI] [PubMed] [Google Scholar]
  • 325.Sinha R, et al. Stress-induced cocaine craving and hypothalamic-pituitary-adrenal responses are predictive of cocaine relapse outcomes. Arch. Gen. Psychiatry. 2006;63:324–331. doi: 10.1001/archpsyc.63.3.324. [DOI] [PubMed] [Google Scholar]
  • 326.Cooney NL, et al. Alcohol cue reactivity, negative-mood reactivity, and relapse in treated alcoholic men. J. Abnorm. Psychol. 1997;106:243–250. doi: 10.1037//0021-843x.106.2.243. [DOI] [PubMed] [Google Scholar]
  • 327.Junghanns K, Backhaus J, Tietz U. Impaired serum cortisol stress response is a predictor of early relapse. Alcohol Alcohol. 2003;38:189–193. doi: 10.1093/alcalc/agg052. [DOI] [PubMed] [Google Scholar]
  • 328.Brady KT, et al. Cold pressor task reactivity: predictors of alcohol use among alcohol-dependent individuals with and without comorbid posttraumatic stress disorder. Alcohol Clin. Exp. Res. 2006;30:938–946. doi: 10.1111/j.1530-0277.2006.00097.x. [DOI] [PubMed] [Google Scholar]
  • 329.Breese GR, et al. Stress enhancement of craving during sobriety and the risk of relapse. Alcoholism: Clin. Exp. Res. 2005;29:185–195. doi: 10.1097/01.alc.0000153544.83656.3c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Sinha R, Kimmerling A, Doebrick C. Effects of lofexidine on stress-induced and cue-induced opioid craving and opioid abstinence rates: preliminary findings. Psychopharmacology. 2007;190:569–574. doi: 10.1007/s00213-006-0640-8. [DOI] [PubMed] [Google Scholar]

RESOURCES