Altered Vulnerability to Acute Opiate Withdrawal Following Stress: Roles of N-Methyl-D-Aspartate and Glucocorticoid Receptors

Size: px
Start display at page:

Download "Altered Vulnerability to Acute Opiate Withdrawal Following Stress: Roles of N-Methyl-D-Aspartate and Glucocorticoid Receptors"

Transcription

1 Behavioral Neuroscience Copyright 2005 by the American Psychological Association 2005, Vol. 119, No. 5, /05/$12.00 DOI: / Altered Vulnerability to Acute Opiate Withdrawal Following Stress: Roles of N-Methyl-D-Aspartate and Glucocorticoid Receptors Gavan P. McNally and Sara Lam University of New South Wales Five experiments studied the modulation of acute opiate withdrawal by restraint stress. Rats were subjected to a 2-hr restraint stress, and 1, 3, or 7 days later they received a single injection of morphine followed by injection of naloxone. Naloxone precipitated a withdrawal syndrome. This syndrome was enhanced when it occurred 1 day after stress but was reduced when it occurred 7 days after stress. The enhancement of withdrawal by restraint stress was prevented by treatment with the N-methyl-D-aspartate (NMDA) receptor antagonist MK801 or the glucocorticoid receptor antagonist RU486 prior to stress. Together these experiments show that restraint stress alters vulnerability to opiate withdrawal and identify activation of NMDA and glucocorticoid receptors as causal to this vulnerability. Keywords: stress, withdrawal, sensitization, MK801, RU486 Gavan P. McNally and Sara Lam, School of Psychology, University of New South Wales, Sydney, New South Wales, Australia. This research was supported by Project Grant from the National Health and Medical Research Council. Correspondence concerning this article should be addressed to Gavan P. McNally, School of Psychology, University of New South Wales, Sydney, New South Wales 2052, Australia. g.mcnally@unsw.edu.au Stress has a profound influence on opiate and psychostimulant dependence. Animal models have consistently shown that exposure to stressors such as intermittent footshock (Goeders & Guerin, 1994; Shaham & Stewart, 1994), restraint (Deroche et al., 1992), food restriction (Deroche et al., 1993), social isolation (Deroche, Piazza, LeMoal, & Simon, 1994), and social defeat (Covington & Miczek, 2001; Haney, Maccari, LeMoal, Simon, & Piazza, 1995) facilitate the behavioral impact as well as the intravenous selfadministration of opiates and psychostimulants. Moreover, such stressors can induce relapse to drug seeking in animals previously trained to self-administer opiates or psychostimulants (Ahmed & Koob, 1997; Erb, Shaham, & Stewart, 1996; Mantch & Goeders, 1999; Shaham & Stewart, 1995). Although these interactions have typically been observed with prolonged or multiple exposures, there is evidence that similar interactions can also occur following acute exposure to stress and drugs of abuse. For example, Will, Watkins, and Maier (1998) exposed rats to a single session of 100 inescapable tailshocks and commenced conditioned place preference training based on injection of morphine at varying intervals following these shocks. Their results showed that the single session of inescapable shock facilitated acquisition of a morphineinduced place preference when that acquisition occurred up to 7 days after shock. These interactions occur, at least in part, because of shared neural mechanisms between stress and drugs of abuse (e.g., Lu, Shepard, Hall, & Shaham, 2003; Marinelli & Piazza, 2002; Piazza & LeMoal, 1998). Opioid peptides (e.g., Zurita, Martijena, Cuadra, Brandao, & Molina, 2000), noradrenaline (e.g., Shaham, Highfield, Delfs, Leung, & Stewart, 1997), corticotropin-releasing hormone (e.g., Erb & Stewart, 1999), and glucocorticoids (e.g., Piazza & LeMoal, 1997) are all critical for the interactions between stress and drugs of abuse. For example, surgical and pharmacological suppression of glucocorticoids prevents the enhancement of morphine-conditioned place preference by uncontrollable stress (Der-Avakian et al., 2005). Moreover, there is evidence that these interactions occur because of common effects on neurotransmission in the mesolimbic dopamine pathway. For example, Saal, Dong, Bonci, and Malenka (2003) reported that exposure to either drugs of abuse (e.g., morphine, cocaine, or amphetamine) or forced swim stress increased the strength of excitatory amino-acid synapses at rat midbrain dopamine neurons. This potentiation of excitatory amino-acid synapses by stress could be prevented by pretreatment with either the noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist MK801 or the glucocorticoid receptor antagonist RU486 (Saal et al., 2003). These results were interpreted to mean that stress and drugs of abuse produce similar alterations in dopamine neurotransmission so that prior exposure to stress may prime, sensitize, or otherwise increase this neurotransmission in response to drugs of abuse. In contrast to the well-documented interactions between stress and opiate reward, considerably less attention has been directed toward possible interactions between stress and opiate withdrawal. Drug withdrawal can play an important role in maintaining persistent drug taking (e.g., Hutcheson, Everitt, Robbins, & Dickinson, 2001). It follows that manipulations that alter the severity of drug withdrawal will have an important impact on drug selfadministration and persistent drug taking. Consistent with interactions between stress and opiate withdrawal, Shaham (1993) reported that rats repeatedly exposed to restraint stress prior to drug self-administration showed augmented drug self-administration and augmented withdrawal. Likewise, Williams, Drugan, and Maier (1984) reported that rats that received two daily sessions of 80 inescapable shocks showed enhanced withdrawal behaviors 24 hr later when injected with morphine followed by a naloxone challenge. Indeed, prolonged exposure to stress, and consequent prolonged activation of endogenous opioid systems, may itself 1215

2 1216 MCNALLY AND LAM produce expression of withdrawal-like behaviors. For example, Miczek, Thompson, and Shuster (1986) subjected mice to 7 days of stress by using the resident intruder preparation. These mice displayed opiate withdrawal-like behaviors when administered an opioid receptor antagonist in the absence of any opiate agonist. Together these and other (Manik & Katz, 1984) findings clearly support the possibility that stress may prime, sensitize, or otherwise increase vulnerability to opiate withdrawal. The aim of the present series of experiments was to characterize this relationship between stress and opiate withdrawal. Opiate-naive rats were first subjected to 2-hr restraint stress. Twenty-four hours later they were injected with morphine followed 30 min later by naloxone. We selected an acute opiate withdrawal preparation to study stress withdrawal interactions because prior research has shown that a history of opiate exposures alters responsivity to restraint stress (Blatchford, Diamond, Westbrook, & McNally, in press). In this way, it was possible to ensure that any differences between groups in withdrawal severity were not due to the effects of the opiate history on stress responsivity (i.e., increasing vulnerability to the stress). In Experiment 1, we studied the effects of a 2-hr exposure to restraint stress on opiate withdrawal precipitated 24 hr after stress. In Experiment 2, we studied the time course of the modulation of opiate withdrawal by stress. The role of NMDA receptors in the modulation of opiate withdrawal by stress was studied in Experiment 3. In Experiment 4, we studied the role of glucocorticoid receptors in the modulation of opiate withdrawal by stress. Finally, in Experiment 5, we studied the effects of NMDA and glucocorticoid receptor antagonism on opiate withdrawal in the absence of restraint stress to ensure that the effects of these drugs were specific to the interaction between stress and opiate withdrawal. Subjects Method Adult male Wistar rats ( g) were obtained from a commercial supplier (Gore Hill Research Laboratories, Sydney, New South Wales, Australia). After arrival, rats were housed in groups of 6 8 in plastic cages maintained on a 12:12-hr light dark cycle (lights on at 7:00 a.m.) and were allowed access to water and food ad libitum. The rats were handled (1 2 min per rat per day) for 3 days to habituate them to the experimenter. The procedures used in these experiments were approved by the Animal Care and Ethics Committee at the University of New South Wales. Apparatus and Drugs Four buckets (29 cm diameter 50 cm height) were used as test chambers. Four Plexiglas cylinders (6 cm diameter; 21 cm length) were used as restrainers. Morphine HCl (a generous gift from Glaxo-Smith- Kline, Melbourne, Victoria, Australia) was dissolved in sterile 0.9% (wt/ vol) saline to obtain a concentration of 15 mg/ml and was injected subcutaneously in a volume of 1 ml/kg. Naloxone HCl (Sigma, Sydney, New South Wales, Australia) was dissolved in sterile 0.9% (wt/vol) saline to obtain a concentration of 5 mg/ml and was injected subcutaneously in a volume of 1 ml/kg. MK801 (Sigma, Sydney, New South Wales, Australia) was dissolved in sterile 0.9% (wt/vol) saline to obtain a concentration of 0.1 mg/ml and was injected intraperitoneally in a volume of 1 ml/kg. RU486 (Sigma, Sydney, New South Wales, Australia) was dissolved in dimethyl sulfoxide (DMSO) to obtain a concentration of 40 mg/ml and was injected subcutaneously in a volume of 1 ml/kg. Procedure Experiment 1: Interactions between stress and opiate withdrawal. Thirty-two rats were randomly allocated to one of four groups: no stress saline, stress saline, no stress morphine, and stress morphine (n 8 per group). In the morning and afternoon of Day 1, rats were transported to the laboratory and placed in the test chambers for 30 min. This was done to familiarize the animals with the test chambers. On Day 2, all rats were transported to a different laboratory. Rats in the stress groups were placed in restrainers for 2 hr, whereas rats in the no-stress groups were briefly handled and returned to their home cages. On Day 3, rats were transported to the test laboratory. Upon arrival, all rats were briefly removed from their home cage, injected, and then returned to their home cage. Rats in the morphine groups were injected with 15 mg/kg morphine, whereas rats in the saline groups were injected with saline. Thirty minutes later, all rats were removed from their home cages, injected with 5 mg/kg naloxone, and then placed in the test chambers. Rats were observed continuously for signs of opiate withdrawal for 15 min. An observer scored behavior for the following signs of opiate withdrawal: wet dog shakes, head shakes, teeth chattering, diarrhea, salivation, secretions from the eyes or nose, ptosis, ejaculation or genital grooming, postural abnormalities (lying on the belly), and irritability on handling (Blasig, Herz, Reinhold, & Zieglgansberger, 1973). All signs were counted. Salivation, secretions from the eyes or nose, ptosis, and postural abnormalities, if observed, counted once only within each 5-min period as a result of difficulties in distinguishing individual bouts. The total number of withdrawal signs observed was calculated. Experiment 2: Time course of interactions between stress and opiate withdrawal. Thirty-two rats were randomly allocated to one of four groups: control, 1 day, 3 days, and 7 days (n 8 per group). Twice on Day 1, rats were transported to the laboratory and placed in the test chambers for 30 min as described above. On Day 2, rats in the 1-day, 3-days, and 7-days groups were placed in restrainers for 2 hr. Rats in the control group were briefly handled before being returned to their home cage. Rats were transported to the test laboratory either 1 day, 3 days, or 7 days later according to their group allocations. Rats in the control group were transported to the test laboratory 1 day (n 2), 3 days (n 3), or 7 days (n 3) later. Upon arrival, rats were briefly removed from their home cage, injected with 15 mg/kg morphine, and then returned to their home cage. Thirty minutes later, they were removed from their home cages, injected with 5 mg/kg naloxone, and then placed in the test chambers. Rats were observed continuously for signs of opiate withdrawal for 15 min as described above. Experiment 3: Role of NMDA receptors in interactions between stress and opiate withdrawal. Forty rats were randomly allocated to one of five groups: control, 1 day saline, 1 day MK801, 7 days saline, and 7 days MK801 (n 8 per group). Twice on Day 1, rats were transported to the laboratory and placed in the test chambers for 30 min as described above. On Day 2, rats in the 1 day saline and 7 days saline groups were injected intraperitoneally with saline, whereas rats in the 1 day MK801 and 7 days MK801 groups were injected intraperitoneally with 0.1 mg/kg MK801. Thirty minutes later, rats in these groups were placed in restrainers for 2 hr. Rats in the control group were briefly handled before being returned to their home cages. Rats were transported to the laboratory and tested either 1 day or 7 days later according to their group allocations. Half of the rats in the control group were tested at 1 day, and the remainder were tested at 7 days. Upon arrival, rats in all groups were briefly removed from their home cage, injected with 15 mg/kg morphine, and then returned to their home cage. Thirty minutes later, rats were removed from their home cages, injected with 5 mg/kg naloxone, and then placed in the test chambers. Rats were observed continuously for signs of opiate withdrawal for 15 min as described above. Experiment 4: Role of glucocorticoid receptors in the interaction between stress and opiate withdrawal. Thirty-five rats were randomly allocated to one of five groups: control, 1 day vehicle, 1 day RU486, 7 days vehicle, and 7 days RU486 (n 7 per group). Twice on Day 1, rats

3 ACUTE OPIATE WITHDRAWAL FOLLOWING STRESS IN RATS 1217 were transported to the laboratory and placed in the test chambers for 30 min as described above. On Day 2, rats in the 1 day vehicle and 7 days vehicle groups were injected subcutaneously with DMSO vehicle, whereas rats in the 1 day RU486 and 7 days RU486 groups were injected subcutaneously with 40 mg/kg RU486. This dose was chosen because it has previously been shown to prevent the effects of swim stress at excitatory amino-acid synapses in the midbrain (Saal et al., 2003). Thirty minutes later, rats in these groups were placed in restrainers for 2 hr. Rats in the control group were briefly handled before being returned to their home cage. Rats were transported to the test laboratory either 1 day or 7 days later according to their group allocations. Rats in the control group were transported to the laboratory and tested either 1 day (n 4) or 7 days (n 3) later. Upon arrival, rats in all groups were briefly removed from their home cage, injected with 15 mg/kg morphine, and then returned to their home cage. Thirty minutes later, rats were removed from their home cages, injected with 5 mg/kg naloxone, and then placed in the test chambers. Rats were observed continuously for signs of opiate withdrawal for 15 min as described above. Experiment 5: The effects of MK801 and RU486 on opiate withdrawal. Thirty-two rats were randomly allocated to one of four groups: saline, MK801, DMSO, and RU486. Twice on Day 1, rats were transported to the laboratory and placed in the test chambers for 30 min as described above. On Day 2, the rats were injected subcutaneously with DMSO or 40 mg/kg RU486 or intraperitoneally with saline or 0.1 mg/kg MK801. The following day, rats were tested. They were transported to the laboratory, and upon arrival, they were briefly removed from their home cage, injected with 15 mg/kg morphine, and then returned to their home cage. Thirty minutes later, rats were removed from their home cage, injected with 5 mg/kg naloxone, and then placed in the test chambers. Rats were observed continuously for signs of opiate withdrawal for 15 min as described above. Statistical Analysis The data were analyzed by means of planned orthogonal contrasts controlling the decision-wise error rate ( ) at 0.05 with the methods described by Harris (1994). Results Experiment 1: Interactions Between Stress and Opiate Withdrawal test is shown in Figure 1. From inspection, it is clear that challenge Figure 1. Stress enhanced opiate withdrawal. Rats were subjected to a 2-hr restraint stress (stress groups) or not (control group). Twenty-four hours later, rats received an injection of morphine (15 mg/kg) or saline followed 30 min later by an injection of naloxone (5 mg/kg). * significant difference from the no stress morphine group. injection of naloxone among animals injected with morphine precipitated an acute withdrawal syndrome (the no stress morphine group) as compared with animals not injected with morphine (the no stress saline group). This acute precipitated withdrawal syndrome differed from that previously described in animals chronically exposed to morphine (e.g., McNally & Akil, 2002b). The acute withdrawal syndrome was characterized predominantly by teeth chattering, whole body or head shakes, genital grooming, and postural abnormalities (e.g., lying on the belly). In contrast to the withdrawal syndrome precipitated from chronic morphine, there were no instances of escape jumping, salivation, or secretions from the eyes and nose, and there were only a few instances of ptosis or diarrhea. There was a significant main effect for morphine versus saline, F(1, 28) 94.5, p.05. Thus, injection of naloxone precipitated signs of opiate withdrawal that were significantly greater among rats previously injected with morphine than rats previously injected with saline. There was a significant main effect for stress versus no stress, F(1, 28) 9.3, p.05. Thus, injection of naloxone precipitated signs of opiate withdrawal that were significantly greater among rats previously subjected to restraint stress. Finally, there was a significant interaction between stress versus no stress and morphine versus saline, F(1, 28) 7.9, p.05. In other words, the effect of restraint stress was to selectively enhance the withdrawal syndrome among the stress morphine group. This was confirmed by a subsequent analysis that showed a significant difference between the stress morphine and no stress morphine groups, F(1, 28) 17.3, p.05. This enhancement was especially pronounced for teeth chattering, postural abnormalities, genital grooming, and ptosis and was less pronounced for body or head shakes. Experiment 2: Time Course of Interactions Between Stress and Opiate Withdrawal test is shown in Figure 2. From inspection, it is apparent that restraint stress enhanced withdrawal from morphine precipitated 1 day but not 3 or 7 days later. Indeed, the opiate withdrawal syndrome appeared to be reduced when it occurred 7 days after restraint stress. These observations were confirmed by the statistical analysis. There was a significant increase in withdrawal among rats in the 1-day group compared with rats in the control group, F(1, 28) 22.4, p.05. This enhancement was especially pronounced for teeth chattering, genital grooming, and ptosis and was less pronounced for postural abnormalities for body or head shakes. There was no significant difference in withdrawal among rats in the 3-days group as compared with rats in the control group, F(1, 28) 1, p.05. Finally, there was a significant decrease in withdrawal among rats in the 7-days group as compared with rats in the control group, F(1, 28) 7.7, p.05. This reduction was especially pronounced for teeth chattering, genital grooming, and postural abnormalities. Experiment 3: Role of NMDA Receptors in the Interaction Between Stress and Opiate Withdrawal test is shown in Figure 3. From inspection, it can be seen that

4 1218 MCNALLY AND LAM There was, however, no significant difference in the number of withdrawal signs observed among the 7 days MK801 group as compared with the 7 days saline group, F(1, 34) 1, p.05. Thus, NMDA receptor antagonism during restraint stress did not prevent the reduction of withdrawal 7 days later. Experiment 4: Role of Glucocorticoid Receptors in the Interaction Between Stress and Opiate Withdrawal Figure 2. The time course of stress modulation of opiate withdrawal. Rats were subjected to a 2-hr restraint stress (1, 3, and 7 days groups) or not (control [Ctrl] group). One, 3, or 7 days later, rats received an injection of morphine (15 mg/kg) followed by an injection of naloxone (5 mg/kg). Stress enhanced withdrawal at 1 day, did not affect withdrawal at 3 days, and reduced withdrawal at 7 days. * significant difference from the control group. withdrawal from morphine was enhanced when it was precipitated 1 day after restraint but was reduced when it occurred 7 days after restraint. Injection of the noncompetitive NMDA receptor antagonist MK801 prior to restraint stress appeared to abolish the enhancement of opiate withdrawal but left unaffected the reduction in opiate withdrawal. The statistical analysis confirmed these observations. There was a significant increase in the number of withdrawal signs observed among the 1 day saline group as compared with the control group, F(1, 34) 7.2, p.05, but a significant decrease in the number of withdrawal signs observed among the 7 days saline group as compared with the control group, F(1, 34) 5.0, p.05. There was a significant decrease in the number of withdrawal signs observed among the 1 day MK801 group as compared with the 1 day saline group, F(1, 34) 21.2, p.05. Thus, NMDA receptor antagonism during restraint stress prevented the facilitation of withdrawal 1 day later. test is shown in Figure 4. From inspection, it can be seen that withdrawal from morphine was enhanced when it was precipitated 1 day after restraint but was reduced when it occurred 7 days after restraint. Injection of the glucocorticoid receptor antagonist RU486 prior to restraint stress appeared to abolish the enhancement of opiate withdrawal observed 1 day after restraint but left unaffected the reduction in opiate withdrawal observed 7 days after restraint. The statistical analysis confirmed these observations. There was a significant increase in the number of withdrawal signs observed among the 1 day saline group as compared with the control group, F(1, 30) 4.8, p.05, but a significant decrease in the number of withdrawal signs observed among the 7 days saline group as compared with the control group, F(1, 30) 7.9, p.05. There was also a significant decrease in the number of withdrawal signs observed among the 1 day RU486 group as compared with the 1 day saline group, F(1, 30) 7.7, p.05. Thus, glucocorticoid receptor antagonism during restraint stress prevented the enhancement of withdrawal 1 day later. There was, however, no significant difference in the number of withdrawal signs observed among the 7 days RU486 group as compared with the 7 days saline group, F(1, 30) 1, p.05. Thus, glucocorticoid receptor antagonism during restraint stress did not prevent the reduction of withdrawal 7 days later. Figure 3. N-methyl-D-aspartate receptors mediated the enhancement of withdrawal by stress. Rats were injected with saline (saline groups) or MK801 (0.1 mg/kg; MK801 groups) and then subjected to a 2-hr restraint stress (1 and 7 days groups) or not (control [Ctrl] group). Either 1 or 7 days later, rats received an injection of morphine (15 mg/kg) followed by an injection of naloxone (5 mg/kg). Stress enhanced withdrawal at 1 day, and this enhancement was not observed in rats previously injected with MK801. Stress reduced withdrawal at 7 days, and this reduction was observed among rats injected with MK801. * significant difference from control group; # significant difference from the 1 day saline group. Figure 4. Glucocorticoid receptors mediated the enhancement of withdrawal by stress. Rats were injected with saline (saline groups) or RU486 (40 mg/kg; RU486 groups) and then subjected to a 2-hr restraint stress (1 and 7 days groups) or not (control [Crtl] group). Either 1 or 7 days later, rats received an injection of morphine (15 mg/kg) followed by an injection of naloxone (5 mg/kg). Stress enhanced withdrawal at 1 day, and this enhancement was not observed in rats injected with RU486 prior to stress. Stress reduced withdrawal at 7 days, and this reduction was observed among rats injected with RU486 prior to stress. * significant difference from control group; # significant difference from the 1 day saline group.

5 ACUTE OPIATE WITHDRAWAL FOLLOWING STRESS IN RATS 1219 Experiment 5: Effects of MK801 and RU486 on Expression of Opiate Withdrawal test is shown in Figure 5. From inspection, it can be seen that injection of either MK801 or RU hr prior to withdrawal failed to alter opiate withdrawal. There was no significant difference in withdrawal between rats injected with MK801 versus saline 1 day prior to test, F(1, 28) 1, p.05. There was also no significant difference in withdrawal between rats injected with RU486 versus vehicle 1 day prior to test, F(1, 28) 1, p.05. Together these results show that the effects of MK801 and RU486 are specific to preventing the enhancement of withdrawal by restraint stress. Discussion Injection of naloxone among morphine-treated animals precipitated an acute opiate withdrawal syndrome characterized predominantly by teeth chattering, whole body or head shakes, genital grooming, and postural abnormalities. This withdrawal syndrome was specific to the interaction between naloxone and morphine because it was not observed in animals injected with naloxone but not injected with morphine. Acute opiate withdrawal was significantly enhanced by a 2-hr period of restraint stress when that stress occurred 1 day prior to opiate withdrawal. This enhancement was selective to an interaction between stress and opiate withdrawal because it was not observed among rats subject to a 2-hr period of restraint stress and injected 1 day later with saline followed by naloxone. These results show that prior exposure to stress can increase the severity of opiate withdrawal. The present results are consistent with the results of Williams et al. (1984), who showed that prior exposures to inescapable tailshock increased the severity of opiate withdrawal. Moreover, the present results are also consistent with the large body of literature showing that exposure to stress typically increases the behavioral and neurobiological effects of drugs of abuse (Lu et al., 2003). It is interesting to note that the present results show that an increase in the behavioral effects of drugs of abuse is not an inevitable consequence of prior to exposure to stress. The results of Experiment 2 revealed no change Figure 5. N-methyl-D-aspartate and glucocorticoid receptor antagonists did not modulate opiate withdrawal in the absence of stress. Rats were injected with saline, dimethyl sulfoxide (vehicle), MK801, or RU hr prior to receiving an injection of morphine (15 mg/kg) followed by an injection of naloxone (5 mg/kg). Pretreatment with MK801 or RU486 did not alter opiate withdrawal. in opiate withdrawal when withdrawal was precipitated 3 days after restraint stress. More important, the results of Experiments 2 4 consistently showed that opiate withdrawal was significantly reduced by a 2-hr period of restraint stress when that withdrawal was precipitated 7 days after restraint. This ability of a brief episode of restraint stress to exert long-lasting (up to 7 days) effects on responding to opiates is surprising and to the best of our knowledge is the first demonstration that an environmental event can decrease the severity of opiate withdrawal. These interactions between stress and opiate withdrawal were mediated, at least in part, by excitatory amino-acid and glucocorticoid receptors because administrations of either MK801 or RU486 prior to restraint stress prevented the facilitation of opiate withdrawal observed 1 day later. There was no evidence here that MK801 or RU486 prevented the stress-induced enhancement of opiate withdrawal because they had some long-lasting (i.e., 24 hr) nonspecific effect on the behavioral expression of acute withdrawal. The results of Experiment 5 showed that injection of MK801 or RU hr prior to acute withdrawal had no effect on the severity of withdrawal. This finding is consistent with prior work showing that MK801 at the dose used here does not disrupt the expression of withdrawal from chronic morphine (Trujillo & Akil, 1991). Rather, our results indicate that MK801 and RU486 acted selectively to prevent the enhancement of withdrawal by restraint stress. Thus, the present experiments show that activation of NMDA and glucocorticoid receptors during restraint stress is critical for the ability of that restraint stress to enhance opiate withdrawal. This role of NMDA and glucocorticoid receptors in determining the interaction between stress and opiates is consistent with demonstrations that these receptors mediate the effects of stress and opiates on other behavioral and neuronal responses (e.g., Lu et al., 2003; Saal et al., 2003). It is interesting to note that the reduction in withdrawal severity 7 days after restraint stress was unaffected by either NMDA or glucocorticoid receptor antagonism. This finding was surprising and indicates a dissociation in the mechanisms for stress-induced increases and decreases in opiate withdrawal severity. The mechanisms for this 7-day reduction in sensitivity to acute opiate withdrawal as well as a full characterization of the time course of stress-withdrawal interactions warrant further investigation. Together, these findings indicate an important, but complex, interaction between stress and opiate withdrawal so that stress increases vulnerability to opiate withdrawal in the short-term via NMDA and glucocorticoid receptor-dependent mechanisms but reduces the severity of opiate withdrawal in the long-term via different mechanisms. It is not immediately clear why the same event (restraint stress) should both facilitate and inhibit opiate withdrawal. One possibility is that that these interactions represent the operation of nonassociative forms of learning namely, sensitization and habituation. That is, how the animal responds to one biologically significant event at Time 2 (e.g., withdrawal from a drug of abuse) is influenced by both feedforward (e.g., sensitization) and feedback (e.g., habituation) mechanisms from other biologically significant events experienced previously at Time 1. The present data could be interpreted to mean that restraint stress produces a short-term sensitization of opiate withdrawal via NMDA and glucocorticoid mechanisms and a long-term habituation of opiate withdrawal via different mechanisms. Irrespective of the fate of this line of reasoning regarding the roles of habituation

6 1220 MCNALLY AND LAM and sensitization in interactions between stressors and drugs of abuse, the present data show that these interactions extend beyond drug reward and highlight the need for models of these interactions that also extend beyond drug reward. Although the present experiments document important roles for NMDA and glucocorticoid receptors in stress-withdrawal interactions, other neurotransmitter and neuropeptide systems are also likely to be important. Of particular interest are the endogenous opioid peptides and their receptors (for review, see McNally & Akil, 2002a). It is well documented that exposure to restraint alters the endogenous opioid system. This stress-induced activation of the endogenous opioid system could have important implications for responsivity to exogenous opiates in the hours to days that follow, which might contribute to altered vulnerability to withdrawal. For example, Adams, Andrews, Hiller, Simon, and Holtzmann (1987) showed that a single 60-min exposure to restraint reduced sensitivity to morphine 24 hr later and that this reduction in sensitivity could be prevented by antagonism of opioid receptors prior to restraint. Consistent with a role for stress-induced activity in the opioid system altering subsequent withdrawal, Williams et al. (1984) showed that facilitation of opiate withdrawal by inescapable stress was prevented if rats were pretreated with naloxone prior to stress. However, it should be noted that dose of naloxone used in this experiment (14 mg/kg) was high and likely to have nonopioid effects. The role of the endogenous opioids in the altered vulnerability to withdrawal observed here warrants further investigation. Finally, it is worth emphasizing that the present experiments studied withdrawal from a single injection of morphine. Antagonist-precipitated withdrawal from a single exposure to opiates has been reported in humans (e.g., Heishman, Stitzer, Bigelow, & Liebson, 1989) and other animals. In rats, this acute withdrawal syndrome has been indexed by somatic signs of withdrawal (e.g., Williams et al., 1984), conditioned place aversions (e.g., Azar, Jones, & Schulteis, 2003), conditioned taste aversion (e.g., McDonald, Parker, & Siegel, 1997), and increases in pain sensitivity (hyperalgesia) (Kim, Fields, & Barbaro, 1990). However, because we studied acute opiate withdrawal in these experiments, it follows that the role of NMDA and glucocorticoid receptors in interactions between stress and withdrawal from chronically administered morphine remains unknown. It was necessary to study withdrawal from acutely administered morphine in these experiments because a history of opiate exposures themselves increases vulnerability to restraint stress (Blatchford et al., in press). It is not possible, therefore, to assess interactions between stress and opiate withdrawal in opiate-experienced animals unconfounded by this vulnerability. However, the present results, taken in combination with previous results (Blatchford et al., in press), document a bidirectional interaction between stress and opiates. A history of opiate exposures increases vulnerability to stress, and a history of stress increases vulnerability to opiate withdrawal. This bidirectional interaction between responding to opiates and stress may offer novel insights into the behavioral and neural adaptations underpinning drug abuse. References Adams, J. U., Andrews, J. S., Hiller, J. M., Simon, E. J., & Holtzmann, S. G. (1987). Effects of stress and beta-funaltrexamine pretreatment on morphine analgesia and opioid binding in rats. Life Sciences, 41, Ahmed, S. H., & Koob, G. F. (1997). Cocaine but not food seeking behavior is reinstated by stress after extinction. Psychopharmacology, 132, Azar, M. R., Jones, B. C., & Schulteis, G. (2003). Conditioned place aversion is a highly sensitive index of acute opioid dependence and withdrawal. Psychopharmacology, 170, Blasig, J., Herz, A., Reinhold, K., & Zieglgansberger, S. (1973). Development of physical dependence on morphine in respect to time and dosage and quantification of the precipitated withdrawal syndrome in rats. Psychopharmacologia, 33, Blatchford, K. E., Diamond, K., Westbrook, R. F., & McNally, G. P. (in press). Increased vulnerability to stress following opiate exposures: Behavioral and autonomic correlates. Behavioral Neuroscience. Covington, H. E., III, & Miczek, K. A. (2001). Repeated social-defeat stress, cocaine or morphine. Effects on behavioral sensitization and intravenous cocaine self-administration binges. Psychopharmacology, 158, Der-Avakian, A., Will, M. J., Bland, S. T., Deak, T., Nguyen, K. T., Schmid, M. J., et al. (2005). Surgical and pharmacological suppression of glucocorticoids prevents the enhancement of morphine conditioned place preference by uncontrollable stress in rats. Psychopharmacology, 179, Deroche, V., Piazza, P. V., Casolini, P., Maccari, S., LeMoal, M., & Simon, H. (1992). Stress-induced sensitization to amphetamine and morphine psychomotor effects depend on stress-induced corticosterone secretion. Brain Research, 598, Deroche, V., Piazza, P. V., Casolini, P., Maccari, S., LeMoal, M., & Simon, H. (1993). Sensitization to the psychomotor effects of amphetamine and morphine induced by food restriction depends on corticosterone secretion. Brain Research, 611, Deroche, V., Piazza, P. V., LeMoal, M., & Simon, H. (1994). Social isolation-induced enhancement of the psychomotor effects of morphine depends on corticosterone secretion. Brain Research, 640, Erb, S., Shaham, Y., & Stewart, J. (1996). Stress reinstates cocaine seeking after prolonged extinction and drug-free periods. Psychopharmacology, 128, Erb, S., & Stewart, J. (1999). A role for the bed nucleus of the stria terminalis, but not the amygdala, in the effects of corticotropin-releasing factor on stress-induced reinstatement of cocaine seeking. Journal of Neuroscience, 19, RC35. Goeders, N. E., & Guerin, G. F. (1994). Non-contingent electric shock facilitates the acquisition of intravenous cocaine self administration in rats. Neuroendocrinology, 64, Haney, M., Maccari, S., LeMoal, M., Simon, H., & Piazza, P. V. (1995). Social stress increases the acquisition of cocaine self-administration behavior in male and female rats. Brain Research, 698, Harris, R. J. (1994). ANOVA: An analysis of variance primer. Itasca, IL: F. E. Peacock. Heishman, S. J., Stitzer, M. L., Bigelow, G. E., & Liebson, I. A. (1989). Acute opioid physical dependence in postaddict humans: Naloxone dose effects after brief morphine exposure. Journal of Pharmacological and Experimental Therapeutics, 248, Hutcheson, D. M., Everitt, B. J., Robbins, T. W., & Dickinson, A. (2001). The role of withdrawal in heroin addiction: Enhances reward or promotes avoidance? Nature Neuroscience, 4, Kim, D. H., Fields, H. L., & Barbaro, N. M. (1990). Morphine analgesia and acute physical dependence: Rapid onset of two opposing doserelated processes. Brain Research, 516, Lu, L., Shepard, J. D., Hall, F. S., & Shaham, Y. (2003). Effect of environmental stressors on opiate and psychostimulant reinforcement, reinstatement, and discrimination in rats: A review. Neuroscience and Biobehavioral Reviews, 27,

7 ACUTE OPIATE WITHDRAWAL FOLLOWING STRESS IN RATS 1221 Manik, C. P., & Katz, R. J. (1984). Stress induced alteration of opiate withdrawal. Drug and Alcohol Dependence, 13, Mantch, J. R., & Goeders, N. E. (1999). Ketoconazole blocks the stressinduced reinstatement of cocaine-seeking behavior in rats: Relationship to the discriminative effects of cocaine. Psychopharmacology, 142, Marinelli, M., & Piazza, P. V. (2002). Interactions between glucocorticoid hormones, stress, and psychostimulant drugs. European Journal of Neuroscience, 16, McDonald, R. V., Parker, L. A., & Siegel, S. (1997). Conditioned sucrose aversions produced by naloxone-precipitated withdrawal from acutely administered morphine. Pharmacology Biochemistry and Behavior, 58, McNally, G. P., & Akil, H. (2002a). Opioid peptides and their receptors: Overview and function in pain modulation. In K. Davis, D. Charney, J. T. Coyle, & C. Nemeroff (Eds.), Neuropsychopharmacology: A fifth generation of progress (pp ). New York: Lippincott Williams & Wilkins. McNally, G. P., & Akil, H. (2002b). Role of corticotropin-releasing hormone in the amygdala and bed nucleus of the stria terminalis in the behavioral, pain modulatory, and endocrine consequences of opiate withdrawal. Neuroscience, 112, Miczek, K. A., Thompson, M. L., & Shuster, L. (1986). Analgesia following defeat in an aggressive encounter: Development of tolerance and changes in opioid receptors. In D. D. Kelly (Ed.), Annals of the New York Academy of Sciences: Vol Stress-induced analgesia (pp ). New York: New York Academy of Sciences. Piazza, P. V., & LeMoal, M. (1997). Glucocorticoids as a biological substrate of reward: Physiological and pathophysiological implications. Brain Research Reviews, 25, Piazza, P. V., & LeMoal, M. (1998). The role of stress in drug selfadministration. Trends in Pharmacological Science, 19, Saal, D., Dong, Y., Bonci, A., & Malenka, R. C. (2003). Drugs of abuse and stress trigger a common synaptic adaptation in dopamine neurons. Neuron, 37, Shaham, Y. (1993). Immobilization stress-induced oral opioid selfadministration and withdrawal in rats: Role of conditioning factors and the effect of stress on relapse to opiod drugs. Psychopharmacology, 111, Shaham, Y., Highfield, D., Delfs, J. M., Leung, S., & Stewart, J. (1997). Clonidine blocks stress-induced reinstatement of heroin seeking in rats: An effect independent of the locus coeruleus noradrenergic neurons. European Journal of Neuroscience, 12, Shaham, Y., & Stewart, J. (1994). Exposure to mild stress enhances the reinforcement efficacy of intravenous heroin self-administration in rats. Psychopharmacology, 114, Shaham, Y., & Stewart, J. (1995). Stress reinstates heroin selfadministration behavior in drug-free animals: An effect mimicking heroin, not withdrawal. Psychopharmacology, 119, Trujillo, K. A., & Akil, H. (1991, January 4). Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801. Science, 51, Will, M. J., Watkins, L. R., & Maier, S. F. (1998). Uncontrollable stress potentiates morphine s rewarding properties. Pharmacology Biochemistry and Behavior, 60, Williams, J. L., Drugan, R. C., & Maier, S. F. (1984). Exposure to uncontrollable stress alters withdrawal from morphine. Behavioral Neuroscience, 98, Zurita, A., Martijena, I., Cuadra, G., Brandao, M. L., & Molina, V. (2000). Early exposure to chronic stress facilitates the occurrence of anhedonia and enhanced emotional reactions to novel stressors: Reversal by naltrexone pretreatment. Behavioural Brain Research, 117, Received May 9, 2005 Revision received June 21, 2005 Accepted June 23, 2005

Effects of contextual or olfactory cues previously paired with morphine withdrawal on behavior and pain sensitivity in the rat

Effects of contextual or olfactory cues previously paired with morphine withdrawal on behavior and pain sensitivity in the rat Psychopharmacology (2001) 156:381 387 DOI 10.1007/s002130100743 ORIGINAL INVESTIGATION Gavan P. McNally Huda Akil Effects of contextual or olfactory cues previously paired with morphine withdrawal on behavior

More information

Conditioned Stimulus Familiarity Determines Effects of MK-801 on Fear Extinction

Conditioned Stimulus Familiarity Determines Effects of MK-801 on Fear Extinction Behavioral Neuroscience 2009 American Psychological Association 2009, Vol. 123, No. 2, 303 314 0735-7044/09/$12.00 DOI: 10.1037/a0014988 Conditioned Stimulus Familiarity Determines Effects of MK-801 on

More information

MOLECULAR BIOLOGY OF DRUG ADDICTION. Sylvane Desrivières, SGDP Centre

MOLECULAR BIOLOGY OF DRUG ADDICTION. Sylvane Desrivières, SGDP Centre 1 MOLECULAR BIOLOGY OF DRUG ADDICTION Sylvane Desrivières, SGDP Centre Reward 2 Humans, as well as other organisms engage in behaviours that are rewarding The pleasurable feelings provide positive reinforcement

More information

Adolescent Prozac Exposure Enhances Sensitivity to Cocaine in Adulthood INTRODUCTION

Adolescent Prozac Exposure Enhances Sensitivity to Cocaine in Adulthood INTRODUCTION INTRODUCTION Epidemiologic reports indicate that mood disorders in children and adolescents are quite common, with up to 70% of depressed children and adolescents experiencing a recurrence within 5 years

More information

Temporal-Difference Prediction Errors and Pavlovian Fear Conditioning: Role of NMDA and Opioid Receptors

Temporal-Difference Prediction Errors and Pavlovian Fear Conditioning: Role of NMDA and Opioid Receptors Behavioral Neuroscience Copyright 2007 by the American Psychological Association 2007, Vol. 121, No. 5, 1043 1052 0735-7044/07/$12.00 DOI: 10.1037/0735-7044.121.5.1043 Temporal-Difference Prediction Errors

More information

Relationship Between Stress and Substance Use Disorders: Neurobiologic Interface

Relationship Between Stress and Substance Use Disorders: Neurobiologic Interface Relationship Between Stress and Substance Use Disorders: Neurobiologic Interface Kathleen Brady, M.D., Ph.D. Professor of Psychiatry Associate Dean of Clinical and Translational Research Medical University

More information

Introduction ORIGINAL INVESTIGATION

Introduction ORIGINAL INVESTIGATION Psychopharmacology (2005) 179: 409 417 DOI 10.1007/s00213-004-2041-1 ORIGINAL INVESTIGATION Andre Der-Avakian. Matthew J. Will. Sondra T. Bland. Terrence Deak. Kien T. Nguyen. Megan J. Schmid. Robert L.

More information

Ghrelin mediates stressinduced. behavior in mice. Chuang et al 2011 L3: Love, Lust, Labor

Ghrelin mediates stressinduced. behavior in mice. Chuang et al 2011 L3: Love, Lust, Labor Ghrelin mediates stressinduced food-reward behavior in mice Chuang et al 2011 L3: Love, Lust, Labor Agenda Introduction What is Ghrelin? Previous Models New model Methods Results Discussion Conclusion

More information

Drug Addiction NROD66H3. (Friday 10:00-12:00 pm; AA206) COURSE DESCRIPTION

Drug Addiction NROD66H3. (Friday 10:00-12:00 pm; AA206) COURSE DESCRIPTION Drug Addiction NROD66H3 (Friday 10:00-12:00 pm; AA206) Instructor: Suzanne Erb Office: SW-627A Office hours: Tues 1-3 pm E-mail: erb@utsc.utoronto.ca COURSE DESCRIPTION This course is designed to provide

More information

Mk-801 Administration in Adolescent Male Rats and Cocaine Conditioned Place

Mk-801 Administration in Adolescent Male Rats and Cocaine Conditioned Place Mk-801 Administration in Adolescent Male Rats and Cocaine Conditioned Place Preference Stephanie Willis, Jonnique Adjmul, Shabaaz Sandhu, Antoniette M. Maldonado-Devincci, Cheryl Kirsten ABSTRACT The present

More information

Recent Advances in Energy, Environment, Biology and Ecology

Recent Advances in Energy, Environment, Biology and Ecology Acute and long-term effects elicited by psychoactive drugs on 50-kHz ultrasonic vocalizations in rats: development of a new experimental tool for the study of drug-mediated reward NICOLA SIMOLA Department

More information

Opioid Receptors Regulate the Extinction of Pavlovian Fear Conditioning

Opioid Receptors Regulate the Extinction of Pavlovian Fear Conditioning Behavioral Neuroscience Copyright 2003 by the American Psychological Association, Inc. 2003, Vol. 117, No. 6, 1292 1301 0735-7044/03/$12.00 DOI: 10.1037/0735-7044.117.6.1292 Opioid Receptors Regulate the

More information

Neurobiology of Addiction JeanAnne Johnson Talbert, DHA, APRN BC, FNP, CARN AP

Neurobiology of Addiction JeanAnne Johnson Talbert, DHA, APRN BC, FNP, CARN AP Neurobiology of Addiction JeanAnne Johnson Talbert, DHA, APRN BC, FNP, CARN AP Disclosures This speaker has no conflicts of interest to disclose Objectives Define drug abuse and addiction Identify the

More information

If you give any person a prescription of something like Valium and have them take it on

If you give any person a prescription of something like Valium and have them take it on As always I am happy to do this presentation, which is my favorite topic in addiction medicine. I am an internist, and I have done healthcare for the homeless in Springfield as well as been the medical

More information

Making a bad thing worse: adverse effects of stress on drug addiction

Making a bad thing worse: adverse effects of stress on drug addiction Review series Making a bad thing worse: adverse effects of stress on drug addiction Jessica N. Cleck and Julie A. Blendy Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia,

More information

BRAIN MECHANISMS OF REWARD AND ADDICTION

BRAIN MECHANISMS OF REWARD AND ADDICTION BRAIN MECHANISMS OF REWARD AND ADDICTION TREVOR.W. ROBBINS Department of Experimental Psychology, University of Cambridge Many drugs of abuse, including stimulants such as amphetamine and cocaine, opiates

More information

Ultra-low-dose naltrexone suppresses rewarding effects of opiates and aversive effects of opiate withdrawal in rats

Ultra-low-dose naltrexone suppresses rewarding effects of opiates and aversive effects of opiate withdrawal in rats Psychopharmacology (2005) 181: 576 581 DOI 10.1007/s00213-005-0022-7 ORIGINAL INVESTIGATION Mary C. Olmstead. Lindsay H. Burns Ultra-low-dose naltrexone suppresses rewarding effects of opiates and aversive

More information

Opioid Receptor Antagonism and Prodynorphin Gene Disruption Block Stress-Induced Behavioral Responses

Opioid Receptor Antagonism and Prodynorphin Gene Disruption Block Stress-Induced Behavioral Responses 5674 The Journal of Neuroscience, July 2, 2003 23(13):5674 5683 Behavioral/Systems/Cognitive Opioid Receptor Antagonism and Prodynorphin Gene Disruption Block Stress-Induced Behavioral Responses Jay P.

More information

Understanding Addiction and Its Impact on the Brain. SDSMA Webinar Matthew Stanley, DO

Understanding Addiction and Its Impact on the Brain. SDSMA Webinar Matthew Stanley, DO Understanding Addiction and Its Impact on the Brain SDSMA Webinar Matthew Stanley, DO Estimated Economic Cost to Society Due to Substance Abuse and Addiction: Illegal drugs: Alcohol: Tobacco: $181 billion/year

More information

Drug Addiction NROD66H3. (Friday 10:00-12:00 pm; AA 204) COURSE DESCRIPTION

Drug Addiction NROD66H3. (Friday 10:00-12:00 pm; AA 204) COURSE DESCRIPTION Drug Addiction NROD66H3 (Friday 10:00-12:00 pm; AA 204) Instructor: Suzanne Erb Office: Temporary location (Sept 2010 until further notice), SW-414B; Permanent location, SW-531 Office hours: Monday 2:30-4:30

More information

Actigraphy-based sleep parameters during the reinstatement of methamphetamine self-administration in rhesus monkeys.

Actigraphy-based sleep parameters during the reinstatement of methamphetamine self-administration in rhesus monkeys. Actigraphy-based sleep parameters during the reinstatement of methamphetamine self-administration in rhesus monkeys. Laís F. Berro, Emory University Monica L. Andersen, Universidade Federal de São Paulo

More information

Understanding Addiction: Why Can t Those Affected Just Say No?

Understanding Addiction: Why Can t Those Affected Just Say No? Understanding Addiction: Why Can t Those Affected Just Say No? 1 The Stigma of Addiction There continues to be a stigma surrounding addiction even among health care workers. Consider the negative opinions

More information

At a Glance. Background Information. Lesson 3 Drugs Change the Way Neurons Communicate

At a Glance. Background Information. Lesson 3 Drugs Change the Way Neurons Communicate Lesson 3 Drugs Change the Way Neurons Communicate Overview Students build upon their understanding of neurotransmission by learning how different drugs of abuse disrupt communication between neurons. Students

More information

by popular demand: Addiction II

by popular demand: Addiction II by popular demand: Addiction II PSY/NEU338: Animal learning and decision making: Psychological, computational and neural perspectives drug addiction huge and diverse field of research (many different drugs)

More information

Basic and Clinical Spring 2012, Volume 3, Number 3

Basic and Clinical Spring 2012, Volume 3, Number 3 The Effect of Nicotine Administration on Physical and Psychological Signs of Withdrawal Syndrome Induced by Single or Frequent Doses of Morphine in Rats Ahmad Taghavi Rafsanjani 1, Abbas Haghparast 2,

More information

Supplementary Online Content

Supplementary Online Content Supplementary Online Content Xue Y-X, Deng J-H, Chen Y-Y, et al. Effect of selective inhibition of reactivated nicotineassociated memories with propranolol on nicotine craving. JAMA Psychiatry. Published

More information

Food restriction: enhancing effects on drug reward and striatal cell signaling

Food restriction: enhancing effects on drug reward and striatal cell signaling Food restriction: enhancing effects on drug reward and striatal cell signaling K.D. Carr Departments of Psychiatry & Pharmacology NYU School of Medicine Common Neural Substrates for Incentive-Motivating

More information

Stress-induced reinstatement of cocaine seeking is mediated by the kappa opioid system

Stress-induced reinstatement of cocaine seeking is mediated by the kappa opioid system DOI 10.1007/s00213-008-1122-y ORIGINAL INVESTIGATION Stress-induced reinstatement of cocaine seeking is mediated by the kappa opioid system Van A. Redila & Charles Chavkin Received: 16 October 2007 / Accepted:

More information

The future of pharmacological treatment.

The future of pharmacological treatment. The future of pharmacological treatment. Anne Lingford-Hughes Professor of Addiction Biology, Imperial College. Hon Consultant CNWL NHS Foundation Trust. What substances and when? What Nicotine Alcohol

More information

NEUROBIOLOGY ALCOHOLISM

NEUROBIOLOGY ALCOHOLISM NEUROBIOLOGY ALCOHOLISM THERE HAS BEEN A MAJOR THEORETICAL SHIFT IN MEDICATION DEVELOPMENT IN ALCOHOLISM Driven by animal models of intermittent ethanol administration followed by termination, then access

More information

NMDA-Receptor Antagonists and Opioid Receptor Interactions as Related to Analgesia and Tolerance

NMDA-Receptor Antagonists and Opioid Receptor Interactions as Related to Analgesia and Tolerance Vol. 19 No. 1(Suppl.) January 2000 Journal of Pain and Symptom Management S7 Proceedings Supplement NDMA-Receptor Antagonists: Evolving Role in Analgesia NMDA-Receptor Antagonists and Opioid Receptor Interactions

More information

Alcohol: Its effects on the brain and implications for future treatment. Dr Mark Daglish

Alcohol: Its effects on the brain and implications for future treatment. Dr Mark Daglish Alcohol: Its effects on the brain and implications for future treatment Senior Lecturer in Addiction Psychiatry University of Queensland Dr Mark Daglish Director of Addiction Psychiatry Royal Brisbane

More information

Council on Chemical Abuse Annual Conference November 2, The Science of Addiction: Rewiring the Brain

Council on Chemical Abuse Annual Conference November 2, The Science of Addiction: Rewiring the Brain Council on Chemical Abuse Annual Conference November 2, 2017 The Science of Addiction: Rewiring the Brain David Reyher, MSW, CAADC Behavioral Health Program Director Alvernia University Defining Addiction

More information

Incubation of sucrose craving: effects of reduced training and sucrose pre-loading

Incubation of sucrose craving: effects of reduced training and sucrose pre-loading Physiology & Behavior 84 (2005) 73 79 Incubation of sucrose craving: effects of reduced training and sucrose pre-loading Jeffrey W. Grimm*, Amber M. Fyall, Dan P. Osincup Department of Psychology, Western

More information

Behavioral and Biological Factors Associated With Individual Vulnerability to Psychostimulant Abuse

Behavioral and Biological Factors Associated With Individual Vulnerability to Psychostimulant Abuse Behavioral and Biological Factors Associated With Individual Vulnerability to Psychostimulant Abuse Pier Vincenzo Piazza, Véronique Deroche, Françoise Rougé- Pont, and Michel Le Moal INDIVIDUAL VULNERABILITY

More information

Notes: Synapse. Overview. PSYC Summer Professor Claffey PDF. Conversion from an signal to a signal - electrical signal is the

Notes: Synapse. Overview. PSYC Summer Professor Claffey PDF. Conversion from an signal to a signal - electrical signal is the PSYC 170 - Summer 2013 - Professor Claffey Notes: Synapse PDF Overview Conversion from an signal to a signal - electrical signal is the - chemical signal is the Presynaptic - refers to that sends/receives

More information

BASIC VOLUME. Elements of Drug Dependence Treatment

BASIC VOLUME. Elements of Drug Dependence Treatment BASIC VOLUME Elements of Drug Dependence Treatment Module 3 Principles of CBT and relapse prevention strategies Introduction to Cognitive Behavioural Therapy Basics of pharmacological treatment Workshop

More information

Repeated Episodes of Heroin Cause Enduring Alterations of Circadian Activity in Protracted Abstinence

Repeated Episodes of Heroin Cause Enduring Alterations of Circadian Activity in Protracted Abstinence Brain Sci. 2012, 2, 421-433; doi:10.3390/brainsci2030421 Article OPEN ACCESS brain sciences ISSN 2076-3425 www.mdpi.com/journal/brainsci/ Repeated Episodes of Heroin Cause Enduring Alterations of Circadian

More information

What are Substance Use Disorders?

What are Substance Use Disorders? What are Substance Use Disorders? Sanchit Maruti, MD Michael Goedde, MD University of Vermont Medical Center 1 Disclosures } Drs. Maruti and Goedde receive compensation as consultants to the American Academy

More information

Subjects. Thirty-five adult male Lister Hooded rats (Charles River, Kent, UK),

Subjects. Thirty-five adult male Lister Hooded rats (Charles River, Kent, UK), Supplemental Material: Subjects. Thirty-five adult male Lister Hooded rats (Charles River, Kent, UK), weighing between 280 300 g at the beginning of the experiment, were housed singly in holding rooms

More information

Does nicotine alter what is learned about non-drug incentives?

Does nicotine alter what is learned about non-drug incentives? East Tennessee State University Digital Commons @ East Tennessee State University Undergraduate Honors Theses 5-2014 Does nicotine alter what is learned about non-drug incentives? Tarra L. Baker Follow

More information

The role of beta-endorphin in the acute motor stimulatory and rewarding actions of cocaine in mice

The role of beta-endorphin in the acute motor stimulatory and rewarding actions of cocaine in mice Psychopharmacology (2008) 197:443 448 DOI 10.1007/s00213-007-1053-z ORIGINAL INVESTIGATION The role of beta-endorphin in the acute motor stimulatory and rewarding actions of cocaine in mice Paul Marquez

More information

NEUROPSYCHOPHARMACOLOGY 2002 VOL. 27, NO American College of Neuropsychopharmacology

NEUROPSYCHOPHARMACOLOGY 2002 VOL. 27, NO American College of Neuropsychopharmacology Conditioned Locomotion Is Not Correlated with Behavioral Sensitization to Cocaine: An Intra-Laboratory Multi-Sample Analysis Gregory Hotsenpiller, Ph.D., and Marina E. Wolf, Ph.D. Pre-clinical and clinical

More information

General introduction. Chapter 1

General introduction. Chapter 1 General introduction Chapter 1 General introduction Historical aspects of drug use Religious, medicinal and recreational use of mind-altering substances by humans has a history of thousands of years 1.

More information

Long term pharmacotherapy for Alcohol Dependence: Anti Craving agents

Long term pharmacotherapy for Alcohol Dependence: Anti Craving agents Long term pharmacotherapy for Alcohol Dependence: Anti Craving agents Myth or Reality? Complete Recovery means a medication-free state True or False? Treatment of Alcoholism Assessment Motivation Alcohol

More information

Effects of lesions of the nucleus accumbens core and shell on response-specific Pavlovian i n s t ru mental transfer

Effects of lesions of the nucleus accumbens core and shell on response-specific Pavlovian i n s t ru mental transfer Effects of lesions of the nucleus accumbens core and shell on response-specific Pavlovian i n s t ru mental transfer RN Cardinal, JA Parkinson *, TW Robbins, A Dickinson, BJ Everitt Departments of Experimental

More information

University of Colorado-Boulder

University of Colorado-Boulder University of Colorado-Boulder Activated Glia as Culprits in Opposing Opioid Analgesia & Driving Tolerance, Dependence & Reward: Role of a Non-classical Non-classical Opioid Receptor Linda R. Watkins Psychology

More information

The Role of Corticotropin-Releasing Factor in the Median Raphe Nucleus in Relapse to Alcohol

The Role of Corticotropin-Releasing Factor in the Median Raphe Nucleus in Relapse to Alcohol The Journal of Neuroscience, September 15, 2002, 22(18):7844 7849 Brief Communication The Role of Corticotropin-Releasing Factor in the Median Raphe Nucleus in Relapse to Alcohol A. D. Lê, 1,2,3 S. Harding,

More information

Pharmacotherapy for Substance Use Disorders

Pharmacotherapy for Substance Use Disorders Pharmacotherapy for Substance Use Disorders Vanessa de la Cruz, MD Chief of Psychiatry Mental Health and Substance Abuse Services Santa Cruz County Health Services Agency 1400 Emeline Avenue Santa Cruz,

More information

Addiction to alcohol and other. The Influence of Stress on the Transition From Drug Use to Addiction. What Is Stress? Gary Wand, M.D.

Addiction to alcohol and other. The Influence of Stress on the Transition From Drug Use to Addiction. What Is Stress? Gary Wand, M.D. The Influence of Stress on the Transition From Drug Use to Addiction Gary Wand, M.D. Stress that is, any type of stimulus that challenges the organism s normal internal balance induces a physiologic response

More information

Addiction in the Brain - Latest Research. Gary M. Henschen, MD, LFAPA Chief Behavioral Health Officer Magellan Healthcare, Inc.

Addiction in the Brain - Latest Research. Gary M. Henschen, MD, LFAPA Chief Behavioral Health Officer Magellan Healthcare, Inc. Addiction in the Brain - Latest Research Gary M. Henschen, MD, LFAPA Chief Behavioral Health Officer Magellan Healthcare, Inc. The Neurobiology of Addiction About the speaker Gary M. Henschen, M.D. is

More information

Substance Abuse and Addictions Substance abuse: a pattern of substance use that produces clinically significant impairment or distress.

Substance Abuse and Addictions Substance abuse: a pattern of substance use that produces clinically significant impairment or distress. Drug Addiction Substance Abuse and Addictions Substance abuse: a pattern of substance use that produces clinically significant impairment or distress. Most recognize it as harmful but continue the addictive

More information

Inhibition of the Acquisition of Conditioned Place Aversion by Dopaminergic Lesions of the Central Nucleus of the Amygdala in Morphine-Treated Rats

Inhibition of the Acquisition of Conditioned Place Aversion by Dopaminergic Lesions of the Central Nucleus of the Amygdala in Morphine-Treated Rats Physiol. Res. 61: 437-442, 2012 RAPID COMMUNICATION Inhibition of the Acquisition of Conditioned Place Aversion by Dopaminergic Lesions of the Central Nucleus of the Amygdala in Morphine-Treated Rats W.

More information

Initial experience of heroin use under a two-chained operant schedule influences drug-seeking behavior after one month of abstinence

Initial experience of heroin use under a two-chained operant schedule influences drug-seeking behavior after one month of abstinence (2010) 31: 387 392 2010 CPS and SIMM All rights reserved 1671-4083/10 $32.00 Original Article Initial experience of heroin use under a two-chained operant schedule influences drug-seeking behavior after

More information

Introduction SHORT COMMUNICATION. Irma V. Belozertseva Anton Yu. Bespalov

Introduction SHORT COMMUNICATION. Irma V. Belozertseva Anton Yu. Bespalov Naunyn-Schmiedeberg s Arch Pharmacol (1998) 358:270 274 Springer-Verlag 1998 SHORT COMMUNICATION Irma V. Belozertseva Anton Yu. Bespalov Effects of NMDA receptor channel blockers, dizocilpine and memantine,

More information

Repeated stress exposure causes strain-dependent shifts in the behavioral economics of cocaine in rats

Repeated stress exposure causes strain-dependent shifts in the behavioral economics of cocaine in rats bs_bs_banneraddiction Biology PRECLINICAL STUDY doi:10.1111/adb.12123 Repeated stress exposure causes strain-dependent shifts in the behavioral economics of cocaine in rats Peter A. Groblewski 1, Chad

More information

Brain Imaging studies in substance abuse. Jody Tanabe, MD University of Colorado Denver

Brain Imaging studies in substance abuse. Jody Tanabe, MD University of Colorado Denver Brain Imaging studies in substance abuse Jody Tanabe, MD University of Colorado Denver NRSC January 28, 2010 Costs: Health, Crime, Productivity Costs in billions of dollars (2002) $400 $350 $400B legal

More information

Kappa Opioid Receptors Regulate Stress-Induced Cocaine Seeking and Synaptic Plasticity

Kappa Opioid Receptors Regulate Stress-Induced Cocaine Seeking and Synaptic Plasticity Article Kappa Opioid Receptors Regulate Stress-Induced Cocaine Seeking and Synaptic Plasticity Nicholas M. Graziane, 1,3 Abigail M. Polter, 1 Lisa A. Briand, 2 R. Christopher Pierce, 2 and Julie A. Kauer

More information

Kathleen T. Brady, M.D., Ph.D., and Susan C. Sonne, Pharm.D.

Kathleen T. Brady, M.D., Ph.D., and Susan C. Sonne, Pharm.D. The Role of Stress in Alcohol Use, Alcoholism Treatment, and Relapse Kathleen T. Brady, M.D., Ph.D., and Susan C. Sonne, Pharm.D. Addiction to alcohol or other drugs (AODs) is a complex problem determined

More information

Using Neuroimaging to Explore Addiction in Animal Models

Using Neuroimaging to Explore Addiction in Animal Models Using Neuroimaging to Explore Addiction in Animal Models NHPs in neuroimaging can be used for Radiotracer development / Occupancy studies Modeling human drug addiction and other brain diseases Radiotracer

More information

The Neurobiology of Drug Addiction

The Neurobiology of Drug Addiction National Institute on Drug Abuse (NIDA) The Neurobiology of Drug Addiction Last Updated January 2007 https://www.drugabuse.gov 1 Table of Contents The Neurobiology of Drug Addiction Section I: Introduction

More information

Effects of Alprazolam and Fluoxetine on Morphine Sensitization in Mice

Effects of Alprazolam and Fluoxetine on Morphine Sensitization in Mice Physiol. Res. 51: 417-423, 2002 Effects of Alprazolam and Fluoxetine on Sensitization in Mice M. VOTAVA, M. KRŠIAK, V. MORAVEC Department of Pharmacology, Third Faculty of Medicine, Charles University,

More information

NIH Public Access Author Manuscript Future Neurol. Author manuscript; available in PMC 2011 March 1.

NIH Public Access Author Manuscript Future Neurol. Author manuscript; available in PMC 2011 March 1. NIH Public Access Author Manuscript Published in final edited form as: Future Neurol. 2010 May 1; 5(3): 393 401. doi:10.2217/fnl.10.14. Neurobiology of dysregulated motivational systems in drug addiction

More information

The Nervous System Mark Stanford, Ph.D.

The Nervous System Mark Stanford, Ph.D. The Nervous System Functional Neuroanatomy and How Neurons Communicate Mark Stanford, Ph.D. Santa Clara Valley Health & Hospital System Addiction Medicine and Therapy Services The Nervous System In response

More information

Negative Reinforcing Properties of Morphine-Antagonists in Naive Rhesus Monkeys* **

Negative Reinforcing Properties of Morphine-Antagonists in Naive Rhesus Monkeys* ** Psychopharmacologia (Berl.) 33, 247--258 (1973) 9 by Springer-Verlag 1973 Negative Reinforcing Properties of Morphine-Antagonists in Naive Rhesus Monkeys* ** F. Hoffmeister and W. Wuttke Institute of Pharmacology,

More information

Supporting Online Material for

Supporting Online Material for www.sciencemag.org/cgi/content/full/328/5983/1288/dc1 Supporting Online Material for Induction of Fear Extinction with Hippocampal-Infralimbic BDNF Jamie Peters, Laura M. Dieppa-Perea, Loyda M. Melendez,

More information

Neurobiological Correlates of Individual Differences in Novelty- Seeking Behavior in the Rat: Differential Expression of Stress- Related Molecules

Neurobiological Correlates of Individual Differences in Novelty- Seeking Behavior in the Rat: Differential Expression of Stress- Related Molecules The Journal of Neuroscience, September 15, 2000, 20(18):6983 6988 Neurobiological Correlates of Individual Differences in Novelty- Seeking Behavior in the Rat: Differential Expression of Stress- Related

More information

The Biology of Addiction

The Biology of Addiction The Biology of Addiction Risk factors for addiction: Biological/Genetic Family history of addiction Being male Having mental illness Exposure to substances in utero * The genes that people are born with

More information

Synapse. Structure & Function. Neurotransmitter Sequence. Integration. History: 10/4/12 original version

Synapse. Structure & Function. Neurotransmitter Sequence. Integration. History: 10/4/12 original version Synapse History: 10/4/12 original version Structure & Function (This content is covered in Sinjin's presentation, see link in calendar) Neurotransmitters Synaptic cleft Post-synaptic potential Excitation

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION SUPPLEMENTARY INFORMATION doi:10.1038/nature12024 entary Figure 1. Distribution of the number of earned cocaine Supplementary Figure 1. Distribution of the number of earned cocaine infusions in Shock-sensitive

More information

C ommon to most descriptions of drug addiction or substance dependence is the idea of a compulsion to

C ommon to most descriptions of drug addiction or substance dependence is the idea of a compulsion to The Neurobiology of Drug Addiction George F. Koob, Ph.D. Eric J. Nestler, M.D., Ph.D. Animal models have begun to provide insights into the neurobiological basis of reinforcement in drug addiction. The

More information

Draft 2 Neuroscience DRAFT work in progress

Draft 2 Neuroscience DRAFT work in progress Foresight: Brain Science, Addiction and Drugs project 19018 Neuroscience of Drugs and Addiction Authors: Trevor W. Robbins 1 Rudolf N. Cardinal 1 Patricia DiCiano 1 Department of Experimental Psychology

More information

Behavioral Neuroscience: Fear thou not. Rony Paz

Behavioral Neuroscience: Fear thou not. Rony Paz Behavioral Neuroscience: Fear thou not Rony Paz Rony.paz@weizmann.ac.il Thoughts What is a reward? Learning is best motivated by threats to survival? Threats are much better reinforcers? Fear is a prime

More information

Brief Communication. Xiu Liu and Friedbert Weiss Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California 92037

Brief Communication. Xiu Liu and Friedbert Weiss Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California 92037 The Journal of Neuroscience, September 15, 2002, 22(18):7856 7861 Brief Communication Additive Effect of Stress and Drug Cues on Reinstatement of Ethanol Seeking: Exacerbation by History of Dependence

More information

A single cocaine exposure enhances both opioid reward and aversion through a ventral tegmental area-dependent mechanism

A single cocaine exposure enhances both opioid reward and aversion through a ventral tegmental area-dependent mechanism A single cocaine exposure enhances both opioid reward and aversion through a ventral tegmental area-dependent mechanism Joseph A. Kim, Kelly A. Pollak, Gregory O. Hjelmstad, and Howard L. Fields* Ernest

More information

Corticosterone-Induced Potentiation of Cocaine Seeking: A Potential Role for Organic Cation Transporter 3

Corticosterone-Induced Potentiation of Cocaine Seeking: A Potential Role for Organic Cation Transporter 3 Marquette University e-publications@marquette Dissertations (2009 -) Dissertations, Theses, and Professional Projects Corticosterone-Induced Potentiation of Cocaine Seeking: A Potential Role for Organic

More information

Behavioral Activation Induced by D 2 -Like Receptor Stimulation during Opiate Withdrawal 1

Behavioral Activation Induced by D 2 -Like Receptor Stimulation during Opiate Withdrawal 1 0022-3565/00/2942-0531$03.00/0 THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS Vol. 294, No. 2 Copyright 2000 by The American Society for Pharmacology and Experimental Therapeutics Printed in

More information

Neurobiology of Addiction

Neurobiology of Addiction Neurobiology of Addiction Domenic A. Ciraulo, MD Director of Alcohol Pharmacotherapy Research Center for Addiction Medicine Department of Psychiatry Massachusetts General Hospital Disclosure Neither I

More information

Sensitization of Salt Appetite Is Associated With Increased Wanting but Not Liking of a Salt Reward in the Sodium-Deplete Rat

Sensitization of Salt Appetite Is Associated With Increased Wanting but Not Liking of a Salt Reward in the Sodium-Deplete Rat Behavioral Neuroscience Copyright 2006 by the American Psychological Association 2006, Vol. 120, No. 1, 206 210 0735-7044/06/$12.00 DOI: 10.1037/0735-7044.120.1.206 Sensitization of Salt Appetite Is Associated

More information

Serotonergic mechanisms of MDMA self-administration. Susan Schenk Victoria University of Wellington School of Psychology

Serotonergic mechanisms of MDMA self-administration. Susan Schenk Victoria University of Wellington School of Psychology Serotonergic mechanisms of MDMA self-administration Susan Schenk Victoria University of Wellington School of Psychology My lab works in the broad area of Behavioural Pharmacology We test the effects of

More information

The Neurobiology of Addiction

The Neurobiology of Addiction The Neurobiology of Addiction Jodi Gilman, Ph.D. Center for Addiction Medicine Massachusetts General Hospital Associate Professor, Department of Psychiatry Harvard Medical School What is Addiction? commonly

More information

The Role of AMPAR Trafficking Mediated by Neuronal Pentraxins in Cocaine-induced Neuroadaptations

The Role of AMPAR Trafficking Mediated by Neuronal Pentraxins in Cocaine-induced Neuroadaptations PharmSight TM DOI: 10.4255/mcpharmacol.09.08 Molecular and Cellular Pharmacology www.mcpharmacol.com The Role of AMPAR Trafficking Mediated by Neuronal Pentraxins in Cocaine-induced Neuroadaptations Alejandra

More information

Environmental regulation of the development of mesolimbic dopamine systems: a neurobiological mechanism for vulnerability to drug abuse?

Environmental regulation of the development of mesolimbic dopamine systems: a neurobiological mechanism for vulnerability to drug abuse? Psychoneuroendocrinology 27 (2002) 127 138 www.elsevier.com/locate/psyneuen Environmental regulation of the development of mesolimbic dopamine systems: a neurobiological mechanism for vulnerability to

More information

Running head: ENVIRONMENTAL ENRICHMENT AND ALCOHOL ADDICTION 1

Running head: ENVIRONMENTAL ENRICHMENT AND ALCOHOL ADDICTION 1 Running head: ENVIRONMENTAL ENRICHMENT AND ALCOHOL ADDICTION 1 Environmental Enrichment and Reinstatement of Alcohol Addiction in Mice Julie Rutter This thesis is submitted in partial fulfillment of the

More information

ASSOCIATED WITH NALORPHINE IN

ASSOCIATED WITH NALORPHINE IN JOURNAL OF THE EXPERIMENTAL ANALYSIS OF BEHAVIOR CONDITIONED SUPPRESSION BY A STIMULUS ASSOCIATED WITH NALORPHINE IN MORPHINE-DEPENDENT MONKEYS1 STEVEN R. GOLDBERG AND CHARLES R. SCHUSTER UNIVERSITY OF

More information

MeCP2 and psychostimulantinduced behavioral adaptations. Anne E. West, M.D., Ph.D. Department of Neurobiology Duke University Medical Center

MeCP2 and psychostimulantinduced behavioral adaptations. Anne E. West, M.D., Ph.D. Department of Neurobiology Duke University Medical Center MeCP2 and psychostimulantinduced behavioral adaptations Anne E. West, M.D., Ph.D. Department of Neurobiology Duke University Medical Center Psychostimulants and antidepressants slowly change behavior Psychostimulants

More information

b 1 Adrenergic Receptors in the Bed Nucleus of Stria Terminalis Mediate Differential Responses to Opiate Withdrawal

b 1 Adrenergic Receptors in the Bed Nucleus of Stria Terminalis Mediate Differential Responses to Opiate Withdrawal (27) 32, 589 599 & 27 Nature Publishing Group All rights reserved 893-133X/7 $3. www.neuropsychopharmacology.org b 1 Adrenergic Receptors in the Bed Nucleus of Stria Terminalis Mediate Differential Responses

More information

Behavioral Neuroscience: Fear thou not. Rony Paz

Behavioral Neuroscience: Fear thou not. Rony Paz Behavioral Neuroscience: Fear thou not Rony Paz Rony.paz@weizmann.ac.il Thoughts What is a reward? Learning is best motivated by threats to survival Threats are much better reinforcers Fear is a prime

More information

Role of the bed nucleus of the stria terminalis versus the amygdala in fear, stress, and anxiety

Role of the bed nucleus of the stria terminalis versus the amygdala in fear, stress, and anxiety European Journal of Pharmacology 463 (2003) 199 216 www.elsevier.com/locate/ejphar Role of the bed nucleus of the stria terminalis versus the amygdala in fear, stress, and anxiety David L. Walker*, Donna

More information

Serotonin System May Have Potential as a Target for Cocaine Medications

Serotonin System May Have Potential as a Target for Cocaine Medications NIDA - Publications - NIDA Notes - Vol. 21, No. 3 - Research Findings of 4 http://www.drugabuse.gov/nida_notes/nnvol21n3/serotonin.html 9/26/2011 3:45 PM NIDA NEWS NIDA Home > Publications > NIDA Notes

More information

Deprivation state determines the motivational effects of neuroleptics in rats

Deprivation state determines the motivational effects of neuroleptics in rats Psychobiology 1992. 20 (4). 294-299 Deprivation state determines the motivational effects of neuroleptics in rats FRANCES HARRINGTON and DEREK VAN DER KOOY University of Toronto, Toronto, Ontario, Canada

More information

11/3/2014. Opiates: methadone, buprenorphine, heroin, prescription drugs: Vicodin, OxyContin, Percocet

11/3/2014. Opiates: methadone, buprenorphine, heroin, prescription drugs: Vicodin, OxyContin, Percocet Estelle B. Gauda, M.D. Professor of Pediatrics Senior Associate Dean of Faculty Development Johns Hopkins Medical Institutions UNDERSTAND HOW COCAI AND METHAMPHETAMIS HAVE ABUSE POTENTIAL UNDERSTAND WHY

More information

DEGREE (if applicable)

DEGREE (if applicable) NAME: Joyce Besheer OMB No. 0925-0001 and 0925-0002 (Rev. 10/15 Approved Through 10/31/2018) BIOGRAPHICAL SKETCH Provide the following information for the Senior/key personnel and other significant contributors.

More information

No! No! No! No! With the possible exception of humans Public Health Question Does the compound have the potential to be abused? Public Health Question Does the compound have the potential to be abused?

More information

Unit II Problem 7 Pharmacology: Substance Abuse, Dependence and Addiction

Unit II Problem 7 Pharmacology: Substance Abuse, Dependence and Addiction Unit II Problem 7 Pharmacology: Substance Abuse, Dependence and Addiction Drugs are classified as being Used (medically approved) abused (medically NOT approved) - What is the difference between dependence

More information

Addiction and the Brain Antireward System

Addiction and the Brain Antireward System Annu. Rev. Psychol. 2008. 59:29 53 The Annual Review of Psychology is online at http://psych.annualreviews.org This article s doi: 10.1146/annurev.psych.59.103006.093548 Copyright c 2008 by Annual Reviews.

More information

The Nervous System. Chapter 4. Neuron 3/9/ Components of the Nervous System

The Nervous System. Chapter 4. Neuron 3/9/ Components of the Nervous System Chapter 4 The Nervous System 1. Components of the Nervous System a. Nerve cells (neurons) Analyze and transmit information Over 100 billion neurons in system Four defined regions Cell body Dendrites Axon

More information

Insights into the Neural Bases of Addiction. Anthony Phillips University of British Columbia Institute of Mental Health

Insights into the Neural Bases of Addiction. Anthony Phillips University of British Columbia Institute of Mental Health Insights into the Neural Bases of Addiction Anthony Phillips University of British Columbia Institute of Mental Health Drug addiction is a brain disease with the following cardinal features: Compulsive

More information

The Role of Smoking in Cocaine. Addiction

The Role of Smoking in Cocaine. Addiction The Role of Smoking in Cocaine Addiction Luca Colnaghi Eric Kandel Laboratory Columbia University in the City of New York Department of Neuroscience Index 1- The Brain, memory, metaplasticity 2- Cocaine

More information

THE OPIUM POPPY OPIOID PHARMACOLOGY 2/18/16. PCTH 300/305 Andrew Horne, PhD MEDC 309. Papaver somniferum. Poppy Seeds Opiates

THE OPIUM POPPY OPIOID PHARMACOLOGY 2/18/16. PCTH 300/305 Andrew Horne, PhD MEDC 309. Papaver somniferum. Poppy Seeds Opiates OPIOID PHARMACOLOGY PCTH 300/305 Andrew Horne, PhD andrew.horne@ubc.ca MEDC 309 THE OPIUM POPPY Papaver somniferum Sleep-bringing poppy Poppy Seeds Opiates Opium Poppy Straw 1 OPIATES VS. OPIOIDS Opiates:

More information