Neuroplastisiteit in die Mesolimbiese stelsel wat deur natuurlike beloning en gevolglike beloning onthouding veroorsaak word. (2010)

COMMENTS: Study shows neuroplastic and behavioral changes in reward circuit can arise from sexual activity. These include the growth of neuron branches and stronger reaction to drugs. As usual, natural reinforcers and drugs have similar effects on the brain.


VOLLEDIGE STUDIE

Pitchers KK, Balfour ME, Lehman MN, Richtand NM, Yu L, Coolen LM.

Biol Psychiatry. 2010 May 1;67(9):872-9. Epub 2009 Dec 16.

Departement Anatomie en Selbiologie, Schulich Skool vir Geneeskunde en Tandheelkunde, Universiteit van Wes-Ontario, Londen, Ontario, Kanada.

OPSOMMING

AGTERGROND: Natural reward and drugs of abuse converge on the mesolimbic system, where drugs of abuse induce neuronal alterations. Here, we tested plasticity in this system after natural reward and the subsequent impact on drug responses.

METHODS: Effects of sexual experience in male rats on behavioral sensitization and conditioned place preference associated with d-amphetamine (AMPH) and Golgi-impregnated dendrites and spines of nucleus accumbens (NAc) cells were determined. Moreover, the impact of abstinence from sexual behavior in experienced males on these parameters was tested.

RESULTATE: First, repeated sexual behavior induced a sensitized locomotor response to AMPH compared with sexually naive control subjects observed 1, 7, and 28 days after last mating session. Second, sexually experienced animals formed a conditioned place preference for lower doses of AMPH than sexually naive males, indicative of enhanced reward value of AMPH. Finally, Golgi-Cox analysis demonstrated increased numbers of dendrites and spines in the NAc core and shell with sexual experience. The latter two alterations were dependent on a period of abstinence of 7-10 days.

CONCLUSIONS: Sexual experience induces functional and morphological alterations in the mesolimbic system similar to repeated exposure to psychostimulants. Moreover, abstinence from sexual behavior after repeated mating was essential for increased reward for drugs and dendritic arbors of NAc neurons, suggesting that the loss of sexual reward might also contribute to neuroplasticity of the mesolimbic system. These results suggest that some alterations in the mesolimbic system are common for natural and drug reward and might play a role in general reinforcement.

Copyright 2010 Society of Biological Psychiatry. Published by Elsevier Inc. All rights reserved.

sleutelwoorde: dopamine, nucleus accumbens, psychostimulant, sexual behavior, substance abuse, dendritic spine

INLEIDING

The mesolimbic dopamine (DA) system, consisting of dopaminergic neurons in the ventral tegmental area (VTA) with projections to the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC), plays a critical role in the motivating and rewarding aspects of behavior including aggression (1), feeding (2-7), drinking (8), mating (9-11) and social bonding (12-13). Drugs of abuse converge upon the mesolimbic DA system (14-15). Moreover, repeated drug administration can induce neuronal alterations in these pathways, that in turn play a putative role in increasing the susceptibility to drug relapse, or in the transition from drug use to drug addiction (16-18). The behavioral effects of repeated drug administration include a sensitized locomotor response to psychostimulants and opiates (19-21), an enhanced conditioned drug reward (22-24), 'Nnd increased operant responses for cues associated with prior drug intake (25). Furthermore, repeated drug administration results in long-lasting changes in dendritic morphology and spine density throughout the mesolimbic circuit (16, 26-31), and induces gene expression changes (32-35). Finally, repeated drug administration alters synaptic strength at excitatory and inhibitory synapses on midbrain dopamine neurons (36-41), and neurons in the NAc (42-44). It is currently unclear if similar alterations in the mesolimbic system occur with repeated exposure to natural rewards. Determining whether such alterations overlap with or are unique to drugs of abuse may lead to a better understanding of the cellular mechanisms underlying the differences between normal reward reinforcement versus compulsive seeking of a particular reward.

Supporting the hypothesis that stimuli other than drugs can cause neuronal alterations in the mesolimbic system are findings that stressful stimuli activate dopamine systems (45-47), and cause psychomotor stimulant sensitization (21, 48-50) and relapse in self-administration models (51-54). However, few studies have investigated whether natural rewarding behaviors can also produce functional changes in the DA system (6, 55-56). Therefore, the hypothesis was tested that male sexual experience causes neuronal alterations within the mesolimbic DA system via analysis of effects of sexual experience on locomotor sensitization, conditioned place preference, and dendrite morphology of NAc neurons. Furthermore, we hypothesized that an abstinence period from sexual behavior (sexual reward) is critical for the onset of these changes, based on recent observations that abstinence from drugs play a key role in the development of neural plasticity associated with repeated drug exposure (40, 57-59).

METODES

diere

Adult male Sprague Dawley rats (210–250 grams) were obtained from Harlan Laboratories (Indianapolis, IN, USA) or Charles River Laboratories (Senneville, QC, Canada) and housed in Plexiglas cages with tunnel tubes. Males were housed in same sex pairs throughout the experiments (experiments 2–5), except for experiment 1 in which males were single housed at the onset of the study. The temperature-regulated colony room was maintained on 12/12 hr light dark cycle with food and water available ad libitum except during behavioral testing. Stimulus females (210–220 grams) for mating behavior sessions were bilaterally ovariectomized and received a subcutaneous implant containing 5% estradiol benzoate and 95% cholesterol. Sexual receptivity was induced by administration of 500μg progesterone in 0.1 ml sesame oil approximately 4 hours before testing. All procedures were approved by the Animal Care and Use Committees of the University of Cincinnati and the University of Western Ontario, and conformed to NIH and CCAC guidelines involving vertebrate animals in research.

Dwelm Behandeling

D-Amphetamine (AMPH) sulfate (Sigma, St. Louis, MO) was dissolved in sterile 0.9% saline (SAL). Animals received AMPH doses ranging 0.5–5.0 mg/kg body weight, calculated based upon the free base, in a volume of 1mL/kg body weight. Control animals received SAL. All injections were given subcutaneously during the first half of the light phase (2–6 hours after lights on), immediately prior to placement into the behavioral apparatus.

Locomotor Activity Testing

Locomotor activity was measured using custom-designed locomotor activity chambers (LACs), modeled on chambers designed by Segal and Kuczenski (60). Locomotor activity was measured using a 16×16 photobeam array (San Diego Instruments, San Diego, CA) and expressed as crossovers per minute(s). A crossover was recorded each time the animal entered any of the “active zones” of the chamber, depicted as shaded areas in Figuur 1A (61).

Figuur 1     

Locomotor response of sexually experienced and naïve animals to saline or amphetamine administration. A is a schematic diagram of the zone map used to measure locomotor activity. A crossover is recorded each time the animal enters one of the black ...

Sexual behavior testing

In all experiments, sexually naïve male rats were randomly divided into groups that either gained sexual experience or remained naïve. For experience, all mating tests were conducted during the first half of the dark phase (3–8 hours after lights off) under dim red light. Animals that remained sexually naïve were handled and housed in the same rooms as sexually experienced males, hence exposed to similar levels of disturbance, environment novelty and distant female odors as experienced animals. For all experiments, groups of sexually experienced males were matched for sexual experience (based on numbers of ejaculations, and latencies to ejaculation and intromission during last mating session).

eksperiment 1

Experiments 1 and 2 utilized different paradigms to test the effects of intermittent mating and environment. In experiment 1, animals in the sexually experienced groups received 5 intermittent mating sessions spaced 3–4 days apart, during which they mated in their home cages with a receptive females for 3 copulatory series (including ejaculation) or 60 minutes, whichever came first. Animals that completed more than five cumulative copulatory series were considered sexually experienced. Sexually naïve animals did not receive female partners. One week following the last mating session, sexually experienced and naïve animals were subdivided into groups receiving AMPH (0.5 mg/kg) or SAL for a total of four groups (Naïve Amphetamine: NA; Experienced Amphetamine: EA; Naïve Saline: NS; and Experienced Saline: ES; n = 6 each).

eksperiment 2

This experiment differed from experiment 1 in three ways: 1. Animals mated to one ejaculation during consecutive days; 2. Animals mated in the same cage as in which they received AMPH (in the LACs); 3. Locomotor activity following AMPH was analyzed at three different times following sexual experience. The sexually experienced animals received 7 consecutive daily mating sessions in the LACs and locomotor activity was recorded during the 15 minutes between placement in LACs and introduction of female. The sexually naïve animals were placed in the LACs for seven consecutive sessions without mating. The day following the final mating session (Day 8 of the experiment), animals were placed in the LACs immediately following injection of AMPH (0.5 mg/kg) or SAL (Naïve Amphetamine: NA; Experienced Amphetamine: EA; Naïve Saline: NS; and Experienced Saline: ES; n = 8–9 each) and locomotor activity was recorded. The animals were tested in the LACs again one week following the final mating session (Day 14). Animals that received AMPH on Day 8 received SAL on Day 14, and animals that received SAL on Day 8 received AMPH on Day 14. Half of the naïve and experienced animals were sacrificed one day later for RNA extraction (data not included in this report). One month following the final mating session (Day 35), the remaining half of the animals (Naïve, n=8; Experienced, n=9) received AMPH and locomotor activity was recorded.

Data-analise

Locomotor Activity

Data were collected in 3-minute bins for 90 minutes following AMPH or SAL injection. Results are shown as the mean ± SEM for each group and analyzed using two-way ANOVA (experiment 1, experiment 2 days 8–14: factors: sexual experience, drug treatment), or one-way ANOVA (experiment 2 day 35 and activity before mating sessions; factor: sexual experience). Post-hoc comparisons were made using Fisher LSD tests with significance set at p-value < 0.05.

Conditioned Place Preference (CPP) Testing

Apparaat

CPP was performed in a three-compartment apparatus (Med Associates Inc., St. Albans, VT, USA) which consisted of two larger, outer chambers (28 × 22 × 21cm) distinguishable by visual and tactile cues, separated by a small central compartment (13 × 12 × 21cm). The apparatus was equipped with photo-beams for automated analysis of tracking and measuring locomotor activity.

Conditioning and Testing

CPP conditioning and testing was conducted during the first half of the light period. A pretest was conducted to determine each animal’s initial preference. No significant differences were detected between the times spent in either chamber. On the following day, the male rats were either confined to the AMPH-paired chamber or to the SAL-paired chamber for 30 minutes. Rats received the opposite treatment the following day in a counterbalanced manner. A posttest that was procedurally identical to the pretest was conducted on the final day.

eksperiment 3

Animals in the sexually experienced groups received 5 consecutive daily mating sessions in test cages. Day 1 was assigned to the first mating day. Control males remained sexually naïve, but were placed into a clean test cage for 1 hour each day for 5 consecutive days. Animals were divided into groups receiving different doses of AMPH (mg/kg; s.c.) (Naïve: N0.5, N1.0, N2.5 or N5.0, n = 7–8 each; Experience: E0, E0.5, E1.0, E2.5 or E5.0, n = 6–9 each). Pretest occurred on day 14, conditioning trials on days 15 and 16, and posttest on day 17. This schedule allowed for 10 days of abstinence from sexual behavior before conditioning.

eksperiment 4

Sexually experienced males gained sexual experience through 5 consecutive days of mating identical to Experiment 3. The key difference with experiment 4 was that CPP testing occurred while animals were gaining sexual experience, thus there was no period of abstinence from sexual behavior. Instead, conditioning trials began following the first 3 mating sessions. Animals were divided into groups receiving different doses of AMPH (mg/kg; s.c.) (Naïve: N0.5, N1.0, N2.5 or N5.0, n = 6–8 each; Experience: E0, E0.5, E1.0, E2.5 or E5.0, n = 7–11 each).

Data-analise

CPP scores were calculated for each an imal as the time spent (sec) in the paired chamber during the posttest minus the pretest. Group means were calculated and compared to the SAL-treated group (E0) using unpaired t-tests. For all experiments significance was set at a p-value < 0.05.

Golgi Experiment

eksperiment 5

Males in the sexually experienced groups were placed in a test cage with a receptive female and allowed to mate until one ejaculation or 60 minutes, whichever occurred first, during 7 consecutive days. Control males remained sexually naïve, but were taken from their home cage and placed into the clean test cage for 30 minutes each day for seven consecutive days. Groups of experienced or naïve animals were sacrificed either one day (N1; n = 5; E1; n = 7) or 7 days (N7, E7; n = 5 each) following the last mating session or exposure to the test cage. Sexually experienced groups did not differ in experience.

Weefselverwerking

One day or one week following the last mating session or exposure to test cage, animals were given an overdose of sodium pentobarbital (i.p.) and were perfused with 500 mL of saline. The brains were processed for Golgi-Cox staining using a method adapted from Pugh and Rossi (62). For further details see Aanvulling 1.

Data-analise

Camera Lucida drawings were made of 5–7 neurons in the caudal NAc core and shell subregions in each animal. Cells were selected for which the entire or the majority of the dendritic branches were visible and easy to distinguish from neighboring cells. Dendritic branches were quantified by centrifugal order (63) and averages per animal were calculated. Dendritic spines were quantified on a 40 μm length of two second order dendrites per cell (4–7 cells per animal). Group means were compared using a two-way ANOVA (factors: sex experience and abstinence period) and Fisher LSD tests for post hoc analise.

RESULTATE

eksperiment 1

The goal of Experiment 1 was to determine if sexual experience affects the locomotor response to AMPH in male rats. Locomotor activity during a 90-minute period was measured in sexually experienced and naïve rats following treatment with 0.5 mg/kg AMPH or SAL. Results from Experiment 1 are illustrated in Figuur 1. Both sex experience (F1,22=15.88; p=0.0006) and drug treatment (F1,22=45.00; p<0.0001) had significant effects on locomotor activity and a two-way interaction between sexual experience and drug treatment was observed (F1,1,22=14.27; p=0.0010). Specifically, both naïve and experienced animals showed a significantly increased locomotor response to AMPH compared to the appropriate SAL controls. Moreover, sexually experienced rats displayed an increased locomotor response to AMPH compared to naïve animals. Sexually experienced and naïve rats did not differ in their responses to SAL.

Analysis of the locomotor responses to AMPH in smaller time intervals of 30 minutes and 3 minutes are illustrated in Figuur 1, panels C-F. Sexually experienced males displayed an increased locomotor response to AMPH compared to naïve rats throughout the 90-minute test period. Moreover, sexually experienced rats showed an increased locomotor response to AMPH compared to their SAL controls throughout the 90-minute test period, while naïve animals only displayed a significantly higher locomotor response during the last 30-minute interval (Figuur 1; p-values are listed in figure legend).

eksperiment 2

The goal of experiment 2 was to test if sexual experience results in locomotor sensitization in animals that mated during consecutive days, and in the same environment as in which they are exposed to AMPH. Exposure to the sex-paired environment caused increased locomotor activity during the 15 minutes prior to each mating session (Figure S1 in Supplement 1), illustrating the learned association between sexual behavior and environment. In addition, experiment 2 investigated the temporal pattern of locomotor sensitization to AMPH in sexually experienced male rats. The locomotor response to AMPH or SAL was measured one day (Day 8), one week (Day 14) and one month (Day 35) following the last mating session. As in experiment 1, sexually experienced rats displayed a greater locomotor response to AMPH compared to naïve animals. Moreover, this effect was evident on all three testing days. Figuur 2 illustrates locomotor activity during the last 60 minutes of the tests during which the most robust differences were observed, and data for first 30 minutes are shown in Figure S2 (Supplement 1). Naïve and experienced animals did not differ in their response to SAL on any of the testing days, and rats that received AMPH displayed increased locomotor activity when compared to their SAL controls (Figuur 2; p-values are listed in figure legend).

Figuur 2     

Locomotor response of sexually experienced and naïve animals to saline or amphetamine administered one day (Day 8; A,B), one week (Day 14; C,D) or one month (Day 35; E,F) following the last mating session. Mean +/− SEM of total number ...

eksperiment 3

Experiment 3 investigated the effect sexual experience on conditioned AMPH reward. AMPH CPP was tested in sexually naïve and experienced males 10 days following the final mating session. Sexually experienced animals show an enhanced conditioned AMPH reward. Specifically, sexually experienced males formed a strong preference for the AMPH-paired chamber with the lower doses of 0.5 and 1.0 mg/kg but not with the higher doses 2.5 or 5.0 mg/kg. In contrast, sexually naïve males only formed a strong preference for the AMPH-paired chamber with the higher doses, 2.5 and 5.0 mg/kg, and not the lower doses (Figuur 3A; p-values are listed in figure legend).

Figuur 3     

Conditioned place preference of sexually experienced and naive animals in response to amphetamine either 10 days following (A) or during (B) mating sessions. Mean +/− SEM of CPP score, defined as the time spent in the AMPH-paired chamber in the ...

eksperiment 4

Experiment 3 demonstrated that sexual experience followed by a period of abstinence resulted in an enhanced conditioned AMPH reward. Experiment 4 investigated whether the effect of sexual experience on conditioned AMPH reward was dependent on this period of abstinence. Results indicated that sexually experienced animals did not show an increased conditioned reward value of AMPH. Sexually experienced and naïve animals showed a strong preference for the AMPH-paired chamber with the higher doses of 2.5 and 5.0 mg/kg. However, neither sexually experienced or naïve males showed an increased CPP score with the lower doses of 0.5 and 1.0 mg/kg dose. The lowest dosage of 0.5 mg/kg even caused an aversion response, but this reached significance only in the sexually experienced animals for the AMPH-paired chamber (Figuur 3B; p-values are listed in figure legend).

eksperiment 5

The purpose of Experiment 5 was to examine morphological alterations in the mesolimbic system, specifically the NAc, following sexual experience. Morphological alterations were evident one week (Figure 4H, J and L; p-values are listed in the figure legend), but not one day (Figure 4G, I and K), following the last mating session. In particular, significant increases in numbers of dendrites (indicative of increased dendritic branching) were detected in NAc core and shell (Figure 4H and J). In addition, numbers of dendritic spines were significantly increased in both the shell and core regions, one week, but not one day, after sex experience (Figure 4L).

Figuur 4     

Dendritic morphology in the NAc of sexually experienced and naïve animals. Sexual experience caused an increase in numbers of dendrites and dendritic spines, illustrated by images (A,B) and camera lucida drawings (C,D) of representative NAc shell ...

BESPREKING

This study demonstrates that sexual experience and the post-experience abstinence from sexual behavior induce functional and morphological alterations in the mesolimbic system of male rats. Functional changes were evident in the form of a sensitized locomotor response and an enhanced conditioned reward with AMPH following sexual experience.

The sensitized locomotor response was observed as early as 1 day and maintained up to 28 days after last mating session. By contrast, the enhanced conditioned AMPH reward was only evident following an abstinence period from sexual behavior. Morphological alterations in both core and shell subregions of NAc were observed 7 days, but not 1 day, following the last mating session in sexually experienced animals. Together these data demonstrate that sexual experience induces plasticity in the mesolimbic system and that an abstinence period from mating is critical for the development of some, but not all mesolimbic system changes.

It is well recognized that natural rewarding behaviors and drugs of abuse act within the same neural pathways (64). Indeed, drugs of abuse have been demonstrated to affect the expression of rewarding behaviors (65-67), including male rat sexual behavior (67-70). The alterations in sexual behavior and motivation caused by repeated drug administration are dependent on a withdrawal or abstinence period from drug, as well as the environment in which the drug was presented. The current study showed that exposure to sexual behavior alters responsiveness to drugs of abuse. It was determined that sexually experienced male rats are sensitized to the locomotor effects of AMPH, and that this phenomenon is long-lasting and independent of an abstinence period from mating. Moreover, the sensitized locomotor response was independent of mating schedule or mating environment and was observed following either consecutive or intermittent mating sessions that occurred in the same or different environment as drug exposure. Studies conducted in female hamsters showed that sexually experienced female hamsters display a more rapid onset of AMPH-induced locomotor response compared to sexually naïve controls (71). However, rodents display sexual dimorphic responses to psychostimulants (72-73). Thus, the current studies expand the findings in female hamsters and demonstrate in male rats, the fast onset and the long duration of the enhanced locomotor responses to psychostimulants following sexual behavior.

It is unclear from the current studies which elements of sexual behavior contribute to the AMPH locomotor sensitization and if social interactions are sufficient. Animals in experiment 2 that failed to reach the criteria for sexual experience (displayed mounts and intromissions, but did not copulate to 5 ejaculations during the mating sessions) did not show a sensitized response (Figure S3 in Supplement 1). Therefore, an additional experiment was performed during which males were exposed to a receptive female without physical interaction, or displayed mounts and intromissions, neither of which resulted in sensitized locomotor responses to AMPH (Figure S4 in Supplement 1). Thus, social interactions do not appear to contribute to the effects of sexual experience on AMPH sensitization, but rather copulation including ejaculation appears essential for this form of plasticity.

In addition to a sensitized behavioral response, sexual experience enhances the conditioned reward value of AMPH, but only following abstinence from sexual reward. Previous work using CPP has shown that repeated exposures to psychostimulants or opiates augment drug-induced rewarding effects in line with the drug-induced locomotor sensitization (22-24). Repeated administration for 5 days of either cocaine (10 mg/kg), d- amphetamine (0.5 mg/kg) or morphine (5 mg/kg) sensitizes the rewarding effects of cocaine when tested 3 days following the cessation of drug pre-treatment. The sensitized effect was displayed by observing a conditioned preference with fewer conditioning trials (from 3 to 2) and with lower drug doses compared to SAL pre-treated control animals. The sensitized conditioned reward caused by repeated cocaine was found 7 days, but not 14 days, after final pretreatment of cocaine (23). A similar study utilizing 5 days of morphine (5.0 mg/kg) shows an augmented conditioned reward response to morphine when conditioning started 3, 10, or 21 days after drug pre-treatment. This augmented response was absent 1 day following morphine pre-treatment (24). Such findings suggest that a period of drug withdrawal of at least 3 days is required for the sensitized or cross-sensitized conditioned reward for both psychostimulants and opiates. Sexual experience, like repeated drug administration, may be instilling similar neuroadaptations in the mesolimbic system responsible for this sensitized drug responsiveness once the reward has been removed. It is currently unclear if reward abstinence is associated with stress and thus acts as a psychological stressor contributing to the observed alterations.

Clearly, there is an interplay between the effects of natural and drug reward. Reward cross-sensitization suggests that the long-lasting effects of both sexual behavior and drugs are mediated by common cellular or molecular mechanisms. Therefore, it is hypothesized that the sex behavior-induced alterations regulate the reinforcing components of sexual behavior and thus may be critical for positive reinforcement of rewarding behaviors in general. However, a subsequent abstinence from sexual reward may induce a state of increased reward seeking, or vulnerability to the effects of addictive substances similar to the effects of abstinence an ‘incubation of drug craving’ (25, 33, 74). In general, sexual behavior in male rodents does not causes compulsive seeking for sex, shown using copulation-malaise associative conditioning experiments (75), although the influence of abstinence has not been tested.

Dendritic morphology has been examined in depth in the fields of learning and memory (76-77) en verslawing (59, 78-79), and is known to be influenced by environmental (80) and hormonal factors (81-82). Since the synaptic inputs are predominantly on dendrites or dendritic spines, they are the most likely target of experience-induced neuroplasticity (26, 83). Natural fluctuations or administration of gonadal hormones have been found to cause dendritic changes within several hours (84-87) 'Nlso, perturbations to the system, such stress (88) or chronic cocaine (79), cause dendritic alterations detectable within 24 hours.

Here, changes to dendritic morphology of medium spiny neurons in both the NAc core and shell were not observed within 24 hours, and instead required a period of abstinence following sexual experience. The structural alterations induced by sexual experience and subsequent abstinence resemble those seen following repeated exposure to psychostimulants (16-17, 26, 30). By contrast, DA depletion in the NAc results in a decreased number of dendrites and complexity in the shell (18, 89). Hence sexual experience-induced changes may be dependent on endogenous DA action in the NAc. However, mating-induced morphological alterations were only evident 7 days following the last mating session and coincide with the enhanced conditioned AMPH reward in sexually experienced animals. These data suggest that these increases in dendritic arborization and spines are not required for the expression of short-term locomotor sensitization to AMPH, yet may play a role in the maintenance and long-term expression of the sensitization. Previous studies of repeated drug administration have also noted a disconnect between long-term sensitization and morphological alterations in the NAc (89-94). It remains unclear what the functional relevance is of the morphological alterations, but it may play a role in the long term changes in function and gene expression.

In summary, the data presented here demonstrate that sexual behavior – a natural rewarding stimulus – can induce long-lasting neuroadaptations in the mesolimbic system. Our findings suggest that behavioral plasticity, particularly a sensitized locomotor response, is an immediate and long-term outcome of sexual experience. Moreover, an abstinence period may allow for neuroadaptations critical for observed morphological changes in the NAc and subsequent enhanced conditioned drug reward. This behavioral and neural plasticity follows a similar, but not identical, profile as seen in drug-sensitized animals. These data are of particular interest since we show that an abstinence from the natural reward induces a vulnerable state to drug administration. Understanding how both natural behaviors and drugs of abuse activate these systems causing neuroadaptations may provide us with a better understanding of reinforcement and reward in general, and provide further insight into the mechanisms of drug addiction.

Aanvullende materiaal

Erkennings

Dr. Richtand has received grant funding from NIH and the Department of Veterans Affairs Medical Research Service. Dr. Richtand reports having served as a consultant to Forest Pharmaceuticals, Bristol-Meyers Squibb, and Gerson Lehrman Group; on the speaker’s bureau of Bristol Meyer’s Squibb and Schering-Plough Corporation; Grand Rounds Presentations to: Sanford School of Medicine of the University of South Dakota and Scius, LLC; and has received grant support from: Janssen Pharmaceutics Research Foundation and Astra Zeneca Pharmaceuticals (study drug only). All other authors report no biomedical financial interests or potential conflicts of interest, except for the following grants to support this research: National Institutes of Health (R01 DA014591), Canadian Institutes of Health Research (RN 014705), and National Sciences and Engineering Research Council of Canada (NSERC) Discovery Grant (341710) to Dr. Lique Coolen, and PGS-M scholarship from NSERC (360696) to Kyle Pitchers. We thank Ms. Maureen 1 Fitzgerald for her assistance with Golgi processing and Dr. Christine Tenk for assistance with supplementary experiment 1.

voetnote

Disclaimer van die uitgewer: Hierdie is 'n PDF-lêer van 'n ongeredigeerde manuskrip wat aanvaar is vir publikasie. As 'n diens aan ons kliënte voorsien ons hierdie vroeë weergawe van die manuskrip. Die manuskrip sal kopieëring, tikwerk en hersiening van die gevolglike bewys ondergaan voordat dit in sy finale citable vorm gepubliseer word. Let asseblief daarop dat tydens die produksieproses foute ontdek kan word wat die inhoud kan beïnvloed, en alle wettige disklaimers wat van toepassing is op die tydskrif betrekking het.

Verwysings

1. Pucilowski O, Kostowski W. Aggressive behaviour and the central serotonergic systems. Gedragsbreinnavorsing. 1983;9: 33-48. [PubMed]
2. Hernandez L, Hoebel BG. Voedings- en hipotalamastimulasie verhoog dopamienomset in die accumbens. Fisiologie en gedrag. 1988;44: 599-606. [PubMed]
3. Noel MB, Wise RA. Ventral tegmental injections of a selective mu or delta opioid enhance feeding in food-deprived rats. Breinnavorsing. 1995;673: 304-312. [PubMed]
4. Martel P, Fantino M. Influence of the amount of food ingested on mesolimbic dopaminergic system activity: a microdialysis study. Farmakologie, biochemie en gedrag. 1996;55: 297-302.
5. Martel P, Fantino M. Mesolimbic dopaminergic system activity as a function of food reward: a microdialysis study. Farmakologie, biochemie en gedrag. 1996;53: 221-226.
6. Avena NM, Rada P, Hoebel BG. Bewyse vir suikerverslawing: gedrags- en neurochemiese effekte van intermitterende, oormatige suiker inname. Neurowetenskap en biobehaviorale resensies. 2008;32: 20-39. [PMC gratis artikel] [PubMed]
7. Avena NM, Rada P, Hoebel BG. Sugar and fat bingeing have notable differences in addictive-like behavior. Die Joernaal van voeding. 2009;139: 623-628. [PMC gratis artikel] [PubMed]
8. Yoshida M, Yokoo H, Mizoguchi K, Kawahara H, Tsuda A, Nishikawa T, et al. Eating and drinking cause increased dopamine release in the nucleus accumbens and ventral tegmental area in the rat: measurement by in vivo microdialysis. Neurowetenschappen briewe. 1992;139: 73-76. [PubMed]
9. Pfaus JG, Damsma G, Nomikos GG, Wenkstern DG, Blaha CD, Phillips AG, et al. Sexual behavior enhances central dopamine transmission in the male rat. Breinnavorsing. 1990;530: 345-348. [PubMed]
10. Balfour ME, Yu L, Coolen LM. Seksuele gedrag en seksverwante omgewingswyses aktiveer die mesolimbiese stelsel in manlike rotte. Neuropsychopharmacology. 2004;29: 718-730. [PubMed]
11. Kohlert JG, Meisel RL. Seksuele ervaring sensitiwiteit paring-verwante kern accumbens dopamien response van vroulike Siriese hamsters. Gedragsbreinnavorsing. 1999;99: 45-52. [PubMed]
12. Young LJ, Lim MM, Gingrich B, Insel TR. Cellular mechanisms of social attachment. Hormone en gedrag. 2001;40: 133-138. [PubMed]
13. Young LJ, Wang Z. The neurobiology of pair bonding. Natuur neurowetenskap. 2004;7: 1048-1054.
14. Wise RA, Bozarth MA. 'N Psigomotoriese stimulerende teorie van verslawing. Psychol Eerw. 1987;94: 469-492. [PubMed]
15. Di Chiara G, Imperato A. Geneesmiddels wat deur mense misbruik word, verhoog die sinaptiese dopamien konsentrasies in die mesolimbiese stelsel van vrybewegende rotte. Verrigtinge van die Nasionale Akademie van Wetenskappe van die Verenigde State van Amerika. 1988;85: 5274-5278. [PMC gratis artikel] [PubMed]
16. Robinson TE, Kolb B. Veranderinge in die morfologie van dendriete en dendritiese stekels in die nukleusakkels en prefrontale korteks na herhaalde behandeling met amfetamien of kokaïen. Die Europese tydskrif van neurowetenskap. 1999;11: 1598-1604. [PubMed]
17. Robinson TE, Gorny G, Mitton E, Kolb B. Kokaïen-selfadministrasie verander die morfologie van dendriete en dendritiese stekels in die nucleus accumbens en neocortex. Synapse (New York, NY. 2001;39: 257-266.
18. Meredith GE, Ypma P, Zahm DS. Effects of dopamine depletion on the morphology of medium spiny neurons in the shell and core of the rat nucleus accumbens. J Neurosci. 1995;15: 3808-3820. [PubMed]
19. Post RM, Rose H. Increasing effects of repetitive cocaine administration in the rat. Die natuur. 1976;260: 731-732. [PubMed]
20. Segal DS, Mandell AJ. Long-term administration of d-amphetamine: progressive augmentation of motor activity and stereotypy. Farmakologie, biochemie en gedrag. 1974;2: 249-255.
21. Kalivas PW, Stewart J. Dopamien-oordrag in die inisiasie en uitdrukking van dwelm- en stres-geïnduseerde sensibilisering van motoriese aktiwiteit. Brain Res Brain Res Ds. 1991;16: 223-244. [PubMed]
22. Lett BT. Repeated exposures intensify rather than diminish the rewarding effects of amphetamine, morphine, and cocaine. Psigofarmakologie. 1989;98: 357-362. [PubMed]
23. Shippenberg TS, Heidbreder C. Sensitization to the conditioned rewarding effects of cocaine: pharmacological and temporal characteristics. Die Tydskrif vir farmakologie en eksperimentele terapeutika. 1995;273: 808-815. [PubMed]
24. Shippenberg TS, Heidbreder C, Lefevour A. Sensitization to the conditioned rewarding effects of morphine: pharmacology and temporal characteristics. Europese joernaal van farmakologie. 1996;299: 33-39. [PubMed]
25. Crombag HS, Bossert JM, Koya E, Shaham Y. Review. Context-induced relapse to drug seeking: a review. Philosophical transactions of the Royal Society of London. 2008;363: 3233-3243. [PMC gratis artikel] [PubMed]
26. Robinson TE, Kolb B. Strukturele plastisiteit wat verband hou met blootstelling aan dwelmmiddels. Neuro Farmacologie. 2004;47(Suppl 1): 33-46. [PubMed]
27. Li Y, Kolb B, Robinson TE. Die ligging van volgehoue ​​amfetamien-geïnduceerde veranderinge in die digtheid van dendritiese stekels op medium stekelneurone in die nukleus accumbens en caudate-putamen. Neuropsychopharmacology. 2003;28: 1082-1085. [PubMed]
28. Robinson TE, Gorny G, Savage VR, Kolb B. Widespread but regionally specific effects of experimenter- versus self-administered morphine on dendritic spines in the nucleus accumbens, hippocampus, and neocortex of adult rats. Synapse (New York, NY. 2002;46: 271-279.
29. Brown RW, Kolb B. Nicotine sensitization increases dendritic length and spine density in the nucleus accumbens and cingulate cortex. Breinnavorsing. 2001;899: 94-100. [PubMed]
30. Robinson TE, Kolb B. Volgehoue ​​strukturele veranderinge in kernklemme en prefrontale korteksneurone wat geproduseer word deur vorige ervaring met amfetamien. J Neurosci. 1997;17: 8491-8497. [PubMed]
31. Sarti F, Borgland SL, Kharazia VN, Bonci A. Acute cocaine exposure alters spine density and long-term potentiation in the ventral tegmental area. Die Europese tydskrif van neurowetenskap. 2007;26: 749-756. [PubMed]
32. Bowers MS, McFarland K, Lake RW, Peterson YK, Lapish CC, Gregory ML, et al. Activator of G protein signaling 3: a gatekeeper of cocaine sensitization and drug seeking. Neuron. 2004;42: 269-281. [PubMed]
33. Lu L, Hope BT, Dempsey J, Liu SY, Bossert JM, Shaham Y. Central amygdala ERK signaling pathway is critical to incubation of cocaine craving. Natuur neurowetenskap. 2005;8: 212-219.
34. McClung CA, Nestler EJ. Regulering van geenuitdrukking en kokaïenbeloning deur CREB en DeltaFosB. Natuur neurowetenskap. 2003;6: 1208-1215.
35. McClung CA, Nestler EJ. Neuroplastisiteit gemedieer deur veranderde geenuitdrukking. Neuropsychopharmacology. 2008;33: 3-17. [PubMed]
36. Saal D, Dong Y, Bonci A, Malenka RC. Dwelms van mishandeling en stres lei tot 'n algemene sinaptiese aanpassing in dopamienneurone. Neuron. 2003;37: 577-582. [PubMed]
37. Nugent FS, Kauer JA. LTP of GABAergic synapses in the ventral tegmental area and beyond. The Journal of physiology. 2008;586: 1487-1493. [PMC gratis artikel] [PubMed]
38. Nugent FS, Penick EC, Kauer JA. Opioids block long-term potentiation of inhibitory synapses. Die natuur. 2007;446: 1086-1090. [PubMed]
39. Kauer JA. Addictive drugs and stress trigger a common change at VTA synapses. Neuron. 2003;37: 549-550. [PubMed]
40. Kauer JA, Malenka RC. Sinaptiese plastisiteit en verslawing. Natuur resensies. 2007;8: 844-858.
41. Liu QS, Pu L, Poo MM. Repeated cocaine exposure in vivo facilitates LTP induction in midbrain dopamine neurons. Die natuur. 2005;437: 1027-1031. [PMC gratis artikel] [PubMed]
42. Thomas MJ, Beurrier C, Bonci A, Malenka RC. Langdurige depressie in die kernklem: 'n neurale korrelaat van gedragsensensitiasie vir kokaïen. Natuur neurowetenskap. 2001;4: 1217-1223.
43. Thomas MJ, Malenka RC. Synaptic plasticity in the mesolimbic dopamine system. Philosophical transactions of the Royal Society of London. 2003;358: 815-819. [PMC gratis artikel] [PubMed]
44. Thomas MJ, Malenka RC, Bonci A. Modulation of long-term depression by dopamine in the mesolimbic system. J Neurosci. 2000;20: 5581-5586. [PubMed]
45. Thierry AM, Tassin JP, Blanc G, Glowinski J. Selective activation of mesocortical DA system by stress. Die natuur. 1976;263: 242-244. [PubMed]
46. de Jong JG, Wasilewski M, van der Vegt BJ, Buwalda B, Koolhaas JM. A single social defeat induces short-lasting behavioral sensitization to amphetamine. Fisiologie en gedrag. 2005;83: 805-811. [PubMed]
47. Tidey JW, Miczek KA. Social defeat stress selectively alters mesocorticolimbic dopamine release: an in vivo microdialysis study. Breinnavorsing. 1996;721: 140-149. [PubMed]
48. Mathews IZ, Mills RG, McCormick CM. Chronic social stress in adolescence influenced both amphetamine conditioned place preference and locomotor sensitization. Dev Psychobiol. 2008;50: 451-459. [PubMed]
49. Yap JJ, Covington HE, 3rd, Gale MC, Datta R, Miczek KA. Behavioral sensitization due to social defeat stress in mice: antagonism at mGluR5 and NMDA receptors. Psigofarmakologie. 2005;179: 230-239. [PubMed]
50. Miczek KA, Covington HE, 3rd, Nikulina EM, Jr, Hammer RP. Aggression and defeat: persistent effects on cocaine self-administration and gene expression in peptidergic and aminergic mesocorticolimbic circuits. Neurowetenskap en biobehaviorale resensies. 2004;27: 787-802. [PubMed]
51. Robinson TE, Berridge KC. Die neurale basis van dwelmverslawing: 'n aansporing-sensibiliseringsteorie van verslawing. Brain Res Brain Res Ds. 1993;18: 247-291. [PubMed]
52. Leri F, Flores J, Rodaros D, Stewart J. Blockade of stress-induced but not cocaine-induced reinstatement by infusion of noradrenergic antagonists into the bed nucleus of the stria terminalis or the central nucleus of the amygdala. J Neurosci. 2002;22: 5713-5718. [PubMed]
53. Marinelli M, Piazza PV. Interaction between glucocorticoid hormones, stress and psychostimulant drugs. Die Europese tydskrif van neurowetenskap. 2002;16: 387-394. [PubMed]
54. Piazza PV, Le Moal M. The role of stress in drug self-administration. Neigings Pharmacol Sci. 1998;19: 67-74. [PubMed]
55. Meisel RL, Mullins AJ. Seksuele ondervinding in vroulike knaagdiere: sellulêre meganismes en funksionele gevolge. Breinnavorsing. 2006;1126: 56-65. [PMC gratis artikel] [PubMed]
56. Wallace DL, Vialou V, Rios L, Carle-Florence TL, Chakravarty S, Kumar A, et al. The influence of DeltaFosB in the nucleus accumbens on natural reward-related behavior. J Neurosci. 2008;28: 10272-10277. [PMC gratis artikel] [PubMed]
57. Wolf ME, Son X, Mangiavacchi S, Chao SZ. Psigomotoriese stimulante en neuronale plastisiteit. Neuro Farmacologie. 2004;47(Suppl 1): 61-79. [PubMed]
58. Thomas MJ, Kalivas PW, Shaham Y. Neuroplasticity in the mesolimbic dopamine system and cocaine addiction. Britse tydskrif van farmakologie. 2008;154: 327-342. [PMC gratis artikel] [PubMed]
59. Nestler EJ. Molekulêre basis van langtermyn-plastisiteit onderliggende verslawing. Natuur resensies. 2001;2: 119-128.
60. Segal DS, Kuczenski R. Individual differences in responsiveness to single and repeated amphetamine administration: behavioral characteristics and neurochemical correlates. Die Tydskrif vir farmakologie en eksperimentele terapeutika. 1987;242: 917-926. [PubMed]
61. Pritchard LM, Logue AD, Hayes S, Welge JA, Xu M, Zhang J, et al. 7-OH-DPAT and PD 128907 selectively activate the D3 dopamine receptor in a novel environment. Neuropsychopharmacology. 2003;28: 100-107. [PubMed]
62. Pugh BC, Rossi ML. A paraffin wax technique of Golgi-Cox impregnated CNS that permits the joint application of other histological and immunocytochemical techniques. J Neurale Transm Suppl. 1993;39: 97-105. [PubMed]
63. Uylings HB, van Pelt J. Measures for quantifying dendritic arborizations. Network (Bristol, England) 2002;13: 397-414.
64. Hyman SE, Malenka RC, Nestler EJ. Neurale meganismes van verslawing: die rol van beloningsverwante leer en geheue. Jaarlikse hersiening van neurowetenskap. 2006;29: 565-598.
65. Della Maggiore V, Ralph MR. The effect of amphetamine on locomotion depends on the motor device utilized. The open field vs. the running wheel. Farmakologie, biochemie en gedrag. 2000;65: 585-590.
66. Aragona BJ, Detwiler JM, Wang Z. Amphetamine reward in the monogamous prairie vole. Neurowetenschappen briewe. 2007;418: 190-194. [PMC gratis artikel] [PubMed]
67. Avena NM, Hoebel BG. Amphetamine-sensitized rats show sugar-induced hyperactivity (cross-sensitization) and sugar hyperphagia. Farmakologie, biochemie en gedrag. 2003;74: 635-639.
68. Fiorino DF, Phillips AG. Facilitation of sexual behavior and enhanced dopamine efflux in the nucleus accumbens of male rats after D-amphetamine-induced behavioral sensitization. J Neurosci. 1999;19: 456-463. [PubMed]
69. Barr AM, Fiorino DF, Phillips AG. Effects of withdrawal from an escalating dose schedule of d-amphetamine on sexual behavior in the male rat. Farmakologie, biochemie en gedrag. 1999;64: 597-604.
70. Mitchell JB, Stewart J. Facilitation of sexual behaviors in the male rat in the presence of stimuli previously paired with systemic injections of morphine. Farmakologie, biochemie en gedrag. 1990;35: 367-372.
71. Bradley KC, Meisel RL. Seksuele gedragsinduksie van c-Fos in die nucleus accumbens en amfetamien gestimuleerde lokomotoriese aktiwiteit word sensitief gemaak deur vorige seksuele ervaring in vroulike Siriese hamsters. J Neurosci. 2001;21: 2123-2130. [PubMed]
72. Castner SA, Xiao L, Becker JB. Seksverskille in striatale dopamien: in vivo mikrodialise en gedragstudies. Breinnavorsing. 1993;610: 127-134. [PubMed]
73. Becker JB, Molenda H, Hummer DL. Gender differences in the behavioral responses to cocaine and amphetamine. Implications for mechanisms mediating gender differences in drug abuse. Annale van die New York Academy of Sciences. 2001;937: 172-187. [PubMed]
74. Grimm JW, Hoop BT, Wise RA, Shaham Y. Neuro adaptation. Inkubasie van kokaïen drang na onttrekking. Die natuur. 2001;412: 141-142. [PMC gratis artikel] [PubMed]
75. Agmo A. Kopulasie-voorwaardelike afwykende kondisionering en seksuele aansporingsmotivering in manlike rotte: bewyse vir 'n twee-stadium proses van seksuele gedrag. Fisiologie en gedrag. 2002;77: 425-435. [PubMed]
76. Chang FL, Greenough WT. Lateralized effects of monocular training on dendritic branching in adult split-brain rats. Breinnavorsing. 1982;232: 283-292. [PubMed]
77. Van Reempts J, Dikova M, Werbrouck L, Clincke G, Borgers M. Synaptic plasticity in rat hippocampus associated with learning. Gedragsbreinnavorsing. 1992;51: 179-183. [PubMed]
78. Nestler EJ, Aghajanian GK. Molecular and cellular basis of addiction. Wetenskap. 1997;278: 58-63. [PubMed]
79. Norrholm SD, Bibb JA, Nestler EJ, Ouimet CC, Taylor JR, Greengard P. Kokaïen-geïnduceerde proliferatie van dendritiese wervelkolven in nucleus accumbens is afhanklik van die aktiwiteit van cyclin-afhanklike kinase-5. Neuroscience. 2003;116: 19-22. [PubMed]
80. Rosenzweig MR, Bennett EL. Psychobiology of plasticity: effects of training and experience on brain and behavior. Gedragsbreinnavorsing. 1996;78: 57-65. [PubMed]
81. Adams VL, Goodman RL, Salm AK, Coolen LM, Karsch FJ, Lehman MN. Morphological plasticity in the neural circuitry responsible for seasonal breeding in the ewe. Endokrinologie. 2006;147: 4843-4851. [PubMed]
82. Jansen HT, Cutter C, Hardy S, Lehman MN, Goodman RL. Seasonal plasticity within the gonadotropin-releasing hormone (GnRH) system of the ewe: changes in identified GnRH inputs and glial association. Endokrinologie. 2003;144: 3663-3676. [PubMed]
83. Lamprecht R, LeDoux J. Structural plasticity and memory. Natuur resensies. 2004;5: 45-54.
84. Gould E, Woolley CS, Frankfurt M, McEwen BS. Gonadal steroids regulate dendritic spine density in hippocampal pyramidal cells in adulthood. J Neurosci. 1990;10: 1286-1291. [PubMed]
85. Woolley CS, Gould E, Frankfurt M, McEwen BS. Naturally occurring fluctuation in dendritic spine density on adult hippocampal pyramidal neurons. J Neurosci. 1990;10: 4035-4039. [PubMed]
86. de Castilhos J, Forti CD, Achaval M, Rasia-Filho AA. Dendritic spine density of posterodorsal medial amygdala neurons can be affected by gonadectomy and sex steroid manipulations in adult rats: a Golgi study. Breinnavorsing. 2008;1240: 73-81. [PubMed]
87. Schwarz JM, Liang SL, Thompson SM, McCarthy MM. Estradiol induces hypothalamic dendritic spines by enhancing glutamate release: a mechanism for organizational sex differences. Neuron. 2008;58: 584-598. [PMC gratis artikel] [PubMed]
88. Dalla C, Whetstone AS, Hodes GE, Shors TJ. Stressful experience has opposite effects on dendritic spines in the hippocampus of cycling versus masculinized females. Neurowetenschappen briewe. 2009;449: 52-56. [PMC gratis artikel] [PubMed]
89. Robinson TE, Kolb B. Morfien verander die struktuur van neurone in die kern accumbens en neocortex van rotte. Synapse (New York, NY. 1999;33: 160-162.
90. Vezina P, Giovino AA, Wise RA, Stewart J. Environment-specific cross-sensitization between the locomotor activating effects of morphine and amphetamine. Farmakologie, biochemie en gedrag. 1989;32: 581-584.
91. Cunningham ST, Finn M, Kelley AE. Sensitization of the locomotor response to psychostimulants after repeated opiate exposure: role of the nucleus accumbens. Neuropsychopharmacology. 1997;16: 147-155. [PubMed]
92. Spanagel R, Shippenberg TS. Modulation of morphine-induced sensitization by endogenous kappa opioid systems in the rat. Neurowetenschappen briewe. 1993;153: 232-236. [PubMed]
93. Singer BF, Tanabe LM, Gorny G, Jake-Matthews C, Li Y, Kolb B, et al. Amphetamine-Induced Changes in Dendritic Morphology in Rat Forebrain Correspond to Associative Drug Conditioning rather than Non-Associative Drug Sensitization. Biol Psyc 2009
94. Pulipparacharuvil S, Renthal W, Hale CF, Taniguchi M, Xiao G, Kumar A, et al. Cocaine regulates MEF2 to control synaptic and behavioral plasticity. Neuron. 2008;59: 621-633. [PMC gratis artikel] [PubMed]