Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Dec 20.
Published in final edited form as: Neurosci Biobehav Rev. 2010 Apr 14;35(2):315–333. doi: 10.1016/j.neubiorev.2010.04.003

The Role of Progestins in the Behavioral Effects of Cocaine and Other Drugs of Abuse: Human and Animal Research

Justin J Anker 1, Marilyn E Carroll 1
PMCID: PMC3243052  NIHMSID: NIHMS205385  PMID: 20398693

Abstract

This review summarizes findings from human and animal research investigating the influence of progesterone and its metabolites allopreganolone and pregnanolone (progestins) on the effects of cocaine and other drugs of abuse. Since a majority of these studies have used cocaine, this will be the primary focus; however, the influence of progestins on other drugs of abuse will also be discussed. Collectively, findings from these studies support a role for progestins in: 1) attenuating the subjective and physiological effects of cocaine in humans, 2) blocking the reinforcing and other behavioral effects of cocaine in animal models of drug abuse, and 3) influencing behavioral responses to other drugs of abuse such as alcohol and nicotine in animals. Administration of several drugs of abuse in both human and nonhuman animals significantly increased progestin levels, and this is explained in terms of progestins acting as homeostatic regulators that decrease and normalize heightened stress and reward responses which lead to increased drug craving and relapse. The findings discussed here highlight the complexity of progestin-drug interactions, and they suggest a possible use for these agents in understanding the etiology and developing treatments for drug abuse.

Keywords: Progesterone, Allopregnanolone, Drug dependence, Cocaine, Animal models, Sex differences

Introduction

Research with humans indicates that females, compared to males, exhibit greater vulnerability to several drugs of abuse at stages of the addiction process that mark transitions in drug use such as initiation, bingeing, withdrawal, and relapse. Women are more likely to initiate cocaine use at an earlier age than men (Chen and Kandel, 2002) and they are more vulnerable to binge-like patterns of alcohol intake (Mann et al., 2005; Randall et al., 1999). Women also report greater difficulty in quitting cigarette smoking (Carpenter et al., 2006) and report greater craving after quitting cocaine (Robbins et al., 1999) than men. They are more sensitive to stress-induced craving of cocaine (Fox and Sinha, 2009), are more likely to relapse (Ignjatova and Raleva, 2009), and also take more cocaine following relapse (Gallop et al., 2007).

Animal models of drug abuse add further support to enhanced female vulnerability across these stages of the addiction process by indicating that female rats acquire drug self-administration at a faster rate (Chaudhri et al., 2005; Fattore et al., 2007; Jackson et al., 2006; Lynch 2008; Lynch and Carroll, 1999), exhibit greater binge-like patterns of drug intake (Roth and Carroll, 2004), and are more vulnerable to relapse of drug-seeking behavior (Anker and Carroll, 2010; Kerstetter et al., 2008; Kippin et al., 2005; Lynch and Carroll, 2000). The animal literature suggests that females are more vulnerable to several aspects of drug abuse than males, possibly due to an underlying biological predisposition. Thus, it is important to not only identify biological vulnerability factors that could contribute to the onset and progression of drug abuse in women but to consider possible sex-specific treatments as well.

An increasing number of findings indicate that these sex differences are influenced by female gonadal hormones. Specifically, increases in drug-related responses in humans are associated with higher levels of endogenous estrogen (EST) (Evans et al., 2002; Sofuoglu et al., 1999). Systemic administration of EST also increases measures of drug-seeking behavior in animal models of drug abuse (Anker et al., 2007, 2009; Becker, 1990b; Jackson et al., 2006; Larson and Carroll, 2007; Larson et al., 2005, 2007). Thus, EST has emerged as a major facilitator of drug effects in females. Progesterone (PROG) has received less attention than EST, but recent findings indicate that PROG, and its metabolites allopregnanolone (ALLO) and pregnanolone, collectively termed progestins (see Frye, 2007), may also be important in altering drug effects during critical stages of the drug abuse process. Of the abused drugs that have been studied, results with cocaine have been the most consistent (Terner and de Wit, 2006); however, other drugs such as alcohol and nicotine have also been examined.

Evidence suggests that hormones may interact with stress and reward systems to affect drug-taking/-seeking behavior. Stress is a major vulnerability factor in drug addiction and it is regulated by several neurobiological systems including the hypothalamic-pituitary-adrenal (HPA) axis (Sinha, 2008). The reward system, which includes dopamine circuitry in the nucleus accumbens (NAc) and ventral tegmental areas of the brain, is also integral to the formation and persistence of addictive behaviors associated with drug abuse (Brieter et al., 1997; Spangel and Wiess, 1999; Volkow et al., 1999).

The purpose of this review is to discuss human and animal research examining the influence of progestins on behavior maintained by cocaine and other drugs of abuse. The paper begins with a small section describing the biosynthesis of progestins and follows with sections devoted to research on the influence of endogenous and exogenously-administered progestins on cocaine-induced subjective and physiological effects in humans. These sections are followed by a discussion of findings from animal research examining cocaine self-administration during critical phases of the animal model of drug abuse under different hormonal conditions, either during the rat or monkey hormone cycle or following systemic progestin administration. Human and animal research examining the interaction between progestin’s and other drugs of abuse such as alcohol and nicotine will then be considered. Finally, mechanisms that may underlie the influence of progestins on behaviors induced by drugs of abuse are described in the final section of the paper. Particular attention is given to the effects of progestins on systems that are thought to underlie drug abuse, such as the stress and reward systems.

Biosynthesis of progestins

In the female mammalian body, the primary source of EST is secretion by the ovaries (Frye, 2007), while the ovaries and the adrenal glands synthesize PROG. ALLO is also secreted by the ovaries as well as the adrenal glands, and it is manufactured in glial cells (Kabbadj et al., 1993). However, the primary source of ALLO and pregnanolone is through metabolism of peripheral sources of PROG (Frye, 2007). The conversion of PROG into its metabolites occurs in a series of metabolic steps. Pregnenolone is converted to PROG which is then reduced to 5-alpha-dihydroprogesterone (DHP) and 5-beta-DHP by 5-alpha- and -beta-reductase, respectively. ALLO and pregnanolone are subsequently synthesized from the 5-alpha and 5-beta forms of DHP by 3-alpha-hydroxysteroid oxidoreductase (Mellon, 2004). For clarification, pregnenolone with an e is the precursor to PROG and pregnanolone with an a is a metabolite of PROG.

Progestin effects on cocaine-induced behavior: human research

Two methods are primarily used in human research to investigate the effects of progestins and other female hormones on drug-related behaviors. The first compares drug-elicited responses across different phases of the female hormone cycle, while the second involves direct manipulation of hormone levels by their systemic administration. Most studies have employed the former method and have focused on menstrual cycle interactions with stimulant-elicited subjective and physiological responses. The 28-day human menstrual cycle is composed of two main phases: follicular and luteal, each characterized by differing levels of the female hormones, EST and PROG (see Figure 1). During the follicular phase (~14 days) PROG levels remain low (10 ng/ml), similar those of men (10 ng/ml), and EST levels increase until they peak just prior to ovulation (210 pg/ml). Ovulation occurs right after the follicular phase and is characterized by declining levels of EST, low levels of PROG, and a surge of luteinizing hormone and follicle stimulating hormone (not shown). After ovulation, during the luteal phase (~ 7 days), EST levels continue to decline (50 pg/ml) and remain relatively low while PROG levels continue to increase (200 ng/ml), surpassing levels in men (Carter, 1994; Genazzani et al., 1998; Pearson Murphy and Allison, 2000; Redei and Freeman, 1995). The luteal phase also marks the period of largest increases of the PROG metabolites, ALLO and pregnanolone (Bixo et al., 1997).

Figure 1.

Figure 1

Plasma concentrations of EST (solid line) and PROG (dashed line) across the human and nonhuman menstrual cycle

Progesterone effects on cocaine across the menstrual cycle

In human research, fluctuations of PROG during the menstrual cycle correspond to differences in the physiological and subjective effects of cocaine. Cardiovascular and/or positive subjective responses to cocaine (Evans and Foltin, 2006; Evans et al., 2002; Sofuoglu et al., 1999) were reduced during the luteal phase of the menstrual cycle (when PROG levels are high) compared to the follicular phase in women.

These results have also been extended to measures of stress reactivity and craving elicited by cocaine-related stimuli in cocaine-dependent women (Sinha et al., 2007). In this study, participants were separated into three groups that had high, medium, or low levels of circulating PROG, corresponding to their midluteal, early luteal, and follicular phases of their menstrual cycles. Groups were then exposed to imagery related to a personally stressful event, or to cocaine-related cues, and compared on measures of cocaine craving, subjective anxiety, and diastolic and systolic blood pressure. Following the drug-cue exposure, women with high levels of PROG showed lower diastolic and systolic blood pressure responses and scored lower on self-reported measures of anxiety and drug craving than women with low levels of PROG. Heightened plasma PROG levels were also associated with less severe stress-induced craving for cocaine (Sinha et al., 2007). This result is particularly important, as stress has long been considered a primary vulnerability factor in addiction: drug abusers often report taking drugs in order to cope with stressful situations and to protect against potentially distressing withdrawal effects (Baker et al., 2004; Khantzian, 1985). Together these results indicate that the phase of the menstrual cycle in which PROG levels are at the highest was associated with low positive affective responses to cocaine and diminished cue- and stress-induced cocaine craving.

In contrast, others have found no differences in the physiological and subjective responses to cocaine during the menstrual cycle (Lukas et al., 1996; Mendelson et al., 1999). However, methodological differences related to the route of cocaine administration may account for these discrepancies. Unlike findings with smoked cocaine (Evans et al., 2002; Sofuoglu et al., 1999), menstrual cycle phase failed to alter subjective or physiological measures following intranasal cocaine administration in women (Collins et al., 2007; Lukas et al., 1996). This lack of an effect may be related to menstrual cycle-dependent changes in nasal secretion. For example, high levels of EST are associated with nasal secretion (Philpott et al., 2004), which may decrease the absorption of cocaine into the nasal mucosa and lower the effects of intranasal cocaine during the EST-dominant follicular phase.

Systemic progesterone administration

The influence of systemic PROG administration on the subjective and physiological effects of cocaine has been examined in several studies using humans. In these experiments, PROG was administered in the micronized (extended release) form at a concentration that produces a physiological level comparable to the midluteal phase of the menstrual cycle. Micronization of PROG results in a decrease in the size of PROG particles and substantially increases the bioavailability of the hormone (Morville et al., 1982). Results from a study conducted by Sofuoglu et al. (2002) indicated that women treated with PROG showed a decrease in the positive subjective effects of smoked cocaine compared with placebo-treated controls (Sofuoglu et al., 2002). This study was subsequently extended to examine PROG’s effects on iv cocaine self-administration in female and male participants (Sofuoglu et al., 2004), and PROG failed to alter iv cocaine self-administration in both groups. This lack of a difference may have been due to a ceiling effect, as both groups self-administered their total allotment of cocaine infusions. However, physiological measures such as cocaine-induced increases in diastolic blood pressure and subjective ratings of “feeling high” and “feeling the effects of the last administered dose” were diminished in PROG-treated vs placebo-treated participants (Sofuouglu et al., 2004).

Similar results were also found in a study conducted by Evans and Foltin (2006). However, the inhibitory effects of PROG were variable and dependent on the cocaine dose as well as the sex of the participant. For example, PROG attenuated cocaine-induced increases in heart rate in women across all of the cocaine doses, but this effect was dose-specific in males. Subjective ratings of “feeling high,” “overall drug quality,” and “willingness to pay for an experimenter-administered dose of cocaine” were also decreased following PROG treatment in females, while PROG had little or no effect on these measures in male patients (Evans and Foltin, 2006). The lack of an effect in males was further supported by research in which PROG failed to decrease outpatient cocaine use in male cocaine users (Sofuoglu et al., 2007). Overall, results from human research indicated that endogenous and exogenous (i.e., systemically administered) PROG decreased certain cocaine-induced subjective and physiological responses, and this effect was generally specific to women.

Few studies using humans as subjects have examined the influence of PROG on motivational aspects of cocaine abuse (cf. Sofuoglu et al., 2004), and none have examined the influence of PROG’s metabolites (ALLO or pregnanolone) on these measures. This would be of interest given that levels of these metabolites show significant increases during the luteal phase of the menstrual cycle when PROG levels are high (Bixo et al., 1997) or following systemic PROG injections (Soderpalm et al., 2004). Table 1 summarizes results across the menstrual cycle and PROG’s effects on amphetamine- and cocaine-induced subjective measures in women.

Table 1.

Summary of the effects of endogenous and exogenous PROG on subjective responses to amphetamine and cocaine in human research

Drug Independent Variable Dependent Measure Findingsa Reference
Cocaine Menstrual cycle state Subjective measures No phase differences Lukas et al., 1996
Mendelson et al., 1999
Feel high, want more drug L < F Sofuoglu et al., 1999
Positive effect, want more drug L < F Evans et al., 2002
Good drug effect L < F Evans and Foltin 2006
Craving following cues L < F Sinha et al., 2007
Subjective measures No phase differences Collins et al., 2007
Systemic PROG administration during the follicular phase Feel the effect of last dose F+P < F Sofuoglu et al., 2002
Feel the effect of last dose, feeling high F+P < F Sofuoglu et al., 2004
Good drug effect, drug quality F+P < F Evans and Foltin 2006
a

L: luteal phase; F: follicular phase; P: PROG

Progestin effects on cocaine-induced behavior during different phases of drug abuse: animal models

There is typically close agreement between results from animal and human research in the screening, prevention, and treatment of drug abuse; thus, animal models of drug self-administration have translational value for the treatment of many aspects of this disorder (Carroll et al., 2009). Studies using animal models of drug self-administration have extended findings from human research and have examined PROG and its metabolite ALLO across critical phases of the drug abuse process that are unable to be studied in humans. As previously mentioned, these stages of drug abuse include initiation, bingeing, withdrawal, and relapse, and they are modeled in animals using acquisition, escalation, withdrawal/extinction, and relapse procedures (Carroll et al., 2009). The following section highlights major findings from animal research examining the effects of PROG and its metabolite ALLO on these important phases of cocaine abuse in addition to progestin effects on other cocaine-induced behavior.

Maintenance of cocaine-seeking behavior across the nonhuman primate menstrual cycle

In animal research, the use of monkeys provides an invaluable model for the human menstrual cycle due the similarity in its duration and hormonal changes. Similar to findings from human research, research with nonhuman primates indicates hormone cycle-dependent alterations of stimulant-related behaviors. For example, during the maintenance of cocaine self-administration under a progressive-ratio (PR) schedule, cynomologus female monkeys reached significantly lower breakpoints (i.e., decreased motivation) for a low dose of cocaine (0.0032 mg/kg/iv injection) during the luteal phase of the menstrual cycle, when PROG levels are high compared with the follicular phase when PROG levels are low (Mello et al., 2007).

Cocaine-seeking behavior across the rodent estrous cycle

While the rodent estrous cycle is much shorter than the primate menstrual cycle (approximately 4–5 days vs 28 days), changes in cocaine self-administration have been noted across estrous phases. The rodent cycle consists of four distinct phases (metestrus, diestrus, proestrus, and estrus), each characterized by varying levels of EST and PROG (Figure 2). During early metestrus, EST (10 pg/ml) and PROG (20 ng/ml) levels remain relatively low until late metestrus when PROG levels rise (28 ng/ml) until they decrease again during early diestrus (8 ng/ml). EST levels slowly increase during both metestrus and diestrus until mid proestrus when they increase dramatically and peak (35 pg/ml). Shortly thereafter, PROG levels reach their highest level in mid proestrus (50 ng/ml). Both hormones reach their lowest levels during the estrus phase of the female rat cycle (EST: 5 pg/ml; PROG: 5 ng/ml) (Brown-Grant et al., 1970; Feltenstein and See, 2006; Naftolin et al., 1972; Roth et al., 2002; Shaikh, 1971). Despite challenges associated with rapid estrous phase transitions, animal research supports human and nonhuman primate work suggesting a role for endogenously circulating PROG in the attenuation of cocaine-related responses during several phases of the drug abuse process.

Figure 2.

Figure 2

Plasma concentrations of EST (solid line) and PROG (dashed line) across the rodent estrous cycle

Maintenance

Similar to findings with nonhuman primates (Mello et al., 2007), research with rodents indicates hormonal cycle effects in the maintenance of cocaine self-administration under a PR schedule. Breakpoints under a PR schedule were higher (increased motivation) for iv cocaine in female (vs male) rats (Hecht et al., 1999; Roberts et al., 1989), and in other studies rats consumed more drug (Feltenstein and See, 200; Lynch, 2008) during the estrus phase, when both EST and PROG levels were at their lowest, than at all other phases of the estrous cycle. These results suggested that phases of the female hormone cycle associated with lower PROG levels corresponded with higher motivation to self-administer cocaine.

Extinction/reinstatement

The extinction and reinstatement (relapse) phases of drug abuse have been modeled in rats, and they are considered to be representative and predictive of abstinence and relapse in humans. Female rats in the estrus phase (low PROG) were more resistant to extinction of lever responding that was previously reinforced with iv cocaine than rats in other estrous phases (Feltenstein and See, 2007; Kerstetter et al., 2008). During reinstatement, females in estrus also exhibited higher levels of cocaine-primed responding on a lever previously associated with cocaine delivery compared to rats in other phases of the estrous cycle (Feltenstein and See, 2007; Kerstetter et al., 2008; Kippin et al., 2005). Moreover, plasma PROG levels in free-cycling female rats were negatively correlated with cocaine seeking during extinction and cocaine-primed reinstatement (Feltenstein and See, 2007). These results did not extend to cue-induced reinstatement, as female rats responded similarly following a cue previously paired with cocaine self-administration during all phases of the estrous cycle (Fuchs et al., 2005).

Systemic progestin administration

Due to the relatively short duration of the rodent estrous cycle and the rapid shifts of EST and PROG across cycle phases, it can be difficult to adequately differentiate the relative contributions of individual female gonadal hormones on cocaine-related effects. An added difficulty is interpreting contributions of hormone metabolites to these effects. As a consequence, direct manipulation of hormone and metabolite levels can be employed to examine dose-dependent effects of individual hormones and/or their metabolites. In animal research, this generally involves depleting hormone levels by ovariectomy (OVX), waiting for the endogenous hormones to deplete (e.g., 5 days), and then administering the hormone or metabolite of interest. The following sections describe recent findings from animal research using surgical methods and/or pharmacological manipulation to examine progestins’ effects on cocaine-maintained behavior during the acquisition, escalation, and reinstatement phases of the drug abuse process.

Acquisition

In a study conducted by Jackson et al. (2006), OVX female rats treated with vehicle (VEH), EST, or EST+PROG were compared during the acquisition of cocaine self-administration. Results indicated that EST treatment facilitated the acquisition of cocaine self-administration in OVX female rats (but not castrated male rats) relative to VEH-treated controls, as previously reported (Lynch et al., 2001), and PROG attenuated this EST-induced facilitation. Cocaine intake was also reduced in female rats treated with EST+PROG relative to OVX rats treated with EST alone (Jackson et al., 2006). These results suggested an important role for PROG in limiting the initiation of cocaine-seeking behavior.

Escalation

In humans, bingeing on drugs is a sign of out-of-control, compulsive, dysregulated behavior that is a hallmark of the progression from drug use to dependence (Koob and Le Moal, 2008). Bingeing on a drug is modeled in animals by increasing access to the self-administered drug (e.g., increasing session length from 2 h to 6 h) for an extended period of time (e.g., 21 days) and then measuring the subsequent escalation of drug intake in the long access vs short access groups (Ahmed and Koob, 1998). Animal research indicates that the development of escalation of cocaine self-administration in rats is influenced by female sex hormones (Larson et al., 2007). In one study rats that were OVX or sham-operated and treated with VEH, EST, PROG, or EST + PROG were compared on the escalation of their cocaine intake over 21 days. Groups did not differ in cocaine self-administration when access was restricted to 2 h (short access); however, when access was increased to 6 h (long access), intact rats, or those treated with EST, progressively increased cocaine intake across the 21-day period, while OVX or PROG-treated sham-operated rats’ intake did not increase (see Figure 3A). Additionally, short-access (2 h) cocaine intake that followed long access (6 h) was higher than the initial short-access intake, except in the PROG-treated rats and VEH-treated sham-operated rats (data not shown).

Figure 3.

Figure 3

Data represent the mean (± SEM) cocaine infusions self-administered each day of the LgA phase. Horizontal lines indicate the 3-day intervals during which there were significant group differences in drug deliveries (p<0.05). Panel A: * = p<0.05 block 1<blocks 3–7; block 2<blocks 5–7 in the OVX-EST group, # = p<0.05 block 1<blocks 4–7 in the SH-VEH group, and @ = p<0.05 block 1<blocks 5–7; block 2<blocks 6 and7; block 3<block 7 in the OVX-VEH group. Panel B:†= p<0.05 block 1< blocks 3–7 in the VEH group.

Recently, these findings were extended to ALLO. In this study, rats were administered ALLO (15 mg/kg) or VEH and allowed to self-administer cocaine under the same experimental conditions described above (6 h/day, 21 days) (Anker and Carroll, unpublished data). Similar to the findings with PROG, ALLO-treated rats did not show escalation of cocaine self-administration during the 21 days, and they earned fewer infusions than VEH-treated controls (see Figure 3B). Additionally, ALLO-treated rats did not show an increase in short-access cocaine self-administration after the long-access period (Anker and Carroll, unpublished data). Relatively few studies have examined pharmacological interventions in the escalation of drugs of abuse using animal models (Hansen and Mark, 2007; Morgan et al., 2002; Specio et al., 2008). However, findings from the studies described above suggest that PROG, and/or its metabolite ALLO, may be important inhibitory agents in the escalation of cocaine use in females.

The rewarding effects of cocaine that lead to excessive or binge-like patterns of cocaine intake during long access (escalation) can result in aversive and potentially lethal consequences. Individuals that consume excessive amounts of cocaine run the risk of overdose, which can take the form seizures, hyperthermia, coma, headache, intracranial hemorrhage and ultimately death (Brody et al., 1990; Lange 2001; Pozzi et al., 2008; Rothman et al., 2001). Kaminski et al. (2003) examined the protective effect of PROG’s metabolites ALLO and pregnanolone against cocaine-kindled seizures in mice. Both treatments attenuated the occurrence of cocaine-precipitated seizures, while ALLO prevented the development of kindling, as reflected by the number of mice exhibiting seizures following repeated cocaine administration (Kaminski et al., 2003; also see Leskiewicz et al., 2003). These results further emphasize the potential value of progestins not only for preventing the escalation of cocaine use but for protection from the negative consequences of cocaine administration.

Reinstatement

Systemic administration of PROG or ALLO has also been implicated in the attenuation of cocaine- and stress-induced reinstatement of drug seeking in female rats. In one study, PROG treatment in sham-operated (i.e., gonadally intact) female rats decreased reinstatement responding following an injection of cocaine relative to sham-operated rats treated with VEH (Figure 4A). In contrast, EST treatment in OVX female rats potentiated cocaine-primed reinstatement responding relative to OVX rats treated with VEH (Figure 4B), and PROG counteracted the potentiating effects of EST in a separate group (Figure 4B). In a similar study, PROG administration during the estrus phase of freely cycling female rats also decreased reinstatement responding following a cocaine priming injection relative to VEH-treated rats (Feltenstein et al., 2009).

Figure 4.

Figure 4

Black bars represent the mean (± SEM) responses on the cocaine-paired lever following a 10 mg/kg cocaine priming injection and white bars refer to responses on the same lever after a saline priming injection. All groups responded more on the cocaine-paired lever after a 10 mg/kg cocaine i.p. than after a saline i.p. (* = p<0.05). Significant group differences in cocaine-primed responding are represented by # (p<0.05). Panel C: †= p<0.05 VEH>ALLO in the female group.

In a separate study, the effects of ALLO were examined in gonadally intact female and male rats. Finasterdie (FIN), a compound that blocks the conversion of PROG into ALLO, was administered with PROG in female rats to determine if PROG’s attenuating effects on cocaine-primed reinstatement were due to its metabolism into ALLO. Hormone treatments were given to intact rats; 2 male groups: 1) VEH and 2) ALLO (30 mg/kg) and 5 groups of females: 1) VEH, 2) ALLO (15 mg/kg), 3) ALLO (30 mg/kg), 4) PROG, and 5) PROG + FIN. Compared to VEH, ALLO (30 mg/kg) decreased responding on a previously active lever in gonadally intact female rats (Figure 4C) at doses that do not interfere with locomotor activity or food maintained responding (Anker et al., 2009). However, ALLO (30 mg/kg) had no effect on cocaine-primed reinstatement in gonadally intact male rats (Figure 4D) in the same study (Anker et al., 2009, Figure 4D). As previously shown (Anker et al., 2007, Figure 4A), PROG also reduced reinstatement in female rats, and this effect was reversed by FIN, suggesting that PROG may act through ALLO to attenuate cocaine-primed reinstatement (Anker et al., 2009, see Figure 4C). These data suggest that the suppressant effects of PROG on cocaine-seeking behavior are specific to females and could be attributed in part to the PROG metabolite ALLO (Anker et al., 2009).

While many of the reinstatement studies have been conducted with ip priming injections of cocaine, other events such as drug-associated exteroceptive cues or stress-evoking stimuli have been used in animal models of relapse. Stress has emerged as a major factor that predicts cocaine craving (Sinha et al., 2003) and relapse to cocaine abuse (Sinha, 2008; Stewart, 2000) in humans as well as the reinstatement of cocaine seeking in rats (Brown and Erb, 2007; Boutrel et al., 2005; Feltenstein and See, 2006; Shalev et al., 2003).

Since previous findings with humans showed attenuated stress-induced craving during the progestin-dominant luteal phase (Sinha et al., 2007), ALLO was tested in gonadally intact female and male rats for its ability to suppress the reinstatement of cocaine seeking precipitated by the anxiogenic drug yohimbine. In this study yohimbine reliably reinstated cocaine-seeking behavior as previously reported (Feltenstein and See, 2006); however, ALLO decreased the yohimbine-induced reinstatement in female rats (Figure 5A). Similar to findings with cocaine-primed reinstatement (Anker et al., 2009), ALLO did not attenuate yohimbine-induced reinstatement in male rats (Anker and Carroll, 2010, see Figure 5B). That ALLO mediates stress-induced reinstatement of cocaine seeking suggests that PROG’s inhibitory effects on stress-elicited craving in female cocaine addicts may be related to the PROG metabolite ALLO (Sinha et al., 2007). Table 2 summarizes findings from animal research examining the effects of endogenous and exogenous progestins on cocaine-seeking behavior across several phases of the drug abuse process.

Figure 5.

Figure 5

Mean (± SEM) responses on the previously drug-paired lever following saline (S) or YOH (Y) priming injections during the reinstatement procedure. Asterisks indicate significantly greater responding following YOH compared to S priming injections (p<0.05). # = a significant difference in YOH compared to responding following A+Y in the female group (p<0.05), and †= a significant sex difference (females>males) in responding following the first YOH injection (p<0.05).

Table 2.

Summary of the effects of endogenous and exogenous progestins on behavioral responses to cocaine across phases of the drug abuse process

Independent Variable Dependent Measure Findinga Reference
Menstrual cycle state (Monkey) Maintenance (PR) BPs: L < F Mello et al., 2007
Estrous cycle state Maintenance (PR) BPs: E < all other phases Roberts et al., 1989
Maintenance (FR) Intake: E < all other phases Feltenstein and See 2007
Lynch 2008
Extinction Resistance to extinction: E < all other phases Feltenstein and See 2007
Kerstetter et al., 2008
Cue-induced Reinstatement no phase differences Fuchs et al., 2005
Cocaine-primed reinstatement E < all other phases Kippin et al., 2005
Feltenstein and See 2007
Kerstetter et al., 2008
Systemic PROG administration Acquisition Days to criteria: OVX+EB+P < OVX+EB Jackson et al., 2006
Maintenance (PR) Intake: P < V Anker and Carroll, unpublished data
Intake: No effect Larson et al., 2005
Escalation Intake: OVX+EB+P < OVX+EB; sham+P < sham+V Larson et al., 2007
Cocaine-primed reinstatement OVX+EB+P < OVX+EB; sham+P < sham+V Anker et al., 2007
P < V Feltenstein et al., 2009
P < V; FIN+P = V Anker et al., 2009
Systemic ALLO administration Escalation Intake: A < V Anker and Carroll, unpublished data
Overdose Seizures and death: A & Preg < V Kaminski et al., 2003
Leskiewicz et al., 2003
Cocaine-primed reinstatement A < V & P Anker et al., 2009
Stress-induced reinstatement A < V Anker and Carroll, 2010
a

L: luteal phase; F: follicular phase; E: estrus phase; EB: EST; P: PROG; V: vehicle; FIN: finasteride; A: ALLO; Preg: pregnanolone

Effects of progestins on other cocaine-induced behaviors related to drug seeking

PROG and ALLO have been implicated in other behavioral responses to cocaine, such as cocaine-induced conditioned place preference (CPP), locomotor activity, and cocaine discrimination (see Table 3). These behaviors are thought to measure different effects of drugs of abuse associated with their conditioned rewarding effects (CPP), behavioral-activating effects (locomotor activity), and their discriminative-stimulus effects (drug discrimination).

Table 3.

Summary of the effects of progestins on other behaviors elicited by cocaine

Independent Variable Dependent Measure Decreased Response a Reference
Systemic PROG administration Cocaine-induced CPP P < V Russo et al., 2003
Romieu et al., 2003
Russo et al., 2008
Cocaine-induced locomotor activity No effect Sircar and Kim 1999
Quinones-Jenab et al., 2000b
Perrotti et al., 2001
Yang et al., 2007
P (24 hr after EB)+EB > V; P (<24 hr after EB)+EB < V Perrotti et al., 2003
P+EB > V; P+EB < V Niyomachi et al., 2008
EB+P > V Yang et al., 2007
P < V Sell et al., 2000
Systemic pregnanolone Cocaine discrimination Blocked by Preg Quinton et al., 2006
a

P: PROG; V: vehicle; EB: EST; Preg: pregnanolone

Similar to its effects on cocaine self-administration (Jackson et al., 2006), PROG attenuated cocaine-induced CPP in female rats (Russo et al., 2003, 2008) and in male mice (Romieu et al., 2003) at doses that did not affect ambulatory or rearing behaviors. However, despite the relatively consistent findings concerning PROG’s influence on other cocaine-related behaviors, the effects of PROG on cocaine-induced locomotor activity have been mixed. PROG increased (Niyomchai et al., 2008; Perrotti et al., 2001; Quinones-Jenab et al., 2000b; Sircar and Kim, 1999), decreased (Niyomchai et al., 2008; Sell et al., 2000; Yang et al., 2007), or had no effect (Perrotti et al., 2001; Quinones-Jenab et al., 2000b; Sircar and Kim, 1999; Yang et al., 2007) on locomotor activity in OVX female rats following cocaine administration. Discrepancies in these results may be explained by the timing, dose, and/or route of administration of PROG and the presence or absence of EST. Thus, results from locomotor activity studies failed to support previous work showing an attenuating effect of progestins on the conditioned rewarding effects of cocaine, suggesting that progestins may specifically inhibit the direct rewarding effects of cocaine.

Drugs of abuse serve as powerful discriminative stimuli, and the translational value of the drug discrimination paradigm is high, as drug discrimination in rodents and nonhuman primates has closely agreed with self-reported subjective effects in humans (Dykstra et al., 1997; Kamien et al., 1993; Schuster and Johanson, 1988). Pharmacological agents that block drug discrimination may also decrease the drug’s reinforcing effects; thus, drug discrimination has been useful for screening possible treatments of drug abuse (Solinas et al., 2006). There has been only one study examining the effects of a progestin on the discrimination of cocaine. Quinton et al. (2006) trained rats to discriminate between saline or two doses of cocaine (5.6 or 10 mg/kg) for food presentation under an operant schedule. Pregnanolone failed to substitute for cocaine in the task, but when it was administered prior to cocaine, the discriminative stimulus effects of cocaine were attenuated. One explanation of this finding may be related to state-dependent learning and a stimulus-generalization decrement. For example, stimulant discrimination is dependent on behavioral and neurochemical responses to stress (Kamien and Woolverton, 1989; Johanson and Barrett, 1993; Spealman, 1995; Mantsch and Goeders, 1998), while the anxiolytic pregnanolone exerts a mitigating effect on stress-related responses (Bitran et al., 1991). Thus, discrimination may have been attenuated when tested in a “nonstressed” state because it was learned in a cocaine-induced “stressed” state.

Effects of progestins on behavior maintained by other drugs of abuse

An additional question to be considered is whether PROG’s effects on cocaine-reinforced behaviors are specific to cocaine or if they occur with other drugs of abuse. Research indicates that progestins mediate responses to other abused drugs; however, the available literature is not as extensive as for cocaine, and there is relatively little data on progestins’s effects during different phases of the drug abuse process. The reported effects also vary depending on the subjects (human or animal), drug, and/or methodology (hormone cycle monitoring or systemic administration) used in the studies. The following section discusses findings from human and animal research on the effects of progestins on behavioral responses to other drugs of abuse. Particular attention is given to alcohol, as it has been one of the most widely studied drugs.

Progestins, other drugs, and the human menstrual cycle

Compared to results with stimulants, behavioral/subjective responses to other drugs of abuse do not consistently vary with phase of the menstrual cycle in humans (see Table 4). During the luteal phase in women, it was reported that alcohol (Garcia-Closas et al., 2002; Harvey and Beckman, 1985) and nicotine (DeBon et al., 1995; Mello et al., 1987; Snively et al., 2000) intake increased (compared to the follicular phase), while in other studies there was either a decrease or no menstrual cycle effect on alcohol (Holdstock and de Wit, 2000; Pomerleau et al., 1994), nicotine (Allen et al., 1996, 1999,Allen et al., 2009a, b; Marks et al., 1999; Pomerleau et al., 1994) marijuana (Griffin et al., 1986), and opioid (Sener et al., 2005) consumption.

Table 4.

The effects of endogenous and exogenous progestins on the intake of and subjective responses to other drugs of abuse

Drug Independent Variable Dependent Measure Finding a Reference
Amphetamine Menstrual cycle phase Feeling high L < F Justice and de Wit 1999
Want more drug L < F Justice and de Wit 2000
White et al., 2002
Nicotine Menstrual cycle phase Intake and positive subjective measures L > F Mello et al., 1987
Intake and/or withdrawal L > F DeBon et al., 1995
Allen et al., 2009b
Intake L > F Snively et al., 2000
Withdrawal L > F Allen et al., 1999
Withdrawal L > F Perkins et al., 2000
Craving L > F Franklin et al., 2004
F >L Carpenter et al., 2008
Intake and subjective measures No phase differences Pomerleau et al., 1994
Allen et al., 1996, 1999, 2009a
Marks et al., 1999
Pomerleau et al., 2000
Allen et al., 2004
Franklin et al., 2008
Abstinence F > L Carpenter et al., 2008
Franklin et al., 2008
Systemic PROG administration during the follicular phase Positive subjective measures P < placebo Sofuoglu et al., 2001, 2009
Alcohol Menstrual cycle phase Intake L > F Garcia-Closes et al., 2002
Harvey and Beckman 1985
Intake and positive subjective measures No phase differences Hay et al., 1984
Sutker et al., 1987
Freitag and Adesso 1993
Pomerleau et al., 1994
Holdstock and de Wit 2000
Nyberg et al., 2004
Marijuana Menstrual cycle phase Intake and subjective measures of positive effect, intoxication, and craving No phase differences Lex et al., 1984
Griffin et al., 1986
Opiods Menstrual cycle phase Intake and subjective measures of positive effect, intoxication, and craving No phase differences Gear et al., 1996
Sener et al., 2005
a

L: luteal phase; F: follicular phase; P: PROG

These findings also extend to other drug-related measures. For example, similar to drug intake, research has been unable to establish a connection between menstrual cycle and self-reported measures of positive affect, intoxication, and craving. Subjective measures were insensitive to menstrual cycle effects following alcohol (Freitag and Adesso, 1993; Hay et al., 1984; Holdstock and de Wit, 2000; Nyberg et al., 2004; Sutker et al., 1987), nicotine (Allen et al., 1999, 2004; Pomerleau et al., 1992, 2000; Snively et al., 2000), marijuana (Lex et al., 1984), and opioid (Gear et al., 1996) administration in women; however, similar to results with cocaine (Evans and Foltin, 2006; Evans et al., 2002; Sofuoglu et al., 1999), women reported decreased positive subjective effects following amphetamine administration during the luteal phase compared to follicular phase of the menstrual cycle (Justice and de Wit, 1999, 2000; White et al., 2002). Overall, these findings indicate a lack of an effect of menstrual cycle phase on behavioral/physiological and subjective measures of alcohol, nicotine, marihuana, and opioid use in humans.

Despite the lack of a consistent effect on these measures, recent work with nicotine indicates that menstrual cycle phase plays an important role in the likelihood of abstaining from smoking in women. In a study by Carpenter et al. (2008), women reported their menstrual cycle phase and were randomly placed into groups that had quit dates assigned to either the luteal or follicular phase. This study found that abstinence was higher in the group that quit during the follicular phase; however, this group also experienced more withdrawal symptoms and cravings suggesting multiple determinants of drug use cessation. In a similar study in which menstrual cycle phase was self-reported, Franklin et al. (2008) found that the success rates of females in the follicular phase of their menstrual cycle was significantly higher than that of females in the luteal group, and these differences lasted for up to nine weeks following the study even as women progressed through other phases of their menstrual cycle (Franklin et al., 2008). Opposite results were reported by Allen and colleagues (2009a). In this study, menstrual cycle phase was biochemically verified, and the results indicated that women were more likely to successfully quit smoking during the luteal phase of the menstrual cycle. Regardless of the inconsistent results, these studies demonstrated that timing quit attempts around periods of the menstrual cycle might improve abstinence from drug abuse.

Systemic progesterone administration: human research

In contrast to research with cocaine, relatively few studies using humans have assessed the effects of PROG administration on responses to other drugs of abuse. In one study, Sofuoglu and colleagues (2001) administered PROG or a placebo to women during the early follicular phase and measured behavioral and subjective responses to cigarettes. Women treated with PROG (vs placebo-treated controls) showed a marked decrease in craving before smoking, and after having two puffs they reported an additional decrease in cigarette-induced positive affect. However, there were no reported effects of PROG on the number of cigarettes smoked. In a follow-up study, Sofuoglu et al. (2009) reported that PROG treatment decreased self-reported measures of “drug liking” following iv nicotine administration in female smokers. Results from these studies indicated differential effects of PROG on measures of subjective responses vs reinforcing effects of smoking, and they support previously mentioned findings showing a similar dissociation of subjective effects with iv cocaine self-administration (Sofuoglu et al., 2004). Table 4 summarizes findings from human research examining menstrual cycle effects and the effects of systemically-administered PROG on drug intake and subjective responses following amphetamine, nicotine, alcohol, marijuana, and opioid administration in human studies.

Effects of estrous cycle on behavior maintained by other drugs of abuse

Similar to findings with humans, in rodents there does not appear to be a consistent effect of circulating PROG on the behavioral effects of drugs of abuse other than cocaine (see Table 5). However, when noted, these effects are specific to the drug, behavioral measures, and phases of the hormone cycle. For example, during the diestrus phase of the estrous cycle (associated with low progestin levels), female rats were more sensitive to the reinforcing effects of ethanol under an operant self-administration procedure than during the estrus and proestrus phases (higher levels of progestins) (Roberts et al., 1998), a finding similar to that reported by Forger and Morin (1982). However, in another study ethanol intake by rats was unaffected by estrous cycle, but the duration of self-administration bouts was longer during proestrus compared to all other phases (Ford et al., 2002).

Table 5.

Summary of hormone cycle effects on behaviors elicited by other drugs of abuse in animals.

Drug Independent Variable Dependent Measure Finding a Reference
Alcohol Menstrual cycle phase (monkey) Maintenance (FR) Intake lower during menstruation Mello et al., 1986
Estrous cycle phase Maintenance (FR) Intake: D > all other phases Forger and Morin 1982
Roberts et al., 1998
Total intake: no phase differences; Duration of intake bout: Pro > all other phases Ford et al., 2002
Nicotine Estrous cycle phase Maintenance (FR and PR) No phase differences (FR) Donny et al., 2000
Intake: E > all other phases (PR) Lynch 2009
Withdrawal No phase differences Hamilton et al., 2009
PCP Menstrual cycle phase (monkey) Maintenance (FR) L < F Newman et al., 2006
a

D: diestrus phase; E: estrus phase; Pro: proestrus phase; L: luteal phase; F: follicular phase

In research with female rhesus monkeys, menstrual cycle phase was not related to ethanol intake under a second order schedule of reinforcement except during menstruation (low PROG and EST) when alcohol intake was significantly lower than all other phases of the cycle (Mello et al., 1986). These results are opposite to those found with rhesus monkeys when orally self-administered PCP was the reinforcer. In this study PCP intake increased during the luteal vs the follicular phase (Newman et al., 2006), a result that agrees with human studies showing increased alcohol (Garcia-Closas et al., 2002; Harvey and Beckman, 1985) and nicotine (DeBon et al., 1995; Mello et al., 1987; Snively et al., 2000) intake during the luteal phase.

Results from animal studies using nicotine as a reinforcer are inconclusive. For example, Donny and colleagues (2000) reported that nicotine self-administration was insensitive to estrous cycle changes at various response requirements under a FR and PR schedule for a range of nicotine doses, and others found no effect of the rodent estrous cycle on behaviors related to nicotine withdrawal (e.g., whole-body shakes, diarrhea, and empty-mouth chewing) (Hamilton et al., 2009). In contrast, Lynch (2009) demonstrated that nicotine infusions, earned under a PR schedule, were negatively correlated with plasma PROG levels in female rats. Together, these results provide little support that nicotine reinforcement and withdrawal are affected by hormone cycle phase in animals.

Systemic progestin administration: animal research

The following section discusses further research on the effects of PROG and its metabolites on drug-induced behaviors in animals. There have been many studies on this topic; however, only research investigating behaviors related to drug reinforcement (drug seeking), withdrawal, and overdose will be discussed, as they are the most representative of drug abuse (see Table 6).

Table 6.

Summary of the effects of ALLO administration on behaviors elicited by other drugs of abuse in animals.

Drug Independent Variable Dependent Measure Finding a Reference
Nicotine ALLO administration Nicotine- precipitated seizure A < V Luntz-Leybmann et al., 1990
Martin-Garcia and Pallares 2005
Alcohol ALLO administration Maintenance A > V in rats with a brief history of alcohol intake Janak et al., 1998
VanDoren et al., 2000
Morrow et al., 2001
No effect in rats with a brief history of alcohol intake Martin-Garcia et al., 2007
A < V in rats with a long history of alcohol intake Morrow et al., 2001
Ford et al., 2005
Martin-Garcia et al., 2007
Withdrawal A < V Hirani et al., 2002
Martin-Garcia and Pallares 2005
Reinstatement Reinstates alcohol seeking: A > V Finn et al., 2008
Morphine ALLO administration Withdrawal A < V Reddy and Kulkarni 1997b
Benzodiazepine ALLO administration Withdrawal A < V Reddy and Kulkarni 1997a

A: ALLO; V: vehicle

Alcohol

Drug seeking

Despite a lack of evidence indicating hormone cycle effects on alcohol reinforcement, a host of studies using controlled progestin manipulation techniques suggest a strong relationship between alcohol and progestins, particularly ALLO. In these studies, the influence of systemic progestin administration on alcohol reinforcement is dependent on the history of alcohol intake (Morrow et al., 2001). Rodents with a brief history of ethanol exposure show an increase (Janak et al., 1998; Morrow et al., 2001; VanDoren et al., 2000) or no change (Martin-Garcia et al., 2007) in ethanol self-administration following treatment with a low dose of ALLO; whereas, rats and mice with an extensive history of ethanol exposure show a decrease in ethanol self-administration (Ford et al., 2005; Martin-Garcia et al., 2007; Morrow et al., 2001). Differences in the effects of ALLO on these measures may be related to the differential effects of chronic vs acute ethanol exposure on GABAA receptor subunit expression and their subsequent function in mesolimbic areas of the brain that underlie the rewarding effects of ethanol (Papadeas et al., 2001).

Blocking ALLO biosynthesis, through the administration of finasteride, forestalls the acquisition and decreases the maintenance (Ford et al., 2008) of ethanol-seeking behavior in rodents. However, the administration of a priming injection of ALLO reinstates ethanol seeking following a period without alcohol (Finn et al., 2008). These results indicate a role of ALLO in the discrimination of alcohol and they support previous human research showing a correlation between progestin increases and alcohol-induced intoxication and positive subjective effects (Torres and Oretag, 2003, 2004; Pierucci-Lagha et al., 2005). Results with cocaine are different, and this illustrates the pharmacological specificity of ALLO’s effects on drug seeking. Like alcohol, cocaine increases plasma levels of ALLO (Quinones-Jenab et al., 2008); however, ALLO did not reinstate cocaine-seeking behavior when injected alone but rather decreased this behavior (Anker et al., 2009). This indicates that ALLO’s effects on drug seeking may be dependent on the pharmacological class of the self-administered drug.

Withdrawal

ALLO and pregnanolone have also been studied for their potential in treating symptoms associated with alcohol withdrawal. In animal research, decreases in PROG and ALLO (Finn et al., 2000) levels were found during ethanol withdrawal, and further decreasing ALLO levels by administering finasteride (Gorin-Meyer et al., 2007) or by performing an adrenalectomy (Strong et al., 2009) potentiated the severity of these withdrawal effects. Interestingly, increasing ALLO levels alleviated ethanol-induced withdrawal effects in rats (Hirani et al., 2002; Martin-Garcia and Pallares, 2005; Uzbay et al., 2004). These findings are supported by human research indicating that decreased biosynthesis of ALLO contributed to the severity of withdrawal symptoms in alcoholics (Hill et al., 2005; Romeo et al., 1996), while the reinstatement of baseline levels of PROG and ALLO produced psychosomatic stability in patients receiving alcohol detoxification therapy (Hill et al., 2005). These findings suggest a possible role for progestins in the alleviation of alcohol withdrawal symptoms in alcoholics.

Progestin-alcohol interactions may be related to their similar neurobiological and behavioral profiles during tolerance and withdrawal states (Deitrich et al., 1989; Frye et al., 1981). Exposure to ethanol resulted in a decrease in receptor sensitivity to GABA (Chandler et al., 1998; Faingold et al., 1998; Grobin et al., 1998) and behavioral tolerance (Allan and Harris, 1987). In addition, following discontinuation of ethanol use, a withdrawal state characterized by depression (McKeon et al., 2008), anxiety, and seizure susceptibility occurred (McKeon et al., 2008; Ueno et al., 2001). Similarly, prolonged exposure to progestins reduced the efficacy of GABAA in neuronal cultures (Friedman et al., 1993, 1996; Yu and Ticku, 1995; Yu et al., 1996), precipitated tolerance to their anticonvulsant effects (Czlonkowska et al., 2001), and like alcohol, produced a withdrawal state characterized by depression (as measured by the forced swim test) (Beckley and Finn, 2007), anxiety (Smith et al., 1998), and susceptibility to seizures (Frye and Bayon, 1998). These results highlight the close relationship between the neurobiological and behavioral effects of progestins and alcohol. It is possible that in the aforementioned studies ALLO contributed to the behavioral and neurobiological effects of alcohol, as ALLO levels dramatically increased following alcohol administration (Torres and Oretag, 2003, 2004; Pierucci-Lagha et al., 2005)

Overall, research suggests that PROG and its metabolite ALLO are important determinants of behaviors associated with alcohol abuse and withdrawal. The effects of progestins on alcohol administration differ depending on the history of alcohol exposure and the neurobiological and behavioral profiles of progestins and alcohol are similar. Results from these studies indicate a possible etiological relationship between progestins and alcohol abuse, and they suggest that knowledge of the progestin-alcohol relationship may lead to novel methods for the treatment of alcohol dependence.

Other drugs of abuse

Progestins are also involved in the attenuation of behavioral responses to nicotine, benzodiazepines, and morphine; however, relatively few studies have examined the effects of progestins on these drugs of abuse across different phases of the drug abuse process. Studies examining these effects indicate that treatment with PROG’s precursor (pregnenolone), its metabolite (ALLO), or PROG itself, blocked the development of tolerance and the expression of withdrawal signs following benzodiazepine (Reddy and Kulkarni, 1997a) and morphine administration (Reddy and Kulkarni, 1997b) in mice. In addition, similar to its effects on cocaine-induced seizures (Kaminski et al., 2003; Leskiewicz et al., 2003), systemic (Luntz-Leybmann et al., 1990) or intrahippocampal (Martin-Garcia and Pallares, 2005) administration of ALLO resulted in an attenuation of seizures precipitated by large doses of nicotine, purportedly through its mediation of the GABAergic neurotransmitter system (Frye, 1995; Frye et al., 2000).

This section expanded the results with cocaine and discussed human and animal research examining the interaction between other drugs of abuse and progestins. One conclusion is that there are relatively few consistent reports in humans and/or animals concerning the influence of progestins on the positive subjective and reinforcing effects of drugs of abuse other than those reported with cocaine. However, variations in the methodology employed in studies examining drugs other than the stimulants may preclude an accurate comparison of their findings.

Mechanisms of progestins’ effects on drug-mediated behavior

Progestins influence a range of neurobiological mechanisms that are related to drug abuse. In addition, the effects of progestins on drug-related behaviors might be dependent on EST levels and/or their influence on the pharmacokinetics of drugs of abuse. The following section attempts to improve our understanding of the effects of drugs of abuse on progestin levels, as that may clarify the mechanisms by which progestins interact with drug-induced behaviors. Particular attention is given to the progestin interaction with neurobiological systems that regulate cocaine abuse, as research on this topic is the most abundant.

Neurobiological mechanisms of progestins on drug seeking

Progestins influence several stress- and reward-related neurobiological substrates that underlie drug abuse such as GABAA (Schumacher and McEwen, 1989; Schumacher et al., 1989; for reviews, see Lambert et al., 1995; Gasior et al., 1999), 5-HT3 (Wetzel et al., 1998), and nicotinic acetylcholine (Bullock et al., 1997) receptors in addition to HPA activation (Drugan et al., 1993; Owens et al., 1992; Patchev et al., 1994; Purdy et al., 1991) and dopamine signaling (Barrot et al., 1999; Jaworska-Feil et al., 1998; Laconi et al., 2007; Rouge-Pont et al., 2002). The following section discusses the more commonly accepted neurobiological mechanisms that are thought to underlie the effects of progestins on drug-related behaviors. These mechanisms most likely do not operate independently but in concert to influence drug abuse. Their interactions are assumed to be complex and have not been extensively studied. Thus, the following sections will focus primarily on how progestins mediate each mechanism related to drug abuse, and neurobiological interactions (e.g., GABA’s effects on dopamine transmission) will be discussed when supported by relevant research.

HPA axis

As suggested by Koob and Volkow (2009), drug taking is initially regulated by positive reinforcement through interaction with the mesolimbic dopamine system. However, over time and with repeated binge and withdrawal cycles, this behavior is increasingly regulated by negative reinforcement through heightened withdrawal, craving, and increased sensitivity to stress that may further enhance vulnerability to drug bingeing and/or relapse. In both humans and animals the administration and subsequent withdrawal from several drugs of abuse (e.g., cocaine, nicotine, alcohol, cannabinoids, opiates) increases extracellular CRF, enhances HPA activation, and produces anxiogenic-like behavior (Fox and Sinha, 2009, George et al., 2007; Knapp et al., 2004; Overstreet et al., 2004; Rodriguez de Fonseca et al., 1997; Sarnyai et al., 1995). Furthermore, stress and HPA activity disruption are predictors of craving (Piasecki et al., 2003; Stine et al., 2002), treatment outcomes, and relapse (Sinha and Fox, 2008) in humans (Fox and Sinha, 2009) and are associated with drug bingeing and relapse in animal models of drug abuse (Harris and Aston-Jones, 2003; Hutcheson et al., 2001; Shaham et al., 1996; Valdez et al., 2002). Therefore, agents that normalize drug-induced disruption in the stress system may be useful treatments for drug abuse.

Interestingly, PROG and/or ALLO alleviate anxiogenic-like behaviors (Bitran et al., 1995; Brot et al., 1997; Laconi et al., 2001), normalize overactive HPA activity by attenuating the release of CRF following exposure to stress (Drugan et al., 1993; Frye et al., 2006; Owens et al., 1992; Patchev et al., 1994; Purdy et al., 1991), and as previously mentioned, inhibit the potentiating effects of stress on cocaine-related responses. For example, increases in PROG were associated with decreased stress-induced craving in women addicted to cocaine (Sinha et al., 2007). Furthermore, reinstatement of cocaine-seeking behavior induced by a stressful stimulus (yohimbine) was significantly attenuated by ALLO in female rats (Anker and Carroll, 2010). Thus, one mechanism that may explain the effects of PROG and ALLO on cocaine-elicited behavior involves their attenuation of stress- and cocaine-induced HPA activation.

The effects of EST on HPA activation and HPA-mediated behaviors are opposite to those of PROG and its metabolite, ALLO. In rats, EST potentiated the release of CRF (Patchev et al., 1995; Swanson and Simmons, 1989) and increased adrenocorticotropic hormone (ACTH) and corticosterone (Burgress and Handa, 1992), leading to increased HPA activity (Dallman et al., 2004). These effects also extended to cocaine-induced HPA activation. In a study by Niyomchai and colleagues (2005), EST increased cocaine-induced corticosterone levels relative to VEH-treated controls. Behavioral studies provided further support for the opposing effects of PROG and EST. Estrogen facilitated fear-potentiated startle in OVX female rats relative to OVX rats treated with VEH, while the administration of PROG attenuated this facilitation (Hiroi and Neumaier, 2006; Toufexis et al., 2004). Further work is needed to examine the interaction between EST and progestins in relation to their effects on stress-related drug abuse. One question for further research is whether PROG or ALLO can block EST’s potentiating effect on cocaine-induced corticosterone release and whether this effect is related to cocaine seeking.

Dopamine

Dopamine mediates stress (Deutch and Roth, 1990; Imperato et al., 1991; Inglis and Moghaddam, 1999; Koob, 2009; Koob and Le Moal, 2005, 2008) in addition to the rewarding effects associated with cocaine (Koob and Kreek, 2007; Pierce and Kumaresan, 2006; Spanagel and Weiss, 1999). These effects occur in the NAc and ventral tegmental areas of the brain (Spangel and Wiess, 1999) that are thought to play a crucial role in cocaine craving in humans (Brieter et al., 1997; Volkow et al., 1999). These regions are also implicated in drug self-administration (Carelli, 2002; Kiyatkin and Stein, 1995) and in the reinstatement of drug seeking in rats (Anderson et al., 2003, 2006; Schmidt et al., 2006). Research with rodents indicated that progestins modulated dopamine in these brain areas. For example, ALLO attenuated stress-induced increases in dopamine (Dazzi et al., 2002) and altered dopamine release in the striatum and NAc (Barrott et al., 1999; Jaworska-Feil et al., 1998; Laconi et al., 2007; Rouge-Pont et al., 2002). PROG also modulated dopamine levels in the striatum; however, results from these studies were equivocal and dependent on the time of testing, manner of administration, and presence or absence of EST (Dluzen and Ramirez, 1984; Fernandez-Ruiz et al., 1989). Nevertheless, evidence suggests that PROG and ALLO may interact with dopamine to produce attenuating effects on drug-seeking behavior.

GABA

Progestins’ effects on GABA neurotransmission may be an additional mechanism influencing drug-induced responses. Pharmacological agents that enhance GABA neurotransmission (e.g., topiramate, tiagabine, and baclofen) decrease dopamine transmission in areas of the brain that support the rewarding effects of drugs of abuse and have shown promise in treating drug dependence in humans (Kalivas, 2007; Karila et al., 2008). Furthermore, there is reduced craving in humans and/or cocaine seeking in animals following treatment with the GABAB agonist baclofen (Brebner et al., 2002; Campbell et al., 1999, 2002; Ling et al., 1998; Shoptaw et al., 2003), the GABA upregulator topiramate (Reis et al., 2008), and the irreversible GABA transaminase inhibitor vigabatrin (Fechtner et al., 2006; Kushner et al., 1999; Peng et al., 2008). Given that ALLO and pregnanolone are potent positive allosteric modulators of GABAA receptors (Gasior et al., 1999; Schumacher and McEwen, 1989; Schumacher et al., 1989; for a review see Lambert et al., 1995), their actions on the GABA receptor system may partially explain their attenuating effects on drug seeking.

Progesterone receptors and sigma receptors

PROG receptors are present in areas of the brain implicated in behavioral responses to drugs of abuse such as prefrontal cortex, ventral tegmental area, and hippocampus (Parsons et al., 1982; Guerra-Araiza, 2000; Frye, 2001). In one study, Wu et al., (2006) demonstrated that PROG receptor protein levels and DNA binding complexes in rats were significantly increased in the NAc following increases in PROG serum levels.

PROG is an antagonist of sigma1 (σ1) receptors that are important neurosubstrates of learning and memory processes (Maurice et al., 2002). σ1 receptors are also thought to regulate drug-seeking behaviors such as withdrawal and relapse (Romieu et al., 2004). For example, the administration of σ1 antagonists blocked the formation and reinstatement of cocaine-induced conditioned place preference in mice (Romieu et al., 2002, 2004). Thus, PROG receptors and σ1 receptors may be implicated in the effect of PROG on drug-seeking behavior.

Progestins’ dependence on estrogen

Findings suggest that the attenuating effects of progestins on cocaine reinforcement may be dependent on the interaction of the progestin with EST. For example, PROG, when coadministered with EST, blocked EST-induced enhancement of cocaine-seeking behavior in OVX female rats (Anker et al., 2007, 2009; Jackson et al., 2006; Larson et al., 2007), but it failed to alter cocaine-induced reinstatement in OVX female rats in the absence of EST (Anker and Carroll, unpublished data). These findings suggest that PROG decreased cocaine-induced responses to the levels of those found in OVX rats, without decreasing them below response levels seen under conditions where EST is low (e.g., following OVX).

Results with cocaine-elicited CPP in OVX female rats are opposite to those reported for cocaine-seeking behavior (Anker and Carroll, unpublished data; Anker et al., 2007, 2009; Jackson et al., 2006; Larson et al., 2007). For example, cocaine-induced CPP was potentiated when PROG was coadministered with EST (Russo et al., 2003) but was blocked when PROG was administered alone. One explanation for these divergent findings may be related to the different learning mechanisms that mediate CPP and drug seeking. For example, drug seeking is guided by response-outcome contingencies (operant conditioning), but it is unknown whether the expression of CPP is mediated by operant and/or stimulus-outcome contingencies (classical conditioning) (Bardo and Bevins, 2000). A comparison of CPP and drug self-administration studies draws further distinction and indicates dissociation between the neurobiological mechanisms that mediate both behaviors (for review see Bardo and Bevins, 2000). Indeed, endogenous levels of PROG and EST, or their systemic injection following OVX in female rats, enhanced performance on learning and memory tasks such as the object recognition and object placement tasks (Frye, 2007; Paris and Frye, 2008), while the administration of both hormones decreased operant behavior maintained by a nondrug food reinforcer (Rodriguez-Sierra et al., 1984). Thus, EST and PROG may facilitate the recall of stimulus-outcome contingencies that guide cocaine-induced CPP (Russo et al., 2003) while they attenuate response-outcome contingencies underlying operant responding and cocaine seeking (Anker et al., 2007; Jackson et al., 2006; Larson et al., 2005, 2007).

In contrast to results with CPP and cocaine seeking, findings concerning EST-dependent effects of progestins on cocaine-induced locomotor activity are equivocal. For example, locomotor activity following cocaine administration increased (Sircar and Kim, 1999; Perrotti et al., 2001), decreased (Sell et al., 2000), or was not affected (Peris et al., 1991; Perrotti et al., 2001) by coadministration of PROG and EST in OVX female rats relative to OVX VEH-treated controls. Furthermore, administration of PROG alone in OVX females decreased cocaine-induced stereotypy (Niyomchai et al., 2005), but it had no effect on cocaine-induced locomotor activity (Quinones-Jenab et al., 2000b; Perrotti et al., 2001; Sell et al., 2000). Inconsistencies in these reports may be related to differences in the hormone dosing regimens as well as strain and age differences of the subjects between the studies.

An additional mechanism that may underlie EST-dependent effects of PROG on cocaine seeking may be related to GABA neurotransmission. Decreased GABA release in the ventral pallidum is associated with increased dopamine release in addition to heightened drug-seeking behavior in rats (Tang et al., 2005; Callie and Parsons, 2004), while the administration of GABA agonists decrease dopamine release and cocaine seeking (Campbell et al., 1999, 2002). This suggests that decreased GABA is associated with heightened vulnerability toward cocaine abuse, while increased GABA may attenuate drug-taking behavior. Previous work indicates that EST inhibited activation of medium spiny GABAergic neurons in the striatum (Mermelstein et al., 1996), increased striatal GABA release (Hu et al., 2006), and enhanced dopamine metabolism and turnover (Di Paolo et al., 1985; Shimizu and Bray, 1993) and stimulant-elicited dopamine release in the striatum (Becker and Beer, 1986; Becker, 1990a, b; Castner et al., 1993). Conversely, PROG and its metabolites promoted striatal-GABA activity (Schumacher and McEwen, 1989, Schumacher et al., 1989) and decreased dopamine (Dazzi et al., 2002; Dluzen and Ramirez, 1984; Jaworska-Feil et al., 1998; Laconi et al., 2007; Shimizu and Bray, 1993) in the striatum. Thus, the different effects of ALLO and EST on cocaine seeking may be explained by their opposite effects on GABA and/or dopamine neurotransmission in areas of the brain that regulate drug seeking.

Progestins’ interaction with cocaine pharmacokinetics

It is possible that PROG and/or its metabolites may interact with drug pharmacokinetics to affect drug-seeking behavior. Most of this research has been conducted with cocaine. However, previous findings do not support this notion and indicate little to no effect of the menstrual cycle on plasma cocaine levels in women following iv or smoked cocaine (Evans and Foltin, 2004; Mendelson et al., 1999; Sofuoglu et al., 1999). This suggests that PROG’s suppressant effect on cocaine-induced behavior does not involve an interaction with cocaine metabolism. While studies examining the effects of menstrual cycle on cocaine metabolism following iv and smoked cocaine agree that pharmacokinetics is not an issue, findings from studies using intranasal cocaine administration have been inconsistent. For example, Lukas et al., (1996) reported lower peak plasma cocaine levels during the PROG-dominant luteal phase than during the follicular phase of the menstrual cycle (Lukas et al., 1996), while Collins et al., (2007) found no phase differences in plasma cocaine. Differences in experimental procedures such as cocaine dose, timing of cocaine administration, and number of self-administered doses may have accounted for these varied results. For example, Collins and co-workers (2007) used two doses separated by 40 min, while Lukas et al. (1996) used only one dose.

Findings showing a lack of effect of female gonadal hormones on blood cocaine levels are also supported by research in animals. There was no interaction between menstrual cycle phase and cocaine levels in plasma in nonhuman primates (Evans and Foltin, 2004). Similarly, studies using hormone replacement in OVX rats failed to show a consistent role of PROG on plasma levels of cocaine’s metabolite benzoylecgonine (Quinones-Jenab et al., 2000a; Perrotti et al., 2001; Niyomchai et al., 2006). Taken together most studies to date indicate that hormones such as PROG are most likely not exerting their inhibitory effects via alteration of cocaine pharmacokinetics.

Cocaine’s effects on progestin levels

Studies have revealed that administration of several drugs of abuse increase circulating progestin levels. Plasma PROG levels were significantly higher during early cocaine abstinence in women (Fox et al., 2008). Also, in male and female rats PROG levels (Chin et al., 2002; Festa and Quinones-Jenab, 2004; Grobin et al., 2005; Quinones-Jenab et al., 2000a, 2008; Walker et al., 2001) and PROG receptor activation (Wu et al., 2006) increased following cocaine administration. Furthermore, cocaine-induced increases in PROG in freely-cycling female rats were dependent on the estrous cycle stage, with the highest levels occurring during proestrus when EST is high (Quinones-Jenab et al., 2000a; Walker et al., 2001). These results indicated that cocaine-induced elevation of PROG was independent of sex (occurs in both males and females) and was possibly influenced by EST in females. However, other research suggested that the effects of cocaine on plasma levels of ALLO were sex-dependent. For example, cocaine-induced increases in plasma levels of PROG’s metabolite ALLO have been reported for female (Quinones-Jenab et al., 2008) but not male (Quinones-Jenab et al., 2008; Grobin et al., 2005) rats. These results indicated that cocaine affects PROG synthesis and receptor expression regardless of sex, while cocaine’s influence on ALLO levels was specific to females.

Similar to cocaine, cortical and/or plasma concentrations of PROG and ALLO were elevated following administration of other drugs of abuse such as alcohol (Barbaccia et al., 1999; Morrow et al., 2001; VanDoren et al., 2000), nicotine (Concas et al., 2006; Porcu et al., 2003), morphine (Concas et al., 2006; Grobin et al., 2005), and tetrahydrocannabinol (Porcu et al., 2003). All of these drugs activate the HPA access and elicite stress responses following their administration and/or during their withdrawal (Adinoff et al., 2005; Fox and Sinha, 2009; Junghanns et al., 2003; Lee et al., 2001; Shaham et al., 2000; Stewart, 2000). Thus, increases in PROG and ALLO may serve as a mechanism to restore homeostasis in the brain following drug-induced HPA dysregulation (Grobin et al., 2005). This is supported by a finding demonstrating that cocaine-induced increases in progestin levels were blocked following adrenalectomy (Walker et al., 2001). Findings also suggest that prolonged drug intake may suppress the ability of progestins to attenuate responses to stress. For example, Childs and de Wit (2009) demonstrated that following a public speaking procedure (i.e., the Trier Social Stress Test), chronic cigarette smokers scored higher on self-reported measures of stress and had lower baseline levels of ALLO in the plasma in addition to lower stress-induced increases of PROG compared to non-smokers. Thus, continued drug use may produce a dysregulation in the body’s ability to cope with stress, and this may lead to further drug use.

Baseline measures of PROG and ALLO were also present to a greater extent in females compared to males during the luteal phase of the menstrual cycle (but are similar between sexes when women are in the follicular phase) (Genazzani et al., 1998; Pearson Murphy and Allison, 2000), and this may partially explain why females, compared to males, showed fewer withdrawal effects across numerous drugs such as alcohol (Alele and Devaud, 2007; Cicero et al., 2002; Devaud and Chadda, 2001; Varlinskaya and Spear, 2004; Woodstock-Striley et al., 2004), pentobarbital (Suzuki et al., 1985), methaqualone (Suzuki et al., 1988), and PCP (Carroll et al., 2009). These results suggest that increased levels of progestins may afford protection against the aversive affects of drugs of abuse in women.

Conclusion

Findings from human and animal research indicate a role for PROG and its metabolites in several aspects of drug abuse. This work highlights the importance of hormonal status of women in regulating behaviors maintained by drugs of abuse, particularly with respect to cocaine. In human research, phases of the menstrual cycle associated with high endogenous levels of progestins, or their systemic injection, corresponded with decreases in the subjective and physiological effects of cocaine. Similar results were found in animal research on the effects of endogenous and exogenous progestins on the rewarding (cocaine seeking) and aversive effects of cocaine across several phases of the drug abuse process. Findings from animal research have further explored PROG’s actions, implicating its metabolite ALLO in the attenuation of many behavioral effects of cocaine. Thus, human research may benefit from examining the effects of ALLO on cocaine-induced subjective and physiological responses.

Of particular interest is the influence of progestins’ on stress as it relates to drug abuse. Physiological and behavioral responses to stress were associated with rewarding and aversive aspects of cocaine abuse and results from human and animal research discussed here indicated a role for progestins in attenuating these responses. Stressful events or the administration of drugs of abuse increased levels of circulating progestins and a relationship between hormones, stress, and cocaine abuse was substantiated by findings from human and animal research.

Human and animal studies suggest that progestins influence responses to other drugs of abuse as well as cocaine. For example, progestins, especially ALLO, have a strong relationship with subjective and behavioral effects of alcohol. However, these effects are opposite to those of cocaine in that ALLO plasma levels were correlated with increased alcohol-induced positive-subjective effects and intoxication in women, while systemic administration of ALLO increased levels of alcohol-seeking behavior in rats. These results indicate that ALLO may be responsible for regulating the reinforcing effects of alcohol. Despite this finding, there is little support that progestins affect reinforcement maintained by other drugs of abuse. However, results indicate an important role for progestins in the alleviation of negative aspects of many of these drugs.

Similar to results with cocaine, findings from various studies using alcohol, benzodiazepines, nicotine, and morphine indicate that progestins afford protection against the aversive consequences of drug abuse such as withdrawal and the potentially lethal effects of overdose. This suggests their role as possible compensatory mechanisms in the regulation of negative aspects of drug use, and this may explain why females are more vulnerable than males to cocaine seeking across numerous phases of drug abuse (i.e., they are less likely to experience the negative consequences).

As discussed earlier, there is little support indicating that the inhibitory effects of progestins are due to their pharmacokinetic effects on cocaine. Rather, there is support that progestins alter drug-maintained behavior through their influence on neurochemical processes related to drug reward. This review presented findings indicating that neurochemical profiles associated with enhanced HPA activation, increased levels of dopamine, and decreased GABA in the striatum coincided with the expression of drug reward and/or drug-seeking behavior, while decreased HPA activity, decreased levels of dopamine, and increased GABA were associated with an attenuation of these behaviors. Interestingly, EST has been shown to produce effects similar to the former profile (increased HPA and dopamine, decreased GABA) while PROG and its metabolites produce effects that fit the latter profile (decreased HPA and dopamine, increased GABA). Thus, the fact that EST increases and progestins decrease measures indicative of drug abuse may be related to their opposite effects on these neurochemical substrates.

An important consideration from the findings discussed in this review is that addictive disorders affect women differently than men. The hormones discussed here are present to a much greater degree in women, and symptomology associated with drug abuse disorders fluctuates with increases and decreases of progestins and EST in females. These fluctuations may mark periods of increased vulnerability to, or protection against, drug abuse in women. The findings presented in this review highlight the importance of sex-specific treatment strategies that take into account the unique interactions between female gonadal hormones (and their metabolites) and drug abuse. The feasibility of this strategy has been demonstrated in previous human research. These studies indicate that scheduling smoking quit dates around periods of the menstrual cycle that mark periods of increased vulnerability to relapse may be beneficial to treatment outcome in woman smokers (Franklin et al., 2008; Carpenter et al., 2008). Similar strategies could be implemented in the treatment of other excessive behaviors maintained by drug and nondrug rewards that are known to be influenced by menstrual cycle such as cocaine and amphetamine dependence.

In conclusion, the findings discussed in this review indicate that women and men respond differently to the effects of drugs of abuse. For women, fluctuations of progestins may mark periods of increased or decreased vulnerability to the development of drug dependence, reflected by their effects on both the reinforcing and negative aspects of this disorder. However, progestins’ potential for therapeutic applications has yet to be examined for problems of drug dependence and other addictive disorders involving nondrug substances/events (e.g., food, gambling). The influence of progestins on addictive behaviors is a promising area of scientific inquiry, as the research presented here suggests the potential for progestins in the treatment of drug abuse.

Footnotes

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

References

  1. Adinoff B, Junghanns K, Kiefer F, Krishnan-Sarin S. Suppression of the HPA axis stress-response: implications for relapse. Alcohol Clin Exp Res. 2005;29:1351–1355. doi: 10.1097/01.ALC.0000176356.97620.84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Ahmed SH, Koob GF. Transition from moderate to excessive drug intake: change in hedonic set point. Science. 1998;282:298–300. doi: 10.1126/science.282.5387.298. [DOI] [PubMed] [Google Scholar]
  3. Alele PE, Devaud LL. Sex differences in steroid modulation of ethanol withdrawal in male and female rats. J Pharmacol Exp Ther. 2007;320:427–436. doi: 10.1124/jpet.106.107896. [DOI] [PubMed] [Google Scholar]
  4. Allan AM, Harris RA. Acute and chronic ethanol treatments alter GABA receptor-operated chloride channels. Pharmacol Biochem Behav. 1987;27:665–670. doi: 10.1016/0091-3057(87)90192-4. [DOI] [PubMed] [Google Scholar]
  5. Allen SS, Allen AM, Lunos S, Hatsukami DK. Patterns of self-selected smoking cessation attempts and relapse by menstrual phase. Addict Behav. 2009a;34:928–931. doi: 10.1016/j.addbeh.2009.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Allen SS, Allen AM, Pomerleau CS. Influence of phase-related variability in premenstrual symptomatology, mood, smoking withdrawal, and smoking behavior during ad libitum smoking, on smoking cessation outcome. Addict Behav. 2009b;34:107–111. doi: 10.1016/j.addbeh.2008.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Allen SS, Hatsukami D, Christianson D, Nelson D. Symptomatology and energy intake during the menstrual cycle in smoking women. J Subst Abuse. 1996;8:303–319. doi: 10.1016/s0899-3289(96)90170-4. [DOI] [PubMed] [Google Scholar]
  8. Allen SS, Hatsukami DK, Bade T, Center B. Transdermal nicotine use in postmenopausal women: does the treatment efficacy differ in women using and not using hormone replacement therapy? Nicotine Tob Res. 2004;6:777–788. doi: 10.1080/1462220042000274239. [DOI] [PubMed] [Google Scholar]
  9. Allen SS, Hatsukami DK, Christianson D, Nelson D. Withdrawal and pre-menstrual symptomatology during the menstrual cycle in short-term smoking abstinence: effects of menstrual cycle on smoking abstinence. Nicotine Tob Res. 1999;1:129–142. doi: 10.1080/14622299050011241. [DOI] [PubMed] [Google Scholar]
  10. Anderson SM, Bari AA, Pierce RC. Administration of the D1-like dopamine receptor antagonist SCH-23390 into the medial nucleus accumbens shell attenuates cocaine priming-induced reinstatement of drug-seeking behavior in rats. Psychopharmacology (Berl) 2003;168:132–138. doi: 10.1007/s00213-002-1298-5. [DOI] [PubMed] [Google Scholar]
  11. Anderson SM, Schmidt HD, Pierce RC. Administration of the D2 dopamine receptor antagonist sulpiride into the shell, but not the core, of the nucleus accumbens attenuates cocaine priming-induced reinstatement of drug seeking. Neuropsychopharmacology. 2006;31:1452–1461. doi: 10.1038/sj.npp.1300922. [DOI] [PubMed] [Google Scholar]
  12. Anker JJ, Carroll ME. Sex differences in the effects of allopregnanolone on yohimbine-induced reinstatement of cocaine seeking in rats. Drug Alcohol Depend. 2010;107:264–267. doi: 10.1016/j.drugalcdep.2009.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Anker JJ, Holtz NA, Zlebnik N, Carroll ME. Effects of allopregnanolone on the reinstatement of cocaine-seeking behavior in male and female rats. Psychopharmacology (Berl) 2009;203:63–72. doi: 10.1007/s00213-008-1371-9. [DOI] [PubMed] [Google Scholar]
  14. Anker JJ, Larson EB, Gliddon LA, Carroll ME. Effects of progesterone on the reinstatement of cocaine-seeking behavior in female rats. Exp Clin Psychopharmacol. 2007;15:472–480. doi: 10.1037/1064-1297.15.5.472. [DOI] [PubMed] [Google Scholar]
  15. Barbaccia ML, Affricano D, Trabucchi M, Purdy RH, Colombo G, Agabio R, Gessa GL. Ethanol markedly increases “GABAergic” neurosteroids in alcohol-preferring rats. Eur J Pharmacol. 1999;384:R1–2. doi: 10.1016/s0014-2999(99)00678-0. [DOI] [PubMed] [Google Scholar]
  16. Baker TB, Piper ME, McCarthy DE, Majeskie MR, Fiore MC. Addiction motivation reformulated: An affective processing model of negativ reinforcement. Psychol Rev. 2004;111:33–51. doi: 10.1037/0033-295X.111.1.33. [DOI] [PubMed] [Google Scholar]
  17. Bardo MT, Bevins RA. Conditioned place preference: what does it add to our preclinical understanding of drug reward? Psychopharmacology (Berl) 2000;153:31–43. doi: 10.1007/s002130000569. [DOI] [PubMed] [Google Scholar]
  18. Barrot M, Vallee M, Gingras MA, Le Moal M, Mayo W, Piazza PV. The neurosteroid pregnenolone sulphate increases dopamine release and the dopaminergic response to morphine in the rat nucleus accumbens. Eur J Neurosci. 1999;11:3757–3760. doi: 10.1046/j.1460-9568.1999.00816.x. [DOI] [PubMed] [Google Scholar]
  19. Becker JB. Direct effect of 17 beta-estradiol on striatum: sex differences in dopamine release. Synapse. 1990a;5:157–164. doi: 10.1002/syn.890050211. [DOI] [PubMed] [Google Scholar]
  20. Becker JB. Estrogen rapidly potentiates amphetamine-induced striatal dopamine release and rotational behavior during microdialysis. Neurosci Lett. 1990b;118:169–171. doi: 10.1016/0304-3940(90)90618-j. [DOI] [PubMed] [Google Scholar]
  21. Becker JB, Beer ME. The influence of estrogen on nigrostriatal dopamine activity: behavioral and neurochemical evidence for both pre- and postsynaptic components. Behav Brain Res. 1986;19:27–33. doi: 10.1016/0166-4328(86)90044-6. [DOI] [PubMed] [Google Scholar]
  22. Bitran D, Hilvers RJ, Kellogg CK. Anxiolytic effects of 3 alpha-hydroxy-5 alpha[beta]-pregnan-20-one: endogenous metabolites of progesterone that are active at the GABAA receptor. Brain Res. 1991;561:157–161. doi: 10.1016/0006-8993(91)90761-j. [DOI] [PubMed] [Google Scholar]
  23. Bitran D, Shiekh M, McLeod M. Anxiolytic effect of progesterone is mediated by the neurosteroid allopregnanolone at brain GABAA receptors. J Neuroendocrinol. 1995;7:171–177. doi: 10.1111/j.1365-2826.1995.tb00744.x. [DOI] [PubMed] [Google Scholar]
  24. Bixo M, Andersson A, Winblad B, Purdy RH, Backstrom T. Progesterone, 5alpha-pregnane-3,20-dione and 3alpha-hydroxy-5alpha-pregnane-20-one in specific regions of the human female brain in different endocrine states. Brain Res. 1997;764:173–178. doi: 10.1016/s0006-8993(97)00455-1. [DOI] [PubMed] [Google Scholar]
  25. Boutrel B, Kenny PJ, Specio SE, Martin-Fardon R, Markou A, Koob GF, de Lecea L. Role for hypocretin in mediating stress-induced reinstatement of cocaine-seeking behavior. Proc Natl Acad Sci U S A. 2005;102:19168–19173. doi: 10.1073/pnas.0507480102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Brebner K, Childress AR, Roberts DC. A potential role for GABA(B) agonists in the treatment of psychostimulant addiction. Alcohol Alcohol. 2002;37:478–484. doi: 10.1093/alcalc/37.5.478. [DOI] [PubMed] [Google Scholar]
  27. Brody SL, Slovis CM, Wrenn KD. Cocaine-related medical problems: consecutive series of 233 patients. Am J Med. 1990;88:325–331. doi: 10.1016/0002-9343(90)90484-u. [DOI] [PubMed] [Google Scholar]
  28. Brot MD, Akwa Y, Purdy RH, Koob GF, Britton KT. The anxiolytic-like effects of the neurosteroid allopregnanolone: interactions with GABA(A) receptors. Eur J Pharmacol. 1997;325:1–7. doi: 10.1016/s0014-2999(97)00096-4. [DOI] [PubMed] [Google Scholar]
  29. Brown ZJ, Erb S. Footshock stress reinstates cocaine seeking in rats after extended post-stress delays. Psychopharmacology (Berl) 2007;195:61–70. doi: 10.1007/s00213-007-0846-4. [DOI] [PubMed] [Google Scholar]
  30. Brown-Grant K, Exley D, Naftolin F. Peripheral plasma oestradiol and luteinizing hormone concentrations during the oestrous cycle of the rat. J Endocrinol. 1970;48:295–296. doi: 10.1677/joe.0.0480295. [DOI] [PubMed] [Google Scholar]
  31. Bullock AE, Clark AL, Grady SR, Robinson SF, Slobe BS, Marks MJ, Collins AC. Neurosteroids modulate nicotinic receptor function in mouse striatal and thalamic synaptosomes. J Neurochem. 1997;68:2412–2423. doi: 10.1046/j.1471-4159.1997.68062412.x. [DOI] [PubMed] [Google Scholar]
  32. Burgess LH, Handa RJ. Chronic estrogen-induced alterations in adrenocorticotropin and corticosterone secretion, and glucocorticoid receptor-mediated functions in female rats. Endocrinology. 1992;131:1261–1269. doi: 10.1210/endo.131.3.1324155. [DOI] [PubMed] [Google Scholar]
  33. Campbell UC, Lac ST, Carroll ME. Effects of baclofen on maintenance and reinstatement of intravenous cocaine self-administration in rats. Psychopharmacology (Berl) 1999;143:209–214. doi: 10.1007/s002130050937. [DOI] [PubMed] [Google Scholar]
  34. Campbell UC, Morgan AD, Carroll ME. Sex differences in the effects of baclofen on the acquisition of intravenous cocaine self-administration in rats. Drug Alcohol Depend. 2002;66:61–69. doi: 10.1016/s0376-8716(01)00185-5. [DOI] [PubMed] [Google Scholar]
  35. Carelli RM. Nucleus accumbens cell firing during goal-directed behaviors for cocaine vs. ‘natural’ reinforcement. Physiol Behav. 2002;76:379–387. doi: 10.1016/s0031-9384(02)00760-6. [DOI] [PubMed] [Google Scholar]
  36. Carpenter MJ, Saladin ME, Leinbach AS, Larowe SD, Upadhyaya HP. Menstrual phase effects on smoking cessation: a pilot feasibility study. J Womens Health (Larchmt) 2008;17:293–301. doi: 10.1089/jwh.2007.0415. [DOI] [PubMed] [Google Scholar]
  37. Carpenter MJ, Upadhyaya HP, LaRowe SD, Saladin ME, Brady KT. Menstrual cycle phase effects on nicotine withdrawal and cigarette craving: a review. Nicotine Tob Res. 2006;8:627–638. doi: 10.1080/14622200600910793. [DOI] [PubMed] [Google Scholar]
  38. Carroll ME, Anker JJ. Sex differences and ovarian hormones in animal models of drug dependence. Horm Behav. 2009 doi: 10.1016/j.yhbeh.2009.10.001. [DOI] [PubMed] [Google Scholar]
  39. Carter CS. Neuroendocrinology of sexual behavior in the female. In: Knobil E, Neill JD, editors. The physiology of reproduction. 2. Raven Press, Ltd; New York, NY: 1994. pp. 711–733. [Google Scholar]
  40. Castner SA, Xiao L, Becker JB. Sex differences in striatal dopamine: in vivo microdialysis and behavioral studies. Brain Res. 1993;610:127–134. doi: 10.1016/0006-8993(93)91225-h. [DOI] [PubMed] [Google Scholar]
  41. Chandler LJ, Harris RA, Crews FT. Ethanol tolerance and synaptic plasticity. Trends Pharmacol Sci. 1998;19:491–495. doi: 10.1016/s0165-6147(98)01268-1. [DOI] [PubMed] [Google Scholar]
  42. Chaudhri N, Caggiula AR, Donny EC, Booth S, Gharib MA, Craven LA, Allen SS, Sved AF, Perkins KA. Sex differences in the contribution of nicotine and nonpharmacological stimuli to nicotine self-administration in rats. Psychopharmacology (Berl) 2005;180:258–266. doi: 10.1007/s00213-005-2152-3. [DOI] [PubMed] [Google Scholar]
  43. Chen K, Kandel D. Relationship between extent of cocaine use and dependence among adolescents and adults in the United States. Drug Alcohol Depend. 2002;68:65–85. doi: 10.1016/s0376-8716(02)00086-8. [DOI] [PubMed] [Google Scholar]
  44. Childs E, de Wit H. Hormonal, cardiovascular, and subjective responses to acute stress in smokers. Psychopharmacology (Berl) 2009;203:1–12. doi: 10.1007/s00213-008-1359-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Chin J, Sternin O, Wu HB, Burrell S, Lu D, Jenab S, Perrotti LI, Quinones-Jenab V. Endogenous gonadal hormones modulate behavioral and neurochemical responses to acute and chronic cocaine administration. Brain Res. 2002;945:123–130. doi: 10.1016/s0006-8993(02)02807-x. [DOI] [PubMed] [Google Scholar]
  46. Cicero TJ, Nock B, Meyer ER. Gender-linked differences in the expression of physical dependence in the rat. Pharmacol Biochem Behav. 2002;72:691–697. doi: 10.1016/s0091-3057(02)00740-2. [DOI] [PubMed] [Google Scholar]
  47. Collins SL, Evans SM, Foltin RW, Haney M. Intranasal cocaine in humans: effects of sex and menstrual cycle. Pharmacol Biochem Behav. 2007;86:117–124. doi: 10.1016/j.pbb.2006.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Concas A, Sogliano C, Porcu P, Marra C, Brundu A, Biggio G. Neurosteroids in nicotine and morphine dependence. Psychopharmacology (Berl) 2006;186:281–292. doi: 10.1007/s00213-005-0111-7. [DOI] [PubMed] [Google Scholar]
  49. Czlonkowska AI, Krzascik P, Sienkiewicz-Jaros H, Siemiqtkowski M, Szyndler J, Maciejak P, Bidzinski A, Plaznik A. Rapid down-regulation of GABA-A receptors after pretreatment of mice with progesterone. Pol J Pharmacol. 2001;53:385–388. [PubMed] [Google Scholar]
  50. Dallman MF, Akana SF, Strack AM, Scribner KS, Pecoraro N, La Fleur SE, Houshyar H, Gomez F. Chronic stress-induced effects of corticosterone on brain: direct and indirect. Ann N Y Acad Sci. 2004;1018:141–150. doi: 10.1196/annals.1296.017. [DOI] [PubMed] [Google Scholar]
  51. Dazzi L, Serra M, Vacca G, Ladu S, Latrofa A, Trapani G, Biggio G. Depletion of cortical allopregnanolone potentiates stress-induced increase in cortical dopamine output. Brain Res. 2002;932:135–139. doi: 10.1016/s0006-8993(02)02290-4. [DOI] [PubMed] [Google Scholar]
  52. DeBon M, Klesges RC, Klesges LM. Symptomatology across the menstrual cycle in smoking and nonsmoking women. Addict Behav. 1995;20:335–343. doi: 10.1016/0306-4603(94)00070-f. [DOI] [PubMed] [Google Scholar]
  53. Deitrich RA, Dunwiddie TV, Harris RA, Erwin VG. Mechanism of action of ethanol: initial central nervous system actions. Pharmacol Rev. 1989;41:489–537. [PubMed] [Google Scholar]
  54. Deutch AY, Roth RH. The determinants of stress-induced activation of the prefrontal cortical dopamine system. Prog Brain Res. 1990;85:367–402. doi: 10.1016/s0079-6123(08)62691-6. discussion 402–363. [DOI] [PubMed] [Google Scholar]
  55. Devaud LL, Chadda R. Sex differences in rats in the development of and recovery from ethanol dependence assessed by changes in seizure susceptibility. Alcohol Clin Exp Res. 2001;25:1689–1696. [PubMed] [Google Scholar]
  56. Di Paolo T, Rouillard C, Bedard P. 17 beta-Estradiol at a physiological dose acutely increases dopamine turnover in rat brain. Eur J Pharmacol. 1985;117:197–203. doi: 10.1016/0014-2999(85)90604-1. [DOI] [PubMed] [Google Scholar]
  57. Dluzen DE, Ramirez VD. Bimodal effect of progesterone on in vitro dopamine function of the rat corpus striatum. Neuroendocrinology. 1984;39:149–155. doi: 10.1159/000123971. [DOI] [PubMed] [Google Scholar]
  58. Donny EC, Caggiula AR, Rowell PP, Gharib MA, Maldovan V, Booth S, Mielke MM, Hoffman A, McCallum S. Nicotine self-administration in rats: estrous cycle effects, sex differences and nicotinic receptor binding. Psychopharmacology (Berl) 2000;151:392–405. doi: 10.1007/s002130000497. [DOI] [PubMed] [Google Scholar]
  59. Drugan RC, Park R, Kaufman L, Holmes PV. Etiology of the sexual dimorphism in renal peripheral benzodiazepine receptor response to stress in rats. Horm Behav. 1993;27:348–365. doi: 10.1006/hbeh.1993.1026. [DOI] [PubMed] [Google Scholar]
  60. Dykstra LA, Preston KL, Bigelow GE. Discriminative stimulus and subjective effects of opioids with mu and kappa activity: data from laboratory animals and human subjects. Psychopharmacology (Berl) 1997;130:14–27. doi: 10.1007/s002130050208. [DOI] [PubMed] [Google Scholar]
  61. Evans SM, Foltin RW. Pharmacokinetics of intravenous cocaine across the menstrual cycle in rhesus monkeys. Neuropsychopharmacology. 2004;29:1889–1900. doi: 10.1038/sj.npp.1300486. [DOI] [PubMed] [Google Scholar]
  62. Evans SM, Foltin RW. Exogenous progesterone attenuates the subjective effects of smoked cocaine in women, but not in men. Neuropsychopharmacology. 2006;31:659–674. doi: 10.1038/sj.npp.1300887. [DOI] [PubMed] [Google Scholar]
  63. Evans SM, Haney M, Foltin RW. The effects of smoked cocaine during the follicular and luteal phases of the menstrual cycle in women. Psychopharmacology (Berl) 2002;159:397–406. doi: 10.1007/s00213-001-0944-7. [DOI] [PubMed] [Google Scholar]
  64. Faingold CL, N’Gouemo P, Riaz A. Ethanol and neurotransmitter interactions--from molecular to integrative effects. Prog Neurobiol. 1998;55:509–535. doi: 10.1016/s0301-0082(98)00027-6. [DOI] [PubMed] [Google Scholar]
  65. Fattore L, Spano MS, Altea S, Angius F, Fadda P, Fratta W. Cannabinoid self-administration in rats: sex differences and the influence of ovarian function. Br J Pharmacol. 2007;152:795–804. doi: 10.1038/sj.bjp.0707465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Fechtner RD, Khouri AS, Figueroa E, Ramirez M, Federico M, Dewey SL, Brodie JD. Short-term treatment of cocaine and/or methamphetamine abuse with vigabatrin: ocular safety pilot results. Arch Ophthalmol. 2006;124:1257–1262. doi: 10.1001/archopht.124.9.1257. [DOI] [PubMed] [Google Scholar]
  67. Feltenstein MW, Byrd EA, Henderson AR, See RE. Attenuation of cocaine-seeking by progesterone treatment in female rats. Psychoneuroendocrinology. 2009;34:343–352. doi: 10.1016/j.psyneuen.2008.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Feltenstein MW, See RE. Potentiation of cue-induced reinstatement of cocaine-seeking in rats by the anxiogenic drug yohimbine. Behav Brain Res. 2006;174:1–8. doi: 10.1016/j.bbr.2006.06.039. [DOI] [PubMed] [Google Scholar]
  69. Feltenstein MW, See RE. Plasma progesterone levels and cocaine-seeking in freely cycling female rats across the estrous cycle. Drug Alcohol Depend. 2007;89:183–189. doi: 10.1016/j.drugalcdep.2006.12.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Fernandez-Ruiz JJ, Amor JC, Ramos JA. Time-dependent effects of estradiol and progesterone on the number of striatal dopaminergic D2-receptors. Brain Res. 1989;476:388–395. doi: 10.1016/0006-8993(89)91266-3. [DOI] [PubMed] [Google Scholar]
  71. Festa ED, Quinones-Jenab V. Gonadal hormones provide the biological basis for sex differences in behavioral responses to cocaine. Horm Behav. 2004;46:509–519. doi: 10.1016/j.yhbeh.2004.04.009. [DOI] [PubMed] [Google Scholar]
  72. Finn DA, Gallaher EJ, Crabbe JC. Differential change in neuroactive steroid sensitivity during ethanol withdrawal. J Pharmacol Exp Ther. 2000;292:394–405. [PubMed] [Google Scholar]
  73. Finn DA, Mark GP, Fretwell AM, Gililland-Kaufman KR, Strong MN, Ford MM. Reinstatement of ethanol and sucrose seeking by the neurosteroid allopregnanolone in C57BL/6 mice. Psychopharmacology (Berl) 2008;201:423–433. doi: 10.1007/s00213-008-1303-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Ford MM, Eldridge JC, Samson HH. Microanalysis of ethanol self-administration: estrous cycle phase-related changes in consumption patterns. Alcohol Clin Exp Res. 2002;26:635–643. [PubMed] [Google Scholar]
  75. Ford MM, Nickel JD, Phillips TJ, Finn DA. Neurosteroid modulators of GABA(A) receptors differentially modulate Ethanol intake patterns in male C57BL/6J mice. Alcohol Clin Exp Res. 2005;29:1630–1640. doi: 10.1097/01.alc.0000179413.82308.6b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Ford MM, Yoneyama N, Strong MN, Fretwell A, Tanchuck M, Finn DA. Inhibition of 5alpha-reduced steroid biosynthesis impedes acquisition of ethanol drinking in male C57BL/6J mice. Alcohol Clin Exp Res. 2008;32:1408–1416. doi: 10.1111/j.1530-0277.2008.00718.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Forger NG, Morin LP. Reproductive state modulates ethanol intake in rats: effects of ovariectomy, ethanol concentration, estrous cycle and pregnancy. Pharmacol Biochem Behav. 1982;17:323–331. doi: 10.1016/0091-3057(82)90087-9. [DOI] [PubMed] [Google Scholar]
  78. Fox HC, Hong KI, Siedlarz K, Sinha R. Enhanced sensitivity to stress and drug/alcohol craving in abstinent cocaine-dependent individuals compared to social drinkers. Neuropsychopharmacology. 2008;33:796–805. doi: 10.1038/sj.npp.1301470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Fox HC, Sinha R. Sex differences in drug-related stress-system changes: implications for treatment in substance-abusing women. Harv Rev Psychiatry. 2009;17:103–119. doi: 10.1080/10673220902899680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Franklin TR, Ehrman R, Lynch KG, Harper D, Sciortino N, O’Brien CP, Childress AR. Menstrual cycle phase at quit date predicts smoking status in an NRT treatment trial: a retrospective analysis. J Womens Health (Larchmt) 2008;17:287–292. doi: 10.1089/jwh.2007.0423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Freitag WJ, Adesso VJ. Mood effects of alcohol and expectancies across the menstrual cycle. Alcohol. 1993;10:291–298. doi: 10.1016/0741-8329(93)90008-c. [DOI] [PubMed] [Google Scholar]
  82. Friedman L, Gibbs TT, Farb DH. Gamma-aminobutyric acidA receptor regulation: chronic treatment with pregnanolone uncouples allosteric interactions between steroid and benzodiazepine recognition sites. Mol Pharmacol. 1993;44:191–197. [PubMed] [Google Scholar]
  83. Friedman LK, Gibbs TT, Farb DH. gamma-aminobutyric acidA receptor regulation: heterologous uncoupling of modulatory site interactions induced by chronic steroid, barbiturate, benzodiazepine, or GABA treatment in culture. Brain Res. 1996;707:100–109. doi: 10.1016/0006-8993(95)01226-5. [DOI] [PubMed] [Google Scholar]
  84. Frye CA. The neurosteroid 3 alpha, 5 apha-THP has antiseizure and possible neuroprotective effects in an animal model of epilepsy. Brain Res. 1995;696:113–120. doi: 10.1016/0006-8993(95)00793-p. [DOI] [PubMed] [Google Scholar]
  85. Frye CA. Progestins influence motivation, reward, conditioning, stress, and/or response to drugs of abuse. Pharmacol Biochem Behav. 2007;86:209–219. doi: 10.1016/j.pbb.2006.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Frye CA. The role of neurosteroids and non-genomic effects of progestins and androgens in mediating sexual receptivity of rodents. Brain Res Rev. 2001;37:201–222. doi: 10.1016/s0165-0173(01)00119-9. [DOI] [PubMed] [Google Scholar]
  87. Frye CA, Bayon LE. Seizure activity is increased in endocrine states characterized by decline in endogenous levels of the neurosteroid 3 alpha,5 alpha-THP. Neuroendocrinology. 1998;68:272–280. doi: 10.1159/000054375. [DOI] [PubMed] [Google Scholar]
  88. Frye CA, Murphy RE, Platek SM. Anti-sense oligonucleotides, for progestin receptors in the VMH and glutamic acid decarboxylase in the VTA, attenuate progesterone-induced lordosis in hamsters and rats. Behav Brain Res. 2000;115:55–64. doi: 10.1016/s0166-4328(00)00242-4. [DOI] [PubMed] [Google Scholar]
  89. Frye CA, Sumida K, Dudek BC, Harney JP, Lydon JP, O’Malley BW, Pfaff DW, Rhodes ME. Progesterone’s effects to reduce anxiety behavior of aged mice do not require actions via intracellular progestin receptors. Psychopharmacology (Berl) 2006;186:312–322. doi: 10.1007/s00213-006-0309-3. [DOI] [PubMed] [Google Scholar]
  90. Frye GD, Chapin RE, Vogel RA, Mailman RB, Kilts CD, Mueller RA, Breese GR. Effects of acute and chronic 1,3-butanediol treatment on central nervous system function: a comparison with ethanol. J Pharmacol Exp Ther. 1981;216:306–314. [PubMed] [Google Scholar]
  91. Fuchs RA, Evans KA, Mehta RH, Case JM, See RE. Influence of sex and estrous cyclicity on conditioned cue-induced reinstatement of cocaine-seeking behavior in rats. Psychopharmacology (Berl) 2005;179:662–672. doi: 10.1007/s00213-004-2080-7. [DOI] [PubMed] [Google Scholar]
  92. Gallop RJ, Crits-Christoph P, Ten Have TR, Barber JP, Frank A, Griffin ML, Thase ME. Differential transitions between cocaine use and abstinence for men and women. J Consult Clin Psychol. 2007;75:95–103. doi: 10.1037/0022-006X.75.1.95. [DOI] [PubMed] [Google Scholar]
  93. Garcia-Closas M, Herbstman J, Schiffman M, Glass A, Dorgan JF. Relationship between serum hormone concentrations, reproductive history, alcohol consumption and genetic polymorphisms in pre-menopausal women. Int J Cancer. 2002;102:172–178. doi: 10.1002/ijc.10651. [DOI] [PubMed] [Google Scholar]
  94. Gasior M, Carter RB, Witkin JM. Neuroactive steroids: potential therapeutic use in neurological and psychiatric disorders. Trends Pharmacol Sci. 1999;20:107–112. doi: 10.1016/s0165-6147(99)01318-8. [DOI] [PubMed] [Google Scholar]
  95. Gear RW, Miaskowski C, Gordon NC, Paul SM, Heller PH, Levine JD. Kappa-opioids produce significantly greater analgesia in women than in men. Nat Med. 1996;2:1248–1250. doi: 10.1038/nm1196-1248. [DOI] [PubMed] [Google Scholar]
  96. Genazzani AR, Petraglia F, Bernardi F, Casarosa E, Salvestroni C, Tonetti A, Nappi RE, Luisi S, Palumbo M, Purdy RH, Luisi M. Circulating levels of allopregnanolone in humans: gender, age, and endocrine influences. J Clin Endocrinol Metab. 1998;83:2099–2103. doi: 10.1210/jcem.83.6.4905. [DOI] [PubMed] [Google Scholar]
  97. George O, Ghozland S, Azar MR, Cottone P, Zorrilla EP, Parsons LH, O’Dell LE, Richardson HN, Koob GF. CRF-CRF1 system activation mediates withdrawal-induced increases in nicotine self-administration in nicotine-dependent rats. Proc Natl Acad Sci U S A. 2007;104:17198–17203. doi: 10.1073/pnas.0707585104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Gorin-Meyer RE, Wiren KM, Tanchuck MA, Long SL, Yoneyama N, Finn DA. Sex differences in the effect of finasteride on acute ethanol withdrawal severity in C57BL/6J and DBA/2J mice. Neuroscience. 2007;146:1302–1315. doi: 10.1016/j.neuroscience.2007.02.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Griffin ML, Mendelson JH, Mello NK, Lex BW. Marihuana use across the menstrual cycle. Drug Alcohol Depend. 1986;18:213–224. doi: 10.1016/0376-8716(86)90053-0. [DOI] [PubMed] [Google Scholar]
  100. Grobin AC, Matthews DB, Devaud LL, Morrow AL. The role of GABA(A) receptors in the acute and chronic effects of ethanol. Psychopharmacology (Berl) 1998;139:2–19. doi: 10.1007/s002130050685. [DOI] [PubMed] [Google Scholar]
  101. Grobin AC, VanDoren MJ, Porrino LJ, Morrow AL. Cortical 3 alpha-hydroxy-5 alpha-pregnan-20-one levels after acute administration of Delta 9-tetrahydrocannabinol, cocaine and morphine. Psychopharmacology (Berl) 2005;179:544–550. doi: 10.1007/s00213-004-2084-3. [DOI] [PubMed] [Google Scholar]
  102. Guerra-Araiza C, Cerbon MA, Morimoto S, Camacho-Arroyo I. Progesterone receptor isoforms expression pattern in the rat brain during the estrous cycle. Life Sci. 2000;66:1743–1752. doi: 10.1016/s0024-3205(00)00497-5. [DOI] [PubMed] [Google Scholar]
  103. Hamilton KR, Berger SS, Perry ME, Grunberg NE. Behavioral effects of nicotine withdrawal in adult male and female rats. Pharmacol Biochem Behav. 2009;92:51–59. doi: 10.1016/j.pbb.2008.10.010. [DOI] [PubMed] [Google Scholar]
  104. Hansen ST, Mark GP. The nicotinic acetylcholine receptor antagonist mecamylamine prevents escalation of cocaine self-administration in rats with extended daily access. Psychopharmacology (Berl) 2007;194:53–61. doi: 10.1007/s00213-007-0822-z. [DOI] [PubMed] [Google Scholar]
  105. Harris GC, Aston-Jones G. Enhanced morphine preference following prolonged abstinence: association with increased Fos expression in the extended amygdala. Neuropsychopharmacology. 2003;28:292–299. doi: 10.1038/sj.npp.1300037. [DOI] [PubMed] [Google Scholar]
  106. Harvey SM, Beckman LJ. Cyclic fluctuation in alcohol consumption among female social drinkers. Alcohol Clin Exp Res. 1985;9:465–467. doi: 10.1111/j.1530-0277.1985.tb05584.x. [DOI] [PubMed] [Google Scholar]
  107. Hay WM, Nathan PE, Heermans HW, Frankenstein W. Menstrual cycle, tolerance and blood alcohol level discrimination ability. Addict Behav. 1984;9:67–77. doi: 10.1016/0306-4603(84)90008-x. [DOI] [PubMed] [Google Scholar]
  108. Hecht GS, Spear NE, Spear LP. Changes in progressive ratio responding for intravenous cocaine throughout the reproductive process in female rats. Dev Psychobiol. 1999;35:136–145. [PubMed] [Google Scholar]
  109. Hill M, Popov P, Havlikova H, Kancheva L, Vrbikova J, Kancheva R, Pouzar V, Cerny I, Starka L. Altered profiles of serum neuroactive steroids in premenopausal women treated for alcohol addiction. Steroids. 2005;70:515–524. doi: 10.1016/j.steroids.2005.02.013. [DOI] [PubMed] [Google Scholar]
  110. Hirani K, Khisti RT, Chopde CT. Behavioral action of ethanol in Porsolt’s forced swim test: modulation by 3 alpha-hydroxy-5 alpha-pregnan-20-one. Neuropharmacology. 2002;43:1339–1350. doi: 10.1016/s0028-3908(02)00330-1. [DOI] [PubMed] [Google Scholar]
  111. Hiroi R, Neumaier JF. Differential effects of ovarian steroids on anxiety versus fear as measured by open field test and fear-potentiated startle. Behav Brain Res. 2006;166:93–100. doi: 10.1016/j.bbr.2005.07.021. [DOI] [PubMed] [Google Scholar]
  112. Holdstock L, de Wit H. Effects of ethanol at four phases of the menstrual cycle. Psychopharmacology (Berl) 2000;150:374–382. doi: 10.1007/s002130000461. [DOI] [PubMed] [Google Scholar]
  113. Hu M, Watson CJ, Kennedy RT, Becker JB. Estradiol attenuates the K+-induced increase in extracellular GABA in rat striatum. Synapse. 2006;59:122–124. doi: 10.1002/syn.20221. [DOI] [PubMed] [Google Scholar]
  114. Hutcheson DM, Everitt BJ, Robbins TW, Dickinson A. The role of withdrawal in heroin addiction: enhances reward or promotes avoidance? Nat Neurosci. 2001;4:943–947. doi: 10.1038/nn0901-943. [DOI] [PubMed] [Google Scholar]
  115. Ignjatova L, Raleva M. Gender difference in the treatment outcome of patients served in the mixed-gender program. Bratisl Lek Listy. 2009;110:285–289. [PubMed] [Google Scholar]
  116. Imperato A, Puglisi-Allegra S, Casolini P, Angelucci L. Changes in brain dopamine and acetylcholine release during and following stress are independent of the pituitary-adrenocortical axis. Brain Res. 1991;538:111–117. doi: 10.1016/0006-8993(91)90384-8. [DOI] [PubMed] [Google Scholar]
  117. Inglis FM, Moghaddam B. Dopaminergic innervation of the amygdala is highly responsive to stress. J Neurochem. 1999;72:1088–1094. doi: 10.1046/j.1471-4159.1999.0721088.x. [DOI] [PubMed] [Google Scholar]
  118. Jackson LR, Robinson TE, Becker JB. Sex differences and hormonal influences on acquisition of cocaine self-administration in rats. Neuropsychopharmacology. 2006;31:129–138. doi: 10.1038/sj.npp.1300778. [DOI] [PubMed] [Google Scholar]
  119. Janak PH, Redfern JE, Samson HH. The reinforcing effects of ethanol are altered by the endogenous neurosteroid, allopregnanolone. Alcohol Clin Exp Res. 1998;22:1106–1112. [PubMed] [Google Scholar]
  120. Jaworska-Feil L, Budziszewska B, Leskiewicz M, Lason W. Opposite effects of inhibitory and excitatory neurosteroids on [3H]dopamine release from rat nucleus accumbens. Pol J Pharmacol. 1998;50:449–452. [PubMed] [Google Scholar]
  121. Johanson CE, Barrett JE. The discriminative stimulus effects of cocaine in pigeons. J Pharmacol Exp Ther. 1993;267:1–8. [PubMed] [Google Scholar]
  122. Junghanns K, Backhaus J, Tietz U, Lange W, Bernzen J, Wetterling T, Rink L, Driessen M. Impaired serum cortisol stress response is a predictor of early relapse. Alcohol Alcohol. 2003;38:189–193. doi: 10.1093/alcalc/agg052. [DOI] [PubMed] [Google Scholar]
  123. Justice AJ, de Wit H. Acute effects of d-amphetamine during the follicular and luteal phases of the menstrual cycle in women. Psychopharmacology (Berl) 1999;145:67–75. doi: 10.1007/s002130051033. [DOI] [PubMed] [Google Scholar]
  124. Justice AJ, De Wit H. Acute effects of d-amphetamine during the early and late follicular phases of the menstrual cycle in women. Pharmacol Biochem Behav. 2000;66:509–515. doi: 10.1016/s0091-3057(00)00218-5. [DOI] [PubMed] [Google Scholar]
  125. Kabbadj K, el-Etr M, Baulieu EE, Robel P. Pregnenolone metabolism in rodent embryonic neurons and astrocytes. Glia. 1993;7:170–175. doi: 10.1002/glia.440070206. [DOI] [PubMed] [Google Scholar]
  126. Kalivas PW. Neurobiology of cocaine addiction: implications for new pharmacotherapy. Am J Addict. 2007;16:71–78. doi: 10.1080/10550490601184142. [DOI] [PubMed] [Google Scholar]
  127. Kamien JB, Bickel WK, Hughes JR, Higgins ST, Smith BJ. Drug discrimination by humans compared to nonhumans: current status and future directions. Psychopharmacology (Berl) 1993;111:259–270. doi: 10.1007/BF02244940. [DOI] [PubMed] [Google Scholar]
  128. Kamien JB, Woolverton WL. A pharmacological analysis of the discriminative stimulus properties of d-amphetamine in rhesus monkeys. J Pharmacol Exp Ther. 1989;248:938–946. [PubMed] [Google Scholar]
  129. Kaminski RM, Gasior M, Carter RB, Witkin JM. Protective efficacy of neuroactive steroids against cocaine kindled-seizures in mice. Eur J Pharmacol. 2003;474:217–222. doi: 10.1016/s0014-2999(03)02086-7. [DOI] [PubMed] [Google Scholar]
  130. Karila L, Gorelick D, Weinstein A, Noble F, Benyamina A, Coscas S, Blecha L, Lowenstein W, Martinot JL, Reynaud M, Lepine JP. New treatments for cocaine dependence: a focused review. Int J Neuropsychopharmacol. 2008;11:425–438. doi: 10.1017/S1461145707008097. [DOI] [PubMed] [Google Scholar]
  131. Kerstetter KA, Aguilar VR, Parrish AB, Kippin TE. Protracted time-dependent increases in cocaine-seeking behavior during cocaine withdrawal in female relative to male rats. Psychopharmacology (Berl) 2008;198:63–75. doi: 10.1007/s00213-008-1089-8. [DOI] [PubMed] [Google Scholar]
  132. Khantzian EJ. The self-medication hypothesis of addctive disorders: Focus on heroin and cocaine dependence. Am J Psychitary. 1985;142:1259–1264. doi: 10.1176/ajp.142.11.1259. [DOI] [PubMed] [Google Scholar]
  133. Kippin TE, Fuchs RA, Mehta RH, Case JM, Parker MP, Bimonte-Nelson HA, See RE. Potentiation of cocaine-primed reinstatement of drug seeking in female rats during estrus. Psychopharmacology (Berl) 2005;182:245–252. doi: 10.1007/s00213-005-0071-y. [DOI] [PubMed] [Google Scholar]
  134. Kiyatkin EA, Stein EA. Fluctuations in nucleus accumbens dopamine during cocaine self-administration behavior: an in vivo electrochemical study. Neuroscience. 1995;64:599–617. doi: 10.1016/0306-4522(94)00436-9. [DOI] [PubMed] [Google Scholar]
  135. Knapp DJ, Overstreet DH, Moy SS, Breese GR. SB242084, flumazenil, and CRA1000 block ethanol withdrawal-induced anxiety in rats. Alcohol. 2004;32:101–111. doi: 10.1016/j.alcohol.2003.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Koob G, Kreek MJ. Stress, dysregulation of drug reward pathways, and the transition to drug dependence. Am J Psychiatry. 2007;164:1149–1159. doi: 10.1176/appi.ajp.2007.05030503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Koob GF. Neurobiological substrates for the dark side of compulsivity in addiction. Neuropharmacology. 2009;56(Suppl 1):18–31. doi: 10.1016/j.neuropharm.2008.07.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Koob GF, Le Moal M. Plasticity of reward neurocircuitry and the ‘dark side’ of drug addiction. Nat Neurosci. 2005;8:1442–1444. doi: 10.1038/nn1105-1442. [DOI] [PubMed] [Google Scholar]
  139. Koob GF, Le Moal M. Review. Neurobiological mechanisms for opponent motivational processes in addiction. Philos Trans R Soc Lond B Biol Sci. 2008;363:3113–3123. doi: 10.1098/rstb.2008.0094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Koob GF, Volkow ND. Neurocircuitry of Addiction. Neuropsychopharmacology. Kushner, S.A., Dewey, S.L., Kornetsky, C., 1999. The irreversible gamma-aminobutyric acid (GABA) transaminase inhibitor gamma-vinyl-GABA blocks cocaine self-administration in rats. J Pharmacol Exp Ther. 2009;290:797–802. [PubMed] [Google Scholar]
  141. Laconi MR, Casteller G, Gargiulo PA, Bregonzio C, Cabrera RJ. The anxiolytic effect of allopregnanolone is associated with gonadal hormonal status in female rats. Eur J Pharmacol. 2001;417:111–116. doi: 10.1016/s0014-2999(01)00865-2. [DOI] [PubMed] [Google Scholar]
  142. Laconi MR, Reggiani PC, Penissi A, Yunes R, Cabrera RJ. Allopregnanolone modulates striatal dopamingergic activity of rats under different gonadal hormones conditions. Neurol Res. 2007;29:622–627. doi: 10.1179/016164107X166281. [DOI] [PubMed] [Google Scholar]
  143. Lambert JJ, Belelli D, Hill-Venning C, Peters JA. Neurosteroids and GABAA receptor function. Trends Pharmacol Sci. 1995;16:295–303. doi: 10.1016/s0165-6147(00)89058-6. [DOI] [PubMed] [Google Scholar]
  144. Lange RA. Cardiovascular complications of cocaine use. N Engl J Med. 2001;345:351–358. doi: 10.1056/NEJM200108023450507. [DOI] [PubMed] [Google Scholar]
  145. Larson EB, Anker JJ, Gliddon LA, Fons KS, Carroll ME. Effects of estrogen and progesterone on the escalation of cocaine self-administration in female rats during extended access. Exp Clin Psychopharmacol. 2007;15:461–471. doi: 10.1037/1064-1297.15.5.461. [DOI] [PubMed] [Google Scholar]
  146. Larson EB, Carroll ME. Estrogen receptor beta, but not alpha, mediates estrogen’s effect on cocaine-induced reinstatement of extinguished cocaine-seeking behavior in ovariectomized female rats. Neuropsychopharmacology. 2007;32:1334–1345. doi: 10.1038/sj.npp.1301249. [DOI] [PubMed] [Google Scholar]
  147. Larson EB, Roth ME, Anker JJ, Carroll ME. Effect of short- vs. long-term estrogen on reinstatement of cocaine-seeking behavior in female rats. Pharmacol Biochem Behav. 2005;82:98–108. doi: 10.1016/j.pbb.2005.07.015. [DOI] [PubMed] [Google Scholar]
  148. Lee S, Schmidt ED, Tilders FJ, Rivier C. Effect of repeated exposure to alcohol on the response of the hypothalamic-pituitary-adrenal axis of the rat: I. Role of changes in hypothalamic neuronal activity. Alcohol Clin Exp Res. 2001;25:98–105. [PubMed] [Google Scholar]
  149. Leskiewicz M, Budziszewska B, Jaworska-Feil L, Kubera M, Basta-Kaim A, Lason W. Inhibitory effect of some neuroactive steroids on cocaine-induced kindling in mice. Pol J Pharmacol. 2003;55:1131–1136. [PubMed] [Google Scholar]
  150. Lex BW, Mendelson JH, Bavli S, Harvey K, Mello NK. Effects of acute marijuana smoking on pulse rate and mood states in women. Psychopharmacology (Berl) 1984;84:178–187. doi: 10.1007/BF00427443. [DOI] [PubMed] [Google Scholar]
  151. Ling W, Shoptaw S, Majewska D. Baclofen as a cocaine anti-craving medication: a preliminary clinical study. Neuropsychopharmacology. 1998;18:403–404. doi: 10.1016/S0893-133X(97)00128-0. [DOI] [PubMed] [Google Scholar]
  152. Lukas SE, Sholar M, Lundahl LH, Lamas X, Kouri E, Wines JD, Kragie L, Mendelson JH. Sex differences in plasma cocaine levels and subjective effects after acute cocaine administration in human volunteers. Psychopharmacology (Berl) 1996;125:346–354. doi: 10.1007/BF02246017. [DOI] [PubMed] [Google Scholar]
  153. Luntz-Leybman V, Freund RK, Collins AC. 5 alpha-Pregnan-3 alpha-ol-20-one blocks nicotine-induced seizures and enhances paired-pulse inhibition. Eur J Pharmacol. 1990;185:239–242. doi: 10.1016/0014-2999(90)90648-p. [DOI] [PubMed] [Google Scholar]
  154. Lynch WJ. Acquisition and maintenance of cocaine self-administration in adolescent rats: effects of sex and gonadal hormones. Psychopharmacology (Berl) 2008;197:237–246. doi: 10.1007/s00213-007-1028-0. [DOI] [PubMed] [Google Scholar]
  155. Lynch WJ. Sex and ovarian hormones influence vulnerability and motivation for nicotine during adolescence in rats. Pharmacol Biochem Behav. 2009;94:43–50. doi: 10.1016/j.pbb.2009.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Lynch WJ, Carroll ME. Sex differences in the acquisition of intravenously self-administered cocaine and heroin in rats. Psychopharmacology (Berl) 1999;144:77–82. doi: 10.1007/s002130050979. [DOI] [PubMed] [Google Scholar]
  157. Lynch WJ, Carroll ME. Reinstatement of cocaine self-administration in rats: sex differences. Psychopharmacology (Berl) 2000;148:196–200. doi: 10.1007/s002130050042. [DOI] [PubMed] [Google Scholar]
  158. Lynch WJ, Roth ME, Mickelberg JL, Carroll ME. Role of estrogen in the acquisition of intravenously self-administered cocaine in female rats. Pharmacol Biochem Behav. 2001;68:641–646. doi: 10.1016/s0091-3057(01)00455-5. [DOI] [PubMed] [Google Scholar]
  159. Mann K, Ackermann K, Croissant B, Mundle G, Nakovics H, Diehl A. Neuroimaging of gender differences in alcohol dependence: are women more vulnerable? Alcohol Clin Exp Res. 2005;29:896–901. doi: 10.1097/01.alc.0000164376.69978.6b. [DOI] [PubMed] [Google Scholar]
  160. Mantsch JR, Goeders NE. Generalization of a restraint-induced discriminative stimulus to cocaine in rats. Psychopharmacology (Berl) 1998;135:423–426. doi: 10.1007/s002130050531. [DOI] [PubMed] [Google Scholar]
  161. Marks JL, Pomerleau CS, Pomerleau OF. Effects of menstrual phase on reactivity to nicotine. Addict Behav. 1999;24:127–134. doi: 10.1016/s0306-4603(98)00033-1. [DOI] [PubMed] [Google Scholar]
  162. Martin-Garcia E, Darbra S, Pallares M. Intrahippocampal allopregnanolone decreases voluntary chronic alcohol consumption in non-selected rats. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31:823–831. doi: 10.1016/j.pnpbp.2007.01.022. [DOI] [PubMed] [Google Scholar]
  163. Martin-Garcia E, Pallares M. Effects of intrahippocampal nicotine and neurosteroid administration on withdrawal in voluntary and chronic alcohol-drinking rats. Alcohol Clin Exp Res. 2005;29:1654–1663. doi: 10.1097/01.alc.0000179206.01621.4b. [DOI] [PubMed] [Google Scholar]
  164. Maurice T, Martin-Fardon R, Romieu P, Matsumoto RR. Selective sigma1 (o’1) receptor antagonists as a new promising strategy to prevent cocaine-indcued behaviors and toxicity. Neurosci Biobehav Rev. 2002;26:499–527. doi: 10.1016/s0149-7634(02)00017-9. [DOI] [PubMed] [Google Scholar]
  165. McKeon A, Frye MA, Delanty N. The alcohol withdrawal syndrome. J Neurol Neurosurg Psychiatry. 2008;79:854–862. doi: 10.1136/jnnp.2007.128322. [DOI] [PubMed] [Google Scholar]
  166. Mello NK, Bree MP, Mendelson JH. Alcohol and food self-administration by female Macaque monkeys as a function of menstrual cycle phase. Physiol Behav. 1986;36:959–966. doi: 10.1016/0031-9384(86)90460-9. [DOI] [PubMed] [Google Scholar]
  167. Mello NK, Knudson IM, Mendelson JH. Sex and menstrual cycle effects on progressive ratio measures of cocaine self-administration in cynomolgus monkeys. Neuropsychopharmacology. 2007;32:1956–1966. doi: 10.1038/sj.npp.1301314. [DOI] [PubMed] [Google Scholar]
  168. Mello NK, Mendelson JH, Palmieri SL. Cigarette smoking by women: interactions with alcohol use. Psychopharmacology (Berl) 1987;93:8–15. doi: 10.1007/BF02439579. [DOI] [PubMed] [Google Scholar]
  169. Mellon SH. Synthesis, Enzyme Localization, and Regulation of Neurosteroids. In: Smith Sheryl S, editor. Neurosteroid Effects in the Central Nervous System: The Role of the GABAA Receptor. 2004. [Google Scholar]
  170. Mendelson JH, Mello NK, Sholar MB, Siegel AJ, Kaufman MJ, Levin JM, Renshaw PF, Cohen BM. Cocaine pharmacokinetics in men and in women during the follicular and luteal phases of the menstrual cycle. Neuropsychopharmacology. 1999;21:294–303. doi: 10.1016/S0893-133X(99)00020-2. [DOI] [PubMed] [Google Scholar]
  171. Mermelstein PG, Becker JB, Surmeier DJ. Estradiol reduces calcium currents in rat neostriatal neurons via a membrane receptor. J Neurosci. 1996;16:595–604. doi: 10.1523/JNEUROSCI.16-02-00595.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Morgan D, Brebner K, Lynch WJ, Roberts DC. Increases in the reinforcing efficacy of cocaine after particular histories of reinforcement. Behav Pharmacol. 2002;13:389–396. doi: 10.1097/00008877-200209000-00012. [DOI] [PubMed] [Google Scholar]
  173. Morrow AL, VanDoren MJ, Penland SN, Matthews DB. The role of GABAergic neuroactive steroids in ethanol action, tolerance and dependence. Brain Res Brain Res Rev. 2001;37:98–109. doi: 10.1016/s0165-0173(01)00127-8. [DOI] [PubMed] [Google Scholar]
  174. Morville R, Reyneir DF, Barrat J. Biodispnibilite de la progesterone naturelle administree par voie orale. Mesure des concentrations du steroide dans le plasma, l’endometre et le tissu mammaire. J Gynecol Obstet Biol Reprod (Paris) 1982;11:355–363. [PubMed] [Google Scholar]
  175. Naftolin F, Brown-Grant K, Corker CS. Plasma and pituitary luteinizing hormone and peripheral plasma oestradiol concentrations in the normal oestrous cycle of the rat and after experimental manipulation of the cycle. J Endocrinol. 1972;53:17–30. doi: 10.1677/joe.0.0530017. [DOI] [PubMed] [Google Scholar]
  176. Newman JL, Thorne JJ, Batulis DK, Carroll ME. Effects of menstrual cycle phase on the reinforcing effects of phencyclidine (PCP) in rhesus monkeys. Pharmacol Biochem Behav. 2006;85:584–591. doi: 10.1016/j.pbb.2006.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Niyomchai T, Jenab S, Festa ED, Akhavan A, Quinones-Jenab V. Effects of short-and long-term estrogen and progesterone replacement on behavioral responses and c-fos mRNA levels in female rats after acute cocaine administration. Brain Res. 2006;1126:193–199. doi: 10.1016/j.brainres.2006.07.099. [DOI] [PubMed] [Google Scholar]
  178. Niyomchai T, Russo SJ, Festa ED, Akhavan A, Jenab S, Quinones-Jenab V. Progesterone inhibits behavioral responses and estrogen increases corticosterone levels after acute cocaine administration. Pharmacol Biochem Behav. 2005;80:603–610. doi: 10.1016/j.pbb.2005.01.010. [DOI] [PubMed] [Google Scholar]
  179. Niyomchai T, Weirstall K, Jenab S, Quinones-Jenab V. Coadministration of estrogen and progesterone differentially affects locomotor responses to cocaine in rats. Ethn Dis. 2008;18:S2–51–53. [PubMed] [Google Scholar]
  180. Nyberg S, Wahlstrom G, Backstrom T, Sundstrom Poromaa I. Altered sensitivity to alcohol in the late luteal phase among patients with premenstrual dysphoric disorder. Psychoneuroendocrinology. 2004;29:767–777. doi: 10.1016/S0306-4530(03)00121-5. [DOI] [PubMed] [Google Scholar]
  181. Overstreet DH, Knapp DJ, Breese GR. Modulation of multiple ethanol withdrawal-induced anxiety-like behavior by CRF and CRF1 receptors. Pharmacol Biochem Behav. 2004;77:405–413. doi: 10.1016/j.pbb.2003.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Owens MJ, Ritchie JC, Nemeroff CB. 5 alpha-pregnane-3 alpha, 21-diol-20-one (THDOC) attenuates mild stress-induced increases in plasma corticosterone via a non-glucocorticoid mechanism: comparison with alprazolam. Brain Res. 1992;573:353–355. doi: 10.1016/0006-8993(92)90788-b. [DOI] [PubMed] [Google Scholar]
  183. Papadeas S, Grobin AC, Morrow AL. Chronic ethanol consumption differentially alters GABA(A) receptor alpha1 and alpha4 subunit peptide expression and GABA(A) receptor-mediated 36 Cl(−) uptake in mesocorticolimbic regions of rat brain. Alcohol Clin Exp Res. 2001;25:1270–1275. [PubMed] [Google Scholar]
  184. Parsons B, Rainbow TC, MacLusky NJ, McEwen BS. Progestin receptor levels in rat hypothalamic and limbic nuclei. J Neurosci. 1982;10:1446–1452. doi: 10.1523/JNEUROSCI.02-10-01446.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Paris JJ, Frye CA. Estrous cycle, pregnancy, and parity enhance performance of rats in object recognition or object placement tasks. Reproduction. 2008;136:105–115. doi: 10.1530/REP-07-0512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Patchev VK, Hayashi S, Orikasa C, Almeida OF. Implications of estrogen-dependent brain organization for gender differences in hypothalamo-pituitary-adrenal regulation. FASEB J. 1995;9:419–423. doi: 10.1096/fasebj.9.5.7896013. [DOI] [PubMed] [Google Scholar]
  187. Patchev VK, Shoaib M, Holsboer F, Almeida OF. The neurosteroid tetrahydroprogesterone counteracts corticotropin-releasing hormone-induced anxiety and alters the release and gene expression of corticotropin-releasing hormone in the rat hypothalamus. Neuroscience. 1994;62:265–271. doi: 10.1016/0306-4522(94)90330-1. [DOI] [PubMed] [Google Scholar]
  188. Pearson Murphy BE, Allison CM. Determination of progesterone and some of its neuroactive ring A-reduced metabolites in human serum. J Steroid Biochem Mol Biol. 2000;74:137–142. doi: 10.1016/s0960-0760(00)00098-4. [DOI] [PubMed] [Google Scholar]
  189. Peng XQ, Li X, Gilbert JG, Pak AC, Ashby CR, Jr, Brodie JD, Dewey SL, Gardner EL, Xi ZX. Gamma-vinyl GABA inhibits cocaine-triggered reinstatement of drug-seeking behavior in rats by a non-dopaminergic mechanism. Drug Alcohol Depend. 2008;97:216–225. doi: 10.1016/j.drugalcdep.2007.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Peris J, Decambre N, Coleman-Hardee ML, Simpkins JW. Estradiol enhances behavioral sensitization to cocaine and amphetamine-stimulated striatal [3H]dopamine release. Brain Res. 1991;566:255–264. doi: 10.1016/0006-8993(91)91706-7. [DOI] [PubMed] [Google Scholar]
  191. Perrotti LI, Russo SJ, Fletcher H, Chin J, Webb T, Jenab S, Quinones-Jenab V. Ovarian hormones modulate cocaine-induced locomotor and stereotypic activity. Ann N Y Acad Sci. 2001;937:202–216. doi: 10.1111/j.1749-6632.2001.tb03566.x. [DOI] [PubMed] [Google Scholar]
  192. Philpott CM, El-Alami M, Murty GE. The effect of the steroid sex hormones on the nasal airway during the normal menstrual cycle. Clin Otolaryngol Allied Sci. 2004;29:138–142. doi: 10.1111/j.1365-2273.2004.00801.x. [DOI] [PubMed] [Google Scholar]
  193. Piasecki TM, Jorenby DE, Smith SS, Fiore MC, Baker TB. Smoking withdrawal dynamics: I. Abstinence distress in lapsers and abstainers. J Abnorm Psychol. 2003;112:3–13. [PubMed] [Google Scholar]
  194. Pierce RC, Kumaresan V. The mesolimbic dopamine system: the final common pathway for the reinforcing effect of drugs of abuse? Neurosci Biobehav Rev. 2006;30:215–238. doi: 10.1016/j.neubiorev.2005.04.016. [DOI] [PubMed] [Google Scholar]
  195. Pierucci-Lagha A, Covault J, Feinn R, Nellissery M, Hernandez-Avila C, Oncken C, Morrow AL, Kranzler HR. GABRA2 alleles moderate the subjective effects of alcohol, which are attenuated by finasteride. Neuropsychopharmacology. 2005;30:1193–1203. doi: 10.1038/sj.npp.1300688. [DOI] [PubMed] [Google Scholar]
  196. Pomerleau CS, Cole PA, Lumley MA, Marks JL, Pomerleau OF. Effects of menstrual phase on nicotine, alcohol, and caffeine intake in smokers. J Subst Abuse. 1994;6:227–234. doi: 10.1016/s0899-3289(94)90253-4. [DOI] [PubMed] [Google Scholar]
  197. Pomerleau CS, Garcia AW, Pomerleau OF, Cameron OG. The effects of menstrual phase and nicotine abstinence on nicotine intake and on biochemical and subjective measures in women smokers: a preliminary report. Psychoneuroendocrinology. 1992;17:627–638. doi: 10.1016/0306-4530(92)90021-x. [DOI] [PubMed] [Google Scholar]
  198. Pomerleau CS, Mehringer AM, Marks JL, Downey KK, Pomerleau OF. Effects of menstrual phase and smoking abstinence in smokers with and without a history of major depressive disorder. Addict Behav. 2000;25:483–497. doi: 10.1016/s0306-4603(99)00075-1. [DOI] [PubMed] [Google Scholar]
  199. Porcu P, Sogliano C, Cinus M, Purdy RH, Biggio G, Concas A. Nicotine-induced changes in cerebrocortical neuroactive steroids and plasma corticosterone concentrations in the rat. Pharmacol Biochem Behav. 2003;74:683–690. doi: 10.1016/s0091-3057(02)01065-1. [DOI] [PubMed] [Google Scholar]
  200. Pozzi M, Roccatagliata D, Sterzi R. Drug abuse and intracranial hemorrhage. Neurol Sci. 2008;29:269–270. doi: 10.1007/s10072-008-0960-z. [DOI] [PubMed] [Google Scholar]
  201. Purdy RH, Morrow AL, Moore PH, Jr, Paul SM. Stress-induced elevations of gamma-aminobutyric acid type A receptor-active steroids in the rat brain. Proc Natl Acad Sci U S A. 1991;88:4553–4557. doi: 10.1073/pnas.88.10.4553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Quinones-Jenab V, Minerly AC, Niyomchia T, Akahvan A, Jenab S, Frye C. Progesterone and allopregnanolone are induced by cocaine in serum and brain tissues of male and female rats. Pharmacol Biochem Behav. 2008;89:292–297. doi: 10.1016/j.pbb.2007.12.024. [DOI] [PubMed] [Google Scholar]
  203. Quinones-Jenab V, Perrotti LI, Ho A, Jenab S, Schlussman SD, Franck J, Kreek MJ. Cocaine affects progesterone plasma levels in female rats. Pharmacol Biochem Behav. 2000a;66:449–453. doi: 10.1016/s0091-3057(00)00213-6. [DOI] [PubMed] [Google Scholar]
  204. Quinones-Jenab V, Perrotti LI, Mc Monagle J, Ho A, Kreek MJ. Ovarian hormone replacement affects cocaine-induced behaviors in ovariectomized female rats. Pharmacol Biochem Behav. 2000b;67:417–422. doi: 10.1016/s0091-3057(00)00381-6. [DOI] [PubMed] [Google Scholar]
  205. Quinton MS, Gerak LR, Moerschbaecher JM, Winsauer PJ. Effects of pregnanolone in rats discriminating cocaine. Pharmacol Biochem Behav. 2006;85:385–392. doi: 10.1016/j.pbb.2006.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Randall CL, Roberts JS, Del Boca FK, Carroll KM, Connors GJ, Mattson ME. Telescoping of landmark events associated with drinking: a gender comparison. J Stud Alcohol. 1999;60:252–260. doi: 10.15288/jsa.1999.60.252. [DOI] [PubMed] [Google Scholar]
  207. Reddy DS, Kulkarni SK. Chronic neurosteroid treatment prevents the development of morphine tolerance and attenuates abstinence behavior in mice. Eur J Pharmacol. 1997a;337:19–25. doi: 10.1016/s0014-2999(97)01294-6. [DOI] [PubMed] [Google Scholar]
  208. Reddy DS, Kulkarni SK. Neurosteroid coadministration prevents development of tolerance and augments recovery from benzodiazepine withdrawal anxiety and hyperactivity in mice. Methods Find Exp Clin Pharmacol. 1997b;19:395–405. [PubMed] [Google Scholar]
  209. Redei E, Freeman EW. Daily plasma estradiol and progesterone levels over the menstrual cycle and their relation to premenstrual symptoms. Psychoneuroendocrinology. 1995;20:259–267. doi: 10.1016/0306-4530(94)00057-h. [DOI] [PubMed] [Google Scholar]
  210. Reis AD, Castro LA, Faria R, Laranjeira R. Craving decrease with topiramate in outpatient treatment for cocaine dependence: an open label trial. Rev Bras Psiquiatr. 2008;30:132–135. doi: 10.1590/s1516-44462008005000012. [DOI] [PubMed] [Google Scholar]
  211. Robbins SJ, Ehrman RN, Childress AR, O’Brien CP. Comparing levels of cocaine cue reactivity in male and female outpatients. Drug Alcohol Depend. 1999;53:223–230. doi: 10.1016/s0376-8716(98)00135-5. [DOI] [PubMed] [Google Scholar]
  212. Roberts AJ, Smith AD, Weiss F, Rivier C, Koob GF. Estrous cycle effects on operant responding for ethanol in female rats. Alcohol Clin Exp Res. 1998;22:1564–1569. [PubMed] [Google Scholar]
  213. Roberts DC, Bennett SA, Vickers GJ. The estrous cycle affects cocaine self-administration on a progressive ratio schedule in rats. Psychopharmacology (Berl) 1989;98:408–411. doi: 10.1007/BF00451696. [DOI] [PubMed] [Google Scholar]
  214. Rodriguez de Fonseca F, Carrera MR, Navarro M, Koob GF, Weiss F. Activation of corticotropin-releasing factor in the limbic system during cannabinoid withdrawal. Science. 1997;276:2050–2054. doi: 10.1126/science.276.5321.2050. [DOI] [PubMed] [Google Scholar]
  215. Rodriguez-Sierra JF, Howard JL, Pollard GT, Hendricks SE. Effect of ovarian hormones on conflict behavior. Psychoneuroendocrinology. 1984;9:293–300. doi: 10.1016/0306-4530(84)90008-8. [DOI] [PubMed] [Google Scholar]
  216. Romeo E, Brancati A, De Lorenzo A, Fucci P, Furnari C, Pompili E, Sasso GF, Spalletta G, Troisi A, Pasini A. Marked decrease of plasma neuroactive steroids during alcohol withdrawal. Clin Neuropharmacol. 1996;19:366–369. doi: 10.1097/00002826-199619040-00011. [DOI] [PubMed] [Google Scholar]
  217. Romieu P, Martin-Fardon R, Bowen WD, Maurice T. Sigma 1 receptor-related neuroactive steroids modulate cocaine-induced reward. J Neurosci. 2003;23:3572–3576. doi: 10.1523/JNEUROSCI.23-09-03572.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Romieu P, Meunier JC, Garcia D, Zozime N, Martin-Fardon R, Bowen WD, Maurice T. The sigma1 (sigma1) receptor activation is a key step for the reactivation of cocaine conditioned place preference by drug priming. Psychopharmacology. 2004;175:154–162. doi: 10.1007/s00213-004-1814-x. [DOI] [PubMed] [Google Scholar]
  219. Romieu P, Phan V, Martin-Fardon R, Maurice T. Involvment of sigma1 receptor in cocaine-induced conditioned place preference: Possible dependence on dopamine uptake blockade. Neuropsychopharmacology. 2002;26:444–445. doi: 10.1016/S0893-133X(01)00391-8. [DOI] [PubMed] [Google Scholar]
  220. Roth ME, Carroll ME. Sex differences in the escalation of intravenous cocaine intake following long- or short-access to cocaine self-administration. Pharmacol Biochem Behav. 2004;78:199–207. doi: 10.1016/j.pbb.2004.03.018. [DOI] [PubMed] [Google Scholar]
  221. Roth ME, Casimir AG, Carroll ME. Influence of estrogen in the acquisition of intravenously self-administered heroin in female rats. Pharmacol Biochem Behav. 2002;72:313–318. doi: 10.1016/s0091-3057(01)00777-8. [DOI] [PubMed] [Google Scholar]
  222. Rothman RB, Baumann MH, Dersch CM, Romero DV, Rice KC, Carroll FI, Partilla JS. Amphetamine-type central nervous system stimulants release norepinephrine more potently than they release dopamine and serotonin. Synapse. 2001;39:32–41. doi: 10.1002/1098-2396(20010101)39:1<32::AID-SYN5>3.0.CO;2-3. [DOI] [PubMed] [Google Scholar]
  223. Rouge-Pont F, Mayo W, Marinelli M, Gingras M, Le Moal M, Piazza PV. The neurosteroid allopregnanolone increases dopamine release and dopaminergic response to morphine in the rat nucleus accumbens. Eur J Neurosci. 2002;16:169–173. doi: 10.1046/j.1460-9568.2002.02084.x. [DOI] [PubMed] [Google Scholar]
  224. Russo SJ, Festa ED, Fabian SJ, Gazi FM, Kraish M, Jenab S, Quinones-Jenab V. Gonadal hormones differentially modulate cocaine-induced conditioned place preference in male and female rats. Neuroscience. 2003;120:523–533. doi: 10.1016/s0306-4522(03)00317-8. [DOI] [PubMed] [Google Scholar]
  225. Russo SJ, Sun WL, Minerly AC, Weierstall K, Nazarian A, Festa ED, Niyomchai T, Akhavan A, Luine V, Jenab S, Quinones-Jenab V. Progesterone attenuates cocaine-induced conditioned place preference in female rats. Brain Res. 2008;1189:229–235. doi: 10.1016/j.brainres.2007.10.057. [DOI] [PubMed] [Google Scholar]
  226. Sarnyai Z, Biro E, Gardi J, Vecsernyes M, Julesz J, Telegdy G. Brain corticotropin-releasing factor mediates ‘anxiety-like’ behavior induced by cocaine withdrawal in rats. Brain Res. 1995;675:89–97. doi: 10.1016/0006-8993(95)00043-p. [DOI] [PubMed] [Google Scholar]
  227. Schmidt HD, Pierce RC. Cooperative activation of D1-like and D2-like dopamine receptors in the nucleus accumbens shell is required for the reinstatement of cocaine-seeking behavior in the rat. Neuroscience. 2006;142:451–461. doi: 10.1016/j.neuroscience.2006.06.004. [DOI] [PubMed] [Google Scholar]
  228. Schumacher M, Coirini H, McEwen BS. Regulation of high-affinity GABAa receptors in specific brain regions by ovarian hormones. Neuroendocrinology. 1989;50:315–320. doi: 10.1159/000125239. [DOI] [PubMed] [Google Scholar]
  229. Schumacher M, McEwen BS. Steroid and barbiturate modulation of the GABAa receptor. Possible mechanisms. Mol Neurobiol. 1989;3:275–304. doi: 10.1007/BF02740608. [DOI] [PubMed] [Google Scholar]
  230. Schuster CR, Johanson CE. Relationship between the discriminative stimulus properties and subjective effects of drugs. Psychopharmacol Ser. 1988;4:161–175. doi: 10.1007/978-3-642-73223-2_13. [DOI] [PubMed] [Google Scholar]
  231. Sell SL, Scalzitti JM, Thomas ML, Cunningham KA. Influence of ovarian hormones and estrous cycle on the behavioral response to cocaine in female rats. J Pharmacol Exp Ther. 2000;293:879–886. [PubMed] [Google Scholar]
  232. Sener EB, Kocamanoglu S, Cetinkaya MB, Ustun E, Bildik E, Tur A. Effects of menstrual cycle on postoperative analgesic requirements, agitation, incidence of nausea and vomiting after gynecological laparoscopy. Gynecol Obstet Invest. 2005;59:49–53. doi: 10.1159/000081222. [DOI] [PubMed] [Google Scholar]
  233. Shaham Y, Erb S, Stewart J. Stress-induced relapse to heroin and cocaine seeking in rats: a review. Brain Res Brain Res Rev. 2000;33:13–33. doi: 10.1016/s0165-0173(00)00024-2. [DOI] [PubMed] [Google Scholar]
  234. Shaham Y, Rajabi H, Stewart J. Relapse to heroin-seeking in rats under opioid maintenance: the effects of stress, heroin priming, and withdrawal. J Neurosci. 1996;16:1957–1963. doi: 10.1523/JNEUROSCI.16-05-01957.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Shaikh AA. Estrone and estradiol levels in the ovarian venous blood from rats during the estrous cycle and pregnancy. Biol Reprod. 1971;5:297–307. doi: 10.1093/biolreprod/5.3.297. [DOI] [PubMed] [Google Scholar]
  236. Shalev U, Marinelli M, Baumann MH, Piazza PV, Shaham Y. The role of corticosterone in food deprivation-induced reinstatement of cocaine seeking in the rat. Psychopharmacology (Berl) 2003;168:170–176. doi: 10.1007/s00213-002-1200-5. [DOI] [PubMed] [Google Scholar]
  237. Shimizu H, Bray GA. Effects of castration, estrogen replacement and estrus cycle on monoamine metabolism in the nucleus accumbens, measured by microdialysis. Brain Res. 1993;621:200–206. doi: 10.1016/0006-8993(93)90107-x. [DOI] [PubMed] [Google Scholar]
  238. Shoptaw S, Yang X, Rotheram-Fuller EJ, Hsieh YC, Kintaudi PC, Charuvastra VC, Ling W. Randomized placebo-controlled trial of baclofen for cocaine dependence: preliminary effects for individuals with chronic patterns of cocaine use. J Clin Psychiatry. 2003;64:1440–1448. doi: 10.4088/jcp.v64n1207. [DOI] [PubMed] [Google Scholar]
  239. Sinha R. Chronic stress, drug use, and vulnerability to addiction. Ann N Y Acad Sci. 2008;1141:105–130. doi: 10.1196/annals.1441.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Sinha R, Fox H, Hong KI, Sofuoglu M, Morgan PT, Bergquist KT. Sex steroid hormones, stress response, and drug craving in cocaine-dependent women: implications for relapse susceptibility. Exp Clin Psychopharmacol. 2007;15:445–452. doi: 10.1037/1064-1297.15.5.445. [DOI] [PubMed] [Google Scholar]
  241. Sinha R, Talih M, Malison R, Cooney N, Anderson GM, Kreek MJ. Hypothalamic-pituitary-adrenal axis and sympatho-adreno-medullary responses during stress-induced and drug cue-induced cocaine craving states. Psychopharmacology (Berl) 2003;170:62–72. doi: 10.1007/s00213-003-1525-8. [DOI] [PubMed] [Google Scholar]
  242. Sircar R, Kim D. Female gonadal hormones differentially modulate cocaine-induced behavioral sensitization in Fischer, Lewis, and Sprague-Dawley rats. J Pharmacol Exp Ther. 1999;289:54–65. [PubMed] [Google Scholar]
  243. Smith SS, Gong QH, Li X, Moran MH, Bitran D, Frye CA, Hsu FC. Withdrawal from 3alpha-OH-5alpha-pregnan-20-One using a pseudopregnancy model alters the kinetics of hippocampal GABAA-gated current and increases the GABAA receptor alpha4 subunit in association with increased anxiety. J Neurosci. 1998;18:5275–5284. doi: 10.1523/JNEUROSCI.18-14-05275.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Snively TA, Ahijevych KL, Bernhard LA, Wewers ME. Smoking behavior, dysphoric states and the menstrual cycle: results from single smoking sessions and the natural environment. Psychoneuroendocrinology. 2000;25:677–691. doi: 10.1016/s0306-4530(00)00018-4. [DOI] [PubMed] [Google Scholar]
  245. Soderpalm AH, Lindsey S, Purdy RH, Hauger R, Wit de H. Administration of progesterone produces mild sedative-like effects in men and women. Psychoneuroendocrinology. 2004;29:339–354. doi: 10.1016/s0306-4530(03)00033-7. [DOI] [PubMed] [Google Scholar]
  246. Sofuoglu M, Babb DA, Hatsukami DK. Progesterone treatment during the early follicular phase of the menstrual cycle: effects on smoking behavior in women. Pharmacol Biochem Behav. 2001;69:299–304. doi: 10.1016/s0091-3057(01)00527-5. [DOI] [PubMed] [Google Scholar]
  247. Sofuoglu M, Babb DA, Hatsukami DK. Effects of progesterone treatment on smoked cocaine response in women. Pharmacol Biochem Behav. 2002;72:431–435. doi: 10.1016/s0091-3057(02)00716-5. [DOI] [PubMed] [Google Scholar]
  248. Sofuoglu M, Dudish-Poulsen S, Nelson D, Pentel PR, Hatsukami DK. Sex and menstrual cycle differences in the subjective effects from smoked cocaine in humans. Exp Clin Psychopharmacol. 1999;7:274–283. doi: 10.1037//1064-1297.7.3.274. [DOI] [PubMed] [Google Scholar]
  249. Sofuoglu M, Mitchell E, Kosten TR. Effects of progesterone treatment on cocaine responses in male and female cocaine users. Pharmacol Biochem Behav. 2004;78:699–705. doi: 10.1016/j.pbb.2004.05.004. [DOI] [PubMed] [Google Scholar]
  250. Sofuoglu M, Mitchell E, Mooney M. Progesterone effects on subjective and physiological responses to intravenous nicotine in male and female smokers. Hum Psychopharmacol. 2009;24:559–564. doi: 10.1002/hup.1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Sofuoglu M, Poling J, Gonzalez G, Gonsai K, Oliveto A, Kosten TR. Progesterone effects on cocaine use in male cocaine users maintained on methadone: a randomized, double-blind, pilot study. Exp Clin Psychopharmacol. 2007;15:453–460. doi: 10.1037/1064-1297.15.5.453. [DOI] [PubMed] [Google Scholar]
  252. Solinas M, Panlilio LV, Justinova Z, Yasar S, Goldberg SR. Using drug-discrimination techniques to study the abuse-related effects of psychoactive drugs in rats. Nat Protoc. 2006;1:1194–1206. doi: 10.1038/nprot.2006.167. [DOI] [PubMed] [Google Scholar]
  253. Spanagel R, Weiss F. The dopamine hypothesis of reward: past and current status. Trends Neurosci. 1999;22:521–527. doi: 10.1016/s0166-2236(99)01447-2. [DOI] [PubMed] [Google Scholar]
  254. Spealman RD. Noradrenergic involvement in the discriminative stimulus effects of cocaine in squirrel monkeys. J Pharmacol Exp Ther. 1995;275:53–62. [PubMed] [Google Scholar]
  255. Specio SE, Wee S, O’Dell LE, Boutrel B, Zorrilla EP, Koob GF. CRF(1) receptor antagonists attenuate escalated cocaine self-administration in rats. Psychopharmacology (Berl) 2008;196:473–482. doi: 10.1007/s00213-007-0983-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Stewart J. Pathways to relapse: the neurobiology of drug- and stress-induced relapse to drug-taking. J Psychiatry Neurosci. 2000;25:125–136. [PMC free article] [PubMed] [Google Scholar]
  257. Stine SM, Southwick SM, Petrakis IL, Kosten TR, Charney DS, Krystal JH. Yohimbine-induced withdrawal and anxiety symptoms in opioid-dependent patients. Biol Psychiatry. 2002;51:642–651. doi: 10.1016/s0006-3223(01)01292-6. [DOI] [PubMed] [Google Scholar]
  258. Strong MN, Kaufman KR, Crabbe JC, Finn DA. Sex differences in acute ethanol withdrawal severity after adrenalectomy and gonadectomy in Withdrawal Seizure-Prone and Withdrawal Seizure-Resistant mice. Alcohol. 2009;43:367–377. doi: 10.1016/j.alcohol.2009.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Sutker PB, Goist KC, Jr, King AR. Acute alcohol intoxication in women: relationship to dose and menstrual cycle phase. Alcohol Clin Exp Res. 1987;11:74–79. doi: 10.1111/j.1530-0277.1987.tb01266.x. [DOI] [PubMed] [Google Scholar]
  260. Suzuki T, Koike Y, Yoshii T, Yanaura S. Sex differences in the induction of physical dependence on pentobarbital in the rat. Jpn J Pharmacol. 1985;39:453–459. doi: 10.1254/jjp.39.453. [DOI] [PubMed] [Google Scholar]
  261. Swanson LW, Simmons DM. Differential steroid hormone and neural influences on peptide mRNA levels in CRH cells of the paraventricular nucleus: a hybridization histochemical study in the rat. J Comp Neurol. 1989;285:413–435. doi: 10.1002/cne.902850402. [DOI] [PubMed] [Google Scholar]
  262. Tang XC, McFarland K, Cagle S, Kalivas PW. Cocaine-induced reinstatement requires endogenous stimulation of mu-opioid receptors in the ventral pallidum. J Neurosci. 2005;25:4512–4520. doi: 10.1523/JNEUROSCI.0685-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Terner JM, de Wit H. Menstrual cycle phase and responses to drugs of abuse in humans. Drug Alcohol Depend. 2006;84:1–13. doi: 10.1016/j.drugalcdep.2005.12.007. [DOI] [PubMed] [Google Scholar]
  264. Torres JM, Ortega E. Alcohol intoxication increases allopregnanolone levels in female adolescent humans. Neuropsychopharmacology. 2003;28:1207–1209. doi: 10.1038/sj.npp.1300170. [DOI] [PubMed] [Google Scholar]
  265. Torres JM, Ortega E. Alcohol intoxication increases allopregnanolone levels in male adolescent humans. Psychopharmacology (Berl) 2004;172:352–355. doi: 10.1007/s00213-003-1662-0. [DOI] [PubMed] [Google Scholar]
  266. Toufexis DJ, Davis C, Hammond A, Davis M. Progesterone attenuates corticotropin-releasing factor-enhanced but not fear-potentiated startle via the activity of its neuroactive metabolite, allopregnanolone. J Neurosci. 2004;24:10280–10287. doi: 10.1523/JNEUROSCI.1386-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Ueno S, Harris RA, Messing RO, Sanchez-Perez AM, Hodge CW, McMahon T, Wang D, Mehmert KK, Kelley SP, Haywood A, Olive MF, Buck KJ, Hood HM, Blednov Y, Findlay G, Mascia MP. Alcohol actions on GABA(A) receptors: from protein structure to mouse behavior. Alcohol Clin Exp Res. 2001;25:76S–81S. doi: 10.1097/00000374-200105051-00014. [DOI] [PubMed] [Google Scholar]
  268. Uzbay IT, Sag Lam E, Kayir H, Celik T, Beyazyurek M. Effects of fluoxetine on ethanol withdrawal syndrome in rats. J Psychiatr Res. 2004;38:445–450. doi: 10.1016/j.jpsychires.2003.11.007. [DOI] [PubMed] [Google Scholar]
  269. Valdez GR, Roberts AJ, Chan K, Davis H, Brennan M, Zorrilla EP, Koob GF. Increased ethanol self-administration and anxiety-like behavior during acute ethanol withdrawal and protracted abstinence: regulation by corticotropin-releasing factor. Alcohol Clin Exp Res. 2002;26:1494–1501. doi: 10.1097/01.ALC.0000033120.51856.F0. [DOI] [PubMed] [Google Scholar]
  270. VanDoren MJ, Matthews DB, Janis GC, Grobin AC, Devaud LL, Morrow AL. Neuroactive steroid 3alpha-hydroxy-5alpha-pregnan-20-one modulates electrophysiological and behavioral actions of ethanol. J Neurosci. 2000;20:1982–1989. doi: 10.1523/JNEUROSCI.20-05-01982.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  271. Varlinskaya EI, Spear LP. Acute ethanol withdrawal (hangover) and social behavior in adolescent and adult male and female Sprague-Dawley rats. Alcohol Clin Exp Res. 2004;28:40–50. doi: 10.1097/01.ALC.0000108655.51087.DF. [DOI] [PubMed] [Google Scholar]
  272. Volkow ND, Fowler JS, Wang GJ. Imaging studies on the role of dopamine in cocaine reinforcement and addiction in humans. J Psychopharmacol. 1999;13:337–345. doi: 10.1177/026988119901300406. [DOI] [PubMed] [Google Scholar]
  273. Walker QD, Francis R, Cabassa J, Kuhn CM. Effect of ovarian hormones and estrous cycle on stimulation of the hypothalamo-pituitary-adrenal axis by cocaine. J Pharmacol Exp Ther. 2001;297:291–298. [PubMed] [Google Scholar]
  274. Wetzel CH, Hermann B, Behl C, Pestel E, Rammes G, Zieglgansberger W, Holsboer F, Rupprecht R. Functional antagonism of gonadal steroids at the 5-hydroxytryptamine type 3 receptor. Mol Endocrinol. 1998;12:1441–1451. doi: 10.1210/mend.12.9.0163. [DOI] [PubMed] [Google Scholar]
  275. White TL, Justice AJ, de Wit H. Differential subjective effects of D-amphetamine by gender, hormone levels and menstrual cycle phase. Pharmacol Biochem Behav. 2002;73:729–741. doi: 10.1016/s0091-3057(02)00818-3. [DOI] [PubMed] [Google Scholar]
  276. Woodstock-Striley C, Cottler LB, Ben Abdallah A. Females have less physiological dependence to alcohol than men. Paper presented at the meeting of the College of Problems on Drug Dependence; San Juan, Puerto Rico. 2004. [Google Scholar]
  277. Wu HB, Fabian S, Jenab S, Quinones-Jenab V. Progesterone receptors activation after acute cocaine administration. Brain Res. 2006;1126:188–192. doi: 10.1016/j.brainres.2006.09.074. [DOI] [PubMed] [Google Scholar]
  278. Yang H, Zhao W, Hu M, Becker JB. Interactions among ovarian hormones and time of testing on behavioral sensitization and cocaine self-administration. Behav Brain Res. 2007;184:174–184. doi: 10.1016/j.bbr.2007.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  279. Yu R, Hay M, Ticku MK. Chronic neurosteroid treatment attenuates single cell GABAA response and its potentiation by modulators in cortical neurons. Brain Res. 1996;706:160–162. doi: 10.1016/0006-8993(95)01247-8. [DOI] [PubMed] [Google Scholar]
  280. Yu R, Ticku MK. Chronic neurosteroid treatment decreases the efficacy of benzodiazepine ligands and neurosteroids at the gamma-aminobutyric acidA receptor complex in mammalian cortical neurons. J Pharmacol Exp Ther. 1995;275:784–789. [PubMed] [Google Scholar]

RESOURCES