Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Brain Res ; 1711: 16-22, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30629943

ABSTRACT

Methamphetamine is a neurotoxic psychostimulant known to cause cell death and terminal degradation of dopaminergic neurons in the striatum concomitant with memory deficits. However, most of the research studies have not examined the influence of sex on these changes. In this study we compared the effects of a binge regimen of methamphetamine (four injections of 4 mg/kg) on male, female, and ovariectomized (OVX) female Sprague-Dawley rats. We show that male and OVX female animals had a deficit in a novel object recognition task, while intact females did not show this deficit. Neurochemical analysis of the same animals indicated higher levels of FosB protein in caudate-putamen (CPu) and nucleus accumbens (NAc) of the male animals than intact or OVX females. Methamphetamine also increased Bcl-2 protein levels in CPu of all the cohorts. We did not find a significant effect of methamphetamine on the dopamine neuron markers tyrosine hydroxylase (TH) or dopamine transporter (DAT) 7 days after methamphetamine administrations. Our behavioral and neurochemical studies indicate that methamphetamine differentially affects male and female animals and shows sex differences in memory and molecular mechanisms in the striatum of these animals.


Subject(s)
Memory/drug effects , Methamphetamine/adverse effects , Animals , Caudate Nucleus/drug effects , Caudate Nucleus/metabolism , Central Nervous System Stimulants/pharmacology , Corpus Striatum/drug effects , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Female , Male , Methamphetamine/metabolism , Neurotoxicity Syndromes/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Ovariectomy , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Sex Factors , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism
2.
Behav Brain Res ; 317: 218-225, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27664298

ABSTRACT

The aim of this study was to investigate the intracellular responses associated with the acquisition and expression of cocaine-context associations. ERK (extracellular regulated kinase), CREB (cAMP responsive element binding protein), FosB and ΔFosB proteins were of particular interest due to their involvement in cocaine reward and in synaptic plasticity underlying learning and memory. We used the conditioned place preference (CPP) paradigm, which employs a Pavlovian conditioning procedure to establish an association between a drug-paired environment and the drug's rewarding effects, to study the role of these signaling pathways in cocaine-context associations. N-methyl-D-aspartate receptor (NMDAR) antagonism prior to cocaine administration during conditioning blocked the acquisition of cocaine CPP and reduced Nucleus Accumbens (NAc) phosphorylated-ERK (pERK) and phosphorylated CREB (pCREB) levels following the CPP test (drug-free). We also show that cocaine-induced increases in Caudate Putamen (CPu) FosB and ΔFosB levels are decreased after MK-801 pre-treatment during conditioning. In addition, our results provide evidence for the involvement of striatal SIRT (Silent Information Regulator of Transcription) proteins in cocaine-CPP. These results will aid in the advancement of general knowledge about the molecular formation and retrieval of cocaine-associated memories that can be used in the future when designing treatments for cocaine addiction that target both prevention and relapse.


Subject(s)
Cocaine/pharmacology , Conditioning, Classical/drug effects , Dopamine Uptake Inhibitors/pharmacology , Down-Regulation/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/drug effects , Animals , CREB-Binding Protein , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Globus Pallidus/drug effects , Globus Pallidus/metabolism , Locomotion/drug effects , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Inbred F344 , Sirtuin 1/metabolism , Sirtuin 2/metabolism , Time Factors
3.
Psychopharmacology (Berl) ; 233(19-20): 3527-36, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27553823

ABSTRACT

Previous studies have shown sex different patterns in behavioral responses to cocaine. Here, we used between-subject experiment design to study whether sex differences exist in the development of behavioral sensitization and tolerance to repeated cocaine, as well as the role of protein kinase A (PKA) signaling cascade in this process. Ambulatory and rearing responses were recorded in male and female rats after 1 to 14 days of administration of saline or cocaine (15 mg/kg; ip). Correspondent PKA-associated signaling in the nucleus accumbens (NAc) and caudate-putamen (CPu) was measured at each time point. Our results showed that females exhibited higher cocaine-induced behavioral responses and developed behavioral sensitization and tolerance faster than males. Whereas females developed behavioral sensitization to cocaine after 2 days and tolerance after 14 days, male rats developed sensitization after 5 days. In addition, cocaine induced a sexual dimorphic pattern in the progression of neuronal adaptations on the PKA cascade signaling in region (NAc vs. CPu) and time (days of cocaine administration)-dependent manners. In general, more PKA signaling cascade changes were found in the NAc of males on day 5 and in the CPu of females with repeated cocaine injection. In addition, in females, behavioral activities positively correlated with FosB levels in the NAc and CPu and negatively correlated with Cdk5 and p35 in the CPu, while no correlation was observed in males. Our studies suggest that repeated cocaine administration induced different patterns of behavioral and molecular responses in the PKA cascade in male and female rats.


Subject(s)
Behavior, Animal/drug effects , Caudate Nucleus/drug effects , Cocaine/pharmacology , Cyclic AMP-Dependent Protein Kinases/drug effects , Dopamine Uptake Inhibitors/pharmacology , Locomotion/drug effects , Nucleus Accumbens/drug effects , Putamen/drug effects , Animals , Caudate Nucleus/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin-Dependent Kinase 5/drug effects , Cyclin-Dependent Kinase 5/metabolism , Drug Tolerance , Female , Male , Motor Activity/drug effects , Nucleus Accumbens/metabolism , Phosphotransferases/drug effects , Phosphotransferases/metabolism , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Putamen/metabolism , Rats , Sex Factors , Signal Transduction/drug effects
4.
Cytokine ; 72(2): 121-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25647266

ABSTRACT

Although estrogen reduces inflammatory-mediated pain responses, the mechanisms behind its effects are unclear. This study investigated if estrogen modulates inflammatory signaling by reducing baseline or inflammation-induced cytokine levels in the injury-site, serum, dorsal root ganglia (DRG) and/or spinal cord. We further tested whether estrogen effects on cytokine levels are in part mediated through hypothalamic-pituitary-adrenal (HPA) axis activation. Lumbar DRG, spinal cord, serum, and hind paw tissue were analyzed for cytokine levels in 17ß-estradiol-(20%) or vehicle-(100% cholesterol) treated female rats following ovariectomy/sham adrenalectomy (OVX), adrenalectomy/sham ovariectomy (ADX) or ADX+OVX operation at baseline and post formalin injection. Formalin significantly increased pro-inflammatory interleukin (IL)-6 levels in the paw, as well as pro- and anti-inflammatory cytokine levels in the DRG, spinal cord and serum in comparison to naïve conditions. Estrogen replacement significantly increased anti-inflammatory IL-10 levels in the DRG. Centrally, estradiol significantly decreased pro-inflammatory tumor necrosis factor (TNF)-α and IL-1ß levels, as well as IL-10 levels, in the spinal cord in comparison to cholesterol treatment. At both sites, most estradiol modulatory effects occurred irrespective of pain or surgical condition. Estradiol alone had no influence on cytokine release in the paw or serum, indicating that estrogen effects were site-specific. Although cytokine levels were altered between surgical conditions at baseline and following formalin administration, ADX operation did not significantly reverse estradiol's modulation of cytokine levels. These results suggest that estrogen directly regulates cytokines independent of HPA axis activity in vivo, in part by reducing cytokine levels in the spinal cord.


Subject(s)
Cytokines/metabolism , Estradiol/pharmacology , Estrogens/physiology , Ganglia, Spinal/immunology , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Spinal Cord/immunology , Adrenalectomy , Animals , Cytokines/blood , Cytokines/genetics , Estradiol/administration & dosage , Estrogens/deficiency , Formaldehyde/administration & dosage , Inflammation , Interleukin-10/blood , Interleukin-10/immunology , Interleukin-1beta/blood , Interleukin-1beta/immunology , Interleukin-6/blood , Interleukin-6/immunology , Ovariectomy , Pain , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
5.
Brain Res ; 1520: 121-33, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23665060

ABSTRACT

Sex differences in cocaine's mechanisms of action and behavioral effects have been widely reported. However, little is known about how sex influences intracellular signaling cascades involved with drug-environment associations. We investigated whether ERK/CREB intracellular responses in the mesocorticolimbic circuitry underlying cocaine environmental associations are sexually dimorphic. We used a standard 4 day conditioned place preference (CPP) paradigm using 20mg/kg cocaine-a dose that induced CPP in male and female Fischer rats. In the nucleus accumbens (NAc) following CPP expression, cocaine treated animals showed increased phosphorylated ERK (pERK), phosphorylated CREB (pCREB) and ΔFosB protein levels. In the hippocampus (HIP) and caudate putamen (CPu), pERK and FosB/ΔFosB levels were also increased, respectively. Cocaine females had a larger change in HIP pERK and CPu ΔFosB levels than cocaine males; partly due to lower protein levels in saline female rats when compared to saline males. Prefrontal cortex (PfC) pCREB levels increased in cocaine males, but not females, whereas PfC pERK levels were increased in cocaine females, but not males. CPP scores were positively correlated to NAc pERK, HIP pERK and CPu FosB protein levels, suggesting that similar to males, the ERK/CREB intracellular pathway in mesocorticolimbic regions undergoes cocaine induced neuroplasticity in female rats. However, there seem to be intrinsic (basal) sexual dimorphisms in this pathway that may contribute to responses expressed after cocaine-CPP. Taken together, our results suggest that cellular responses associated with the expression of learned drug-environment associations may play an important role in sex differences in cocaine addiction and relapse.


Subject(s)
Brain/drug effects , Cocaine-Related Disorders/metabolism , Sex Characteristics , Signal Transduction/drug effects , Animals , Blotting, Western , Brain/metabolism , Cocaine/pharmacology , Conditioning, Operant/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Dopamine Uptake Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Male , Phosphorylation , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Inbred F344 , Signal Transduction/physiology
6.
ILAR J ; 53(1): 14-22, 2012.
Article in English | MEDLINE | ID: mdl-23520596

ABSTRACT

Cocaine abuse is on the rise among women, and drug addiction studies consistently show greater responses among females than males in various cocaine-related outcomes. Animal and human studies reveal sexually dimorphic patterns in behavioral responses to cocaine in all phases of the cocaine addiction process from initiation to maintenance and relapse. Furthermore, in animal models, females require lower doses of cocaine to develop faster conditioned place preference and cocaine-induced psychomotor behaviors and sensitization. A clear picture is emerging and suggests that the biological basis of sex-specific differences in cocaine addiction lies, in part, in the disparate regulation of the central nervous system by male and female gonadal hormones and, in part, in chromosomal mechanisms that contribute to drug abuse vulnerability. The interactions of the many factors that affect sex differences appear to be complex. For example, in females, estradiol has facilitatory effects overall, whereas progesterone inhibits most cocaine responses. This review presents a discussion of sex differences and the role of gonadal hormones as the biological basis for the sexually dimorphic pattern in behavioral responses to cocaine.


Subject(s)
Cocaine-Related Disorders/physiopathology , Gonadal Hormones/metabolism , Sex Characteristics , Animals , Cocaine-Related Disorders/metabolism , Female , Humans , Male , Models, Animal
7.
Brain Res ; 1382: 181-8, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21281615

ABSTRACT

How exogenous estrogen affects inflammatory responses is poorly understood despite the large numbers of women receiving estrogen-alone hormone therapy. The aim of this study was to determine if estradiol alters injury- or inflammation-induced nociceptive responses after carrageenan administration in females and whether its effects are mediated through cyclo-oxygenase (COX) and prostaglandins (PG). To this end, paw withdrawal latencies and serum levels of PGE2 and PGD2 were measured in rats treated with estradiol (0, 10, 20, and 30%) and/or SC560 (COX-1 inhibitor) or NS398 (COX-2 inhibitor) after intraplantar carrageenan administration. Estradiol significantly increased withdrawal latencies before (baseline condition) and after carrageenan administration to one hindpaw. NS398 was anti-nociceptive only in carrageenan treated animals. SC560 increased withdrawal latencies in both paws at 1 and 5hours after carrageenan administration. Co-administration of estradiol and NS398, but not SC560, was additive except for a prolonged anti-nociceptive effects of estradiol combined with NS398. The anti-nociceptive effect extended beyond that observed with either drug or estradiol alone at the 5-hour time point. Estradiol had no significant effect on PGE2 serum levels, but both COX antagonists decreased them. Although neither estradiol nor the COX inhibitors alone had an effect on PGD2 serum levels, co-administration of NS398 and estradiol significantly elevated PGD2 levels. Taken together, our results suggest that estradiol is anti-nociceptive in the thermal test and reduces carrageenan-induced hyperalgesia. These effects are minimally altered through PG-mediated mechanisms.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Estrogens/pharmacology , Hyperalgesia/drug therapy , Nociceptors/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Carrageenan/antagonists & inhibitors , Disease Models, Animal , Drug Synergism , Estrogens/metabolism , Female , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Nociceptors/physiology , Rats , Rats, Sprague-Dawley
8.
Synapse ; 65(7): 643-51, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21132813

ABSTRACT

Estrogen modulates pain perception but how it does so is not fully understood. The aim of this study was to determine if estradiol reduces nociceptive responses in part via hypothalamic-pituitary-adrenal (HPA) axis regulation of cyclooxygenase (COX)-1/COX-2 activity. The first study examined the effects of estradiol (20%) or vehicle with concurrent injection nonsteroidal antiinflammatory drugs (NSAIDs) on formalin-induced nociceptive responding (flinching) in ovariectomized (OVX) rats. The drugs were ibuprofen (COX-1 and COX-2 inhibitor), SC560 (COX-1 inhibitor), or NS398 (COX-2 inhibitor). In a second study, estradiol's effects on formalin-induced nociception were tested in adrenalectomized (ADX), OVX, and ADX+OVX rats. Serum levels of prostaglandins (PG) PGE(2) and corticosterone were measured. Estradiol significantly decreased nociceptive responses in OVX rats with effects during both the first and the second phase of the formalin test. The nonsteroidal antiinflammatory drugs (NSAIDs) did not alter nociception at the doses used here. Adrenalectomy neither altered flinching responses in female rats nor reversed estradiol-induced antinociceptive responses. Estradiol alone had no effect on corticosterone (CORT) or prostaglandin levels after the formalin test, dissociating the effects of estradiol on behavior and these serum markers. Ibuprofen and NS398 significantly reduced PGE2 levels. CORT was not decreased by OVX surgery or by estradiol below that of ADX. Only IBU significantly increased corticosterone levels. Taken together, our results suggest that estradiol-induced antinociception in female rats is independent of COX activity and HPA axis activation.


Subject(s)
Estradiol/pharmacology , Pain Perception/drug effects , Pain/physiopathology , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Female , Formaldehyde/toxicity , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Irritants/toxicity , Ovariectomy , Pain Perception/physiology , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Rats , Rats, Sprague-Dawley
9.
Ethn Dis ; 20(1 Suppl 1): S1-24-7, 2010.
Article in English | MEDLINE | ID: mdl-20521380

ABSTRACT

INTRODUCTION: Although it is known that female rats have a more robust behavioral response to acute cocaine administration than male rats, the neurobiological mechanisms underlying these differences remain unclear. The purpose of the present study was to determine if there are sex differences in cocaine's regulation of dopamine D1 and D2 receptor mRNA levels. METHODS: Male and female Fischer rats received acute cocaine (20 mg/kg, intraperitoneal) or saline. Ambulatory activity was recorded one hour post drug treatment. Rats were then sacrificed either 1 or 24 hours post drug treatment and D1/D2 DA receptor mRNA levels were measured via solution hybridization assay. RESULTS: Cocaine-induced ambulatory activity was greater in female than male rats. There were no sex differences in baseline levels of D1 and D2 receptor mRNA in the caudate putamen (CPu) or the nucleus accumbens (NAc). Cocaine administration reduced levels of D1 mRNA in the NAc only in male rats. CONCLUSION: Our findings suggest that the regulation of striatal D1 mRNA levels after acute cocaine administration is a sexually dimorphic process. We also hypothesize that the D1 receptor may be an important substrate in the regulation of sex differences in cocaine-induced locomotor activity.


Subject(s)
Cocaine/poisoning , Receptors, Dopamine D1/physiology , Sex Characteristics , Animals , Female , Male , Motor Skills/drug effects , Motor Skills/physiology , RNA, Messenger/analysis , Rats , Rats, Inbred F344 , Receptors, Dopamine D2/physiology
10.
Ethn Dis ; 20(1 Suppl 1): S1-50-4, 2010.
Article in English | MEDLINE | ID: mdl-20521385

ABSTRACT

INTRODUCTION: This study aimed to determine whether the previously reported differential effects of estradiol on inflammation-induced behavioral responses are in part explained through differential activation of the corticosterone-cyclooxygenase (CORT-COX) regulatory pathway. METHODS: Prostaglandin E2 (PGE2), COX, and CORT levels were analyzed before and after a formalin administration (1% vs. 5%, representing different intensities of inflammatory stimuli). RESULTS: In vehicle-treated rats, chronic estradiol administration increased corticosterone, and decreased COX and PGE2. After acute estradiol administration, although corticosterone serum levels were increased, COX protein levels were unchanged. In rats treated with formalin, PGE2 serum levels were higher in rats administered 5% formalin than vehicle- and 1%-treated rats. Significant correlations were observed between PGE2 serum levels, CORT serum levels, and COX protein levels. CONCLUSIONS: Our results suggest that the administration of exogenous estradiol may mediate inflammatory responses by regulating the levels of PGE2 and/or CORT release, thereby mediating the nociceptive response to an inflammatory stimulus.


Subject(s)
Corticosterone/physiology , Dinoprostone/blood , Estradiol/pharmacology , Estrogens/pharmacology , Inflammation/physiopathology , Prostaglandin-Endoperoxide Synthases/physiology , Animals , Blotting, Western , Carrier Proteins/blood , Estradiol/administration & dosage , Estrogens/administration & dosage , Female , Nociceptors/physiology , Rats , Rats, Sprague-Dawley
11.
Ethn Dis ; 20(1 Suppl 1): S1-55-9, 2010.
Article in English | MEDLINE | ID: mdl-20521386

ABSTRACT

INTRODUCTION: This study aimed to determine if endogenous gonadal hormones affect the intracellular mechanisms in the spinal cord that control inflammatory pain responses. METHODS: We analyzed behavioral responses to, and changes in, serum levels of prostaglandin E2, estradiol, progesterone, and corticosterone after administration of 5% formalin in intact and ovariectomized (OVX) female rats. RESULTS: OVX females displayed significantly more flinching than did intact females during Phase I, and after formalin administration their corticosterone levels were significantly lower. No differences were seen across COX-1 and COX-2 protein expression in the spinal cord of either naive or formalin-treated rats. However, subsequent to formalin a main effect of gonadectomy was seen in prostaglandin E2 levels; OVX animals had significantly lower prostaglandin E2 levels than intact animals. CONCLUSIONS: These results indicate that in female rats nociceptive responses to formalin are regulated through the levels of prostaglandin E2, an important mediator in inflammation, whereas protein levels of COX-1 and COX-2 play a more limited role.


Subject(s)
Dinoprostone/metabolism , Estrogens/physiology , Formaldehyde/pharmacology , Pain/physiopathology , Testosterone/physiology , Animals , Blotting, Western , Corticosterone/blood , Cyclooxygenase 1/blood , Cyclooxygenase 2/blood , Dinoprostone/blood , Estradiol/physiology , Estrogens/blood , Female , Inflammation/physiopathology , Membrane Proteins/blood , Ovariectomy , Pain Measurement , Progesterone/blood , Rats , Rats, Sprague-Dawley , Spinal Cord/chemistry
12.
Ethn Dis ; 20(1 Suppl 1): S1-73-7, 2010.
Article in English | MEDLINE | ID: mdl-20521389

ABSTRACT

INTRODUCTION: The present study aimed to determine if, as occurs in female rats, progesterone attenuates cocaine-induced reward and psychomotor responses in male rats. METHODS: The role of progesterone in the acquisition and/or expression of cocaine-induced conditioned place preference (CPP) and locomotor responses of intact male rats was studied. For chronic progesterone treatment, rats received Silastic capsules with either progesterone (100%) or vehicle 1 week prior to conditioning. For acute progesterone treatment, rats received subcutaneous injections of progesterone (500 microg) or vehicle (sesame oil) 4 hours before intraperitoneal injections of saline or cocaine administration (20 mg/kg) on conditioning days (acquisition phase-formation of reward associations) or before testing (expression phase-recall of reward associations). RESULTS: Both progesterone-treatment paradigms produced equivalent progesterone serum levels. Progesterone administered chronically or acutely during the acquisition and expression phases of cocaine conditioning did not block cocaine-induced CPP. Nor did progesterone affect ambulatory or rearing behaviors after cocaine administration. CONCLUSION: These results suggest that, unlike the findings with female rats (in which similar treatment paradigms inhibited the formation and recall of cocaine-induced CPP), progesterone plays a limited role in the cocaine-induced reward or psychomotor responses of male rats.


Subject(s)
Cocaine/pharmacology , Conditioning, Psychological/drug effects , Dopamine Uptake Inhibitors/pharmacology , Memory, Short-Term/drug effects , Motor Activity/drug effects , Progesterone/pharmacology , Progestins/pharmacology , Animals , Behavior, Animal/drug effects , Cocaine-Related Disorders/physiopathology , Exploratory Behavior/drug effects , Male , Memory, Short-Term/physiology , Progesterone/administration & dosage , Rats , Rats, Inbred F344 , Recognition, Psychology/drug effects
13.
Ethn Dis ; 20(1 Suppl 1): S1-83-7, 2010.
Article in English | MEDLINE | ID: mdl-20521391

ABSTRACT

INTRODUCTION: The aim of this study was to determine if progesterone affects spatial and non-spatial working memory in intact male and female rats. METHODS: Rats received subcutaneous injections of progesterone (500 microg) or vehicle (sesame oil). Four hours after hormone treatments, spatial and non-spatial memories were tested using novel object recognition and spatial object recognition tasks. RESULTS: Vehicle-treated female rats had higher progesterone serum levels than males, but progesterone treatment produced equivalent progesterone serum levels in both sexes. In the object recognition task--a non-spatial memory task-females showed better performance than males, and progesterone had no effect on either sex. However, in the object replacement task--a spatial memory task-progesterone significantly impaired the retention in both male and female rats as compared with vehicle-treated groups. CONCLUSION: These results suggest that acute progesterone treatment interferes with spatial working memory consolidation, but not recognition (non-spatial) working memory. As such, the observed sexual incongruities in progesterone's effects on working memory suggest that progesterone-based hormone therapies have a negative impact on cognition.


Subject(s)
Memory/drug effects , Progesterone/pharmacology , Animals , Cognition/drug effects , Estrogen Replacement Therapy , Female , Male , Progesterone/administration & dosage , Rats , Rats, Inbred F344 , Sesame Oil/pharmacology
14.
Ethn Dis ; 20(1 Suppl 1): S1-88-91, 2010.
Article in English | MEDLINE | ID: mdl-20521392

ABSTRACT

INTRODUCTION: Sexually dimorphic behavioral responses to cocaine have been linked to a difference in activation of dopamine receptors. Our study was conducted to determine whether dopamine D2-like receptor-activated G-protein contributes to sex differences in response to cocaine in the medial prefrontal cortex (mPFC). METHOD: In vitro functional autoradiography was performed using dopamine receptor D2 agonist (quinpirole, 100 microM) to stimulate [35S]GTPgammaS binding in brain tissue sections from male and female Fischer rats treated with saline (1 mL/kg) or cocaine (20 mg/kg; i.p.). RESULTS: Overall, quinpirole increased G-protein activation in the caudate-putamen, nucleus accumbens, and frontal cortex in both sexes. Although saline-treated male rats had higher [35S]GTPyS binding in the mPFC than their female counterparts, cocaine-treated females had higher [35S]GTPgammaS binding in the mPFC than cocaine-treated males. CONCLUSIONS: These data suggest that both intrinsic and activational effects of dopamine D2-like receptor-mediated G-protein activation in the mPFC may contribute to the differences between males and females in their response to acute cocaine administration.


Subject(s)
Dopamine Agonists/pharmacology , GTP-Binding Proteins/metabolism , Quinpirole/pharmacology , Receptors, Dopamine D2/physiology , Animals , Autoradiography , Binding, Competitive/physiology , Cocaine/pharmacology , Female , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Male , Motor Activity/drug effects , Prefrontal Cortex/physiology , Rats , Rats, Inbred F344 , Sex Characteristics
15.
Pharmacol Biochem Behav ; 94(3): 404-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19822170

ABSTRACT

Little is known about the physiological and behavioral effects of testosterone when co-administered with cocaine during adolescence. The present study aimed to determine whether exogenous testosterone administration differentially alters psychomotor responses to cocaine in adolescent and adult male rats. To this end, intact adolescent (30-days-old) and adult (60-day-old) male Fisher rats were pretreated with vehicle (sesame oil) or testosterone (5 or 10mg/kg) 45 min prior to saline or cocaine (20mg/kg) administration. Behavioral responses were monitored 1h after drug treatment, and serum testosterone levels were determined. Serum testosterone levels were affected by age: saline- and cocaine-treated adults in the vehicle groups had higher serum testosterone levels than adolescent rats, but after co-administration of testosterone the adolescent rats had higher serum testosterone levels than the adults. Pretreatment with testosterone affected baseline activity in adolescent rats: 5mg/kg of testosterone increased both rearing and ambulatory behaviors in saline-treated adolescent rats. After normalizing data to % saline, an interaction between hormone administration and cocaine-induced behavioral responses was observed; 5mg/kg of testosterone decreased both ambulatory and rearing behaviors among adolescents whereas 10mg/kg of testosterone decreased only rearing behaviors. Testosterone pretreatment did not alter cocaine-induced behavioral responses in adult rats. These findings suggest that adolescents are more sensitive than adults to an interaction between testosterone and cocaine, and, indirectly, suggest that androgen abuse may lessen cocaine-induced behavioral responses in younger cocaine users.


Subject(s)
Behavior, Animal/drug effects , Cocaine/pharmacology , Motor Activity/drug effects , Testosterone/pharmacology , Animals , Male , Rats , Rats, Inbred F344 , Testosterone/blood
16.
Horm Behav ; 58(1): 22-32, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19819242

ABSTRACT

In this review, we summarize literature focused on how progesterone alters cocaine-induced psychomotor, reinforcement, and physiological responses. Clinical studies suggest that progesterone attenuates the subjective effects of cocaine. Similarly, preclinical studies have demonstrated that cocaine-induced reward and psychomotor responses are attenuated after progesterone administration. In rats progesterone also reduces the reinforcement effects of cocaine attenuates acquisition, escalation, reinstatement of cocaine self-administration, and cocaine-seeking behaviors. Progesterone also counteracts the facilitatory effects of estrogen on cocaine self-administration and psychomotor activation. These findings suggest that progesterone has a potential in clinical applications as a treatment for cocaine addiction. Constantly changing progesterone serum levels in female humans and rats affect the female's reinforcement responses to cocaine and may in part contribute to the known sex differences in cocaine responses.


Subject(s)
Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Progesterone/metabolism , Animals , Cocaine/administration & dosage , Cocaine/pharmacokinetics , Dopamine Uptake Inhibitors/administration & dosage , Dopamine Uptake Inhibitors/pharmacokinetics , Humans , Models, Neurological , Motor Activity/drug effects , Motor Activity/physiology , Reinforcement, Psychology , Sex Characteristics
17.
Brain Res Bull ; 80(6): 377-81, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-19716863

ABSTRACT

Numerous studies have shown that biochemical and behavioral effects of cocaine are mediated by dopamine D1 receptor (D1R) and NMDA R1 receptor (NR1)-mediated transmission. In this study, we investigated the physical interactions between D1R and NR1 in response to acute cocaine administration in a time course of 5-60 min. In the caudate-putamen (CPu) of male Fischer rats, a single cocaine injection (30 mg/kg) reduced D1R-NR1 protein-protein interactions 30 min after treatment. In addition, activation or blockade of the NMDA receptor using NMDA (25mg/kg) or MK-801 (0.25mg/kg), respectively, also reduced the D1R-NR1 physical interactions. Acute cocaine administration did not alter total D1R or NR1 protein levels in our time course of study. These results indicate that D1R-NR1 physical interaction rather than total protein levels may regulate the intracellular signaling after acute cocaine administration.


Subject(s)
Caudate Nucleus/drug effects , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Putamen/drug effects , Receptors, Dopamine D1/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Blotting, Western , Caudate Nucleus/metabolism , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Immunoprecipitation , Male , N-Methylaspartate/pharmacology , Putamen/metabolism , Rats , Rats, Inbred F344 , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Time Factors , Tubulin/metabolism
18.
Psychopharmacology (Berl) ; 203(1): 175-83, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18985320

ABSTRACT

RATIONALE: Behavioral and dopamine responses to cocaine are sexually dimorphic: Female rats exhibit higher levels of locomotor and reward-associated behaviors after cocaine administration and dopamine release than do males. Activation of the dopamine- and cAMP-regulated phosphoprotein of Mr 32 kDa (DARPP-32) intracellular cascade mediates responses to cocaine. OBJECTIVE: To examine the possibility that acute cocaine administration alters the DARPP-32 cascade in a sexually dimorphic pattern. MATERIALS AND METHODS: Male and female rats received either saline or cocaine (30 mg/kg). Protein levels of DARPP-32, phosphorylation of DARPP-32 at the Thr34 site (P-Thr34-DARPP-32), protein phosphatase 1 (PP-1), and protein phosphatase 2B (PP-2B) in nucleus accumbens were measured via Western blot analysis. RESULTS: Females had higher protein levels of DARPP-32, P-Thr34-DARPP-32, calcineurin A (CaN-A; catalytic subunit of PP-2B), and calcineurin B (CaN-B; regulatory subunit of PP-2B) than males 5 min after saline treatment. In females, CaN-A protein levels were also higher at 15 min and PP-1 protein levels were higher 30 min after saline administration than males. In male rats, cocaine significantly increased CaN-A protein levels at 30 min and CaN-B protein levels at 15 min. In females, cocaine administration significantly decreased protein levels of DARPP-32, P-Thr34-DARPP-32, and CaN-A at 45 min but increased PP-1 protein levels at 30 min. Overall, males had higher activation of the DARPP-32 pathway after cocaine administration than did females. CONCLUSION: These novel results show that basal and cocaine-induced sex differences in the DARPP-32/PP-1 cascade may be responsible for the sexual dimorphism in acute cocaine-induced behavioral responses.


Subject(s)
Central Nervous System Stimulants/pharmacology , Cocaine/pharmacology , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Nucleus Accumbens/drug effects , Animals , Behavior, Animal/drug effects , Calcineurin/metabolism , Central Nervous System Stimulants/administration & dosage , Cocaine/administration & dosage , Female , Injections, Intraperitoneal , Male , Nucleus Accumbens/enzymology , Nucleus Accumbens/metabolism , Phosphorylation , Protein Phosphatase 1/metabolism , Rats , Rats, Inbred F344 , Sex Factors , Time Factors
19.
Psychopharmacology (Berl) ; 203(3): 641-50, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19052730

ABSTRACT

INTRODUCTION: Alterations in protein kinase (PKA) protein levels have been implicated in the regulation of responses to and development of cocaine addiction. However, the contribution of differences in PKA intracellular cascade to the known sex differences in responses to cocaine is not well understood. This study examined whether there are intrinsic or cocaine-induced alterations in PKA-mediated responses, such as phosphorylation of cyclic AMP response element binding protein, in male and female rats. MATERIALS AND METHODS: To this end, protein levels of PKA and phosphorylated CREB (pCREB) in the caudate putamen (CPu) and nucleus accumbens (NAc) of male and female rats were measured basally or after acute (one 30-mg/kg intraperitoneal injection) or chronic (twice-daily 15-mg/kg injections for 14 days) cocaine administration. Behavioral responses to both cocaine administration paradigms were also studied. RESULTS: Similar to previous findings, ambulatory, rearing, and stereotypic activities were higher in female rats after acute cocaine administration. Sex differences in cocaine-induced responses were also observed after chronic cocaine administration: While males developed a robust sensitization in ambulatory activities to cocaine, females developed tolerance in cocaine-induced rearing and stereotypic activities. In the basal group, females had significantly higher PKA protein levels in the NAc. Regardless of the cocaine administration paradigm, PKA protein levels in the NAc were higher overall in females than in males. Furthermore, after cocaine administration, while pCREB protein levels in male rats were induced for a longer amount of time than in female rats, the magnitude of change on pCREB levels were higher in female than male rats. However, in the CPu, no sex differences in PKA or pCREB protein levels were observed either in the basal group or after acute or chronic cocaine administration. DISCUSSION: Taken together, these findings suggest that sex differences in basal and cocaine-induced alterations in the PKA signaling regulation in the NAc may contribute to sex differences in the psychomotor responses to cocaine.


Subject(s)
Cocaine/adverse effects , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Nucleus Accumbens/drug effects , Sex Characteristics , Animals , Behavior, Animal/drug effects , Female , Male , Nucleus Accumbens/enzymology , Nucleus Accumbens/metabolism , Rats , Rats, Inbred F344
20.
Brain Res ; 1243: 1-9, 2008 Dec 03.
Article in English | MEDLINE | ID: mdl-18822274

ABSTRACT

Cocaine is an addictive psychostimulant that induces immediate early gene (IEG) expression by activating dopamine (DA) D1 and glutamate NMDA receptors in the striatum. In this study, we show that a single cocaine administration (30 mg/kg) time-dependently increases ERK phosphorylation, c-Fos and FosB protein expression, and MKP-1 phosphorylation (p-MKP-1), in the caudate-putamen (CPu) and nucleus accumbens (NAc) of Fischer rats. In the CPu, 1 h after cocaine injection, the increase in c-Fos and FosB protein expressions is totally abolished by pre-administration of DA-D1 receptor antagonist, SCH23390. In the NAc, SCH23390 also inhibits cocaine-induced c-Fos protein expression. The pre-treatment of NMDA receptor antagonist, MK801, partially reduces cocaine-activated c-Fos protein expression in the CPu. Furthermore, the escalation of p-MKP-1 after acute cocaine administration is dependent on both DA-D1 and NMDA receptor activation in both brain regions examined. Our data suggest that cocaine may modulate ERK pathway signaling through the activation of DA-D1 and NMDA receptors, subsequently influencing the IEG protein expression.


Subject(s)
Cocaine/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Proto-Oncogene Proteins c-fos/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cocaine-Related Disorders/genetics , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/physiopathology , Corpus Striatum/physiopathology , Disease Models, Animal , Dopamine/pharmacology , Dopamine Antagonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dual Specificity Phosphatase 1/drug effects , Dual Specificity Phosphatase 1/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, Immediate-Early/drug effects , Genes, Immediate-Early/genetics , Male , Phosphorylation/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Inbred F344 , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D1/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Time Factors , Up-Regulation/drug effects , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...