Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Scott J. Russo is active.

Publication


Featured researches published by Scott J. Russo.


Neuroscience | 2003

Gonadal hormones differentially modulate cocaine-induced conditioned place preference in male and female rats.

Scott J. Russo; Eugene D. Festa; Sosimo J. Fabian; F.M Gazi; M Kraish; Shirzad Jenab; Vanya Quinones-Jenab

There is accumulating evidence that suggests there are sex differences in behavioral and subjective responses to cocaine. However, it is not known whether differences in cocaine reward contribute to sex differences in these responses or whether gonadal hormones affect the rewarding properties of cocaine. In the present study, conditioned place preference (CPP), a measure of non-contingent reward, was used to determine the effects of endogenous gonadal hormones and of estrogen and progesterone replacement on cocaine reward. Neurochemical measurements were also taken to identify monoaminergic substrates which underlie the behavioral phenotype. Although both intact and gonadectomized male and female rats showed a significant CPP for cocaine, ovariectomy attenuated the magnitude of CPP. These alterations coincided with a decrease in serum levels of corticosterone. In ovariectomized rats, pretreatment with progesterone inhibited cocaine CPP while estrogen plus progesterone potentiated the magnitude of CPP. Additionally, gonadectomy and ovarian hormone replacement in female rats affected serotonin/dopamine levels and turnover ratios in the ventral tegmental area and nucleus accumbens shell. While no effects of castration were observed, ovariectomy decreased levels of dopamine and serotonin in the ventral tegmental area. In females, progesterone replacement increased levels of serotonin and dopamine in the ventral tegmental area, while estrogen plus progesterone replacement increased dopamine levels in the nucleus accumbens. Collectively, these results indicate that ovarian hormones may influence cocaine reward by altering monoaminergic systems, which, in turn, may contribute to the current sex disparities in overall cocaine use.


Neuropharmacology | 2004

Sex differences in cocaine-induced behavioral responses, pharmacokinetics, and monoamine levels

Eugene D. Festa; Scott J. Russo; Farhad M. Gazi; Tipyamol Niyomchai; Lynne M. Kemen; Shen-Nan Lin; Rodger L. Foltz; Shirzad Jenab; Vanya Quinones-Jenab

Female rats display a more robust behavioral response to acute cocaine administration than do male rats. However, a clear understanding of the biological mechanisms underlying these differences remains elusive. The present study investigated whether sexual dimorphisms in cocaine-induced motor behavior might be based on monoaminergic levels and/or cocaine pharmacokinetics. An acute injection of cocaine (5, 15, 20 or 30 mg/kg) or saline was administered to male and female rats, and behavioral activity was monitored for 3 h. Following acute cocaine or saline administration motor behavior varied according to dose and sex; overall, female rats displayed greater rearing counts and stereotypic scores, greater total locomotor counts at 15, 20, and 30 mg/kg of cocaine, and greater ambulatory counts at 20 and 30 mg/kg of cocaine than did male rats. Neurochemical determinations in post-mortem tissue showed that both male and female rats had increases in total dopamine (DA) in the caudate putamen (CPu) 15 min following cocaine administration. Additionally, male rats had a decrease in dihydroxyphenylacetic acid (DOPAC)/DA turnover. Female rats showed significant reductions in total levels of DA, DOPAC, HVA, serotonin (5-HT), 5-hydroxyindole acetic acid (5-HIAA), and DOPAC/DA turnover in the nucleus accumbens (NAc). Male rats displayed a reduction only in DOPAC/DA turnover and increases in 5-HT in the NAc following cocaine administration. Furthermore, sex differences in cocaine metabolism were observed where females had greater brain/blood levels of norcocaine and ecgonine methyl ester while male rats had higher blood levels of benzoylecgonine. These results suggest that sex differences in the behavioral responses to cocaine administration could be explained in part by intrinsic differences in both monoaminergic levels and metabolic processes.


Brain Research Bulletin | 2004

The role of D1 and D2 receptors in the cocaine conditioned place preference of male and female rats.

Arbi Nazarian; Scott J. Russo; Eugene D. Festa; Mohammed Kraish; Vanya Quinones-Jenab

The rewarding effects of cocaine have been shown to be sexually dimorphic; female rats develop cocaine conditioned place preference at lower doses and with fewer cocaine pairings than male rats. The present study was conducted to determine whether D1 and D2 receptors contribute to sex differences in cocaine conditioned place preference using a 4-day paradigm. Fifteen minutes prior to receiving saline or cocaine (5mg/kg for females and 20mg/kg for males), rats were pretreated with either SCH 23390, a D1 receptor antagonist, (0.10, 0.25, or 0.50mg/kg), eticlopride, a D2 receptor antagonist, (0.05, 0.10, or 0.25mg/kg), or vehicle (saline). Antagonism of D1 receptors by SCH 23390 fully blocked cocaine conditioned place preference in male rats, while only the two lower doses of SCH 23390 blocked cocaine conditioned place preference in female rats. Conversely, antagonism of D2 receptors using eticlopride had no effect on cocaine conditioned place preference in male or female rats. Due to the known role of D1 receptors in cocaine conditioned place preference, sex differences in D1 receptor sensitivity may explain the differences observed in cocaine reward between male and female rats.


Annals of the New York Academy of Sciences | 2006

Ovarian Hormones Modulate Cocaine-Induced Locomotor and Stereotypic Activity

Linda I. Perrotti; Scott J. Russo; Helen Fletcher; Juliet Chin; Tyler Webb; Shirzad Jenab; Vanya Quinones-Jenab

Abstract: Interactions between ovarian hormones and cocaine may underlie gender and estrous cycle differences in cocaine‐induced behavioral and neurochemical alterations. The aim of this study was to further understand how ovarian hormones modulate cocaine‐induced behavioral alterations. Ovariectomized rats received acute or chronic saline or cocaine (15 mg/kg i.p.) administration and were further subdivided into one of four hormone‐treatment conditions: cholesterol (vehicle‐control), estrogen, progesterone, or estrogen+progesterone. Overall, acute and chronic cocaine administration increased all locomotor measurements (total locomotor, ambulatory, and rearing counts). Estrogen administration augmented cocaine‐induced increases in ambulatory and rearing activity. After chronic cocaine administration, rats in the vehicle‐control group developed behavioral tolerance (exhibited by a decrease in activity) in rearing and ambulatory activity. Estrogen replacement not only prevented the development of tolerance in ambulatory and rearing activities, but also enhanced total locomotor activity (sensitization) in response to chronic cocaine. Progesterone treatment did not alter the behavioral responses to acute or chronic cocaine administration. Estrogen+progesterone‐treated animals had higher counts of locomotor activity in response to chronic cocaine than did vehicle‐control or progesterone‐treated rats. In stereotypic behaviors, the different hormonal treatments did not affect activity in cocaine‐ or saline‐treated rats after acute or chronic drug administration. Plasma levels of cocaine did not change after different hormonal treatments. Interestingly, animals coadministered chronic cocaine and estrogen had higher levels of corticosterone than did nonestrogen cocaine‐treated rats. Thus, it is likely that alterations in HPA activation may underlie the observed behavioral differences. In summary, these data suggest that there are interactions between ovarian hormones and cocaine‐induced behavioral alterations in female rats, and they extend previous results by showing that estrogen and progesterone affect the development of sensitization.


Pharmacology, Biochemistry and Behavior | 2005

Progesterone inhibits behavioral responses and estrogen increases corticosterone levels after acute cocaine administration.

Tipyamol Niyomchai; Scott J. Russo; Eugene D. Festa; Alaleh Akhavan; Shirzad Jenab; Vanya Quinones-Jenab

Accumulating evidence suggests that estrogen and progesterone contribute to the sexually dimorphic behavioral response to cocaine. In this study, we tested the hypothesis that varying the level of estrogen or progesterone affects cocaine-induced locomotive behavior in female rats. Ovariectomized (OVX) rats received estrogen (0, 5, 10, 20, or 50 microg) 48 h or progesterone (0, 50, 100, 250, or 500 microg) 24 h before acute saline or cocaine (15 mg/kg) administration. Although estrogen did not affect cocaine-induced ambulatory and rearing behaviors, it affected stereotypic behaviors regardless of cocaine administration (animals receiving 50 microg had higher stereotypic counts than did the OVX group). In contrast, progesterone affected rearing activity dose-dependently: 50 and 500 microg of progesterone inhibited, whereas 100 microg and 250 microg stimulated, rearing in response to cocaine. That estrogen and progesterone did not affect overall baseline behavioral activity suggests their effects are mediated in part through interactions with cocaine. Progesterone administration did not affect corticosterone levels in saline- or cocaine-treated rats. Estrogen administration, however, affected levels of corticosterone both at baseline and after cocaine treatment. After accounting for baseline differences, we found that rats receiving 5 or 10 microg of estrogen and cocaine had higher percentage increases in serum corticosterone levels than did the control group that did not receive estrogen. On the basis of these observations, we suggest that progesterone fluctuations during the estrous cycle impact cocaine-induced behavioral responses, whereas estrogen may affect activity in the hypothalamic-pituitary-adrenal axis. Thus, dose-dependent effects of gonadal hormones may underlie some of the reported sex differences and reproductive cycle effects of cocaine.


Brain Research | 2008

Progesterone attenuates cocaine-induced conditioned place preference in female rats

Scott J. Russo; Wei Lun Sun; Ana Christina Minerly; Karen Weierstall; Arbi Nazarian; Eugene D. Festa; Tipyamol Niyomchai; Alaleh Akhavan; Victoria N. Luine; Shirzad Jenab; Vanya Quinones-Jenab

Progesterone replacement attenuates the intensity of cocaine-induced conditioned place preference (CPP) behaviors in female rats. The present study aimed to expand that finding by (i) determining the role of progesterone in the acquisition and/or expression of cocaine-induced CPP and (ii) determining if progesterones effects might be meditated through learning and memory. To this end, female rats were administered progesterone during cocaine conditioning or object recognition tasks; rats received subcutaneous injections of progesterone (500 microg) or vehicle (sesame oil) 4 h before saline or cocaine (5 mg/kg) on conditioning days (acquisition phase) or before testing (expression phase or object recognition tasks). Progesterone treatment during both the acquisition and the expression phases of cocaine conditioning blocked cocaine-induced CPP. Progesterone affected neither the number of entrances and explorations in the CPP chambers nor the ambulatory and rearing behaviors. In the object recognition task (a non-spatial learning and memory task), progesterone treatment had no effect. However, in the object placement task (a spatial learning and memory task), progesterone treatment significantly impaired retention in hormone-treated rats as compared with control groups. These results suggest that progesterone treatment interferes with cocaine-induced reward associations, possibly through effects on spatial working memory consolidation The observed effects of acute progesterone treatment on cocaine-induced CPP may in part contribute reported menstrual effects and sex disparities in overall cocaine use and rates of relapse.


Annals of the New York Academy of Sciences | 2006

Endocrinological basis of sex differences in cocaine-induced behavioral responses

Vanya Quinones-Jenab; Linda I. Perrotti; Sosimo J. Fabian; Juliet Chin; Scott J. Russo; Shirzad Jenab

Abstract: Currently, 1.8 million Americans use cocaine, 30% of whom are females. Sex differences in the pattern of cocaine abuse may reside in neuroendocrinological modulations that affect the use of and/or dependence on cocaine. This review discusses sex differences in cocaine‐induced behavioral and molecular alterations in the central nervous system, with emphasis on the role of endocrine responses in the neuronal modulations of this drug. Mechanisms and data supporting the role of the hypothalamic‐gonadal axis in the modulation of cocaine‐induced behavioral and molecular alterations are also provided.


Brain Research Bulletin | 2006

Cocaine-induced sex differences in D1 receptor activation and binding levels after acute cocaine administration

Eugene D. Festa; Shirzad Jenab; Jan Weiner; Arbi Nazarian; Tipyamol Niyomchai; Scott J. Russo; Lynne M. Kemen; Alaleh Akhavan; Hui-Bing K. Wu; Vanya Quinones-Jenab

Although it is established 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 whether dopamine (DA) receptor activation influences sex differences in cocaine-induced behaviors. A second study was performed to determine sex differences in D1/D2 receptor levels prior to and post-cocaine administration. Male and female Fischer rats were pre-treated with the D1 antagonist SCH-23390 (0.05, 0.1, and 0.25 mg/kg, i.p.), the D2 antagonist eticlopride (0.03, 0.1 mg/kg, i.p.), or vehicle (saline) 15 min before acute cocaine (20 mg/kg, i.p.) or saline administration. Cocaine-induced ambulatory and rearing activity was greater in female than male rats. Pre-treatment with SCH-23390 affected cocaine-induced ambulatory, rearing, and stereotypic activity in a sex-dependent manner; cocaine-induced ambulatory and stereotypic behavior in female rats was reduced by the lowest dose of SCH-23390. Eticlopride did not alter behavioral responses to cocaine in male or female rats. These results suggest that in both male and female rats, activation of the D1, but not the D2, receptor modulates cocaines motor effects. There were no sex differences in baseline levels of D1, D2, and DA transporter binding in the caudate putamen (CPu) and the nucleus accumbens (NAc). Cocaine administration reduced D1 binding levels in the CPu only in male rats. Our findings suggest that the regulation of striatal D1 binding levels after acute cocaine administration is a sexually dimorphic process. We also hypothesize that the greater sensitivity to D1 receptor blockade in female rats, as compared to male rats, may contribute to their overall increased hyperactivity in response to acute cocaine. Taken together, the D1 receptor may be an important substrate in the regulation of sex differences to cocaine-induced locomotor activity.


Brain Research Bulletin | 2002

Effects of cocaine on c-fos and preprodynorphin mRNA levels in intact and ovariectomized Fischer rats.

Shirzad Jenab; Tipyamol Niyomchai; Juliet Chin; Eugene D. Festa; Scott J. Russo; Linda I. Perrotti; Vanya Quinones-Jenab

Psychostimulants such as cocaine have been shown to regulate c-fos and opioid gene expression in male rats. However, little information is available on cocaine effects in female rats or how the ovarian hormones, estrogen and progesterone, modulate these effects. In this study we used quantitative solution hybridization assays to measure c-fos and preprodynorphin (PDYN) mRNA levels after cocaine administration in the caudate/putamen of intact male and female rats or ovariectomized (OVX) female rats that were pretreated with vehicle, estrogen and/or progesterone. The c-fos mRNA levels were increased in intact male and female rats after 30min or 3h of one single cocaine injection and after 14 days of single daily cocaine injections. The c-fos mRNA levels were also increased after 30min of a single cocaine injection in OVX female rats that were treated with vehicle, estrogen and/or progesterone. The PDYN mRNA levels did not change after 30min, 3h or 14 days in intact male or female rats. However, PDYN mRNA levels were increased in the caudate/putamen of OVX female rats pretreated with vehicle or a combination of estrogen and progesterone but not in OVX female rats that were pretreated with either estrogen or progesterone alone. Our data suggest hormonal regulation of cocaine effects on PDYN mRNA levels which may modulate cocaine-induced behaviors in female rats.


Molecular Brain Research | 2003

MK-801 attenuates cocaine induction of c-fos and preprodynorphin mRNA levels in Fischer rats.

Shirzad Jenab; Eugene D. Festa; Scott J. Russo; Hui Bing K. Wu; Charles E. Inturrisi; Vanya Quinones-Jenab

This study shows that single (15 mg/kg) or binge (3x15 mg/kg) cocaine administration increases c-fos mRNA levels, but only binge cocaine administration increases preprodynorphin mRNA levels in the caudate/putamen of Fischer rats. This increase in both c-fos and preprodynorphin mRNA levels is attenuated by the noncompetitive NMDA antagonist MK-801 (0.25 mg/kg), suggesting a preferential role for NMDA receptor co-activation in cellular events leading to cocaine-induced behaviors.

Collaboration


Dive into the Scott J. Russo's collaboration.

Top Co-Authors

Avatar

Shirzad Jenab

City University of New York

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eugene D. Festa

City University of New York

View shared research outputs
Top Co-Authors

Avatar

Tipyamol Niyomchai

City University of New York

View shared research outputs
Top Co-Authors

Avatar

Arbi Nazarian

City University of New York

View shared research outputs
Top Co-Authors

Avatar

Juliet Chin

City University of New York

View shared research outputs
Top Co-Authors

Avatar

Linda I. Perrotti

University of Texas at Arlington

View shared research outputs
Top Co-Authors

Avatar

Alaleh Akhavan

City University of New York

View shared research outputs
Top Co-Authors

Avatar

Hui Bing K. Wu

City University of New York

View shared research outputs
Top Co-Authors

Avatar

Lynne M. Kemen

City University of New York

View shared research outputs
Researchain Logo
Decentralizing Knowledge