Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38866486

RESUMEN

We investigated sex differences in dopamine (DA) release in the nucleus accumbens (NAc) and dorsolateral striatum (DLS) using a chronic 16-channel carbon fiber electrode and fast-scan cyclic voltammetry (FSCV). Electrical stimulation (ES; 60  Hz) induced DA release was recorded in the NAc of single or pair-housed male and female rats. When core (NAcC) and shell (NAcS) were recorded simultaneously, there was greater ES DA release in NAcC of pair-housed females compared with single females and males. Housing did not affect ES NAc DA release in males. In contrast, there was significantly more ES DA release from the DLS of female rats than male rats. This was true prior to and after treatment with methamphetamine. Furthermore, in castrated (CAST) males and ovariectomized (OVX) females, there were no sex differences in ES DA release from the DLS, demonstrating the hormone dependence of this sex difference. However, in the DLS of both intact and gonadectomized rats, DA reuptake was slower in females than in males. Finally, DA release following ES of the medial forebrain bundle at 60  Hz was studied over four weeks. ES DA release increased over time for both CAST males and OVX females, demonstrating sensitization. Using this novel 16-channel chronic FSCV electrode, we found sex differences in the effects of social housing in the NAcS, sex differences in DA release from intact rats in DLS, sex differences in DA reuptake in DLS of intake and gonadectomized rats, and we report sensitization of ES-induced DA release in DLS in vivo.Significance Statement Dopamine release is not uniform or fixed. In the nucleus accumbens, pair housing, compared with individual housing, is shown to differentially affect dopamine responsiveness to stimulation in a sex-dependent and region-specific way. There are also sex differences in stimulated dopamine release in the dorsolateral striatum of intact rats, which are not seen in gonadectomized rats, indicating the hormone dependence of this sex difference. However, reuptake of dopamine was slower in females than in males, independent of gonadal hormones. Importantly, the electrical stimulation-induced dopamine release in the dorsolateral striatum of gonadectomized rats demonstrated sensitization of dopamine release in vivo within animals for the first time. Thus, stimulated dopamine release exhibits sex-specific neuroplasticity that is modified in females by the housing conditions.

2.
Neuromodulation ; 27(2): 343-352, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36609088

RESUMEN

OBJECTIVES: There are limited treatment options for female sexual dysfunction (FSD). Percutaneous tibial nerve stimulation (PTNS) has shown improvements in FSD symptoms in neuromodulation clinical studies, but the direct effects on sexual function are not understood. This study evaluated the immediate and long-term effects of PTNS on sexual motivation and receptivity in a rat model of menopausal women. Our primary hypothesis was that long-term PTNS would yield greater changes in sexual behavior than short-term stimulation. MATERIALS AND METHODS: In two experiments, after receiving treatment, we placed ovariectomized female rats in an operant chamber in which the female controls access to a male by nose poking. We used five treatment conditions, which were with or without PTNS and no, partial, or full hormone priming. In experiment 1, we rotated rats through each condition twice with behavioral testing immediately following treatment for ten weeks. In experiment 2, we committed rats to one condition for six weeks and tracked sexual behavior over time. We quantified sexual motivation and sexual receptivity with standard measures. RESULTS: No primary comparisons were significant in this study. In experiment 1, we observed increased sexual motivation but not receptivity immediately following PTNS with partial hormone priming, as compared with priming without PTNS (linear mixed effect models; initial latency [p = 0.34], inter-interval latency [p = 0.77], nose poke frequency [p = 0.084]; eight rats). In experiment 2, we observed trends of increased sexual receptivity (linear correlation for weekly group means; mounts [p = 0.094 for trendline], intromissions [p = 0.073], lordosis quotient [p = 0.58], percent time spent with a male [p = 0.39], decreased percent time alone [p = 0.024]; four rats per condition), and some sexual motivation metrics (linear correlation for weekly group means; nose pokes per interval [p = 0.050], nose poke frequency [p = 0.039], decreased initial latency [p = 0.11]; four rats per condition) when PTNS was applied long-term with partial hormone priming, as compared with hormone-primed rats without stimulation. CONCLUSIONS: PTNS combined with hormone priming shows potential for increasing sexual motivation in the short-term and sexual receptivity in the long-term in rats. Further studies are needed to examine variability in rat behavior and to investigate PTNS as a treatment for FSD in menopausal women.


Asunto(s)
Estimulación Eléctrica Transcutánea del Nervio , Humanos , Masculino , Femenino , Ratas , Animales , Conducta Sexual , Nervio Tibial/fisiología , Motivación , Hormonas , Resultado del Tratamiento
3.
J Neurosci ; 42(45): 8477-8487, 2022 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-36351834

RESUMEN

Sex differences in motivation for food rewards, gambling, and drugs of abuse are modulated by multiple factors, including sensory stimuli, gonadal hormones, and cognitive bias. Cues, drugs of abuse, and a high-fat diet can significantly impact neural signaling in the reward system and functioning of neural systems that regulate executive functions differentially in males and females. Additionally, sex differences in risky decision-making, cognitive bias, and motivation for food and drugs of abuse are mediated by gonadal hormones in both sexes. As neuroscientists analyze data from both sexes, it is becoming apparent that these differences are not simply mediated by hormones in females, but involve sex differences in the specific neural responses to stimuli, including both external stimuli and internal hormonal signals. Understanding sex differences in the mechanisms underlying reward-seeking behaviors and the development of substance use disorders will help uncover potential therapies and treatments that will benefit both men and women. Based on these observations, it is essential that females are included in neuroscience research.


Asunto(s)
Juego de Azar , Femenino , Humanos , Masculino , Juego de Azar/psicología , Motivación , Caracteres Sexuales , Recompensa , Cognición
4.
FASEB J ; 35(2): e21303, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33433026

RESUMEN

In the midst of the current coronavirus pandemic, the United States continues to struggle with an ongoing opioid epidemic, initially fueled by widespread prescribing of opioid medications during the 1990s. The primary reason for prescribing opioids is to treat pain. Women have more acute and chronic pain and have been prescribed these drugs in significantly greater numbers than men. Comparison of women and men with chronic pain also shows that women receive the majority of prescription opioids, and the use of these prescribed medications became the major pathway to misuse and addiction for women. Yet, recognition of the extent of women's exposure to opioids and the attendant consequences has been limited. Attempts to stem the overall tide of the epidemic focused on reducing the availability of prescription opioids. However, as these medications became more difficult to obtain and treatment opportunities were limited, many turned to other synthetic opioids, such as heroin and fentanyl. Thus, the public health crisis of opioid addiction has endured. This paper highlights the importance of understanding differences among women and men in opioid use and its biological and psychosocial effects to advance the gender-based treatment approaches and effective public health policy.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Dolor Crónico/tratamiento farmacológico , Trastornos Relacionados con Opioides/epidemiología , Factores Sexuales , COVID-19 , Femenino , Humanos , Masculino , Trastornos Relacionados con Opioides/terapia , Pandemias , Prescripciones/estadística & datos numéricos , Estados Unidos/epidemiología
5.
Horm Behav ; 130: 104949, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33609527

RESUMEN

There are sex differences in the response to psychomotor stimulants, where females exhibit a greater response than males, due to the presence of the gonadal hormone estradiol (E2). Extensive research has shown that E2 enhances drug-seeking and the rewarding properties of cocaine for females. The role of E2 in male drug-seeking, however, is not well understood. The current study investigated pharmacological manipulation of E2 receptors in the dorsolateral striatum (DLS) on preference for cocaine in gonad-intact male and female rats. In males, activation of G-protein coupled E2 receptor 1 (GPER1), via administration of ICI 182,780 or G1, attenuated conditioned place preference for 10 mg/kg cocaine, while inhibition of GPER1, via G15, enhanced preference at a 5 mg/kg cocaine dose. Similarly, GPER1 activation, via G1, prevented males from forming a preference for 0.1% saccharin (SACC) versus plain water. Surprisingly, activation of GPER1 did not alter preference for cocaine or SACC in females. These studies also examined the quantity of E2 receptor mRNA in the dorsal striatum, using qPCR. No sex differences in relative mRNA expression of ERα, ERß, and GPER1 were observed. However, there was greater GPER1 mRNA, relative to ERα and ERß, in both males and females. The results presented here indicate that E2, acting via GPER1, may be protective against drug preference in male rats.


Asunto(s)
Cocaína , Receptor alfa de Estrógeno , Animales , Cocaína/farmacología , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Proteínas de Unión al GTP , Masculino , Ratas , Receptores de Estradiol , Receptores Acoplados a Proteínas G/metabolismo , Sacarina/farmacología
6.
Pharmacol Rev ; 68(2): 242-63, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26772794

RESUMEN

The purpose of this review is to discuss ways to think about and study sex differences in preclinical animal models. We use the framework of addiction, in which animal models have excellent face and construct validity, to illustrate the importance of considering sex differences. There are four types of sex differences: qualitative, quantitative, population, and mechanistic. A better understanding of the ways males and females can differ will help scientists design experiments to characterize better the presence or absence of sex differences in new phenomena that they are investigating. We have outlined major quantitative, population, and mechanistic sex differences in the addiction domain using a heuristic framework of the three established stages of the addiction cycle: binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation. Female rats, in general, acquire the self-administration of drugs and alcohol more rapidly, escalate their drug taking with extended access more rapidly, show more motivational withdrawal, and (where tested in animal models of "craving") show greater reinstatement. The one exception is that female rats show less motivational withdrawal to alcohol. The bases for these quantitative sex differences appear to be both organizational, in that estradiol-treated neonatal animals show the male phenotype, and activational, in that the female phenotype depends on the effects of gonadal hormones. In animals, differences within the estrous cycle can be observed but are relatively minor. Such hormonal effects seem to be most prevalent during the acquisition of drug taking and less influential once compulsive drug taking is established and are linked largely to progesterone and estradiol. This review emphasizes not only significant differences in the phenotypes of females and males in the domain of addiction but emphasizes the paucity of data to date in our understanding of those differences.


Asunto(s)
Modelos Animales de Enfermedad , Trastornos Relacionados con Sustancias , Animales , Humanos , Caracteres Sexuales , Trastornos Relacionados con Sustancias/metabolismo
7.
FASEB J ; 31(1): 29-34, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27682203

RESUMEN

In June 2015, the National Institutes of Health (NIH) released a Guide notice (NOT-OD-15-102) that highlighted the expectation of the NIH that the possible role of sex as a biologic variable be factored into research design, analyses, and reporting of vertebrate animal and human studies. Anticipating these guidelines, the NIH Office of Research on Women's Health, in October 2014, convened key stakeholders to discuss methods and techniques for integrating sex as a biologic variable in preclinical research. The workshop focused on practical methods, experimental design, and approaches to statistical analyses in the use of both male and female animals, cells, and tissues in preclinical research. Workshop participants also considered gender as a modifier of biology. This article builds on the workshop and is meant as a guide to preclinical investigators as they consider methods and techniques for inclusion of both sexes in preclinical research and is not intended to prescribe exhaustive/specific approaches for compliance with the new NIH policy.-Miller, L. R., Marks, C., Becker, J. B., Hurn, P. D., Chen, W.-J., Woodruff, T., McCarthy, M. M., Sohrabji, F., Schiebinger, L., Wetherington, C. L., Makris, S., Arnold, A. P., Einstein, G., Miller, V. M., Sandberg, K., Maier, S., Cornelison, T. L., Clayton, J. A. Considering sex as a biological variable in preclinical research.


Asunto(s)
Investigación Biomédica/normas , Evaluación Preclínica de Medicamentos , National Institutes of Health (U.S.)/normas , Femenino , Humanos , Masculino , Factores Sexuales , Estados Unidos
8.
Horm Behav ; 104: 119-129, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29626485

RESUMEN

Contribution to Special Issue on Fast effects of steroids. Estradiol and progesterone rapidly induce changes in dopaminergic signaling within the dorsal striatum and nucleus accumbens of female rats. In ovariectomized females, estradiol rapidly enhances dopamine release and modulates binding of dopamine receptors. Progesterone further potentiates the effect of estradiol on dopamine release. The effects of both estradiol and progesterone are time course dependent, with increases in dopamine release immediately after acute hormone administration followed by later inhibition of dopamine release. Importantly, these changes are also seen in naturally cycling females, indicating their importance for normal physiological states and relevant reproductive behaviors. Here, we summarize the literature establishing the rapid effects of estradiol and progesterone on dopamine release and receptor expression in dorsal striatum and nucleus accumbens of both males and females. Integrating this literature with the larger body of work focusing on dopamine regulated behaviors, we propose hypotheses for adaptive reasons (i.e., ultimate causes) as to why changes in ovarian hormones modulate dopamine release. Finally, we note the importance of these studies for understanding sex differences in vulnerability to drug addiction. Research on how dopaminergic systems regulate behavior in both males and females is crucial for developing a full appreciation of dopamine's role in both natural and drug-induced behaviors.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Estradiol/farmacología , Núcleo Accumbens/efectos de los fármacos , Progesterona/farmacología , Animales , Cuerpo Estriado/fisiología , Estradiol/metabolismo , Femenino , Masculino , Núcleo Accumbens/fisiología , Ovario/metabolismo , Progesterona/metabolismo , Ratas , Caracteres Sexuales , Factores de Tiempo
9.
J Neurosci Res ; 95(1-2): 136-147, 2017 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-27870394

RESUMEN

This review discusses alcohol and other forms of drug addiction as both a sociocultural and biological phenomenon. Sex differences and gender are not solely determined by biology, nor are they entirely sociocultural. The interactions among biological, environmental, sociocultural, and developmental influences result in phenotypes that may be more masculine or more feminine. These gender-related sex differences in the brain can influence the responses to drugs of abuse, progressive changes in the brain after exposure to drugs of abuse and whether addiction results from drug-taking experiences. In addition, the basic laboratory evidence for sex differences is discussed within the context of four types of sex/gender differences. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Encéfalo/patología , Caracteres Sexuales , Trastornos Relacionados con Sustancias , Humanos , Factores Sexuales , Trastornos Relacionados con Sustancias/epidemiología , Trastornos Relacionados con Sustancias/patología
10.
Addict Biol ; 21(5): 995-1006, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27029841

RESUMEN

Among mammals, every cell has a biological sex, and the sex of an individual pervades its body and brain. In this review, we describe the processes through which mammals become phenotypically male or female by organizational and activational influences of genes and hormones throughout development. We emphasized that the molecular and cellular changes triggered by sex chromosomes and steroid hormones may generate sex differences in overt physiological functions and behavior, but they may alternatively promote end-point convergences between males and females. Clinical and pre-clinical evidences suggest that sex and gender differences modulate drug consumption as well as of the transition towards drug-promoted pathological states such as dependence and addiction. Additionally, sex differences in drug pharmacokinetics and pharmacodynamics will also influence dependence and addiction as well as side effects of drugs. These effects will further interact with socially gendered factors to result in sex differences in the access to, engagement in and efficacy of any therapeutic attempt. Finally, we maintain that 'sex sameness' is as important as 'sex differences' when building a complete understanding of biology for both males and females and provide a framework with which to classify and guide investigation into the mechanisms mediating sex differences and sex sameness.


Asunto(s)
Conducta Adictiva/fisiopatología , Investigación , Caracteres Sexuales , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Factores Sexuales
11.
Addict Biol ; 21(5): 1052-9, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26935336

RESUMEN

In this review, we discuss the importance of investigating both sex and gender differences in addiction and relapse in studies of humans and in animal models. Addiction is both a cultural and biological phenomenon. Sex and gender differences are not solely determined by our biology, nor are they entirely cultural; they are interactions between biology and the environment that are continuously played out throughout development. Lessons from the historical record illustrate how context and attitudes affect the way that substance use in men and women is regarded. Finally, cultural and environmental influences may differentially affect men and women, and affect how they respond to drugs of abuse and to treatment protocols. We recommend that both animal models and clinical research need to be developed to consider how contextual and social factors may influence the biological processes of addiction and relapse differentially in men and women.


Asunto(s)
Cultura , Caracteres Sexuales , Conducta Social , Trastornos Relacionados con Sustancias/fisiopatología , Femenino , Humanos , Masculino , Factores Sexuales
12.
bioRxiv ; 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-37645814

RESUMEN

We investigated sex differences in dopamine (DA) release in the nucleus accumbens (NAc) and dorsolateral striatum (DLS) using a chronic 16-channel carbon fiber electrode and fast-scan cyclic voltammetry (FSCV). Electrical stimulation (ES; 60Hz) induced DA release was recorded in the NAc of single or pair-housed male and female rats. When core (NAcC) and shell (NAcS) were recorded simultaneously, there was greater ES DA release in NAcC of pair-housed females compared with single females and males. Housing did not affect ES NAc DA release in males. In contrast, there was significantly more ES DA release from the DLS of female rats than male rats. This was true prior to and after treatment with methamphetamine. Furthermore, in castrated (CAST) males and ovariectomized (OVX) females, there were no sex differences in ES DA release from the DLS, demonstrating the hormone dependence of this sex difference. However, in the DLS of both intact and gonadectomized rats, DA reuptake was slower in females than in males. Finally, DA release following ES of the medial forebrain bundle at 60Hz was studied over four weeks. ES DA release increased over time for both CAST males and OVX females, demonstrating sensitization. Using this novel 16-channel chronic FSCV electrode, we found sex differences in the effects of social housing in the NAcS, sex differences in DA release from intact rats in DLS, sex differences in DA reuptake in DLS of intake and gonadectomized rats, and we report sensitization of ES-induced DA release in DLS in vivo.

13.
J Neurosci Methods ; 401: 110003, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37918446

RESUMEN

Recently, many funding agencies have released guidelines on the importance of considering sex as a biological variable (SABV) as an experimental factor, aiming to address sex differences and avoid possible sex biases to enhance the reproducibility and translational relevance of preclinical research. In neuroscience and pharmacology, the female sex is often omitted from experimental designs, with researchers generalizing male-driven outcomes to both sexes, risking a biased or limited understanding of disease mechanisms and thus potentially ineffective therapeutics. Herein, we describe key methodological aspects that should be considered when sex is factored into in vitro and in vivo experiments and provide practical knowledge for researchers to incorporate SABV into preclinical research. Both age and sex significantly influence biological and behavioral processes due to critical changes at different timepoints of development for males and females and due to hormonal fluctuations across the rodent lifespan. We show that including both sexes does not require larger sample sizes, and even if sex is included as an independent variable in the study design, a moderate increase in sample size is sufficient. Moreover, the importance of tracking hormone levels in both sexes and the differentiation between sex differences and sex-related strategy in behaviors are explained. Finally, the lack of robust data on how biological sex influences the pharmacokinetic (PK), pharmacodynamic (PD), or toxicological effects of various preclinically administered drugs to animals due to the exclusion of female animals is discussed, and methodological strategies to enhance the rigor and translational relevance of preclinical research are proposed.


Asunto(s)
Proyectos de Investigación , Caracteres Sexuales , Animales , Masculino , Femenino , Reproducibilidad de los Resultados , Factores Sexuales , Tamaño de la Muestra
14.
Horm Behav ; 64(4): 573-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24013034

RESUMEN

Estradiol is thought to play a critical role in the increased vulnerability to psychostimulant abuse in women. Sex differences in the ability of estradiol to influence cocaine self-administration in adult rats have been hypothesized to depend upon pubertal estradiol exposure. The current study investigated whether the presence of gonadal hormones during puberty affected cocaine self-administration behavior and its sensitivity to adult estradiol treatment in male and female Sprague-Dawley rats. Subjects were gonadectomized or SHAM-operated at postnatal day (PD) 22, and received either OIL or estradiol benzoate (EB) during the approximate time of puberty (PD27 to PD37). Adult rats were subsequently treated with either EB or OIL 30 min before cocaine self-administration (0.3 mg/kg/inf) in order to examine the effects of pubertal manipulations on the estradiol sensitivity of acquisition on a fixed ratio (FR) 1 schedule, total intake on a FR5 schedule and motivation on a progressive ratio schedule. Adult EB treatment only affected cocaine self-administration in females, which is consistent with previous research. Adult EB treatment enhanced acquisition in all females irrespective of puberty manipulations. All females, except those treated with EB during puberty, displayed increased cocaine intake following adult EB treatment. Adult EB treatment only enhanced motivation in females that were intact during puberty, whereas those treated with EB during puberty showed reduced motivation. Therefore, the sensitivities of different self-administration behaviors to adult estradiol treatment are organized independently in females, with pubertal estradiol exerting a greater influence over motivational processes, and negligible effects on learning/acquisition.


Asunto(s)
Cocaína/administración & dosificación , Estradiol/farmacología , Maduración Sexual/efectos de los fármacos , Factores de Edad , Animales , Castración , Trastornos Relacionados con Cocaína/etiología , Trastornos Relacionados con Cocaína/metabolismo , Femenino , Masculino , Ratas , Ratas Sprague-Dawley , Autoadministración , Factores Sexuales
15.
Artículo en Inglés | MEDLINE | ID: mdl-36285173

RESUMEN

Background: Excessive alcohol and tobacco use are risk factors for poor health in both men and women, but use patterns and relationships with diseases and mortality differ between sexes. The impact of substance use on the epigenome, including DNA methylation profiles, may also differ by sex. It is also unknown whether parental substance use during childhood is associated with epigenetic changes that persist into adulthood. This study assessed the sex-specific effects of individuals' alcohol and tobacco use, as well as paternal alcohol and paternal/maternal tobacco use, on offspring's cellular aging as measured by epigenetic age acceleration. Methods: Four measures of epigenetic age acceleration (HorvathAA, HannumAA, PhenoAA, and GrimAA), the difference between chronological age and inferred age based on DNA methylation, were estimated from saliva samples. Linear mixed models tested associations between alcohol/tobacco use and epigenetic age acceleration in parents and offspring. Results: Current tobacco smoking was associated with a 4.61-year increase in GrimAA, and former tobacco smoking was associated with a 3.60-year increase in HannumAA after accounting for multiple testing (p < 0.0125). In males only, current tobacco smoking was nominally associated with a 2.19-year increase in HannumAA (p < 0.05), and this effect was significantly different than the female-specific effect (p < 0.0125). Paternal heavy alcohol use when the offspring was 12 or younger was associated with a 4.43-year increase in GrimAA among offspring (p < 0.0125). Conclusions: This study found evidence of sex-specific effects of alcohol and tobacco use, as well as paternal heavy alcohol use, on epigenetic age acceleration.

16.
Biol Sex Differ ; 12(1): 46, 2021 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-34391470

RESUMEN

BACKGROUND: Estradiol potentiates drug-taking behaviors, including motivation to self-administer cocaine and reinstatement of drug-seeking after extinction in females, but not males. The dorsolateral stratum (DLS) is a region of the brain implicated in mediating drug-seeking behaviors and, more specifically, is a target brain area to study how estradiol regulates these behaviors. The estradiol receptors α, ß, and G protein-coupled estradiol receptor 1 (GPER1) are all present in the DLS. In this study, the effects of activating GPER1 in the DLS on drug-seeking are investigated. METHODS: Gonad-intact male and female rats were trained to self-administer cocaine (0.4 mg/kg/inf) on a fixed-ratio 1 schedule of reinforcement. For 4 weeks, animals underwent testing on a progressive ratio schedule of reinforcement to determine their motivation to attain cocaine. Halfway through progressive ratio testing, a selective agonist targeting GPER1 (G1) was administered intra-DLS to determine the contribution of GPER1 activation on motivation for cocaine. The effects of intra-DLS GPER1 activation on drug-induced reinstatement after extinction were subsequently determined. RESULTS: Activation of GPER1, via intra-DLS G1 administration, potentiated females' motivation to self-administer cocaine. There was no effect of prior G1 treatment on extinction of cocaine-taking in females; however, G1 treatment resulted in greater drug-induced reinstatement (10 mg/kg cocaine, i.p.). There were no effects of intra-DLS GPER1 activation observed on motivation for cocaine or cocaine-induced reinstatement of responding in males. CONCLUSIONS: These results support the conclusion that activation of GPER1 in the DLS enhances cocaine-seeking behaviors for female, but not male rats.


Asunto(s)
Trastornos Relacionados con Cocaína , Cocaína , Preparaciones Farmacéuticas , Animales , Estradiol , Femenino , Proteínas de Unión al GTP , Masculino , Motivación , Ratas , Receptores de Estradiol
17.
Neuropharmacology ; 187: 108491, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33567305

RESUMEN

This article reviews evidence for sex differences in vulnerability to addiction with an emphasis on the neural mechanisms underlying these differences. Sex differences in the way that the gonadal hormone, estradiol, interacts with the ascending telencephalic dopamine system results in sex differences in motivated behaviors, including drug-seeking. In rodents, repeated psychostimulant exposure enhances incentive sensitization to a greater extent in females than males. Estradiol increases females' motivation to attain psychostimulants and enhances the value of drug related cues, which ultimately increases their susceptibility towards spontaneous relapse. This, along with females' dampened ability to alter decisions regarding risky behaviors, enhances their vulnerability for escalation of drug use. In males, recent evidence suggests that estradiol may be protective against susceptibility towards drug-preference. Sex differences in the actions of estradiol are reviewed to provide a foundation for understanding how future research might enhance understanding of the mechanisms of sex differences in addiction-related behaviors, which are dependent on estradiol receptor (ER) subtype and the region of the brain they are acting in. A comprehensive review of the distribution of ERα, ERß, and GPER1 throughout the rodent brain are provided along with a discussion of the possible ways in which these patterns differentially regulate drug-taking between the sexes. The article concludes with a brief discussion of the actions of gonadal hormones on the circuitry of the stress system, including the hypothalamic pituitary adrenal axis and regulation of corticotropin-releasing factor. Sex differences in the stress system can also contribute to females' enhanced vulnerability towards addiction.


Asunto(s)
Encéfalo/metabolismo , Estradiol/metabolismo , Receptores de Estrógenos/metabolismo , Estrés Psicológico/metabolismo , Trastornos Relacionados con Sustancias/metabolismo , Animales , Susceptibilidad a Enfermedades , Dopamina/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Ratones , Sistema Hipófiso-Suprarrenal/metabolismo , Ratas , Receptores Acoplados a Proteínas G/metabolismo , Recompensa , Roedores , Factores Sexuales
18.
J Neurosci ; 29(6): 1897-903, 2009 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-19211896

RESUMEN

Classical estrogen receptor-signaling mechanisms involve estradiol binding to intracellular nuclear receptors [estrogen receptor-alpha (ERalpha) and estrogen receptor-beta (ERbeta)] to promote changes in protein expression. Estradiol can also exert effects within seconds to minutes, however, a timescale incongruent with genomic signaling. In the brain, estradiol rapidly potentiates stimulated dopamine release in the striatum of female rats and enhances spontaneous rotational behavior. Furthermore, estradiol rapidly attenuates the K(+)-evoked increase of GABA in dialysate. We hypothesize that these rapid effects of estradiol in the striatum are mediated by ERalpha located on the membrane of medium spiny GABAergic neurons. This experiment examined whether overexpression of ERalpha in the striatum would enhance the effect of estradiol on rotational behavior and the K(+)-evoked increase in GABA in dialysate. Ovariectomized female rats were tested for rotational behavior or underwent microdialysis experiments after unilateral intrastriatal injections of a recombinant adeno-associated virus (AAV) containing the human ERalpha cDNA (AAV.ERalpha) into the striatum; controls received either the same vector into areas outside the striatum or an AAV containing the human alkaline phosphatase gene into the striatum (AAV.ALP). Animals that received AAV.ERalpha in the striatum exhibited significantly greater estradiol-induced contralateral rotations compared with controls and exhibited behavioral sensitization of contralateral rotations induced by a low-dose of amphetamine. ERalpha overexpression also enhanced the inhibitory effect of estradiol on K(+)-evoked GABA release suggesting that disinhibition of dopamine release from terminals in the striatum resulted in the enhanced rotational behavior.


Asunto(s)
Cuerpo Estriado/metabolismo , Cuerpo Estriado/virología , Estradiol/fisiología , Receptor alfa de Estrógeno/biosíntesis , Receptor alfa de Estrógeno/genética , Regulación de la Expresión Génica/fisiología , Actividad Motora/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Cuerpo Estriado/fisiología , Dependovirus/genética , Estradiol/genética , Receptor alfa de Estrógeno/administración & dosificación , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Actividad Motora/genética , Ratas , Ratas Sprague-Dawley , Conducta Sexual/fisiología
19.
Horm Behav ; 58(1): 8-12, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19769978

RESUMEN

Cocaine self-administration in rodents has been used widely as a preclinical model of cocaine use in humans. In laboratory animals, estradiol enhances behavioral sensitization to cocaine and the acquisition of cocaine self-administration in female rats. The rewarding effect of cocaine has been shown to be enhanced following behavioral sensitization in male rats. This experiment examined whether behavioral sensitization to cocaine would promote cocaine-taking behavior in female rats, and whether estradiol could further modulate cocaine-taking behavior in cocaine-sensitized rats. Ovariectomized female rats were pretreated with either cocaine or saline for 4 days per week for 3 weeks. Self-administration sessions started 2 weeks after the last dose of drug. Female Sprague-Dawley rats received either estradiol or oil 30 min prior to the start of each session and self-administration was carried out 5 days per week for 4 weeks. The dose of cocaine self-administered each week was as follows (in mg/kg/infusion): week 1, 0.1; week 2, 0.1; week 3, 0.15; and week 4, 0.4. The rats that received cocaine pretreatment took fewer days to acquire cocaine self-administration and took more cocaine than rats that received saline pretreatment. Estradiol enhanced cocaine intake during the last six self-administration sessions after acquisition but did not affect acquisition of self-administration at the lowest doses of cocaine used. In conclusion, cocaine sensitization promotes the acquisition of cocaine self-administration in female rats. Furthermore, prior cocaine experience is more powerful than estradiol at enhancing acquisition, while estradiol enhances intake of cocaine after acquisition of self-administration.


Asunto(s)
Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Estradiol/farmacología , Estrógenos/farmacología , Animales , Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Relación Dosis-Respuesta a Droga , Estradiol/administración & dosificación , Estrógenos/administración & dosificación , Femenino , Actividad Motora/efectos de los fármacos , Ovariectomía , Ratas , Ratas Sprague-Dawley , Autoadministración , Caracteres Sexuales , Factores de Tiempo
20.
PLoS One ; 15(3): e0230170, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32163469

RESUMEN

Treatment options are limited for the approximately 40% of postmenopausal women worldwide who suffer from female sexual dysfunction (FSD). Neural stimulation has shown potential as a treatment for genital arousal FSD, however the mechanisms for its improvement are unknown. One potential cause of some cases of genital arousal FSD are changes to the composition of the vaginal microbiota, which is associated with vulvovaginal atrophy. The primary hypothesis of this study was that neural stimulation may induce healthy changes in the vaginal microbiome, thereby improving genital arousal FSD symptoms. In this study we used healthy rats, which are a common animal model for sexual function, however the rat vaginal microbiome is understudied. Thus this study also sought to examine the composition of the rat vaginal microbiota. Treatment rats (n = 5) received 30 minutes of cutaneous electrical stimulation targeting the genital branch of the pudendal nerve, and Control animals (n = 4) had 30-minute sessions without stimulation. Vaginal lavage samples were taken during a 14-day baseline period including multiple estrous periods and after twice-weekly 30-minute sessions across a six-week trial period. Analysis of 16S rRNA gene sequences was used to characterize the rat vaginal microbiota in baseline samples and determine the effect of stimulation. We found that the rat vaginal microbiota is dominated by Proteobacteria, Firmicutes, and Actinobacteria, which changed in relative abundance during the estrous cycle and in relationship to each other. While the overall stimulation effects were unclear in these healthy rats, some Treatment animals had less alteration in microbiota composition between sequential samples than Control animals, suggesting that stimulation may help stabilize the vaginal microbiome. Future studies may consider additional physiological parameters, in addition to the microbiome composition, to further examine vaginal health and the effects of stimulation.


Asunto(s)
Ciclo Estral/fisiología , Nervio Pudendo/fisiología , Roedores/microbiología , Vagina/microbiología , Vagina/fisiología , Animales , Nivel de Alerta/fisiología , Bacterias/genética , Estimulación Eléctrica/métodos , Femenino , Microbiota/genética , ARN Ribosómico 16S/genética , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA