Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
BMC Genomics ; 23(1): 679, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36183097

RESUMEN

BACKGROUND: The importance of fathers' engagement in care and its critical role in the offspring's cognitive and emotional development is now well established. Yet, little is known on the underlying neurobiology due to the lack of appropriate animal models. In the socially monogamous and bi-parental prairie vole (Microtus ochrogaster), while 60-80% of virgin males show spontaneous paternal behaviors (Paternal), others display pup-directed aggression (Attackers). Here we took advantage of this phenotypic dichotomy and used RNA-sequencing in three important brain areas to characterize gene expression associated with paternal behaviors of Paternal males and compare it to experienced Fathers and Mothers. RESULTS: While Paternal males displayed the same range and extent of paternal behaviors as experienced Fathers, we observed structure-specific transcriptomic differences between parental behaviors phenotypes. Using differential expression, gene set expression, as well as co-expression network analyses, we found that phenotypic differences between Paternal males and Attackers were mainly reflected by the lateral septum (LS), and to a lower extent, the nucleus accumbens (NAc), transcriptomes. In the medial preoptic area (MPOA), the profiles of gene expression mainly reflected differences between females and males regardless of their parental behaviors phenotype. Functional enrichment analyses of those gene sets associated with Paternal males or Attackers in the LS and the NAc revealed the involvement of processes related to the mitochondria, RNA translation, protein degradation processes, as well as epigenetic regulation of gene expression. CONCLUSIONS: By leveraging the natural phenotypic differences in parental behaviors in virgin male prairie voles alongside fathers and mothers, we identified a marked structure- and phenotype-specific pattern of gene expression associated with spontaneous paternal behaviors independently from fatherhood and pair-bonding. The LS transcriptome related to the mitochondria, RNA translation, and protein degradation processes was thus highlighted as a primary candidate associated with the spontaneous display of paternal behaviors. Altogether, our observations further characterize the behavioral and transcriptomic signature of parental behaviors in the socially monogamous prairie vole and lay the groundwork to further our understanding of the molecular underpinnings of paternal behavior.


Asunto(s)
Conducta Paterna , Transcriptoma , Animales , Arvicolinae/genética , Epigénesis Genética , Femenino , Pradera , Masculino , Conducta Paterna/fisiología , ARN/metabolismo
2.
J Pharmacol Exp Ther ; 367(3): 393-404, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30213876

RESUMEN

Recent work from our group and others has revealed a higher sensitivity of female rodents to the antidepressant-like effects of the N-methyl d-aspartate receptor antagonist ketamine strongly influenced by circulating estrogen and progesterone levels. However, in the absence of any preclinical studies of pharmacokinetic sex differences using low-dose ketamine in rats, it is unclear whether the effects of sex and hormonal milieu on ketamine's behavioral actions are influenced by differences in ketamine metabolism between male and female rats. Therefore, this work examined whether sex and hormonal status affect ketamine metabolism and distribution in male and female rats using a low antidepressant-like dose selectively effective in females. Intact male rats and female rats in either diestrus (low estrogen, progesterone) or proestrus (high estrogen, progesterone) were administered low-dose ketamine, and their plasma and brains were collected to analyze levels of ketamine and its metabolites norketamine (NK) and dehydronorketamine. Females exhibited greater concentrations of ketamine and NK over the first 30 min following treatment in both brain and plasma, largely accounted for by slower clearance rates and longer half-lives. Interestingly, despite the impact of ovarian hormones on behavioral sensitivity to ketamine, no appreciable differences in pharmacokinetic parameters existed between proestrus and diestrus female rats. This work is the first to demonstrate sex differences in ketamine pharmacokinetics in rats, and suggests that while sex differences in metabolism may influence the amount of ketamine and NK reaching target areas in the brain, the impact of circulating hormone levels here is negligible.


Asunto(s)
Encéfalo/metabolismo , Ketamina/sangre , Ketamina/farmacocinética , Plasma/metabolismo , Animales , Estrógenos/metabolismo , Femenino , Ketamina/análogos & derivados , Ketamina/metabolismo , Masculino , Progesterona/metabolismo , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales
3.
FASEB J ; 31(6): 2492-2506, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28235781

RESUMEN

Cardiovascular dysfunction is highly comorbid with mood disorders, such as anxiety and depression. However, the mechanisms linking cardiovascular dysfunction with the core behavioral features of mood disorder remain poorly understood. In this study, we used mice bearing a knock-in sarcomeric mutation, which is exhibited in human hypertrophic cardiomyopathy (HCM), to investigate the influence of HCM over the development of anxiety and depression. We employed behavioral, MRI, and biochemical techniques in young (3-4 mo) and aged adult (7-8 mo) female mice to examine the effects of HCM on the development of anxiety- and depression-like behaviors. We focused on females because in both humans and rodents, they experience a 2-fold increase in mood disorder prevalence vs. males. Our results showed that young and aged HCM mice displayed echocardiographic characteristics of the heart disease condition, yet only aged HCM females displayed anxiety- and depression-like behaviors. Electrocardiographic parameters of sympathetic nervous system activation were increased in aged HCM females vs. controls and correlated with mood disorder-related symptoms. In addition, when compared with controls, aged HCM females exhibited adrenal gland hypertrophy, reduced volume in mood-related brain regions, and reduced hippocampal signaling proteins, such as brain-derived neurotrophic factor and its downstream targets vs. controls. In conclusion, prolonged systemic HCM stress can lead to development of mood disorders, possibly through inducing structural and functional brain changes, and thus, mood disorders in patients with heart disease should not be considered solely a psychologic or situational condition.-Dossat, A. M., Sanchez-Gonzalez, M. A., Koutnik, A. P., Leitner, S., Ruiz, E. L., Griffin, B., Rosenberg, J. T., Grant, S. C., Fincham, F. D., Pinto, J. R. Kabbaj, M. Pathogenesis of depression- and anxiety-like behavior in an animal model of hypertrophic cardiomyopathy.


Asunto(s)
Ansiedad/genética , Cardiomiopatía Hipertrófica/complicaciones , Depresión/genética , Envejecimiento , Animales , Cardiomiopatía Hipertrófica/genética , Vías Eferentes , Femenino , Técnicas de Sustitución del Gen , Humanos , Ratones , Mutación , Sarcómeros/genética , Sistema Nervioso Simpático/fisiología , Nervio Vago
4.
J Neurosci ; 35(23): 8948-58, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26063926

RESUMEN

Epigenetic mechanisms, such as histone modifications, regulate responsiveness to drugs of abuse, such as cocaine, but relatively little is known about the regulation of addictive-like behaviors by DNA methylation. To investigate the influence of DNA methylation on the locomotor-activating effects of cocaine and on drug-seeking behavior, rats receiving methyl supplementation via chronic l-methionine (MET) underwent either a sensitization regimen of intermittent cocaine injections or intravenous self-administration of cocaine, followed by cue-induced and drug-primed reinstatement. MET blocked sensitization to the locomotor-activating effects of cocaine and attenuated drug-primed reinstatement, with no effect on cue-induced reinstatement or sucrose self-administration and reinstatement. Furthermore, upregulation of DNA methyltransferase 3a and 3b and global DNA hypomethylation were observed in the nucleus accumbens core (NAc), but not in the medial prefrontal cortex (mPFC), of cocaine-pretreated rats. Glutamatergic projections from the mPFC to the NAc are critically involved in the regulation of cocaine-primed reinstatement, and activation of both brain regions is seen in human addicts when reexposed to the drug. When compared with vehicle-pretreated rats, the immediate early gene c-Fos (a marker of neuronal activation) was upregulated in the NAc and mPFC of cocaine-pretreated rats after cocaine-primed reinstatement, and chronic MET treatment blocked its induction in both regions. Cocaine-induced c-Fos expression in the NAc was associated with reduced methylation at CpG dinucleotides in the c-Fos gene promoter, effects reversed by MET treatment. Overall, these data suggest that drug-seeking behaviors are, in part, attributable to a DNA methylation-dependent process, likely occurring at specific gene loci (e.g., c-Fos) in the reward pathway.


Asunto(s)
Encéfalo/efectos de los fármacos , Trastornos Relacionados con Cocaína/fisiopatología , Cocaína/efectos adversos , Metilación de ADN/efectos de los fármacos , Inhibidores de Captación de Dopamina/efectos adversos , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Encéfalo/metabolismo , Trastornos Relacionados con Cocaína/etiología , Condicionamiento Operante/efectos de los fármacos , ADN Metiltransferasa 3A , Modelos Animales de Enfermedad , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Extinción Psicológica , Masculino , Proteínas Proto-Oncogénicas c-fos/genética , Ratas , Ratas Sprague-Dawley , Refuerzo en Psicología , Autoadministración , Sacarosa/administración & dosificación , Edulcorantes/administración & dosificación
5.
J Biol Chem ; 289(33): 23097-23111, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24973218

RESUMEN

The cardiac troponin I (cTnI) R21C (cTnI-R21C) mutation has been linked to hypertrophic cardiomyopathy and renders cTnI incapable of phosphorylation by PKA in vivo. Echocardiographic imaging of homozygous knock-in mice expressing the cTnI-R21C mutation shows that they develop hypertrophy after 12 months of age and have abnormal diastolic function that is characterized by longer filling times and impaired relaxation. Electrocardiographic analyses show that older R21C mice have elevated heart rates and reduced cardiovagal tone. Cardiac myocytes isolated from older R21C mice demonstrate that in the presence of isoproterenol, significant delays in Ca(2+) decay and sarcomere relaxation occur that are not present at 6 months of age. Although isoproterenol and stepwise increases in stimulation frequency accelerate Ca(2+)-transient and sarcomere shortening kinetics in R21C myocytes from older mice, they are unable to attain the corresponding WT values. When R21C myocytes from older mice are treated with isoproterenol, evidence of excitation-contraction uncoupling is indicated by an elevation in diastolic calcium that is frequency-dissociated and not coupled to shorter diastolic sarcomere lengths. Myocytes from older mice have smaller Ca(2+) transient amplitudes (2.3-fold) that are associated with reductions (2.9-fold) in sarcoplasmic reticulum Ca(2+) content. This abnormal Ca(2+) handling within the cell may be attributed to a reduction (2.4-fold) in calsequestrin expression in conjunction with an up-regulation (1.5-fold) of Na(+)-Ca(2+) exchanger. Incubation of permeabilized cardiac fibers from R21C mice with PKA confirmed that the mutation prevents facilitation of mechanical relaxation. Altogether, these results indicate that the inability to enhance myofilament relaxation through cTnI phosphorylation predisposes the heart to abnormal diastolic function, reduced accessibility of cardiac reserves, dysautonomia, and hypertrophy.


Asunto(s)
Señalización del Calcio , Cardiomiopatía Hipertrófica/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diástole , Miocitos Cardíacos/metabolismo , Troponina I/metabolismo , Animales , Cardiomiopatía Hipertrófica/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Transgénicos , Miocitos Cardíacos/patología , Fosforilación/fisiología , Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/metabolismo , Troponina I/genética
6.
Front Neuroendocrinol ; 35(1): 42-57, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24076484

RESUMEN

Compelling evidence exists for pervasive sex differences in pathological conditions, including anxiety and depressive disorders, with females more than twice as likely to be afflicted. Gonadal hormones may be a major factor in this disparity, given that women are more likely to experience mood disturbances during times of hormonal flux, and testosterone may have protective benefits against anxiety and depression. In this review we focus on the effects of testosterone in males and females, revealed in both human and animal studies. We also present possible neurobiological mechanisms underlying testosterone's mostly protective benefits, including the brain regions, neural circuits, and cellular and molecular pathways involved. While the precise underlying mechanisms remain unclear, both activational and organizational effects of testosterone appear to contribute to these effects. Future clinical studies are necessary in order to better understand when and how testosterone therapy may be effective in both sexes.


Asunto(s)
Ansiedad/metabolismo , Depresión/metabolismo , Caracteres Sexuales , Testosterona/metabolismo , Animales , Femenino , Humanos , Masculino , Transducción de Señal/fisiología
7.
J Exp Biol ; 218(Pt 1): 21-31, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25568448

RESUMEN

Major depressive disorder (MDD) is a devastating neuropsychiatric disorder encompassing a wide range of cognitive and emotional dysfunctions. The prevalence of MDD is expected to continue its growth to become the second leading cause of disease burden (after HIV) by 2030. Despite an extensive research effort, the exact etiology of MDD remains elusive and the diagnostics uncertain. Moreover, a marked inter-individual variability is observed in the vulnerability to develop depression, as well as in response to antidepressant treatment, for nearly 50% of patients. Although a genetic component accounts for some cases of MDD, it is now clearly established that MDD results from strong gene and environment interactions. Such interactions could be mediated by epigenetic mechanisms, defined as chromatin and DNA modifications that alter gene expression without changing the DNA structure itself. Some epigenetic mechanisms have recently emerged as particularly relevant molecular substrates, promoting vulnerability or resilience to the development of depressive-like symptoms. Although the role of brain-derived neurotrophic factor (BDNF) in the pathophysiology of MDD remains unclear, its modulation of the efficacy of antidepressants is clearly established. Therefore, in this review, we focus on the epigenetic mechanisms regulating the expression of BDNF in humans and in animal models of depression, and discuss their role in individual differences in vulnerability to depression and response to antidepressant drugs.


Asunto(s)
Antidepresivos/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/genética , Depresión/tratamiento farmacológico , Depresión/genética , Epigénesis Genética , Animales , Antidepresivos/farmacología , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Predisposición Genética a la Enfermedad , Humanos
8.
J Neurosci ; 33(27): 11048-60, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23825410

RESUMEN

Some personality traits, including novelty seeking, are good predictors of vulnerability to stress-related mood disorders in both humans and rodents. While high-novelty-seeking rats [high responders (HRs)] are vulnerable to the induction of depressive-like symptoms by social defeat stress, low-novelty-seeking rats [low responders (LRs)] are not. Here, we show that such individual differences are critically regulated by hippocampal BDNF. While LR animals exhibited an increase in BDNF levels following social defeat, HR individuals did not. This difference in hippocampal BDNF expression promoted the vulnerability of HR and the resilience of LR rats. Indeed, preventing activation of BDNF signaling by infusing the BDNF scavenger TrkB-Fc into the dentate gyrus of the hippocampus of LR rats led to social defeat-induced social avoidance, whereas its activation in HR rats by the TrkB agonist 7,8-dihydroxyflavone promoted social approach. Along with the changes in BDNF expression following defeat, we report in LR animals a downregulation of the inactive BDNF receptor TrkB.T1, associated with an activation of CREB through Akt-mediated signaling, but not MSK1-mediated signaling. In HR animals, none of these molecules were affected by social defeat. Importantly, the BDNF upregulation involved an epigenetically controlled transcription of bdnf exon VI, associated with a coherent regulation of relevant epigenetic factors. Altogether, our data support the importance of hippocampal BDNF regulation in response to stressful events. Moreover, we identify a specific and adaptive regulation of bdnf exon VI in the hippocampus as a critical regulator of stress resilience, and strengthen the importance of epigenetic factors in mediating stress-induced adaptive and maladaptive responses in different individuals.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Epigénesis Genética/fisiología , Conducta Exploratoria/fisiología , Regulación de la Expresión Génica , Individualidad , Actividad Motora/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Predicción , Hipocampo/fisiología , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Conducta Social
9.
J Behav Med ; 37(2): 289-99, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23274763

RESUMEN

The study examined differential effects of preoperative biomarkers (cotisol, C-reactive protein/CRP, and interleukin-6/IL-6) on postoperative symptoms in mental health (depressed mood, anxiety and hostility) 1 month following open-heart surgery, controlling for known predictors. Preoperative and postoperative interviews were conducted on 162 patients. Peripheral venous blood samples were collected between 8 and 10 a.m. prior to surgery. Cardiac indices were obtained from the Society of Thoracic Surgeons' national database. Preoperative anxiety contributed to all outcomes about 1 month postoperatively. Patients with high preoperative plasma IL-6 used more avoidant coping and experienced greater depressed mood. Patients with increased plasma CRP and with hope were less depressed. Elevated plasma cortisol predicted hostility. Finally, medical comorbidities predicted anxiety and hostility. The combination of anxiety and stress-sensitive biomarkers may be one way to predict postoperative symptoms following open-heart surgery. Our findings emphasize the importance of investigating the mind-body interplay to come up with better interventions.


Asunto(s)
Ansiedad/sangre , Proteína C-Reactiva , Procedimientos Quirúrgicos Cardíacos/psicología , Depresión/sangre , Interleucina-6/sangre , Periodo Preoperatorio , Psicofisiología , Adaptación Psicológica , Biomarcadores/sangre , Femenino , Hostilidad , Humanos , Masculino , Persona de Mediana Edad
10.
Peptides ; 181: 171298, 2024 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-39317295

RESUMEN

Cocaine and amphetamine-regulated transcript (CART) mRNA and peptide are vastly expressed in both cortical and subcortical brain areas and are involved in critical cognitive functions. CART peptide (CARTp), described in reward-related brain structures, regulates drug-induced learning and memory, and its role appears specific to psychostimulants. However, many other drugs of abuse, such as alcohol, opiates, nicotine, and caffeine, have been shown to alter the expression levels of CART mRNA and peptides in brain structures directly or indirectly associated with learning and memory processes. However, the number of studies demonstrating the contribution of CARTp in learning and memory is still minimal. Notably, the exact cellular and molecular mechanisms underlying CARTp effects are still unknown. The discoveries that CARTp effects are mediated through a putative G-protein coupled receptor and activation of cellular signaling cascades via NMDA receptor-coupled ERK have enhanced our knowledge about the action of this neuropeptide and allowed us to comprehend better CARTp exact cellular/molecular mechanisms that could mediate drug-induced changes in learning and memory functions. Unfortunately, these efforts have been impeded by the lack of suitable and specific CARTp receptor antagonists. In this review, following a short introduction about CARTp, we report on current knowledge about CART's roles in learning and memory processes and its recently described role in memory-related neurological disorders. We will also discuss the importance of further investigating how CARTp interacts with its receptor(s) and other neurotransmitter systems to influence learning and memory functions. This topic is sure to intrigue and motivate further exploration in the field of neuroscience.

11.
Alcohol ; 120: 169-178, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38290696

RESUMEN

Ceftriaxone is an antibiotic that increases central nervous system (CNS) protein expression of the glutamate transporters GLT-1 and xCT and ameliorates pathological behaviors in rodent models of neurological disease and substance use disorder. However, little ceftriaxone passes through the blood-brain barrier, the CNS binding partner of ceftriaxone is unknown, and ceftriaxone does not consistently upregulate GLT-1 and xCT in cell culture. Ceftriaxone alters the gut microbiome composition in rodents and humans, and the microbiome-gut-brain axis regulates drug-seeking. Thus, here we test the hypothesis that ceftriaxone reduces alcohol intake while ameliorating alcohol-induced disruption of the gut microbiome composition. Male and female Sprague-Dawley rats received intermittent access to alcohol (IAA) while controls received access to only water. Following 17 IAA sessions, ceftriaxone/vehicle treatment was given for 5 days. Analysis of the gut microbiome composition was assessed by 16S rRNA gene amplicon sequencing conducted on fecal pellets collected prior to and after alcohol consumption and following ceftriaxone treatment. Male rats displayed escalated alcohol intake and preference over the course of the 17 sessions; however, total alcohol intake did not differ between the sexes. Ceftriaxone reduced alcohol intake and preference in male and female rats. While alcohol affected a diverse set of amplicon sequencing variants (ASV), ceftriaxone markedly reduced the diversity of microbial communities reflected by a blooming of the Enterococcaceae family. The remaining effects of ceftriaxone, however, encompassed families both affected and unaffected by prior alcohol drinking and highlight the Ruminococcaceae and Muribaculaceae families as bidirectionally modulated by alcohol and ceftriaxone. Altogether, our study confirms that ceftriaxone reduces alcohol intake in rats and partially reverses alcohol-induced dysbiosis.


Asunto(s)
Consumo de Bebidas Alcohólicas , Ceftriaxona , Microbioma Gastrointestinal , Ratas Sprague-Dawley , Animales , Femenino , Masculino , Ceftriaxona/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Ratas , Antibacterianos/farmacología , Etanol/farmacología , Etanol/administración & dosificación , Heces/microbiología
12.
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
13.
Neuropharmacology ; 255: 110008, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38797243

RESUMEN

Ketamine (KET), a non-competitive N-methyl-d-aspartate (NMDA) receptor antagonist, has rapid onset of antidepressant effects in Treatment-Resistant Depression patients and repeated infusions are required to sustain its antidepressant properties. However, KET is an addictive drug, and so more preclinical and clinical research is needed to assess the safety of recurring treatments in both sexes. Thus, the aim of this study was to investigate the reinforcing properties of various doses of KET (0-, 0.125-, 0.25-, 0.5 mg/kg/infusion) and assess KET's cue-induced reinstatement and neuronal activation in both sexes of Long Evans rats. Neuronal activation was assessed using the protein expression of the immediate early gene cFos in the nucleus accumbens (Nac), an important brain area implicated in reward, reinforcement and reinstatement to most drug-related cues. Our findings show that KET has reinforcing effects in both male and female rats, albeit exclusively at the highest two doses (0.25 and 0.5 mg/kg/infusion). Furthermore, we noted sex differences, particularly at the highest dose of ketamine, with female rats displaying a higher rate of self-administration. Interestingly, all groups that self-administered KET reinstated to drug-cues. Following drug cue-induced reinstatement test in rats exposed to KET (0.25 mg/kg/infusion) or saline, there was higher cFos protein expression in KET-treated animals compared to saline controls, and higher cFos expression in the core compared to the shell subregions of the Nac. As for reinstatement, there were no notable sex differences reported for cFos expression in the Nac. These findings reveal some sex and dose dependent effects in KET's reinforcing properties and that KET at all doses induced similar reinstatement in both sexes. This study also demonstrated that cues associated with ketamine induce comparable neuronal activation in the Nac of both male and female rats. This work warrants further research into the potential addictive properties of KET, especially when administered at lower doses which are now being used in the clinic for treating various psychopathologies.


Asunto(s)
Señales (Psicología) , Relación Dosis-Respuesta a Droga , Ketamina , Núcleo Accumbens , Ratas Long-Evans , Refuerzo en Psicología , Animales , Ketamina/farmacología , Ketamina/administración & dosificación , Masculino , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Femenino , Proteínas Proto-Oncogénicas c-fos/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Ratas , Caracteres Sexuales , Autoadministración , Condicionamiento Operante/efectos de los fármacos
14.
Proc Natl Acad Sci U S A ; 107(3): 1217-22, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20080553

RESUMEN

The prairie vole (Microtus ochrogaster) is a socially monogamous rodent species that forms pair bonds after mating, a behavior in which central dopamine (DA) has been implicated. Here, we used male prairie voles to examine the effects of drug exposure on pair bonding and related neural circuitry. In our first experiment, amphetamine (AMPH) motivated behavior was examined using a conditioned place preference (CPP) paradigm and was shown to be mediated by activation of D1-like DA receptors. Next, we examined the effects of repeated AMPH exposure on pair bonding. Intact and saline pretreated control males displayed mating-induced partner preferences, whereas males pretreated with AMPH at the doses effective to induce CPP failed to show mating-induced partner preferences. Such AMPH treatment also enhanced D1, but not D2, DA receptor expression in the nucleus accumbens (NAcc). Furthermore, pharmacological blockade of D1-like DA receptors in the NAcc rescued mating-induced partner preferences in AMPH-treated males. Together, our data indicate that repeated AMPH exposure may narrow the behavioral repertoire of male prairie voles via a DA receptor-specific mechanism in the NAcc, resulting in the impairment of pair bond formation.


Asunto(s)
Anfetamina/farmacología , Arvicolinae/fisiología , Dopamina/metabolismo , Núcleo Accumbens/metabolismo , Conducta Sexual Animal/efectos de los fármacos , Conducta Social , Animales , Western Blotting , Femenino , Masculino
15.
Horm Behav ; 61(5): 678-85, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22426002

RESUMEN

RATIONALE: Affective disorders are twice as likely to occur in women as they are in men suggesting a critical role for gonadal hormones in their etiology. In particular, testosterone has been shown to have protective effects in men. OBJECTIVE: To investigate antidepressant effects and interactions between testosterone and imipramine in socially isolated male and female rats. METHODS: A chronic social isolation model was used to induce an anxiety and depressive-like state in adult gonadectomized (Gnx) male and ovariectomized (Ovx) female rats receiving chronic testosterone and imipramine treatments. Their anxiety and depression-like behaviors were examined using the light-dark box, elevated plus maze, open field, sucrose preference and novelty induced hypophagia tests. RESULTS: In socially isolated rats, the anxiolytic and antidepressant effects of testosterone and imipramine were limited to male rats. Additionally, testosterone enhanced the neurogenic effect of imipramine on hippocampal cell proliferation in male rats. Although female rats exhibited signs of anxiety and depressive-like behaviors following social isolation, testosterone and/or imipramine administration had no anxiolytic or antidepressant effects in Ovx females. CONCLUSIONS: Testosterone and imipramine had anxiolytic and antidepressant effects in socially isolated male, but not female rats. Testosterone enhanced the effect of imipramine on cell proliferation in the hippocampus of male rats.


Asunto(s)
Depresión/prevención & control , Imipramina/farmacología , Aislamiento Social/psicología , Testosterona/farmacología , Animales , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Castración , Evaluación Preclínica de Medicamentos , Femenino , Masculino , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales , Estudios de Validación como Asunto
16.
Curr Top Behav Neurosci ; 54: 283-310, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34595741

RESUMEN

Aggression is a complex behavioral trait modulated by both genetic and environmental influences on gene expression. By controlling gene expression in a reversible yet potentially lasting manner in response to environmental stimulation, epigenetic mechanisms represent prime candidates in explaining both individual differences in aggression and the development of elevated aggressive behaviors following life adversity. In this manuscript, we review the evidence for an epigenetic basis in the development and expression of aggression in both humans and related preclinical animal models. In particular, we discuss reports linking DNA methylation, histone post-translational modifications, as well as non-coding RNA, to the regulation of a variety of genes implicated in the neurobiology of aggression including neuropeptides, the serotoninergic and dopaminergic systems, and stress response related systems. While clinical reports do reveal interesting patterns of DNA methylation underlying individual differences and experience-induced aggressive behaviors, they do, in general, face the challenge of linking peripheral observations to central nervous system regulations. Preclinical studies, on the other hand, provide detailed mechanistic insights into the epigenetic reprogramming of gene expression following life adversities. Although the functional link to aggression remains unclear in most, these studies together do highlight the involvement of epigenetic events driven by DNA methylation, histone modifications, and non-coding RNA in the neuroadaptations underlying the development and expression of aggression.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Agresión/fisiología , Animales , Metilación de ADN/genética , Código de Histonas , ARN no Traducido
17.
Data Brief ; 43: 108338, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35712367

RESUMEN

In this study, we collected electrophysiological data from acute hippocampal slices of male and female Sprague Dawley rats. Rats were exposed to social isolation rearing and then acutely treated with various doses of ketamine in order to rescue hippocampal plasticity deficits induced by isolation stress. We used two different approaches to study neuronal plasticity: Long-Term Potentiation (LTP) which is a well-established cellular model for memory and Paired-Pulse Facilitation (PPF) which is short-term of presynaptic plasticity. The aim of this article is to offer more experimental details about out LTP and PPF procedures.

18.
Neurosci Lett ; 766: 136301, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34688854

RESUMEN

Chronic social isolation stress (SIS) induces lasting negative effects on the brain, including memory deficits, cognitive impairments, and mood alterations such as depression and anxiety. All these symptoms, at least in part, reflect reduced hippocampal function. In both clinical and preclinical studies, subanesthetic doses of the NMDA receptor antagonist, ketamine (KET), was shown to have rapid and lasting antidepressant effects. Animal studies have shown that biological sex and levels of gonadal hormones alter the behavioral effects of KET, with ovarian hormones increasing sensitivity to the antidepressant-like effects of KET. Since the hippocampus plays a key role in mediating some of the effects of SIS, and considering that KET at low doses has been shown to rescue some of the behavioral deficits of isolation rearing this study aimed to assess the effects of isolation stress on pre- and post-synaptic hippocampal functions in male and female rats reared in SIS, as well as determine whether some of the physiological deficits can be rescued with a single injection of sub-anesthetic doses of KET. To do this, Sprague-Dawley rats were raised from weaning in either social isolation or with same-sex cage mate for 5 to 7 weeks. Male and female rats in either diestrus of proestrus received a single injection of KET (0, 2.5, or 5.0 mg/kg) three hours prior to termination and collection of acute hippocampal slices for ex vivo electrophysiological field potential recordings. Long-term potentiation (LTP) and paired pulse facilitation (PPF) outputs were assessed in a canonical CA3-CA1 dorsal hippocampal circuit. Our data show that SIS inhibits hippocampal LTP without affecting PPF in male rats, an effect that was rescued by KET. In female rats, isolation stress did not alter LTP, but did reduce PPF - especially when females were tested in diestrus-, an effect that was rescued by KET at the highest dose. Our data thus suggest sex differences in the contribution of pre-and postsynaptic hippocampal compartments in response to stress and KET.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/farmacología , Hipocampo/efectos de los fármacos , Ketamina/farmacología , Plasticidad Neuronal/fisiología , Caracteres Sexuales , Estrés Psicológico/fisiopatología , Animales , Femenino , Hipocampo/fisiopatología , Masculino , Plasticidad Neuronal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Aislamiento Social/psicología
19.
Biol Psychiatry ; 91(1): 141-151, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33549315

RESUMEN

BACKGROUND: The ability to form enduring social bonds is characteristic of human nature, and impairments in social affiliation are central features of severe neuropsychiatric disorders including autism spectrum disorder and schizophrenia. Owing to its ability to form long-term pair-bonds, the socially monogamous prairie vole has emerged as an excellent model to study the neurobiology of social attachment. Despite the enduring nature of the bond, however, surprisingly few genes have been implicated in the pair-bonding process in either sex. METHODS: Male and female prairie voles (Microtus ochrogaster) were cohabitated with an opposite-sex partner for 24 hours or 3 weeks, and transcriptomic regulations in the nucleus accumbens were measured by RNA sequencing. RESULTS: We found sex-specific response patterns despite similar behavioral indicators of pair-bond establishment. Indeed, 24 hours of cohabitation with an opposite-sex partner induced widespread transcriptomic changes that remained sustained to some extent in females after 3 weeks but returned to baseline before a second set of regulations in males. This led to a highly sexually biased nucleus accumbens transcriptome at 3 weeks related to processes such as neurotransmission, protein turnover, and DNA transcription. In particular, we found sex-specific alterations of mitochondrial dynamics following cohabitation, with a shift toward fission in males. CONCLUSIONS: In addition to identifying the genes, networks, and pathways involved in the pair-bonding process in the nucleus accumbens, our work illustrates the vast extent of sex differences in the molecular mechanisms underlying pair-bonding in prairie voles and paves the way to further our understanding of the complex social bonding process.


Asunto(s)
Trastorno del Espectro Autista , Transcriptoma , Animales , Arvicolinae , Femenino , Pradera , Humanos , Masculino , Apareamiento , Conducta Sexual Animal , Conducta Social
20.
Sci Rep ; 12(1): 1820, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-35110693

RESUMEN

Numerous emotional and cognitive processes mediated by the hippocampus present differences between sexes and can be markedly influenced by hormonal status in males and females of several species. In rodents, the dorsal hippocampus (dHPC) is known to contribute to the rapid antidepressant actions of the NMDA receptor antagonist ketamine. We and others have demonstrated a greater sensitivity to the fast-acting antidepressant ketamine in female versus male rats that is estrogen- and progesterone-dependent. However, the underlying mechanisms remain unclear. Using an acute low dose (2.5 mg/kg) of ketamine that is behaviorally effective in female but not male rats, a label-free phosphoproteomics approach was employed to identify ketamine-induced changes in signaling pathway activation and phosphoprotein abundance within the dHPC of intact adult male rats and female rats in either diestrus or proestrus. At baseline, males and females showed striking dissimilarities in the dHPC proteome and phosphoproteome related to synaptic signaling and mitochondrial function-differences also strongly influenced by cycle stage in female rats. Notably, phosphoproteins enriched in PKA signaling emerged as being both significantly sex-dependent at baseline and also the primary target of ketamine-induced protein phosphorylation selectively in female rats, regardless of cycle stage. Reduced phosphoprotein abundance within this pathway was observed in males, suggesting bi-directional effects of low-dose ketamine between sexes. These findings present biological sex and hormonal milieu as critical modulators of ketamine's rapid actions within this brain region and provide greater insight into potential translational and post-translational processes underlying sex- and hormone-dependent modulation of ketamine's therapeutic effects.


Asunto(s)
Ciclo Estral/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Hipocampo/efectos de los fármacos , Ketamina/farmacología , Fosfoproteínas/metabolismo , Proteoma , Animales , Femenino , Hipocampo/metabolismo , Masculino , Fosforilación , Mapas de Interacción de Proteínas , Proteómica , Ratas Sprague-Dawley , Caracteres Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA