Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 15: 1290523, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38410515

RESUMEN

Severe COVID-19 leads to widespread transcriptomic changes in the human brain, mimicking diminished cognitive performance. As long noncoding RNAs (lncRNAs) play crucial roles in the regulation of gene expression, identification of the lncRNAs differentially expressed upon COVID-19 may nominate key regulatory nodes underpinning cognitive changes. Here we identify hundreds of lncRNAs differentially expressed in the brains of COVID-19 patients relative to uninfected age/sex-matched controls, many of which are associated with decreased cognitive performance and inflammatory cytokine response. Our analyses reveal pervasive transcriptomic changes in lncRNA expression upon severe COVID-19, which may serve as key regulators of neurocognitive changes in the brain.


Asunto(s)
COVID-19 , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , COVID-19/genética , Perfilación de la Expresión Génica , Citocinas/genética , ARN Mensajero/genética
2.
Cell Rep ; 42(12): 113478, 2023 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-37991919

RESUMEN

Coronavirus disease 2019 (COVID-19) remains a significant public health threat due to the ability of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants to evade the immune system and cause breakthrough infections. Although pathogenic coronaviruses such as SARS-CoV-2 and Middle East respiratory syndrome (MERS)-CoV lead to severe respiratory infections, how these viruses affect the chromatin proteomic composition upon infection remains largely uncharacterized. Here, we use our recently developed integrative DNA And Protein Tagging methodology to identify changes in host chromatin accessibility states and chromatin proteomic composition upon infection with pathogenic coronaviruses. SARS-CoV-2 infection induces TP53 stabilization on chromatin, which contributes to its host cytopathic effect. We mapped this TP53 stabilization to the SARS-CoV-2 spike and its propensity to form syncytia, a consequence of cell-cell fusion. Differences in SARS-CoV-2 spike variant-induced syncytia formation modify chromatin accessibility, cellular senescence, and inflammatory cytokine release via TP53. Our findings suggest that differences in syncytia formation alter senescence-associated inflammation, which varies among SARS-CoV-2 variants.


Asunto(s)
COVID-19 , Coronavirus del Síndrome Respiratorio de Oriente Medio , Humanos , SARS-CoV-2 , Cromatina , Proteómica , Senescencia Celular , Células Gigantes , Proteína p53 Supresora de Tumor/genética
3.
bioRxiv ; 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37693555

RESUMEN

COVID-19 remains a significant public health threat due to the ability of SARS-CoV-2 variants to evade the immune system and cause breakthrough infections. Although pathogenic coronaviruses such as SARS-CoV-2 and MERS-CoV lead to severe respiratory infections, how these viruses affect the chromatin proteomic composition upon infection remains largely uncharacterized. Here we used our recently developed integrative DNA And Protein Tagging (iDAPT) methodology to identify changes in host chromatin accessibility states and chromatin proteomic composition upon infection with pathogenic coronaviruses. SARS-CoV-2 infection induces TP53 stabilization on chromatin, which contributes to its host cytopathic effect. We mapped this TP53 stabilization to the SARS-CoV-2 spike and its propensity to form syncytia, a consequence of cell-cell fusion. Differences in SARS-CoV-2 spike variant-induced syncytia formation modify chromatin accessibility, cellular senescence, and inflammatory cytokine release via TP53. Our findings suggest that differences in syncytia formation alter senescence-associated inflammation, which varies among SARS-CoV-2 variants.

4.
Nat Aging ; 2(12): 1130-1137, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-37118539

RESUMEN

As coronavirus disease 2019 (COVID-19) and aging are both accompanied by cognitive decline, we hypothesized that COVID-19 might lead to molecular signatures similar to aging. We performed whole-transcriptome analysis of the frontal cortex, a critical area for cognitive function, in individuals with COVID-19, age-matched and sex-matched uninfected controls, and uninfected individuals with intensive care unit/ventilator treatment. Our findings indicate that COVID-19 is associated with molecular signatures of brain aging and emphasize the value of neurological follow-up in recovered individuals.


Asunto(s)
COVID-19 , Humanos , Envejecimiento/genética , Encéfalo , Lóbulo Frontal , Perfilación de la Expresión Génica
5.
medRxiv ; 2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34845457

RESUMEN

Coronavirus disease 2019 (COVID-19) is predominantly an acute respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and remains a significant threat to public health. COVID-19 is accompanied by neurological symptoms and cognitive decline, but the molecular mechanisms underlying this effect remain unclear. As aging induces distinct molecular signatures in the brain associated with cognitive decline in healthy populations, we hypothesized that COVID-19 may induce molecular signatures of aging. Here, we performed whole transcriptomic analysis of human frontal cortex, a critical area for cognitive function, in 12 COVID-19 cases and age- and sex-matched uninfected controls. COVID-19 induces profound changes in gene expression, despite the absence of detectable virus in brain tissue. Pathway analysis shows downregulation of genes involved in synaptic function and cognition and upregulation of genes involved in immune processes. Comparison with five independent transcriptomic datasets of aging human frontal cortex reveals striking similarities between aged individuals and severe COVID-19 patients. Critically, individuals below 65 years of age exhibit profound transcriptomic changes not observed among older individuals in our patient cohort. Our data indicate that severe COVID-19 induces molecular signatures of aging in the human brain and emphasize the value of neurological follow-up in recovered individuals.

6.
Addict Biol ; 26(3): e12973, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33078503

RESUMEN

Withdrawal from opioid painkillers can produce short-lived physical symptoms and protracted psychological symptoms including anxiety and depressive-like states that often lead to opioid misuse and opioid use disorder (OUD). Studies testing the hypothesis that opioid withdrawal potentiates the reinforcing effects of opioid self-administration (SA) are largely inconclusive and have focused on males. Although some clinical evidence indicates that women are more likely than men to misuse opioids to self-medicate, preclinical studies in both sexes are lacking. Based on clinical reports, we hypothesized that withdrawal from escalating-dose morphine injections that approximates a prescription painkiller regimen would lead to increased oxycodone SA to a greater extent in female compared to male rats. After escalating-dose morphine (5-30 mg/kg or vehicle, twice/day for 12 days), rats underwent a 2-week abstinence period during which withdrawal signs were measured. The impact of this treatment was assessed on oxycodone SA acquisition, maintenance, dose response, and progressive ratio responding, with additional analyses to compare sexes. We found that both sexes expressed somatic withdrawal, whereas only males demonstrated hyperalgesia in the warm water tail flick assay. During SA acquisition, males with prior morphine exposure took significantly more oxycodone than females. Finally, females with prior morphine exposure demonstrated the lowest motivation to SA oxycodone in the progressive ratio test. Contrary to our initial hypothesis, our findings suggest that prior opioid exposure increases vulnerability to initiate misuse more in males and decreases the reinforcing efficacy of oxycodone in females.


Asunto(s)
Morfina/administración & dosificación , Narcóticos/administración & dosificación , Trastornos Relacionados con Opioides/tratamiento farmacológico , Oxicodona/administración & dosificación , Animales , Femenino , Masculino , Ratas , Ratas Sprague-Dawley , Refuerzo en Psicología , Autoadministración , Síndrome de Abstinencia a Sustancias/psicología
7.
Front Mol Neurosci ; 12: 236, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31636537

RESUMEN

Anxiety disorders disproportionately affect women compared to men, which may arise from sex differences in stress responses. MiRNAs are small non-coding RNAs known to regulate gene expression through actions on mRNAs. MiRNAs are regulated, in part, by factors such as stress and gonadal sex, and they have been implicated in the pathophysiology of multiple psychiatric disorders. Here, we assessed putative sex differences in miRNA expression in the bed nucleus of the stria terminalis (BNST) - a sexually dimorphic brain region implicated in anxiety - of adult male and female rats that had been exposed to social isolation (SI) stress throughout adolescence. To assess the translational utility of our results, we assessed if childhood trauma in humans resulted in changes in blood miRNA expression that are similar to those observed in rats. Male and female Sprague-Dawley rats underwent SI during adolescence or remained group housed (GH) and were tested for anxiety-like behavior in the elevated plus maze as adults. Small RNA sequencing was performed on tissue extracted from the BNST. Furthermore, we re-analyzed an already available small RNA sequencing data set from the Grady Trauma Project (GTP) from men and women to identify circulating miRNAs that are associated with childhood trauma exposure. Our results indicated that there were greater anxiogenic-like effects and changes in BNST miRNA expression in SI versus GH females compared to SI versus GH males. In addition, we found nine miRNAs that were regulated in both the BNST from SI compared to GH rats and in blood samples from humans exposed to childhood trauma. These studies emphasize the utility of rodent models in studying neurobiological mechanisms underlying psychiatric disorders and suggest that rodent models could be used to identify novel sex-specific pharmacotherapies for anxiety disorders.

8.
Int J Neuropsychopharmacol ; 22(11): 735-745, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31613314

RESUMEN

BACKGROUND: New treatments for stress-related disorders including depression, anxiety, and substance use disorder are greatly needed. Kappa opioid receptors are expressed in the central nervous system, including areas implicated in analgesia and affective state. Although kappa opioid receptor agonists share the antinociceptive effects of mu opioid receptor agonists, they also tend to produce negative affective states. In contrast, selective kappa opioid receptor antagonists have antidepressant- and anxiolytic-like effects, stimulating interest in their therapeutic potential. The prototypical kappa opioid receptor antagonists (e.g., norBNI, JDTic) have an exceptionally long duration of action that complicates their use in humans, particularly in tests to establish safety. This study was designed to test dose- and time-course effects of novel kappa opioid receptor antagonists with the goal of identifying short-acting lead compounds for future medication development. METHODS: We screened 2 novel, highly selective kappa opioid receptor antagonists (CYM-52220 and CYM-52288) with oral efficacy in the warm water tail flick assay in rats to determine initial dose and time course effects. For comparison, we tested existing kappa opioid receptor antagonists JDTic and LY-2456302 (also known as CERC-501 or JNJ-67953964). RESULTS: In the tail flick assay, the rank order of duration of action for the antagonists was LY-2456302 < CYM-52288 < CYM-52220 << JDTic. Furthermore, LY-2456302 blocked the depressive (anhedonia-producing) effects of the kappa opioid receptor agonist U50,488 in the intracranial self-stimulation paradigm, albeit at a higher dose than that needed for analgesic blockade in the tail flick assay. CONCLUSIONS: These results suggest that structurally diverse kappa opioid receptor antagonists can have short-acting effects and that LY-2456302 reduces anhedonia as measured in the intracranial self-stimulation test.


Asunto(s)
3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Analgésicos no Narcóticos/farmacología , Ansiolíticos/farmacología , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Benzamidas/farmacología , Antagonistas de Narcóticos/farmacología , Piperidinas/farmacología , Pirrolidinas/farmacología , Receptores Opioides kappa/antagonistas & inhibidores , Tetrahidroisoquinolinas/farmacología , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/administración & dosificación , Analgésicos no Narcóticos/administración & dosificación , Animales , Ansiolíticos/administración & dosificación , Antidepresivos/administración & dosificación , Benzamidas/administración & dosificación , Desarrollo de Medicamentos , Evaluación Preclínica de Medicamentos , Masculino , Antagonistas de Narcóticos/administración & dosificación , Piperidinas/administración & dosificación , Pirrolidinas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptores Opioides kappa/agonistas , Tetrahidroisoquinolinas/administración & dosificación
9.
Int J Neuropsychopharmacol ; 22(6): 383-393, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-30989210

RESUMEN

BACKGROUND: There is an urgent need to identify factors that increase vulnerability to opioid addiction to help stem the opioid epidemic and develop more efficient pharmacotherapeutics. MicroRNAs are small non-coding RNAs that regulate gene expression at a posttranscriptional level and have been implicated in chronic drug-taking in humans and in rodent models. Recent evidence has shown that chronic opioid treatment regulates the microRNA miR-9. The present study was designed to test the hypothesis that miR-9 in the nucleus accumbens potentiates oxycodone addictive-like behavior. METHODS: We utilized adeno-associated virus (AAV) to overexpress miR-9 in the nucleus accumbens of male rats and tested the effects on intravenous self-administration of the highly abused prescription opioid, oxycodone, in 1-hour short-access followed by 6-h long-access sessions, the latter of which leads to escalation of drug intake. In separate rats, we assessed the effects of nucleus accumbens miR-9 overexpression on mRNA targets including RE1-silencing transcription factor (REST) and dopamine D2 receptor (DRD2), which have been shown to be regulated by drugs of abuse. RESULTS: Overexpression of miR-9 in the nucleus accumbens significantly increased oxycodone self-administration compared with rats expressing a control, scrambled microRNA. Analysis of the pattern of oxycodone intake revealed that miR-9 overexpression increased "burst" episodes of intake and decreased the inter-infusion interval. Furthermore, miR-9 overexpression decreased the expression of REST and increased DRD2 in the nucleus accumbens at time points that coincided with behavioral effects. CONCLUSIONS: These results suggest that nucleus accumbens miR-9 regulates oxycodone addictive-like behavior as well as the expression of genes that are involved in drug addiction.


Asunto(s)
Conducta Adictiva/fisiopatología , MicroARNs/biosíntesis , MicroARNs/fisiología , Núcleo Accumbens/metabolismo , Oxicodona/farmacología , Animales , Conducta Adictiva/inducido químicamente , Dependovirus/genética , Vectores Genéticos/administración & dosificación , Masculino , Microinyecciones , Ratas , Receptores de Dopamina D2/biosíntesis , Proteínas Represoras/biosíntesis , Autoadministración
10.
Front Neurosci ; 11: 128, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28360832

RESUMEN

Melanocortin neurons conserve body mass in hyper- or hypo-caloric conditions by conveying signals from nutrient sensors into areas of the brain governing appetite and metabolism. In mice, melanocortin-3 receptor (MC3R) deletion alters nutrient partitioning independently of hyperphagia, promoting accumulation of fat over muscle mass. Enhanced rhythms in insulin and insulin-responsive metabolic genes during hypocaloric feeding suggest partial insulin resistance and enhanced lipogenesis. However, exactly where and how MC3Rs affect metabolic control to alter nutrient partitioning is not known. The behavioral phenotypes exhibited by MC3R-deficient mice suggest a contextual role in appetite control. The impact of MC3R-deficiency on feeding behavior when food is freely available is minor. However, homeostatic responses to hypocaloric conditioning involving increased expression of appetite-stimulating (orexigenic) neuropeptides, binge-feeding, food anticipatory activity (FAA), entrainment to nutrient availability and enhanced feeding-related motivational responses are compromised with MC3R-deficiency. Rescuing Mc3r transcription in hypothalamic and limbic neurons improves appetitive responses during hypocaloric conditioning while having minor effects on nutrient partitioning, suggesting orexigenic functions. Rescuing hypothalamic MC3Rs also restores responses of fasting-responsive hypothalamic orexigenic neurons in hypocaloric conditions, suggesting actions that sensitize fasting-responsive neurons to signals from nutrient sensors. MC3R signaling in ventromedial hypothalamic SF1(+ve) neurons improves metabolic control, but does not restore appetitive responses or nutrient partitioning. In summary, desensitization of fasting-responsive orexigenic neurons may underlie attenuated appetitive responses of MC3R-deficient mice in hypocaloric situations. Further studies are needed to identify the specific location(s) of MC3Rs controlling appetitive responses and partitioning of nutrients between fat and lean tissues.

11.
Sci Rep ; 7: 44444, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28294152

RESUMEN

Melanocortin-3 receptors (MC3R) have a contextual role in appetite control that is amplified with hypocaloric conditioning. C57BL/6J (B6) mice subjected to hypocaloric feeding schedules (HFS) exhibit compulsive behavioral responses involving food anticipatory activity (FAA) and caloric loading following food access. These homeostatic responses to calorie-poor environs are attenuated in B6 mice in which Mc3r transcription is suppressed by a lox-stop-lox sequence in the 5'UTR (Mc3rTB/TB). Here, we report that optimization of caloric loading in B6 mice subject to HFS, characterized by increased meal size and duration, is not observed in Mc3rTB/TB mice. Analysis of hypothalamic and neuroendocrine responses to HFS throughout the light-dark cycle suggests uncoupling of hypothalamic responses involving appetite-stimulating fasting-responsive hypothalamic neurons expressing agouti-related peptide (AgRP) and neuropeptide Y (Npy). Rescuing Mc3rs expression in Nkx2.1(+ve) neurons is sufficient to restore normal hypothalamic responses to negative energy balance. In addition, Mc3rs expressed in Nkx2.1(+ve) neurons are also sufficient to restore FAA and caloric loading of B6 mice subjected to HFS. In summary, MC3Rs expressed in Nkx2.1(+ve) neurons are sufficient to coordinate hypothalamic response and expression of compulsive behavioral responses involving meal anticipation and consumption of large meals during situations of prolonged negative energy balance.


Asunto(s)
Proteína Relacionada con Agouti/genética , Metabolismo Energético/genética , Neuropéptido Y/genética , Receptor de Melanocortina Tipo 3/genética , Animales , Apetito/genética , Ingestión de Energía/genética , Homeostasis , Hipotálamo/metabolismo , Ratones , Neuronas/metabolismo , Fotoperiodo , Factor Nuclear Tiroideo 1/genética
12.
Psychopharmacology (Berl) ; 234(6): 977-987, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28127624

RESUMEN

RATIONALE: Oxycodone is one of the most widely prescribed painkillers in the USA. However, its use is complicated by high abuse potential. As sex differences have been described in drug addiction, the present study tested for sex differences in intravenous oxycodone self-administration in rats. METHODS: Male and female Sprague-Dawley rats were implanted with jugular vein catheters and trained to self-administer oxycodone (0.03 mg/kg/infusion) under fixed ratio 1 (FR1), FR2, and FR5 schedules of reinforcement followed by a dose-response study to assess sensitivity to the reinforcing effects of oxycodone. In separate rats, sucrose pellet self-administration was assessed under an FR1 schedule to determine whether sex differences in oxycodone self-administration could be generalized across reinforcers. In separate rats, oxycodone distribution to plasma and brain was measured after intravenous drug delivery. RESULTS: In the first 3 trials under an FR1 schedule of reinforcement, male rats self-administered more oxycodone than females. In contrast, females self-administered more sucrose pellets. Under FR2 and FR5 schedules, no significant sex differences in oxycodone intake were observed, although female rats had significantly more inactive lever presses. Male and female rats showed similar inverted U-shaped dose-effect functions, with females tending to self-administer more oxycodone than males at higher doses. No significant sex differences were observed in plasma or brain oxycodone levels, suggesting that sex differences in oxycodone self-administration behavior were not due to pharmacokinetics. CONCLUSION: Our results suggest subtle sex differences in oxycodone self-administration, which may influence the abuse liability of oxycodone and have ramifications for prescription opioid addiction treatment.


Asunto(s)
Condicionamiento Psicológico , Narcóticos/administración & dosificación , Oxicodona/administración & dosificación , Sacarosa/administración & dosificación , Edulcorantes/administración & dosificación , Animales , Femenino , Infusiones Intravenosas , Masculino , Ratas , Ratas Sprague-Dawley , Esquema de Refuerzo , Refuerzo en Psicología , Autoadministración , Factores Sexuales
13.
Mol Metab ; 5(7): 566-579, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27408780

RESUMEN

OBJECTIVE: Appetitive responses to weight loss are mediated by a nutrient-sensing neural network comprised of melanocortin neurons. The role of neural melanocortin-3 receptors (MC3R) in mediating these responses is enigmatic. Mc3r knockout mice exhibit a paradoxical phenotype of obesity and reduced feeding-related behaviors in situations of nutrient scarcity. Here we examined whether MC3Rs expressed in mesolimbic neurons regulate feeding-related motivational responses. METHODS: Interactions between Mc3r genotype, cognitive function and energy balance on food self-administration were assessed using operant conditioning with fixed- and progressive ratio (FR1/PR1) settings. Inhibition of Mc3r transcription by a loxP-flanked transcriptional blocker (TB) in C57BL/6JN mice (Mc3r (TB/TB) ) was reversed in mesolimbic neurons using DAT-Cre (DAT-MC3R). RESULTS: Caloric restriction (CR) caused 10-15% weight loss and increased motivation to acquire food rewards during training sessions. c-Fos-expression in the nucleus accumbens was increased 1 h following food presentation. While exhibiting weight loss, total food self-administration, enhanced motivation to self-administer food rewards in training sessions held during CR and c-Fos-activation in the nucleus accumbens following re-feeding were all markedly attenuated in Mc3r (TB/TB) mice. In contrast, cognitive abilities were normal in Mc3r (TB/TB) mice. Total food self-administration during FR1 sessions was not rescued in DAT-MC3R mice, however enhanced motivational responses to self-administer food rewards in PR1 conditions were restored. The nutrient-partitioning phenotype observed with Mc3r-deficiency was not rescued in DAT-MC3R mice. CONCLUSIONS: Mesolimbic MC3Rs mediate enhanced motivational responses during CR. However, they are insufficient to restore normal caloric loading when food is presented during CR and do not affect metabolic conditions altering nutrient partitioning.

14.
Cell ; 163(5): 1176-1190, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26590421

RESUMEN

The ghrelin receptor (GHSR1a) and dopamine receptor-1 (DRD1) are coexpressed in hippocampal neurons, yet ghrelin is undetectable in the hippocampus; therefore, we sought a function for apo-GHSR1a. Real-time single-molecule analysis on hippocampal neurons revealed dimerization between apo-GHSR1a and DRD1 that is enhanced by DRD1 agonism. In addition, proximity measurements support formation of preassembled apo-GHSR1a:DRD1:Gαq heteromeric complexes in hippocampal neurons. Activation by a DRD1 agonist produced non-canonical signal transduction via Gαq-PLC-IP3-Ca(2+) at the expense of canonical DRD1 Gαs cAMP signaling to result in CaMKII activation, glutamate receptor exocytosis, synaptic reorganization, and expression of early markers of hippocampal synaptic plasticity. Remarkably, this pathway is blocked by genetic or pharmacological inactivation of GHSR1a. In mice, GHSR1a inactivation inhibits DRD1-mediated hippocampal behavior and memory. Our findings identify a previously unrecognized mechanism essential for DRD1 initiation of hippocampal synaptic plasticity that is dependent on GHSR1a, and independent of cAMP signaling.


Asunto(s)
Dopamina/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Ghrelina/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Hipocampo/metabolismo , Memoria , Ratones , Plasticidad Neuronal , Receptores de Dopamina D1/agonistas
15.
Front Neurosci ; 9: 466, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26733781

RESUMEN

Behavioral, biological, and social sequelae that lead to drug addiction differ between men and women. Our efforts to understand addiction on a mechanistic level must include studies in both males and females. Stress, anxiety, and depression are tightly linked to addiction, and whether they precede or result from compulsive drug use depends on many factors, including biological sex. The neuropeptide dynorphin (DYN), an endogenous ligand at kappa opioid receptors (KORs), is necessary for stress-induced aversive states and is upregulated in the brain after chronic exposure to drugs of abuse. KOR agonists produce signs of anxiety, fear, and depression in laboratory animals and humans, findings that have led to the hypothesis that drug withdrawal-induced DYN release is instrumental in negative reinforcement processes that drive addiction. However, these studies were almost exclusively conducted in males. Only recently is evidence available that there are sex differences in the effects of KOR activation on affective state. This review focuses on sex differences in DYN and KOR systems and how these might contribute to sex differences in addictive behavior. Much of what is known about how biological sex influences KOR systems is from research on pain systems. The basic molecular and genetic mechanisms that have been discovered to underlie sex differences in KOR function in pain systems may apply to sex differences in KOR function in reward systems. Our goals are to discuss the current state of knowledge on how biological sex contributes to KOR function in the context of pain, mood, and addiction and to explore potential mechanisms for sex differences in KOR function. We will highlight evidence that the function of DYN-KOR systems is influenced in a sex-dependent manner by: polymorphisms in the prodynorphin (pDYN) gene, genetic linkage with the melanocortin-1 receptor (MC1R), heterodimerization of KORs and mu opioid receptors (MORs), and gonadal hormones. Finally, we identify several gaps in our understanding of "if" and "how" DYN and KORs modulate addictive behavior in a sex-dependent manner. Future work may address these gaps by building on the mechanistic studies outlined in this review. Ultimately this will enable the development of novel and effective addiction treatments tailored to either males or females.

16.
Int J Neuropsychopharmacol ; 18(6)2014 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-25522428

RESUMEN

BACKGROUND: The endocannabinoid system interacts extensively with other neurotransmitter systems and has been implicated in a variety of functions, including regulation of basal ganglia circuits and motor behavior. The present study examined the effects of repeated administration of the nonselective cannabinoid receptor 1 agonist WIN55,212-2 on locomotor activity and on binding and mRNA levels of dopamine receptors and transporters and GABAA receptors in mesostriatal dopaminergic regions of the rat. METHODS: Rats received systemic injections of WIN55,212-2 (0, 0.1, 0.3, or 1mg/kg, intraperitoneally) for 20 consecutive days. Locomotor activity was measured on days 1, 10, and 20. Following the last measurement, rats were euthanized and prepared for in vitro binding and in situ hybridization experiments. RESULTS: Acutely, 0.3 and 1mg/kg of WIN55,212-2 produced hypolocomotion, which was sustained for the next 2 measurements, compared to vehicle. Repeated administration of WIN55,212-2 decreased the mRNA levels of the D2 autoreceptors in substantia nigra and ventral tegmental area and increased D1 receptor mRNA and binding in nucleus accumbens. Furthermore, both dopamine receptor and transporter binding and mRNA levels were decreased in substantia nigra. Moreover, repeated administration of WIN55,212-2 decreased GABAA receptor binding levels in dorsal striatum and substantia nigra. CONCLUSIONS: Our data indicate that chronic WIN55,212-2 administration results in sustained effects on locomotor activity, similar to those observed after acute administration, and modulates the dopaminergic and GABAergic systems in a region-, dose-, and neurotransmitter-selective manner.


Asunto(s)
Ganglios Basales/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Benzoxazinas/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Morfolinas/farmacología , Actividad Motora/efectos de los fármacos , Naftalenos/farmacología , Receptor Cannabinoide CB1/agonistas , Animales , Ganglios Basales/metabolismo , Benzoxazinas/administración & dosificación , Agonistas de Receptores de Cannabinoides/administración & dosificación , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Neuronas Dopaminérgicas/metabolismo , Relación Dosis-Respuesta a Droga , Inyecciones Intraperitoneales , Masculino , Morfolinas/administración & dosificación , Naftalenos/administración & dosificación , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Factores de Tiempo
17.
Endocrinology ; 155(12): 4843-55, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25211592

RESUMEN

The stomach hormone ghrelin and hypothalamic melanocortin neurons belong to a gut-brain circuit controlling appetite and metabolic homeostasis. Mice lacking melanocortin-3 receptor (Mc3rKO) or growth hormone secretagogue receptor (GhsrKO) genes exhibit attenuated food anticipatory activity (FAA), a rise in locomotor activity anticipating mealtime, suggesting common circuitry regulating anticipatory responses to nutrient loading. To investigate the interaction between Ghsrs and Mc3rs, we compared food anticipatory responses in GhsrKO, Mc3rKO, and double Ghsr;Mc3r knockout (DKO) mice subjected to a hypocaloric restricted feeding (RF) protocol in constant dark or 12-hour light, 12-hour dark settings. DKO are viable, exhibiting no overt behavioral or metabolic phenotypes in ad libitum or fasting conditions. FAA was initially attenuated in all mutant strains in constant darkness. However, GhsrKO eventually exhibited a robust food anticipatory response, suggesting compensation. Mc3rKO and DKO did not compensate, indicating a continued requirement for Mc3rs in maintaining the expression of FAA in situations of RF. Abnormal regulation of hypothalamic agouti-related peptide/neuropeptide Y (AgRP/Npy) neurons previously observed during fasting may contribute to attenuated FAA in Mc3rKO. AgRP and Npy expression measured 1 hour before food presentation correlated positively with FAA. Absence of Mc3rs (but not Ghsrs) was associated with lower AgRP/Npy expression, suggesting attenuated responses to signals of negative energy balance. These observations support the importance of Mc3rs as modulators of anticipatory responses to feeding, with mice able to compensate for loss of Ghsrs. The behavioral deficits of Mc3rKO displayed during RF may be partially explained by reduced hunger sensations owing to abnormal regulation of orexigenic AgRP/Npy neurons.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Apetito/fisiología , Actividad Motora/fisiología , Receptor de Melanocortina Tipo 3/metabolismo , Receptores de Ghrelina/metabolismo , Animales , Composición Corporal , Oscuridad , Metabolismo Energético , Privación de Alimentos , Genotipo , Homeostasis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados
18.
Int J Neuropsychopharmacol ; 17(12): 1981-93, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24852388

RESUMEN

Restless legs syndrome (RLS) and Parkinson's disease (PD) are movement disorders usually accompanied by emotional and cognitive deficits. Although D3/D2 receptor agonists are effective against motor and non-motor deficits in RLS and PD, the exact behavioral and neurochemical effects of these drugs are not clearly defined. This study aimed to evaluate the effects of acute ropinirole (0, 0.1, 1 or 10 mg/kg, i.p.), a preferential D3/D2 receptor agonist, on intracranial self-stimulation (ICSS), spontaneous motor activity, anxiety- and depression-like behaviors, spatial reference and working memory in rats as well as on certain markers of neuronal activity, i.e. induction of immediate early genes, such as c-fos and arc, and crucial phosphorylations on GluA1 subunit of α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors and NA1, NA2A and NA2B subunits of N-methyl-D-aspartate (NMDA) receptors. Ropinirole decreased ICSS thresholds and induced anxiolytic- and antidepressive-like effects without affecting motor activity or spatial memory. The effects on emotionality were associated with a decrease in p-Ser897-NA1 and an increase in p-Tyr1472-NA2B in the ventral striatum as well as an increased induction of c-fos messenger RNA (mRNA) in the prefrontal cortex (PFC) and decreased expression of arc mRNA in the striatum and the shell of the nucleus accumbens. Our data indicate that ropinirole significantly affects emotionality at doses (1-10 mg/kg, i.p.) that exert no robust effects on locomotion or cognition. The data reinforce the use of D3/D2 receptor agonists in the treatment of RLS and PD patients characterized by emotional deficits and suggest that altered NMDA-mediated neurotransmission in the limbic forebrain may underlie some of ropinirole's therapeutic actions.


Asunto(s)
Agonistas de Dopamina/farmacología , Emociones/efectos de los fármacos , Indoles/farmacología , Sistema Límbico/efectos de los fármacos , Prosencéfalo/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Proteínas del Citoesqueleto/metabolismo , Relación Dosis-Respuesta a Droga , Emociones/fisiología , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Sistema Límbico/fisiopatología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Proteínas del Tejido Nervioso/metabolismo , Prosencéfalo/fisiopatología , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Autoestimulación/efectos de los fármacos , Autoestimulación/fisiología , Memoria Espacial/efectos de los fármacos , Memoria Espacial/fisiología , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/fisiopatología , Transmisión Sináptica/fisiología
19.
Eur Neuropsychopharmacol ; 24(4): 630-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24275700

RESUMEN

Bipolar disorder (BD) is a severe pathological condition with impaired reward-related processing. The present study was designed to assess the effects of two commonly used BD medications, the mood stabilizer lithium chloride (LiCl) and the atypical antipsychotic and antimanic agent aripiprazole, in an animal model of reward and motivation and on markers of neuroplasticity in the limbic forebrain in rats. We utilized intracranial self-simulation (ICSS) to assess the effects of acute and chronic administration of LiCl and aripiprazole on brain stimulation reward, and phosphorylation studies to determine their effects on specific cellular neuroplasticity markers, i.e., the phosphorylation of CREB and crucial phosphorylation sites on the GluA1 subunit of AMPA receptors and the NA1 and NA2B subunits of NMDA receptors, in the limbic forebrain. Chronic LiCl induced tolerance to the anhedonic effect of the drug observed after acute administration, while chronic aripiprazole induced a sustained anhedonic effect. These distinct behavioral responses might be related to differences in molecular markers of neuroplasticity. Accordingly, we demonstrated that chronic LiCl, but not aripiprazole, decreased phosphorylation of CREB at the Ser133 site and NA1 at the Ser896 site in the prefrontal cortex and GluA1 at the Ser831 site and NA2B at the Ser1303 site in the ventral striatum. The present study provides evidence for BD medication-evoked changes in reward and motivation processes and in specific markers of neuronal plasticity in the limbic forebrain, promoting the notion that these drugs may blunt dysregulated reward processes in BD by counteracting neuronal plasticity deficits.


Asunto(s)
Antipsicóticos/farmacología , Aripiprazol/farmacología , Tolerancia a Medicamentos , Sistema Límbico/efectos de los fármacos , Cloruro de Litio/farmacología , Neuronas/efectos de los fármacos , Prosencéfalo/efectos de los fármacos , Animales , Antipsicóticos/administración & dosificación , Aripiprazol/administración & dosificación , Biomarcadores/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Estimulación Eléctrica , Sistema Límbico/metabolismo , Cloruro de Litio/administración & dosificación , Masculino , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Prosencéfalo/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Subunidades de Proteína/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Recompensa
20.
Int J Neuropsychopharmacol ; 13(4): 541-8, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20047715

RESUMEN

The atypical antipsychotic aripiprazole has been demonstrated to reduce symptoms of bipolar mania. To further profile the antimanic-like properties of aripiprazole in relevant preclinical models, we examined its efficacy in d-amphetamine-based behavioural models of acute mania in rats. The effects of acute and repeated administration of aripiprazole were assessed in the facilitation of intracranial self-stimulation (ICSS) and hyperlocomotion after acute d-amphetamine, and in the sensitized facilitation of ICSS function and hyperlocomotion after repeated d-amphetamine. Acutely, aripiprazole (0.75, 1.5 and 2.5 mg/kg i.p.) increased ICSS thresholds, attenuated the reward-facilitating effects of d-amphetamine (0.5 mg/kg i.p.), decreased motor activity and prevented d-amphetamine-induced hyperlocomotion. Co-administration of aripiprazole and d-amphetamine for 7 d resulted in aripiprazole counteracting the d-amphetamine-induced sensitization in facilitation of brain reward function and hyperlocomotion. These results indicate the efficacy of aripiprazole in d-amphetamine-based preclinical models of acute mania that are characterized by increased motivational drive and/or hyperfunction of brain reward.


Asunto(s)
Trastorno Bipolar/inducido químicamente , Trastorno Bipolar/tratamiento farmacológico , Dextroanfetamina/farmacología , Modelos Animales de Enfermedad , Piperazinas/farmacología , Quinolonas/farmacología , Autoestimulación/efectos de los fármacos , Animales , Antipsicóticos/administración & dosificación , Antipsicóticos/farmacología , Aripiprazol , Estimulantes del Sistema Nervioso Central/administración & dosificación , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/farmacología , Dextroanfetamina/administración & dosificación , Dextroanfetamina/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Locomoción/efectos de los fármacos , Masculino , Piperazinas/administración & dosificación , Quinolonas/administración & dosificación , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA