Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Brain Behav Immun ; 109: 271-284, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36746342

RESUMEN

Prenatal infections can increase the risk of developing psychiatric disorders such as schizophrenia in the offspring, especially when combined with other postnatal insults. Here, we tested, in a rat model of prenatal immune challenge by the viral mimic polyriboinosinic-polyribocytidilic acid, whether maternal immune activation (MIA) affects the endocannabinoid system and endocannabinoid-mediated modulation of dopamine functions. Experiments were performed during adolescence to assess i) the behavioral endophenotype (locomotor activity, plus maze, prepulse inhibition of startle reflex); ii) the locomotor activity in response to Δ9-Tetrahydrocannabinol (THC) and iii) the properties of ventral tegmental area (VTA) dopamine neurons in vivo and their response to THC; iv) endocannabinoid-mediated synaptic plasticity in VTA dopamine neurons; v) the expression of cannabinoid receptors and enzymes involved in endocannabinoid synthesis and catabolism in mesolimbic structures and vi) MIA-induced neuroinflammatory scenario evaluated by measurements of levels of cytokine and neuroinflammation markers. We revealed that MIA offspring displayed an altered locomotor activity in response to THC, a higher bursting activity of VTA dopamine neurons and a lack of response to cumulative doses of THC. Consistently, MIA adolescence offspring showed an enhanced 2-arachidonoylglycerol-mediated synaptic plasticity and decreased monoacylglycerol lipase activity in mesolimbic structures. Moreover, they displayed a higher expression of cyclooxygenase 2 (COX-2) and ionized calcium-binding adaptor molecule 1 (IBA-1), associated with latent inflammation and persistent microglia activity. In conclusion, we unveiled neurobiological mechanisms whereby inflammation caused by MIA influences the proper development of endocannabinoid signaling that negatively impacts the dopamine system, eventually leading to psychotic-like symptoms in adulthood.


Asunto(s)
Efectos Tardíos de la Exposición Prenatal , Esquizofrenia , Embarazo , Femenino , Ratas , Masculino , Animales , Humanos , Endocannabinoides/metabolismo , Dopamina/metabolismo , Transducción de Señal , Neuronas Dopaminérgicas/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(18): 9991-10002, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32312805

RESUMEN

The initial response to an addictive substance can facilitate repeated use: That is, individuals experiencing more positive effects are more likely to use that drug again. Increasing evidence suggests that psychoactive cannabinoid use in adolescence enhances the behavioral effects of cocaine. However, despite the behavioral data, there is no neurobiological evidence demonstrating that cannabinoids can also alter the brain's initial molecular and epigenetic response to cocaine. Here, we utilized a multiomics approach (epigenomics, transcriptomics, proteomics, and phosphoproteomics) to characterize how the rat brain responds to its first encounter with cocaine, with or without preexposure to the synthetic cannabinoid WIN 55,212-2 (WIN). We find that in adolescent (but not in adult) rats, preexposure to WIN results in cross-sensitization to cocaine, which correlates with histone hyperacetylation and decreased levels of HDAC6 in the prefrontal cortex (PFC). In the PFC, we also find that WIN preexposure blunts the typical mRNA response to cocaine and instead results in alternative splicing and chromatin accessibility events, involving genes such as Npas2 Moreover, preexposure to WIN enhances the effects of cocaine on protein phosphorylation, including ERK/MAPK-targets like gephyrin, and modulates the synaptic AMPAR/GluR composition both in the PFC and the nucleus accumbens (NAcc). PFC-NAcc gene network topological analyses, following cocaine exposure, reveal distinct top nodes in the WIN preexposed group, which include PACAP/ADCYAP1. These preclinical data demonstrate that adolescent cannabinoid exposure reprograms the initial behavioral, molecular, and epigenetic response to cocaine.


Asunto(s)
Conducta Adictiva/genética , Conducta Animal/efectos de los fármacos , Cannabinoides/efectos adversos , Cocaína/efectos adversos , Adolescente , Animales , Conducta Adictiva/inducido químicamente , Conducta Adictiva/patología , Benzoxazinas/efectos adversos , Benzoxazinas/farmacología , Cannabinoides/farmacología , Péptidos y Proteínas de Señalización del Ritmo Circadiano/genética , Cocaína/farmacología , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Regulación de la Expresión Génica/efectos de los fármacos , Histona Desacetilasa 6/genética , Humanos , Proteínas de la Membrana/farmacología , Morfolinas/efectos adversos , Morfolinas/farmacología , Naftalenos/efectos adversos , Naftalenos/farmacología , Fosfoproteínas/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Corteza Prefrontal/efectos de los fármacos , Proteoma/efectos de los fármacos , Ratas , Transcriptoma/efectos de los fármacos
3.
Int J Mol Sci ; 24(1)2022 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-36614142

RESUMEN

The regular use of cannabis during adolescence has been associated with a number of negative life outcomes, including psychopathology and cognitive impairments. However, the exact molecular mechanisms that underlie these outcomes are just beginning to be understood. Moreover, very little is known about the spatio-temporal molecular changes that occur following cannabinoid exposure in adolescence. To understand these changes, we exposed mid-adolescent male rats to a synthetic cannabinoid (WIN 55,212-2 mesylate; WIN) and, following drug abstinence through late adolescence, we subjected the synaptosomal fractions of the prefrontal cortex (PFC) to proteomic analyses. A total of N = 487 differentially expressed proteins were found in WIN-exposed animals compared to controls. Gene ontology analyses revealed enrichment of terms related to the gamma-aminobutyric acid (GABA)-ergic neurotransmitter system. Among the top differentially expressed proteins was the synaptic Ras GTPase-activating protein 1 (SYNGAP1). Using Western blotting experiments, we found that the WIN-induced upregulation of SYNGAP1 was spatio-temporal in nature, arising only in the synaptosomal fractions (not in the cytosol) and only following prolonged drug abstinence (not on abstinence day 1). Moreover, the SYNGAP1 changes were found to be specific to WIN-exposure in adolescence and not adulthood. Adolescent animals exposed to a natural cannabinoid (Δ9-tetrahydrocannabinol; THC) were also found to have increased levels of SYNGAP1 in the PFC. THC exposure also led to a pronounced upregulation of SYNGAP1 in the amygdala, but without any changes in the dorsal striatum, hippocampus, or nucleus accumbens. To our knowledge, this is the first study to uncover a link between cannabinoid exposure and changes in SYNGAP1 that are spatio-temporal and developmental in nature. Future studies are needed to investigate the putative role of SYNGAP1 in the negative behavioral consequences of cannabis use in adolescence.


Asunto(s)
Cannabinoides , Proteínas Activadoras de GTPasa , Animales , Masculino , Ratas , Agonistas de Receptores de Cannabinoides , Cannabinoides/farmacología , Dronabinol/farmacología , Núcleo Accumbens/metabolismo , Corteza Prefrontal/metabolismo , Proteómica , Proteínas Activadoras de GTPasa/metabolismo
4.
Molecules ; 27(13)2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-35807384

RESUMEN

COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a global pandemic that might lead to very serious consequences. Notably, mental status change, brain confusion, and smell and taste disorders along with neurological complaints have been reported in patients infected with SARS-CoV-2. Furthermore, human brain tissue autopsies from COVID-19 patients show the presence of SARS-CoV-2 neuroinvasion, which correlates with the manifestation of meningitis, encephalitis, leukocyte infiltration, and neuronal damage. The olfactory mucosa has been suggested as a way of entry into the brain. SARS-CoV-2 infection is also known to provoke a hyper-inflammatory reaction with an exponential increase in the production of pro-inflammatory cytokines leading to systemic responses, even in the absence of direct infection of brain cells. Angiotensin-converting enzyme 2 (ACE2), the entry receptor of SARS-CoV-2, has been extensively demonstrated to be present in the periphery, neurons, and glial cells in different brain regions. To dissect the details of neurological complications and develop therapies helping COVID-19 survivors regain pre-infection quality of life, the development of robust clinical models is highly warranted. Several human angiotensin-converting enzyme 2 (hACE2) transgenic mouse models have been developed and used for antiviral drug screening and vaccine development, as well as for better understanding of the molecular pathogenetic mechanisms of SARS-CoV-2 infection. In this review, we summarize recent results from the studies involving two such mouse models, namely K18- and CAG-hACE2 transgenics, to evaluate the direct and indirect impact of SARS-CoV-2 infection on the central nervous system.


Asunto(s)
COVID-19 , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/genética , Animales , Modelos Animales de Enfermedad , Melfalán , Ratones , Ratones Transgénicos , Peptidil-Dipeptidasa A , Calidad de Vida , gammaglobulinas
5.
Int J Mol Sci ; 22(4)2021 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-33668469

RESUMEN

Cannabidiol (CBD) is the most abundant non-psychoactive component of cannabis; it displays a very low affinity for cannabinoid receptors, facilitates endocannabinoid signaling by inhibiting the hydrolysis of anandamide, and stimulates both transient receptor potential vanilloid 1 and 2 and serotonin type 1A receptors. Since CBD interacts with a wide variety of molecular targets in the brain, its therapeutic potential has been investigated in a number of neuropsychiatric diseases, including anxiety and mood disorders. Specifically, CBD has received growing attention due to its anxiolytic and antidepressant properties. As a consequence, and given its safety profile, CBD is considered a promising new agent in the treatment of anxiety and mood disorders. However, the exact molecular mechanism of action of CBD still remains unknown. In the present preclinical review, we provide a summary of animal-based studies that support the use of CBD as an anxiolytic- and antidepressant-like compound. Next, we describe neuropharmacological evidence that links the molecular pharmacology of CBD to its behavioral effects. Finally, by taking into consideration the effects of CBD on DNA methylation, histone modifications, and microRNAs, we elaborate on the putative role of epigenetic mechanisms in mediating CBD's therapeutic outcomes.


Asunto(s)
Ansiolíticos/uso terapéutico , Trastornos de Ansiedad , Cannabidiol/uso terapéutico , Epigénesis Genética/efectos de los fármacos , Trastornos del Humor , Animales , Trastornos de Ansiedad/tratamiento farmacológico , Trastornos de Ansiedad/metabolismo , Trastornos de Ansiedad/patología , Humanos , Trastornos del Humor/tratamiento farmacológico , Trastornos del Humor/metabolismo , Trastornos del Humor/patología , Receptor de Serotonina 5-HT1A/metabolismo , Canales Catiónicos TRPV/metabolismo
6.
Int J Eat Disord ; 53(5): 432-446, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32275093

RESUMEN

OBJECTIVE: Both environmental and genetic factors are known to contribute to the development of anorexia nervosa (AN), but the exact etiology remains poorly understood. Herein, we studied the transcriptional regulation of the endocannabinoid system, an interesting target for body weight maintenance and the control of food intake and energy balance. METHOD: We used two well-characterized animal models of AN: (a) the activity-based anorexia (ABA) model in which rats, housed with running wheels and subjected to daily food restriction, show reductions in body weight and increase in physical activity; (b) the genetic anx/anx mouse displaying the core features of AN: low food intake and emaciation. RESULTS: Among the evaluated endocannabinoid system components, we observed a selective and significant down-regulation of the gene encoding for the type 1 cannabinoid receptor (Cnr1) in ABA rats' hypothalamus and nucleus accumbens and, in the latter area, a consistent, significant and correlated increase in DNA methylation at the gene promoter. No changes were evident in the anx/anx mice except for a down-regulation of Cnr1, in the prefrontal cortex. DISCUSSION: Our findings support a possible role for Cnr1 in the ABA animal model of AN. In particular, its regulation in the nucleus accumbens appears to be triggered by environmental cues due to the consistent epigenetic modulation of the promoter. These data warrant further studies on Cnr1 regulation as a possible target for treatment of AN.

7.
Int J Mol Sci ; 21(22)2020 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-33198335

RESUMEN

The accumulation of aggregated α-synuclein (αSyn) is a hallmark of Parkinson's disease (PD). Current evidence indicates that small soluble αSyn oligomers (αSynOs) are the most toxic species among the forms of αSyn aggregates, and that size and topological structural properties are crucial factors for αSynOs-mediated toxicity, involving the interaction with either neurons or glial cells. We previously characterized a human αSynO (H-αSynO) with specific structural properties promoting toxicity against neuronal membranes. Here, we tested the neurotoxic potential of these H-αSynOs in vivo, in relation to the neuropathological and symptomatic features of PD. The H-αSynOs were unilaterally infused into the rat substantia nigra pars compacta (SNpc). Phosphorylated αSyn (p129-αSyn), reactive microglia, and cytokine levels were measured at progressive time points. Additionally, a phagocytosis assay in vitro was performed after microglia pre-exposure to αsynOs. Dopaminergic loss, motor, and cognitive performances were assessed. H-αSynOs triggered p129-αSyn deposition in SNpc neurons and microglia and spread to the striatum. Early and persistent neuroinflammatory responses were induced in the SNpc. In vitro, H-αSynOs inhibited the phagocytic function of microglia. H-αsynOs-infused rats displayed early mitochondrial loss and abnormalities in SNpc neurons, followed by a gradual nigrostriatal dopaminergic loss, associated with motor and cognitive impairment. The intracerebral inoculation of structurally characterized H-αSynOs provides a model of progressive PD neuropathology in rats, which will be helpful for testing neuroprotective therapies.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad de Parkinson/fisiopatología , Sustancia Negra/metabolismo , alfa-Sinucleína/metabolismo , Animales , Citocinas/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Humanos , Inflamación , Masculino , Microglía/metabolismo , Neuronas/metabolismo , Fagocitosis , Fosforilación , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Sustancia Negra/patología
8.
Acta Pharmacol Sin ; 40(3): 309-323, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30050084

RESUMEN

Anandamide is a lipid mediator that acts as an endogenous ligand of CB1 receptors. These receptors are also the primary molecular target responsible for the pharmacological effects of Δ9-tetrahydrocannabinol, the psychoactive ingredient in Cannabis sativa. Several studies demonstrate that anandamide exerts an overall modulatory effect on the brain reward circuitry. Several reports suggest its involvement in the addiction-producing actions of other abused drugs, and it can also act as a behavioral reinforcer in animal models of drug abuse. Importantly, all these effects of anandamide appear to be potentiated by pharmacological inhibition of its metabolic degradation. Enhanced brain levels of anandamide after treatment with inhibitors of fatty acid amide hydrolase, the main enzyme responsible for its degradation, seem to affect the rewarding and reinforcing actions of many drugs of abuse. In this review, we will provide an overview from a preclinical perspective of the current state of knowledge regarding the behavioral pharmacology of anandamide, with a particular emphasis on its motivational/reinforcing properties. We will also discuss how modulation of anandamide levels through inhibition of enzymatic metabolic pathways could provide a basis for developing new pharmaco-therapeutic tools for the treatment of substance use disorders.


Asunto(s)
Ácidos Araquidónicos/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Endocannabinoides/fisiología , Amidohidrolasas/antagonistas & inhibidores , Animales , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Alcamidas Poliinsaturadas , Autoadministración
9.
Int J Eat Disord ; 52(11): 1251-1262, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31456239

RESUMEN

OBJECTIVE: Despite the growing knowledge on the functional relationship between an altered endocannabinoid (eCB) system and development of anorexia nervosa (AN), to date no studies have investigated the central eCB tone in the activity-based anorexia (ABA) model that reproduces key aspects of human AN. METHOD: We measured levels of two major eCBs, anandamide (AEA) and 2-arachidonoylglycerol (2-AG), those of two eCB-related lipids, oleoylethanolamide (OEA) and palmitoylethanolamide (PEA), and the cannabinoid type-1 receptor (CB1R) density in the brain of female ABA rats, focusing on areas involved in homeostatic and rewarding-related regulation of feeding behavior (i.e., prefrontal cortex, nucleus accumbens, caudato putamen, amygdala, hippocampus and hypothalamus). Analysis was carried out also at the end of recovery from the ABA condition. RESULTS: At the end of the ABA induction phase, 2-AG was significantly decreased in ABA rats in different brain areas but not in the caudato putamen. No changes were detected in AEA levels in any region, whereas the levels of OEA and PEA were decreased exclusively in the hippocampus and hypothalamus. Furthermore, CB1R density was decreased in the dentate gyrus of hippocampus and in the lateral hypothalamus. After recovery, both 2-AG levels and CB1R density were partially normalized in some areas. In contrast, AEA levels became markedly reduced in all the analyzed areas. DISCUSSION: These data demonstrate an altered brain eCB tone in ABA rats, further supporting the involvement of an impaired eCB system in AN pathophysiology that may contribute to the maintenance of some symptomatic aspects of the disease.


Asunto(s)
Anorexia Nerviosa/inducido químicamente , Encéfalo/efectos de los fármacos , Endocannabinoides/efectos adversos , Animales , Femenino , Humanos , Ratas , Ratas Sprague-Dawley
10.
Schizophr Res ; 266: 197-204, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38422890

RESUMEN

Remission, relapse prevention, and clinical recovery are crucial areas of interest in schizophrenia (SCZ) research. Although SCZ is a chronic disorder with poor overall outcomes, years of research demonstrated that recovery is possible. There are considerable data linking brain-derived neurotrophic factor (BDNF) to SCZ, however, evidence on the role of BDNF in remission in SCZ is scarce. This secondary analysis of the Longitudinal Assessment of BDNF in Sardinian patients (LABSP) data aimed to investigate the relationship between serum BDNF levels and symptomatic remission, simultaneous clinical and functional remission, and recovery in patients with SCZ. A total of 105 patients with SCZ or schizoaffective disorder were recruited for a longitudinal assessment of BDNF levels over 24 months. Longitudinal data were analyzed using mixed-effects linear regression models. The study found significant associations between use of long acting injectables (χ2 = 7.075, df = 1, p = 0.008), baseline serum BDNF levels (U = 701, z = -2.543, p = 0.011), and "childhood" (U = 475, z = -2.124, p = 0.034) and "general" (U = 55, z = -2.014, p = 0.044) subscales of the Premorbid Adjustment Scale (PAS) with patients maintaining remission and recovery. The diagnosis of SCZ was significantly associated with lower BDNF levels for patients with simultaneous clinical and functional remission (Z = 2.035, p = 0.0419) and recovery (Z = 2.009, p = 0.0445) compared to those without. There were no significant associations between remission in the entire sample and longitudinal serum BDNF levels or genetic variants within the BDNF gene. These findings provide further insight into the complex relationship between BDNF and SCZ.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Trastornos Psicóticos , Esquizofrenia , Humanos , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Trastornos Psicóticos/genética , Trastornos Psicóticos/terapia , Esquizofrenia/genética , Esquizofrenia/terapia , Prevención Secundaria , Inducción de Remisión
11.
Nutrients ; 15(9)2023 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-37432348

RESUMEN

The pathophysiology of Anorexia Nervosa (AN) has not been fully elucidated. Anaplastic lymphoma kinase (ALK) receptor is a protein-tyrosine kinase mainly known as a key oncogenic driver. Recently, a genetic deletion of ALK in mice has been found to increase energy expenditure and confers resistance to obesity in these animals, suggesting its role in the regulation of thinness. Here, we investigated the expression of ALK and the downstream intracellular pathways in female rats subjected to the activity-based anorexia (ABA) model, which reproduces important features of human AN. In the hypothalamic lysates of ABA rats, we found a reduction in ALK receptor expression, a downregulation of Akt phosphorylation, and no change in the extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) phosphorylation. After the recovery from body weight loss, ALK receptor expression returned to the control baseline values, while it was again suppressed during a second cycle of ABA induction. Overall, this evidence suggests a possible involvement of the ALK receptor in the pathophysiology of AN, that may be implicated in its stabilization, resistance, and/or its exacerbation.


Asunto(s)
Anorexia Nerviosa , Humanos , Femenino , Animales , Ratones , Ratas , Quinasa de Linfoma Anaplásico , Anorexia , Proteínas Tirosina Quinasas , Fosforilación
12.
Behav Brain Res ; 444: 114374, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-36863461

RESUMEN

Compelling data support altered dopamine (DA) and serotonin (5-HT) signaling in anorexia nervosa (AN). However, their exact role in the etiopathogenesis of AN has yet to be elucidated. Here, we evaluated the corticolimbic brain levels of DA and 5-HT in the induction and recovery phases of the activity-based anorexia (ABA) model of AN. We exposed female rats to the ABA paradigm and measured the levels of DA, 5-HT, the metabolites 3,4-dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA), 5-hydroxyindoleacetic acid (5-HIAA), and the dopaminergic type 2 (D2) receptors density in feeding- and reward-implicated brain regions (i.e., cerebral cortex, Cx; prefrontal cortex, PFC; caudate putamen, CPu; nucleus accumbens, NAcc; amygdala, Amy; hypothalamus, Hyp; hippocampus, Hipp). DA levels were significantly increased in the Cx, PFC and NAcc, while 5-HT was significantly enhanced in the NAcc and Hipp of ABA rats. Following recovery, DA was still elevated in the NAcc, while 5-HT was increased in the Hyp of recovered ABA rats. DA and 5-HT turnover were impaired at both ABA induction and recovery. D2 receptors density was increased in the NAcc shell. These results provide further proof of the impairment of the dopaminergic and serotoninergic systems in the brain of ABA rats and support the knowledge of the involvement of these two important neurotransmitter systems in the development and progression of AN. Thus, providing new insights on the corticolimbic regions involved in the monoamine dysregulations in the ABA model of AN.


Asunto(s)
Dopamina , Serotonina , Ratas , Femenino , Animales , Dopamina/metabolismo , Serotonina/metabolismo , Encéfalo/metabolismo , Ácido Homovanílico , Núcleo Accumbens/metabolismo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Ácido Hidroxiindolacético/metabolismo
13.
Nutrients ; 15(17)2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37686862

RESUMEN

Anorexia nervosa (AN) is a complex eating disorder characterized by reduced caloric intake to achieve body-weight loss. Furthermore, over-exercise is commonly reported. In recent years, animal models of AN have provided evidence for neuroplasticity changes in specific brain areas of the mesocorticolimbic circuit, which controls a multitude of functions including reward, emotion, motivation, and cognition. The activity-regulated cytoskeleton-associated protein (Arc) is an immediate early gene that modulates several forms of synaptic plasticity and has been linked to neuropsychiatric illness. Since the role of Arc in AN has never been investigated, in this study we evaluated whether the anorexic-like phenotype reproduced by the activity-based anorexia (ABA) model may impact its expression in selected brain regions that belong to the mesocorticolimbic circuit (i.e., prefrontal cortex, nucleus accumbens, and hippocampus). The marker of neuronal activation c-Fos was also assessed. We found that the expression of both markers increased in all the analyzed brain areas of ABA rats in comparison to the control groups. Moreover, a negative correlation between the density of Arc-positive cells and body-weight loss was found. Together, our findings suggest the importance of Arc and neuroplasticity changes within the brain circuits involved in dysfunctional behaviors associated with AN.


Asunto(s)
Anorexia Nerviosa , Animales , Ratas , Anorexia , Modelos Animales , Citoesqueleto , Pérdida de Peso
14.
Eur Psychiatry ; 65(1): e71, 2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36281033

RESUMEN

BACKGROUND AND HYPOTHESIS: Schizophrenia spectrum disorders are among the most debilitating mental disorders and has complex pathophysiological underpinnings. There is growing evidence that brain-derived neurotrophic factor (BDNF) can play a role in its pathogenesis. The present study investigated the longitudinal variation of serum BDNF levels in a 24-month observational prospective cohort study of Sardinian psychotic patients and its relationship with psychopathological and cognitive changes. Furthermore, we examined whether genetic variation within the BDNF gene could moderate these relationships. STUDY DESIGN: Every 6 months, 105 patients were assessed for their BDNF serum levels, as well as for a series of psychopathological, cognitive, and social measures. We performed a targeted analysis of four tag single nucleotide polymorphisms within the BDNF gene that were selected and analyzed using polymerase chain reaction. Longitudinal data were analyzed using mixed-effects linear regression models. STUDY RESULTS: We observed a declining longitudinal trajectory of BDNF levels in psychotic patients in general, and in relation to the severity of depressive and negative symptoms. BDNF serum levels also declined in patients scoring lower in cognitive measures such as attention and speed of information processing and verbal fluency. The rs7934165 polymorphism moderated the significant association between verbal fluency and BDNF levels. CONCLUSIONS: These findings in patients from real-world settings suggest a plausible role of peripheral BDNF levels as a marker of illness burden in schizophrenia spectrum disorders.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Esquizofrenia , Humanos , Factor Neurotrófico Derivado del Encéfalo/genética , Estudios Prospectivos , Esquizofrenia/diagnóstico , Cognición/fisiología , Polimorfismo de Nucleótido Simple
15.
Cells ; 11(17)2022 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-36078036

RESUMEN

Parkinson's disease (PD) is a complex pathology causing a plethora of non-motor symptoms besides classical motor impairments, including cognitive disturbances. Recent studies in the PD human brain have reported microgliosis in limbic and neocortical structures, suggesting a role for neuroinflammation in the development of cognitive decline. Yet, the mechanism underlying the cognitive pathology is under investigated, mainly for the lack of a valid preclinical neuropathological model reproducing the disease's motor and non-motor aspects. Here, we show that the bilateral intracerebral infusion of pre-formed human alpha synuclein oligomers (H-αSynOs) within the substantia nigra pars compacta (SNpc) offers a valid model for studying the cognitive symptoms of PD, which adds to the classical motor aspects previously described in the same model. Indeed, H-αSynOs-infused rats displayed memory deficits in the two-trial recognition task in a Y maze and the novel object recognition (NOR) test performed three months after the oligomer infusion. In the anterior cingulate cortex (ACC) of H-αSynOs-infused rats the in vivo electrophysiological activity was altered and the expression of the neuron-specific immediate early gene (IEG) Npas4 (Neuronal PAS domain protein 4) and the AMPA receptor subunit GluR1 were decreased. The histological analysis of the brain of cognitively impaired rats showed a neuroinflammatory response in cognition-related regions such as the ACC and discrete subareas of the hippocampus, in the absence of any evident neuronal loss, supporting a role of neuroinflammation in cognitive decline. We found an increased GFAP reactivity and the acquisition of a proinflammatory phenotype by microglia, as indicated by the increased levels of microglial Tumor Necrosis Factor alpha (TNF-α) as compared to vehicle-infused rats. Moreover, diffused deposits of phospho-alpha synuclein (p-αSyn) and Lewy neurite-like aggregates were found in the SNpc and striatum, suggesting the spreading of toxic protein within anatomically interconnected areas. Altogether, we present a neuropathological rat model of PD that is relevant for the study of cognitive dysfunction featuring the disease. The intranigral infusion of toxic oligomeric species of alpha-synuclein (α-Syn) induced spreading and neuroinflammation in distant cognition-relevant regions, which may drive the altered neuronal activity underlying cognitive deficits.


Asunto(s)
Disfunción Cognitiva , Enfermedad de Parkinson , Animales , Disfunción Cognitiva/metabolismo , Giro del Cíngulo/metabolismo , Giro del Cíngulo/patología , Humanos , Enfermedades Neuroinflamatorias , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Ratas , Sustancia Negra/metabolismo , alfa-Sinucleína/metabolismo
16.
Brain Sci ; 12(12)2022 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-36552127

RESUMEN

Brain-derived neurotrophic factor (BDNF) is a key modulator of neuroplasticity and has an important role in determining the susceptibility to severe psychiatric disorder with a significant neurodevelopmental component such as major psychoses. Indeed, a potential association between BDNF serum levels and schizophrenia (SCZ) and schizoaffective disorder (SAD) has been tested in diverse studies and a considerable amount of them found reduced BDNF levels in these disorders. Here, we aimed at testing the association of BDNF serum levels with several demographic, clinical, and psychometric measures in 105 patients with SCZ and SAD, assessing the moderating effect of genetic variants within the BDNF gene. We also verified whether peripheral BDNF levels differed between patients with SCZ and SAD. Our findings revealed that BDNF serum levels are significantly lower in patients affected by SCZ and SAD presenting more severe depressive symptomatology. This finding awaits replication in future independent studies and points to BDNF as a possible prognostic indicator in major psychoses.

17.
Methods Mol Biol ; 2201: 221-229, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32975803

RESUMEN

Opioid addiction in humans is a chronically relapsing disorder characterized by discontinuous periods of drug use and abstinence resulting in dependence. With time, the probability of falling into renewed drug consumption becomes particularly high and constitutes a considerable problem in the management of opioid addicts. Opioid addiction represents an important health concern and animal models have been crucial in understanding the neurobiology and pathophysiology of this complex disease. Although animal models of addiction do not fully reproduce the human condition, they do permit investigation of specific elements of the process as well as identification of potential therapeutic targets. In this chapter, we provide a step-by-step description of the morphine-conditioned place preference (CPP) model that represents a useful preclinical animal model extensively used to study the rewarding/aversive effect of drugs.


Asunto(s)
Control de la Conducta/métodos , Condicionamiento Operante/efectos de los fármacos , Trastornos Relacionados con Opioides/fisiopatología , Analgésicos Opioides/farmacología , Animales , Conducta Adictiva/fisiopatología , Condicionamiento Operante/fisiología , Modelos Animales de Enfermedad , Masculino , Morfina/farmacología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Recompensa
18.
Br J Pharmacol ; 178(17): 3476-3497, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33837969

RESUMEN

BACKGROUND AND PURPOSE: Spice/K2 herbal mixtures, containing synthetic cannabinoids such as JWH-018, have been marketed as marijuana surrogates since 2004. JWH-018 has cannabinoid CB1 receptor-dependent reinforcing properties and acutely increases dopaminergic transmission selectively in the NAc shell. Here, we tested the hypothesis that repeated administration of JWH-018 (i) modulates behaviour, (ii) affects dopaminergic transmission and its responsiveness to motivational stimuli, and (iii) is associated with a neuroinflammatory phenotype. EXPERIMENTAL APPROACH: Rats were administered with JWH-018 once a day for 14 consecutive days. We then performed behavioural, electrophysiological, and neurochemical evaluation at multiple time points after drug discontinuation. KEY RESULTS: Repeated JWH-018 exposure (i) induced anxious and aversive behaviours, transitory attentional deficits, and withdrawal signs; (ii) decreased spontaneous activity and number of dopamine neurons in the VTA; and (iii) reduced stimulation of dopaminergic transmission in the NAc shell while potentiating that in the NAc core, in response to acute JWH-018 challenge. Moreover, (iv) we observed a decreased dopamine sensitivity in the NAc shell and core, but not in the mPFC, to a first chocolate exposure; conversely, after a second exposure, dialysate dopamine fully increased in the NAc shell and core but not in the mPFC. Finally, selected dopamine brain areas showed (v) astrogliosis (mPFC, NAc shell and core, VTA), microgliosis (NAc shell and core), and downregulation of CB1 receptors (mPFC, NAc shell and core). CONCLUSION AND IMPLICATIONS: Repeated exposure to JWH-018 may provide a useful model to clarify the detrimental effects of recurring use of Spice/K2 drugs.


Asunto(s)
Dopamina , Naftalenos , Animales , Indoles/farmacología , Naftalenos/farmacología , Neuroglía , Núcleo Accumbens , Ratas
19.
Learn Mem ; 16(5): 332-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19403796

RESUMEN

Inhibitors of fatty acid amide hydrolase (FAAH) increase endogenous levels of anandamide (a cannabinoid CB(1)-receptor ligand) and oleoylethanolamide and palmitoylethanolamide (OEA and PEA, ligands for alpha-type peroxisome proliferator-activated nuclear receptors, PPAR-alpha) when and where they are naturally released in the brain. Using a passive-avoidance task in rats, we found that memory acquisition was enhanced by the FAAH inhibitor URB597 or by the PPAR-alpha agonist WY14643, and these enhancements were blocked by the PPAR-alpha antagonist MK886. These findings demonstrate novel mechanisms for memory enhancement by activation of PPAR-alpha, either directly by administering a PPAR-alpha agonist or indirectly by administering a FAAH inhibitor.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Encéfalo/fisiología , Aprendizaje/fisiología , Memoria/fisiología , PPAR alfa/metabolismo , Animales , Benzamidas/farmacología , Encéfalo/efectos de los fármacos , Carbamatos/farmacología , Inhibidores Enzimáticos/farmacología , Masculino , Memoria/efectos de los fármacos , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/metabolismo
20.
Dialogues Clin Neurosci ; 22(3): 271-279, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-33162770

RESUMEN

Mental disorders represent a significant public health burden worldwide due to their high prevalence, chronically disabling nature, and substantial impact on quality of life. Despite growing knowledge of the pathological mechanisms that underlie the development of these disorders, a high percentage of patients do not respond to first-line clinical treatments; thus, there is a strong need for alternative therapeutic approaches. During the past half-century, after the identification of the endocannabinoid system and its role in multiple physiological processes, both natural and synthetic cannabinoids have attracted considerable interest as putative medications in pathological conditions such as, but not exclusive to, mental disorders. Here, we provide a summary of cannabinoid effects in support of possible therapeutic applications for major depression, bipolar disorder, anxiety, posttraumatic stress disorder, and schizophrenia. Considering this evidence, highlighted benefits and risks of cannabinoid use in the management of these illnesses require further experimental study.
.


Los trastornos mentales representan una carga importante para la salud pública en todo el mundo debido a su alta prevalencia, su naturaleza crónica con discapacidad y su impacto significativo en la calidad de vida. A pesar del creciente conocimiento de los mecanismos patológicos que subyacen al desarrollo de estos trastornos, un alto porcentaje de pacientes no responde a los tratamientos clínicos de primera línea; por lo que existe una gran necesidad de enfoques terapéuticos alternativos. Durante los últimos cincuenta años, después de la identificación del sistema endocannabinoide y su papel en múltiples procesos fisiológicos, tanto los cannabinoides naturales como los sintéticos han concentrado un alto interés como posibles fármacos para trastornos mentales y otras patologías. En este artículo se resumen los efectos de los cannabinoides en apoyo de posibles aplicaciones terapéuticas para la depresión mayor, el trastorno bipolar, la ansiedad, el trastorno por estrés postraumático y la esquizofrenia. Teniendo en cuenta esta evidencia, el destacar tanto los riesgos como los beneficios del empleo de los cannabinoides en el tratamiento de estas enfermedades requiere de más trabajo experimental.


La prévalence élevée des troubles mentaux, leur chronicité et leur impact important sur la qualité de vie pèsent significativement sur la santé publique mondiale. La connaissance des mécanismes pathologiques qui sous-tendent ces troubles a progressé mais, un pourcentage élevé de patients ne répondant pas aux traitements de première ligne, d'autres approches thérapeutiques sont indispensables. L'étude du système endocannabinoïde et la mise en évidence de son rôle dans de multiples processus physiologiques au cours des cinquante dernières années, ont fortement attiré l'attention sur les cannabinoïdes naturels et synthétiques en tant que médicaments potentiels dans certaines pathologies comme, entre autres, les troubles mentaux. Nous résumons dans cet article les effets cannabinoϊdes qui pourraient s'inscrire dans le traitement de la dépression majeure, des troubles bipolaires, de l'anxiété, de l'état de stress post-traumatique et de la schizophrénie. Ces arguments nous incitent à explorer davantage les avantages et les risques des cannabinoïdes dans la prise en charge de ces maladies.


Asunto(s)
Cannabinoides/farmacología , Cannabinoides/uso terapéutico , Trastornos Mentales/tratamiento farmacológico , Animales , Trastornos de Ansiedad/tratamiento farmacológico , Endocannabinoides/fisiología , Humanos , Trastornos Mentales/psicología , Trastornos Psicóticos/tratamiento farmacológico , Calidad de Vida
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA