Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 116(51): 25958-25967, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31796600

RESUMO

Psychostimulant use is an ever-increasing socioeconomic burden, including a dramatic rise during pregnancy. Nevertheless, brain-wide effects of psychostimulant exposure are incompletely understood. Here, we performed Fos-CreERT2-based activity mapping, correlated for pregnant mouse dams and their fetuses with amphetamine, nicotine, and caffeine applied acutely during midgestation. While light-sheet microscopy-assisted intact tissue imaging revealed drug- and age-specific neuronal activation, the indusium griseum (IG) appeared indiscriminately affected. By using GAD67gfp/+ mice we subdivided the IG into a dorsolateral domain populated by γ-aminobutyric acidergic interneurons and a ventromedial segment containing glutamatergic neurons, many showing drug-induced activation and sequentially expressing Pou3f3/Brn1 and secretagogin (Scgn) during differentiation. We then combined Patch-seq and circuit mapping to show that the ventromedial IG is a quasi-continuum of glutamatergic neurons (IG-Vglut1+) reminiscent of dentate granule cells in both rodents and humans, whose dendrites emanate perpendicularly toward while their axons course parallel with the superior longitudinal fissure. IG-Vglut1+ neurons receive VGLUT1+ and VGLUT2+ excitatory afferents that topologically segregate along their somatodendritic axis. In turn, their efferents terminate in the olfactory bulb, thus being integral to a multisynaptic circuit that could feed information antiparallel to the olfactory-cortical pathway. In IG-Vglut1+ neurons, prenatal psychostimulant exposure delayed the onset of Scgn expression. Genetic ablation of Scgn was then found to sensitize adult mice toward methamphetamine-induced epilepsy. Overall, our study identifies brain-wide targets of the most common psychostimulants, among which Scgn+/Vglut1+ neurons of the IG link limbic and olfactory circuits.


Assuntos
Mapeamento Encefálico , Encéfalo/metabolismo , Regulação da Expressão Gênica , Lobo Límbico/metabolismo , Animais , Axônios/metabolismo , Encéfalo/diagnóstico por imagem , Dendritos/metabolismo , Feminino , Glutamato Descarboxilase/genética , Humanos , Interneurônios/metabolismo , Lobo Límbico/anatomia & histologia , Lobo Límbico/efeitos dos fármacos , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Bulbo Olfatório/metabolismo , Fatores do Domínio POU/genética , Fatores do Domínio POU/metabolismo , Secretagoginas/genética , Secretagoginas/metabolismo , Proteína Vesicular 1 de Transporte de Glutamato/genética , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Ácido gama-Aminobutírico/metabolismo
2.
Proc Natl Acad Sci U S A ; 113(16): 4500-5, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27001846

RESUMO

Anxiety disorders peak in incidence during adolescence, a developmental window that is marked by dynamic changes in gene expression, endocannabinoid signaling, and frontolimbic circuitry. We tested whether genetic alterations in endocannabinoid signaling related to a common polymorphism in fatty acid amide hydrolase (FAAH), which alters endocannabinoid anandamide (AEA) levels, would impact the development of frontolimbic circuitry implicated in anxiety disorders. In a pediatric imaging sample of over 1,000 3- to 21-y-olds, we show effects of the FAAH genotype specific to frontolimbic connectivity that emerge by ∼12 y of age and are paralleled by changes in anxiety-related behavior. Using a knock-in mouse model of the FAAH polymorphism that controls for genetic and environmental backgrounds, we confirm phenotypic differences in frontoamygdala circuitry and anxiety-related behavior by postnatal day 45 (P45), when AEA levels begin to decrease, and also, at P75 but not before. These results, which converge across species and level of analysis, highlight the importance of underlying developmental neurobiology in the emergence of genetic effects on brain circuitry and function. Moreover, the results have important implications for the identification of risk for disease and precise targeting of treatments to the biological state of the developing brain as a function of developmental changes in gene expression and neural circuit maturation.


Assuntos
Endocanabinoides/metabolismo , Lobo Frontal/metabolismo , Lobo Límbico/metabolismo , Rede Nervosa/metabolismo , Transdução de Sinais/fisiologia , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Feminino , Lobo Frontal/citologia , Humanos , Lobo Límbico/citologia , Masculino , Camundongos , Camundongos Transgênicos , Rede Nervosa/citologia , Especificidade da Espécie
3.
Cell Mol Neurobiol ; 38(1): 305-316, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28695320

RESUMO

The prelimbic cortex (PL) is an important structure in the neural pathway integrating stress responses. Brain angiotensin is involved in cardiovascular control and modulation of stress responses. Blockade of angiotensin receptors has been reported to reduce stress responses. Acute restraint stress (ARS) is a stress model, which evokes sustained blood pressure increase, tachycardia, and reduction in tail temperature. We therefore hypothesized that PL locally generated angiotensin and angiotensin receptors modulate stress autonomic responses. To test this hypothesis, we microinjected an angiotensin-converting enzyme (ACE) inhibitor or angiotensin antagonists into the PL, prior to ARS. Male Wistar rats were used; guide cannulas were bilaterally implanted in the PL for microinjection of vehicle or drugs. A polyethylene catheter was introduced into the femoral artery to record cardiovascular parameters. Tail temperature was measured using a thermal camera. ARS was started 10 min after PL treatment with drugs. Pretreatment with ACE inhibitor lisinopril (0.5 nmol/100 nL) reduced the pressor response, but did not affect ARS-evoked tachycardia. At a dose of 1 nmol/100 nL, it reduced both ARS pressor and tachycardic responses. Pretreatment with candesartan, AT1 receptor antagonist reduced ARS-evoked pressor response, but not tachycardia. Pretreatment with PD123177, AT2 receptor antagonist, reduced tachycardia, but did not affect ARS pressor response. No treatment affected ARS fall in tail temperature. Results suggest involvement of PL angiotensin in the mediation of ARS cardiovascular responses, with participation of both AT1 and AT2 receptors. In conclusion, results indicate that PL AT1-receptors modulate the ARS-evoked pressor response, while AT2-receptors modulate the tachycardic component of the autonomic response.


Assuntos
Pressão Sanguínea/fisiologia , Córtex Cerebral/metabolismo , Frequência Cardíaca/fisiologia , Receptor Tipo 1 de Angiotensina/fisiologia , Receptor Tipo 2 de Angiotensina/fisiologia , Estresse Psicológico/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Relação Dose-Resposta a Droga , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Lobo Límbico/efeitos dos fármacos , Lobo Límbico/metabolismo , Masculino , Ratos , Ratos Wistar , Restrição Física/fisiologia , Restrição Física/psicologia , Estresse Psicológico/psicologia
4.
Addict Biol ; 23(5): 1032-1045, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-28971565

RESUMO

The hypocretin receptor 1 (HCRTr1) is a critical participant in the regulation of motivated behavior. Previous observations demonstrate that acute pharmacological blockade of HCRTr1 disrupts dopamine (DA) signaling and the motivation for cocaine when delivered systemically or directly into the ventral tegmental area (VTA). To further examine the involvement of HCRTr1 in regulating reward and reinforcement processing, we employed an adeno-associated virus to express a short hairpin RNA designed to knock down HCRTr1. We injected virus into the VTA and examined the effects of HCRTr1 knockdown on cocaine self-administration and DA signaling in the nucleus accumbens (NAc) core. We determined that the viral approach was effective at reducing HCRTr1 expression without affecting the expression of hypocretin receptor 2 or DA-related mRNAs. We next examined the effects of HCRTr1 knockdown on cocaine self-administration, observing delayed acquisition under a fixed-ratio schedule and reduced motivation for cocaine under a progressive ratio schedule. These effects did not appear to be associated with alterations in sleep/wake activity. Using fast-scan cyclic voltammetry, we then examined whether HCRTr1 knockdown alters DA signaling dynamics in the NAc core. We observed reduced DA release and slower uptake rate as well as attenuated cocaine-induced DA uptake inhibition in rats with knockdown of HCRTr1. These observations indicate that HCRTr1 within the VTA influence the motivation for cocaine, likely via alterations in DA signaling in the NAc.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/genética , Cocaína/administração & dosagem , Dopamina/metabolismo , Motivação/genética , Receptores de Orexina/genética , Transdução de Sinais/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo , Animais , Cocaína/genética , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Modelos Animais de Doenças , Lobo Límbico/efeitos dos fármacos , Lobo Límbico/metabolismo , Masculino , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Motivação/efeitos dos fármacos , Motivação/fisiologia , Ratos , Ratos Sprague-Dawley , Reforço Psicológico , Recompensa , Autoadministração , Transdução de Sinais/fisiologia , Área Tegmentar Ventral/efeitos dos fármacos
5.
Development ; 141(21): 4182-93, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25336744

RESUMO

The corpus callosum connects cerebral hemispheres and is the largest axon tract in the mammalian brain. Callosal malformations are among the most common congenital brain anomalies and are associated with a wide range of neuropsychological deficits. Crossing of the midline by callosal axons relies on a proper midline environment that harbors guidepost cells emitting guidance cues to instruct callosal axon navigation. Little is known about what controls the formation of the midline environment. We find that two components of the Hippo pathway, the tumor suppressor Nf2 (Merlin) and the transcriptional coactivator Yap (Yap1), regulate guidepost development and expression of the guidance cue Slit2 in mouse. During normal brain development, Nf2 suppresses Yap activity in neural progenitor cells to promote guidepost cell differentiation and prevent ectopic Slit2 expression. Loss of Nf2 causes malformation of midline guideposts and Slit2 upregulation, resulting in callosal agenesis. Slit2 heterozygosity and Yap deletion both restore callosal formation in Nf2 mutants. Furthermore, selectively elevating Yap activity in midline neural progenitors is sufficient to disrupt guidepost formation, upregulate Slit2 and prevent midline crossing. The Hippo pathway is known for its role in controlling organ growth and tumorigenesis. Our study identifies a novel role of this pathway in axon guidance. Moreover, by linking axon pathfinding and neural progenitor behaviors, our results provide an example of the intricate coordination between growth and wiring during brain development.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Corpo Caloso/embriologia , Neurofibromatose 2/metabolismo , Fosfoproteínas/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular , Corpo Caloso/metabolismo , Células Ependimogliais/citologia , Células Ependimogliais/metabolismo , Feminino , Lobo Límbico/embriologia , Lobo Límbico/metabolismo , Camundongos , Sistema Nervoso , Neurofibromatose 2/genética , Fosfoproteínas/genética , Gravidez , Fatores de Transcrição/genética , Proteínas de Sinalização YAP
6.
Addict Biol ; 21(3): 603-12, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-25899624

RESUMO

Orexins (hypocretins) are hypothalamic neuropeptides that innervate the entire neuraxis, including the prelimbic cortex and ventral tegmental area and have been implicated in ethanol-seeking behaviour. The present study aimed to use the orexin-1 (OX1 ) receptor antagonist SB-334867 to examine the role of prelimbic cortex and ventral tegmental area OX1 receptors in cue-induced reinstatement of ethanol-seeking. Ethanol-preferring rats (iP) rats were trained to self-administer ethanol (10 percent v/v, FR3) or sucrose (0.2-1 percent w/v, FR3) in the presence of reward-associated cues before being implanted with indwelling guide cannulae. Rats then underwent extinction training for 11 days. On test days, rats were given a microinjection of vehicle or SB-334867 (3 µg/side) and presented with reward-associated cues to precipitate reinstatement. Results show SB-334867 infused into the prelimbic cortex attenuated cue-induced reinstatement of ethanol-seeking, but not sucrose-seeking. OX1 antagonism in the ventral tegmental area also attenuated cue-induced reinstatement of ethanol-seeking. These findings suggest that OX1 receptors located in the prelimbic cortex and ventral tegmental area are part of a circuit driving cue-mediated ethanol-seeking behaviour.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Condicionamento Psicológico , Comportamento de Procura de Droga/efeitos dos fármacos , Etanol/farmacologia , Extinção Psicológica , Receptores de Orexina/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Benzoxazóis/farmacologia , Sinais (Psicologia) , Lobo Límbico/metabolismo , Masculino , Naftiridinas , Antagonistas dos Receptores de Orexina/farmacologia , Receptores de Orexina/metabolismo , Ratos , Ratos Endogâmicos , Transdução de Sinais , Ureia/análogos & derivados , Ureia/farmacologia , Área Tegmentar Ventral/metabolismo
7.
Dev Psychobiol ; 57(1): 50-62, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25355379

RESUMO

In mammals, the presence of the mother can reduce or "buffer" stress responses of her young in threatening conditions. We compared the effect of the mother, a familiar littermate, and an unfamiliar adult male on three classes of response shown by guinea pig pups in a novel environment: short latency active behaviors, particularly vocalizing; slower developing passive behaviors that appear mediated by inflammatory mechanisms; and hypothalamic-pituitary-adrenal activity. We also examined Fos induction in the prelimbic cortex, a region hypothesized to mediate buffering effects. Only the mother significantly suppressed all classes of behavior. The greatest selectivity was observed for passive behavioral responses. Contrary to expectations, the adult male reduced plasma cortisol levels of pups as effectively as did the mother. The presence of the male also resulted in increased Fos induction in the prelimbic cortex and high levels of social interaction. Maternal buffering was not associated with prelimbic activity. These results confirm the ability of the mother to reduce active behavioral and HPA responses and suggest a specific maternal buffering effect on the later developing passive behavioral responses. The findings also demonstrate an unexpected ability of adult males to reduce HPA responses and raise the possibility that different social partners buffer HPA activity through different underlying processes.


Assuntos
Animais Recém-Nascidos/fisiologia , Relações Interpessoais , Lobo Límbico/fisiologia , Proteínas Proto-Oncogênicas c-fos/fisiologia , Hormônio Adrenocorticotrópico/sangue , Animais , Animais Recém-Nascidos/psicologia , Comportamento Animal/fisiologia , Química Encefálica/fisiologia , Feminino , Cobaias , Hidrocortisona/sangue , Lobo Límbico/química , Lobo Límbico/metabolismo , Masculino , Proteínas Proto-Oncogênicas c-fos/análise , Proteínas Proto-Oncogênicas c-fos/metabolismo
8.
J Parkinsons Dis ; 14(6): 1271-1276, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38995804

RESUMO

 In progressive supranuclear palsy (PSP), subcortical tau and cortical perfusion can be assessed using the tracer [18F]PI-2620. We investigated if subcortical tau (globus pallidus internus, dentate nucleus) and frontal/limbic perfusion correlate in a cohort of 32 PSP patients. Tau in subcortical regions showed significant negative correlation with perfusion in limbic cortex. Perfusion in frontal regions was negatively associated with tau in both subcortical regions, but the significance threshold was only passed for the dentate nucleus. A reason could be a diaschisis-like phenomenon; that is, subcortical tau could lead to reduced connectivity to frontal regions and, thereby, to decreased perfusion.


In a study of 32 patients with progressive supranuclear palsy (PSP), we used a molecular imaging tracer called [18F]PI-2620 to measure two things: the presence of a protein called tau in deep brain areas (specifically, the globus pallidus internus and dentate nucleus) and the function of the brain's cortex by assessing blood flow (perfusion). We found that higher amounts of tau in these deep brain areas were associated with reduced blood flow in the limbic cortex, which is involved in emotion regulation. Also, the frontal areas of the brain showed reduced blood flow related to tau in these deep brain regions. However, this connection was statistically significant only for the dentate nucleus. This study suggests that the buildup of tau protein in deeper brain areas can disrupt function in parts of the brain's cortex, highlighting the damaging role of tau in PSP.


Assuntos
Lobo Frontal , Paralisia Supranuclear Progressiva , Proteínas tau , Humanos , Paralisia Supranuclear Progressiva/diagnóstico por imagem , Paralisia Supranuclear Progressiva/metabolismo , Paralisia Supranuclear Progressiva/patologia , Paralisia Supranuclear Progressiva/fisiopatologia , Proteínas tau/metabolismo , Masculino , Feminino , Idoso , Lobo Frontal/diagnóstico por imagem , Lobo Frontal/metabolismo , Tomografia por Emissão de Pósitrons , Pessoa de Meia-Idade , Sistema Límbico/diagnóstico por imagem , Sistema Límbico/metabolismo , Sistema Límbico/fisiopatologia , Lobo Límbico/metabolismo , Lobo Límbico/diagnóstico por imagem , Lobo Límbico/fisiopatologia , Lobo Límbico/patologia , Circulação Cerebrovascular/fisiologia
9.
Behav Brain Res ; 384: 112553, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32057826

RESUMO

Chronic social defeat stress (CSDS) has been found to produce different impacts on anxiety-like behaviors, spatial cognitive function and memory in rodents with different susceptibilities. However, the impacts of chronic social defeat on social behaviors in adult male mice with different susceptibilities to social defeat and the underlying mechanisms in the brain remain unclear. In the present study, we found that ten days of social defeat reduced the tendency of susceptible adult male C57 mice to approach an unfamiliar individual and increased their avoidance of an unfamiliar CD-1 mouse but had no effects on resilient individuals. In addition, CSDS enhanced anxiety-like behavior in susceptible animals, but produced no effects in the resilient group. Meanwhile, CSDS increased the number of corticotropin-releasing factor (CRF)-positive neurons in the paraventricular nucleus of the hypothalamus and CRF-R2-positive neurons in the accumbens nucleus shell in both resilient and susceptible animals. CSDS increased the number of CRF-R1-positive neurons and CRF-R1 mRNA expression in the prelimbic cortex (PrL) and the number of CRF-R2-positive neurons in the basolateral amygdala, but reduced the number of CRF-R2-positive neurons and mRNA expression in the PrL in susceptible animals. Therefore, the different effects of CSDS on sociability and anxiety-like behavior in mice with different susceptibilities may be associated with region- and type-specific alterations in CRF receptor levels. These findings help us understand the underlying mechanism by which social stress affects emotion and social behavior and provides an important basis for the treatment of disorders of social and emotional behavior caused by social stress.


Assuntos
Encéfalo/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética , Comportamento Social , Derrota Social , Estresse Psicológico/genética , Animais , Ansiedade/genética , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Aprendizagem da Esquiva , Complexo Nuclear Basolateral da Amígdala/metabolismo , Lobo Límbico/metabolismo , Masculino , Camundongos , Neurônios/metabolismo , Núcleo Accumbens/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , RNA Mensageiro/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia
10.
J Neuroimmune Pharmacol ; 14(4): 661-678, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31372820

RESUMO

In the era of combined antiretroviral therapy, HIV-1 infected individuals are living longer lives; however, longevity is met with an increasing number of HIV-1 associated neurocognitive disorders (HAND) diagnoses. The transactivator of transcription (Tat) is known to mediate the neurotoxic effects in HAND by acting directly on neurons and also indirectly via its actions on glia. The Go/No-Go (GNG) task was used to examine HAND in the Tat transgenic mouse model. The GNG task involves subjects discriminating between two stimuli sets in order to determine whether or not to inhibit a previously trained response. Data reveal inhibitory control deficits in female Tat(+) mice (p = .048) and an upregulation of cannabinoid type 1 receptors (CB1R) in the infralimbic (IL) cortex in the same female Tat(+) group (p < .05). A significant negative correlation was noted between inhibitory control and IL CB1R expression (r = -.543, p = .045), with CB1R expression predicting 30% of the variance of inhibitory control (R2 = .295, p = .045). Furthermore, there was a significant increase in spontaneous excitatory postsynaptic current (sEPSC) frequencies in Tat(+) compared to Tat(-) mice (p = .008, across sexes). The increase in sEPSC frequency was significantly attenuated by bath application of PF3845, a fatty acid amide hydrolase (FAAH) enzyme inhibitor (p < .001). Overall, the GNG task is a viable measure to assess inhibitory control deficits in Tat transgenic mice and results suggest a potential therapeutic treatment for the observed deficits with drugs which modulate endocannabinoid enzyme activity. Graphical Abstract Results of the Go/No-Go operant conditioning task reveal inhibitory control deficits in female transgenic Tat(+) mice without significantly affecting males. The demonstrated inhibitory control deficits appear to be associated with an upregulation of cannabinoid type 1 receptors (CB1R) in the infralimbic (IL) cortex in the same female Tat(+) group.


Assuntos
Complexo AIDS Demência/metabolismo , Modelos Animais de Doenças , HIV-1 , Inibição Psicológica , Receptor CB1 de Canabinoide/biossíntese , Produtos do Gene tat do Vírus da Imunodeficiência Humana/biossíntese , Complexo AIDS Demência/genética , Complexo AIDS Demência/psicologia , Animais , Feminino , Lobo Límbico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transtornos Neurocognitivos/genética , Transtornos Neurocognitivos/metabolismo , Desempenho Psicomotor/fisiologia , Receptor CB1 de Canabinoide/genética , Regulação para Cima/fisiologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
11.
Neurotox Res ; 36(1): 132-143, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30879275

RESUMO

Designer drug mixtures popularized as "bath salts" often contain the synthetic cathinones 3,4 methylenedioxypyrovalerone (MDPV), mephedrone, and methylone in various combinations. However, most preclinical investigations have only assessed the effects of individual bath salt constituents, and little is known about whether co-exposure to MDPV, mephedrone, and methylone produces significant neuropharmacological interactions. This study evaluated and compared how MDPV, mephedrone, and methylone influence discrete brain tissue dopamine (DA) levels and motor stimulant responses in mice when administered alone and as a ternary mixture. Male adolescent Swiss-Webster mice received intraperitoneal injections of saline or 1 or 10 mg/kg doses of MDPV, mephedrone, or methylone, or a cocktail of all three cathinones at doses of 1, 3.3, or 10 mg/kg each. The effect of each treatment on DA and DA metabolite levels in mesolimbic and nigrostriatal brain tissue was quantified 15 min after a single exposure using HPLC-ECD. Additionally, locomotor activity was recorded in mice after acute (day 1) and chronic intermittent (day 7) dosing. MDPV, mephedrone, and methylone produced dose-related increases in mesolimbic and nigrostriatal DA levels that were significantly enhanced following their co-administration. In addition, mice treated with the cathinone cocktail displayed decreased locomotor activity on day 1 that was exacerbated by day 7 and not observed with any of the drugs alone. Our findings demonstrate a significant enhanced effect of MDPV, mephedrone, and methylone on both DA, and these effects on DA result in significant alterations in locomotor activity.


Assuntos
Benzodioxóis/farmacologia , Encéfalo/efeitos dos fármacos , Dopaminérgicos/farmacologia , Dopamina/metabolismo , Metanfetamina/análogos & derivados , Pirrolidinas/farmacologia , Animais , Encéfalo/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Drogas Desenhadas/farmacologia , Dopamina/análise , Relação Dose-Resposta a Droga , Lobo Límbico/efeitos dos fármacos , Lobo Límbico/metabolismo , Locomoção/efeitos dos fármacos , Masculino , Metanfetamina/farmacologia , Camundongos , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo , Catinona Sintética
12.
Schizophr Bull ; 45(5): 1092-1100, 2019 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30388260

RESUMO

Psychotic experiences may be understood as altered information processing due to aberrant neural computations. A prominent example of such neural computations is the computation of prediction errors (PEs), which signal the difference between expected and experienced events. Among other areas showing PE coding, hippocampal-prefrontal-striatal neurocircuits play a prominent role in information processing. Dysregulation of dopaminergic signaling, often secondary to psychosocial stress, is thought to interfere with the processing of biologically important events (such as reward prediction errors) and result in the aberrant attribution of salience to irrelevant sensory stimuli and internal representations. Bayesian hierarchical predictive coding offers a promising framework for the identification of dysfunctional neurocomputational processes and the development of a mechanistic understanding of psychotic experience. According to this framework, mismatches between prior beliefs encoded at higher levels of the cortical hierarchy and lower-level (sensory) information can also be thought of as PEs, with important consequences for belief updating. Low levels of precision in the representation of prior beliefs relative to sensory data, as well as dysfunctional interactions between prior beliefs and sensory data in an ever-changing environment, have been suggested as a general mechanism underlying psychotic experiences. Translating the promise of the Bayesian hierarchical predictive coding into patient benefit will come from integrating this framework with existing knowledge of the etiology and pathophysiology of psychosis, especially regarding hippocampal-prefrontal-striatal network function and neural mechanisms of information processing and belief updating.


Assuntos
Encéfalo/fisiopatologia , Cognição/fisiologia , Aprendizagem/fisiologia , Transtornos Psicóticos/fisiopatologia , Esquizofrenia/fisiopatologia , Psicologia do Esquizofrênico , Teorema de Bayes , Encéfalo/metabolismo , Neurociência Cognitiva , Corpo Estriado/metabolismo , Corpo Estriado/fisiopatologia , Delusões/metabolismo , Delusões/fisiopatologia , Delusões/psicologia , Dopamina/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Lobo Límbico/metabolismo , Lobo Límbico/fisiopatologia , Modelos Neurológicos , Modelos Psicológicos , Vias Neurais/metabolismo , Vias Neurais/fisiopatologia , Neurofisiologia , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiopatologia , Transtornos Psicóticos/metabolismo , Transtornos Psicóticos/psicologia , Reforço Psicológico , Recompensa , Esquizofrenia/metabolismo , Transmissão Sináptica , Lobo Temporal/metabolismo , Lobo Temporal/fisiopatologia
13.
Neurosci Res ; 130: 23-38, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28842244

RESUMO

The presence of substance P (SP) receptor (Neurokinin-1 receptor, NK1R) in the indusium griseum (IG) and anterior hippocampal continuation (AHC) during postnatal development was studied by immunocytochemistry (ICC). NK1R-immunopositive neurons (NK1RIP-n) first appeared in both areas on postnatal day (P) 5. From P5 onward, their distribution pattern was adult-like. In sagittal sections NK1RIP-n formed a narrow strip of neurons and dendrites that were located over the corpus callosum (cc); in coronal sections they were found in a roughly triangular area at the base of the cingulate cortex (Cg) on the dorsal surface of the cc. NK1RIP-n were also found in the AHC, which is considered as a subcallosal extension of the IG, located ventral to the genu of the cc. At all ages studied, IG NK1RIP-n sent dendrites to the contralateral IG, the underlying cc, and the Cg. Moreover, NK1RIP-n located in the Cg and the cc sent dendrites to the IG. The present findings are in line with previous ICC studies describing dopaminergic and serotoninergic afferents to the IG. Together these data suggest that, through NK1R, SP could play an important role in regulating the release mechanisms of these afferents and that it could be an important developmental factor. Notably, IG neurons could be activated by cortical and intracallosal afferents.


Assuntos
Lobo Límbico/crescimento & desenvolvimento , Lobo Límbico/metabolismo , Receptores da Neurocinina-1/metabolismo , Animais , Axônios/metabolismo , Dendritos/metabolismo , Imuno-Histoquímica , Lobo Límbico/citologia , Ratos Sprague-Dawley
14.
Pharmacol Biochem Behav ; 171: 54-65, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29908200

RESUMO

BACKGROUND: Chronic nicotine exposure produces neuroadaptations in brain reward systems and α4ß2 nicotinic acetylcholine receptors (nAChRs) in the corticolimbic brain areas. We previously demonstrated opposite effects of nicotine exposure delivered by self-administration or pumps on brain reward thresholds that can be attributed to the different temporal pattern and contingency of nicotine exposure. We investigated the effects of these two factors on reward thresholds and somatic signs during nicotine withdrawal, and on nAChRs binding in corticolimbic brain areas. METHODS: The intracranial self-stimulation procedure was used to assess reward thresholds in rats prepared with pumps delivering various doses of nicotine continuously or intermittently. Separate group of rats were randomly exposed to nicotine via pumps (non-contingent) or nicotine self-administration (contingent) to determine [125I]-epibatidine binding at α4ß2* nAChRs. RESULTS: Withdrawal from continuous non-contingent nicotine exposure led to significant elevations in thresholds and increases in somatic signs in rats, while there was no significant effect of withdrawal from intermittent non-contingent nicotine exposure at the same doses. nAChRs were upregulated during withdrawal from continuous non-contingent nicotine exposure. α4ß2* nAChRs were upregulated in the ventral tegmental area and prelimbic cortex during withdrawal from non-contingent intermittent exposure and in the nucleus accumbens during withdrawal from contingent intermittent nicotine exposure to the same dose. CONCLUSIONS: During non-contingent nicotine exposure, the temporal pattern of nicotine delivery differentially affected thresholds and somatic signs of withdrawal. Upregulation of α4ß2* nAChRs was brain site-specific and depended on both temporal pattern and contingency of nicotine exposure.


Assuntos
Nicotina/efeitos adversos , Receptores Nicotínicos/metabolismo , Recompensa , Síndrome de Abstinência a Substâncias/diagnóstico , Síndrome de Abstinência a Substâncias/metabolismo , Animais , Autoantígenos , Compostos Bicíclicos Heterocíclicos com Pontes/metabolismo , Esquema de Medicação , Bombas de Infusão Implantáveis , Radioisótopos do Iodo/metabolismo , Lobo Límbico/metabolismo , Masculino , Nicotina/administração & dosagem , Núcleo Accumbens/metabolismo , Piridinas/metabolismo , Ensaio Radioligante , Ratos , Ratos Wistar , Regulação para Cima , Área Tegmentar Ventral/metabolismo
15.
Psychopharmacology (Berl) ; 235(2): 447-458, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29063964

RESUMO

Growing clinical and preclinical evidence suggests a potential role for the phytocannabinoid cannabidiol (CBD) as a pharmacotherapy for various neuropsychiatric disorders. In contrast, delta-9-tetrahydrocannabinol (THC), the primary psychoactive component in cannabis, is associated with acute and neurodevelopmental propsychotic side effects through its interaction with central cannabinoid type 1 receptors (CB1Rs). CB1R stimulation in the ventral hippocampus (VHipp) potentiates affective memory formation through inputs to the mesolimbic dopamine (DA) system, thereby altering emotional salience attribution. These changes in DA activity and salience attribution, evoked by dysfunctional VHipp regulatory actions and THC exposure, could predispose susceptible individuals to psychotic symptoms. Although THC can accelerate the onset of schizophrenia, CBD displays antipsychotic properties, can prevent the acquisition of emotionally irrelevant memories, and reverses amphetamine-induced neuronal sensitization through selective phosphorylation of the mechanistic target of rapamycin (mTOR) molecular signaling pathway. This review summarizes clinical and preclinical evidence demonstrating that distinct phytocannabinoids act within the VHipp and associated corticolimbic structures to modulate emotional memory processing through changes in mesolimbic DA activity states, salience attribution, and signal transduction pathways associated with schizophrenia-related pathology.


Assuntos
Canabinoides/uso terapêutico , Dopamina/metabolismo , Emoções/fisiologia , Hipocampo/metabolismo , Memória/fisiologia , Transtornos Mentais/metabolismo , Animais , Antipsicóticos/farmacologia , Antipsicóticos/uso terapêutico , Canabinoides/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Emoções/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Humanos , Lobo Límbico/efeitos dos fármacos , Lobo Límbico/metabolismo , Memória/efeitos dos fármacos , Transtornos Mentais/tratamento farmacológico , Transtornos Mentais/psicologia , Compostos Fitoquímicos/farmacologia , Compostos Fitoquímicos/uso terapêutico
16.
Brain Res ; 1661: 88-99, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28214522

RESUMO

Neural mechanism underlying memory retrieval has been extensively studied in the hippocampus and amygdala. However, little is known about the role of medial prefrontal cortex in long-term memory retrieval. We evaluate this issue in one-trial step-through inhibitory avoidance (IA) paradigm. Our results showed that, 1) inactivation of mPFC by local infusion of GABAA-receptor agonist muscimol caused severe deficits in retrieval of 1-day and 7-day but had no effects on 2-h inhibitory avoidance memory; 2) the protein level of phosphorylated-ERK1/2 in mPFC were significantly increased following retrieval of 1-day and 7-day IA memory, so did the numbers of phosphorylated-ERK (pERK) and phosphorylated-CREB (pCREB) labeled neurons; 3) intra-mPFC infusion of ERK kinase inhibitor PD98095 significantly reduced phosphorylated ERK1/2 levels and phosphorylated-ERK1/2 and phosphorylated-CREB labeled cells, and severely impaired retrieval of 7-day IA memory when the drugs were administrated 30min prior to test. The present study provides evidence that retrieval of long-lasting memory for inhibitory avoidance requires mPFC and involves the ERK-CREB signaling cascade.


Assuntos
Memória de Longo Prazo/fisiologia , Córtex Pré-Frontal/fisiologia , Tonsila do Cerebelo/fisiologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Proteína de Ligação a CREB , Hipocampo/metabolismo , Lobo Límbico/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Memória/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Muscimol/efeitos adversos , Muscimol/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
17.
Neurotox Res ; 31(2): 289-297, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27832448

RESUMO

During adolescence, the medial prefrontal cortex (mPFC) is still developing. We have previously shown that developmental cocaine exposure alters mPFC's ability to cope with challenging events. In this manuscript, we exposed rats developmentally treated with cocaine to a novelty task and analyzed the molecular changes of mPFC. Rats were exposed to cocaine from post-natal day (PND) 28 to PND 42 and sacrificed at PND 43, immediately after the novel object recognition (NOR) test. Cocaine-treated rats spent more time exploring the novel object than saline-treated counterparts, suggesting an increased response to novelty. The messenger RNA (mRNA) and protein levels of the immediate early gene Arc/Arg3.1 were reduced in both infralimbic (IL) and prelimbic (PL) cortices highlighting a baseline reduction of mPFC neuronal activity as a consequence of developmental exposure to cocaine. Intriguingly, significant molecular changes were observed in the IL, but not PL, cortex in response to the combination of cocaine exposure and test such as a marked upregulation of both Arc/Arg3.1 mRNA and protein levels only in cocaine-treated rats. As for proteins, such increase was observed only in the post-synaptic density and not in the whole homogenate, suggesting psychostimulant-induced changes in trafficking of Arc/Arg3.1 or an increased local translation. Notably, the same profile of Arc/Arg3.1 was observed for post-synaptic density (PSD)-95 leading to the possibility that Arc/Arg3.1 and PSD-95 bridge together to promote aberrant synaptic connectivity in IL cortex following repeated exposure to cocaine during brain development.


Assuntos
Cocaína/efeitos adversos , Proteínas do Citoesqueleto/biossíntese , Lobo Límbico/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Córtex Pré-Frontal/metabolismo , Reconhecimento Psicológico/efeitos dos fármacos , Animais , Proteína 4 Homóloga a Disks-Large , Comportamento Exploratório/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Lobo Límbico/efeitos dos fármacos , Masculino , Proteínas de Membrana/biossíntese , Densidade Pós-Sináptica/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Ratos , Regulação para Cima/efeitos dos fármacos
18.
Artigo em Inglês | MEDLINE | ID: mdl-28351548

RESUMO

Schizophrenia is associated with significant pathophysiological changes to interneurons within the prefrontal cortex (PFC), with mRNA and protein changes associated with the GABA network localized to specific interneuron subtypes. Methamphetamine is a commonly abused psychostimulant that can induce chronic psychosis and symptoms that are similar to schizophrenia, suggesting that chronic METH induced psychosis may be associated with similar brain pathology to schizophrenia in the PFC. The aim of this study, therefore, was to examine mRNA expression of interneuron markers across two regions of the PFC (prelimbic (PRL) and orbitofrontal cortices (OFC)) following METH sensitization, an animal model of METH psychosis. We also studied the association between GABA mRNA expression and interneuronal mRNA expression to identify whether particular changes to the GABA network could be localized to a specific inhibitory cellular phenotype. METH sensitization increased the transcriptional expression of calbindin, calretinin, somatostatin, cholecyctokinin and vasoactive intestinal peptide in the PRL while parvalbumin, calbindin, cholectokinin and vasoactive intestinal peptide were upregulated in the OFC. Based on our previous findings, we also found significant correlations between GAD67, GAT1 and parvalbumin while GAD67, GAD65 and GAT1 were positively correlated with cholecystokinin in the PRL of METH sensitized rats. Within the OFC, the expression of GABAAα1 was positively correlated with somatostatin while GABAAα5 was negatively associated with somatostatin and calbindin. These findings suggest that METH sensitization differentially changes the expression of mRNAs encoding for multiple peptides and calcium binding proteins across the PRL and the OFC. Furthermore, these findings support that changes to the GABA network may also occur within specific cell types. These results, therefore, provide the first evidence that METH sensitization mediates differential interneuronal pathology across the PRL and OFC and such changes could have profound consequences on behavior and cognitive output.


Assuntos
Sensibilização do Sistema Nervoso Central , Interneurônios/metabolismo , Interneurônios/patologia , Lobo Límbico/metabolismo , Metanfetamina/farmacologia , Córtex Pré-Frontal/metabolismo , RNA Mensageiro/metabolismo , Animais , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Glutamato Descarboxilase/metabolismo , Lobo Límbico/patologia , Proteínas do Tecido Nervoso/metabolismo , Peptídeos/metabolismo , Córtex Pré-Frontal/patologia , Ratos , Receptores de GABA-A/biossíntese , Ácido gama-Aminobutírico/metabolismo
19.
Mol Autism ; 8: 30, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28649315

RESUMO

BACKGROUND: People with fragile X syndrome (FXS) often have deficits in social behavior, and a substantial portion meet criteria for autism spectrum disorder. Though the genetic cause of FXS is known to be due to the silencing of FMR1, and the Fmr1 null mouse model representing this lesion has been extensively studied, the contributions of this gene and its protein product, FMRP, to social behavior are not well understood. METHODS: Fmr1 null mice and wildtype littermates were exposed to a social or non-social stimulus. In one experiment, subjects were assessed for expression of the inducible transcription factor c-Fos in response to the stimulus, to detect brain regions with social-specific activity. In a separate experiment, tissue was taken from those brain regions showing differential activity, and RNA sequencing was performed. RESULTS: Immunohistochemistry revealed a significantly greater number of c-Fos-positive cells in the lateral amygdala and medial amygdala in the brains of mice exposed to a social stimulus, compared to a non-social stimulus. In the prelimbic cortex, there was no significant effect of social stimulus; although the number of c-Fos-positive cells was lower in the social condition compared to the non-social condition, and negatively correlated with c-Fos in the amygdala. RNA sequencing revealed differentially expressed genes enriched for molecules known to interact with FMRP and also for autism-related genes identified in the Simons Foundation Autism Research Initiative gene database. Ingenuity Pathway Analysis detected enrichment of differentially expressed genes in networks and pathways related to neuronal development, intracellular signaling, and inflammatory response. CONCLUSIONS: Using the Fmr1 null mouse model of fragile X syndrome, we have identified brain regions, gene networks, and molecular pathways responsive to a social stimulus. These findings, and future experiments following up on the role of specific gene networks, may shed light on the neural mechanisms underlying dysregulated social behaviors in fragile X syndrome and more broadly.


Assuntos
Tonsila do Cerebelo/metabolismo , Modelos Animais de Doenças , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Lobo Límbico/metabolismo , Tonsila do Cerebelo/fisiopatologia , Animais , Comportamento Animal , Biomarcadores/análise , Mapeamento Encefálico , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/diagnóstico , Síndrome do Cromossomo X Frágil/fisiopatologia , Deleção de Genes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Ontologia Genética , Redes Reguladoras de Genes , Humanos , Relações Interpessoais , Lobo Límbico/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Anotação de Sequência Molecular , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Análise de Sequência de RNA , Transdução de Sinais
20.
Transl Psychiatry ; 7(2): e1038, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28221365

RESUMO

Ketamine and deep brain stimulation produce rapid antidepressant effects in humans and rodents. An increased AMPA receptor (AMPA-R) signaling in medial prefrontal cortex (mPFC) has been suggested to mediate these responses. However, little research has addressed the direct effects of enhancing glutamate tone or AMPA-R stimulation in mPFC subdivisions. The current study investigates the behavioral and neurochemical consequences of glutamate transporter-1 (GLT-1) blockade or s-AMPA microinfusion in the infralimbic (IL) and prelimbic (PrL) cortex. Owing to the connectivity between the mPFC and raphe nuclei, the role of serotonin is also explored. The bilateral microinfusion of the depolarizing agent veratridine into IL -but not PrL- of rats evoked immediate antidepressant-like responses. The same regional selectivity was observed after microinfusion of dihydrokainic acid (DHK), a selective inhibitor of GLT-1, present in astrocytes. The DHK-evoked antidepressant-like responses appear to be mediated by an AMPA-R-driven enhancement of serotonergic activity, as (i) they were prevented by NBQX 2,3-dioxo-6-nitro-1,2,3,4-tetrahydrobenzo[f]quinoxaline-7-sulfonamide disodium salt) and mimicked by s-AMPA; (ii) DHK and s-AMPA elevated similarly extracellular glutamate in IL and PrL, although extracellular 5-HT and c-fos expression in the midbrain dorsal raphe increased only when these agents were applied in IL; and (iii) DHK antidepressant-like responses were prevented by 5-HT synthesis inhibition and mimicked by citalopram microinfusion in IL. These results indicate that an acute increase of glutamatergic neurotransmission selectively in IL triggers immediate antidepressant-like responses in rats, likely mediated by the activation of IL-raphe pathways, which then results in a fast increase of serotonergic activity.


Assuntos
Afeto/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Transportador 2 de Aminoácido Excitatório/antagonistas & inibidores , Ácido Caínico/análogos & derivados , Neuroglia/metabolismo , Veratridina/farmacologia , Animais , Citalopram/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Ácido Caínico/farmacologia , Lobo Límbico/citologia , Lobo Límbico/metabolismo , Masculino , Córtex Pré-Frontal/metabolismo , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Quinoxalinas/farmacologia , Núcleos da Rafe/metabolismo , Ratos , Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa