Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
bioRxiv ; 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39229064

RESUMO

Generalization allows for experience to flexibly guide behavior when conditions change. A basic physical unit of memory storage and expression in the brain are sparse, distributed groups of neurons known as ensembles (i.e., the engram). The infralimbic (IL) subregion of the ventral medial prefrontal cortex plays a key role in modulating conditioned defensive responses. How IL neuronal ensembles established during learning contribute to generalized responses is unknown. In this set of experiments, generalization was tested in male and female mice by presenting a novel, ambiguous, tone generalization stimulus following Pavlovian defensive (fear) conditioning. The first experiment was designed to test a role for IL in generalization using chemogenetic manipulations. Results show IL bidirectionally regulates defensive behavior. IL silencing promotes a switch in defensive state from vigilant scanning to generalized freezing, while IL stimulation reduces freezing in favor of scanning. Leveraging activity-dependent tagging technology (ArcCreERT2 x eYFP system), a neuronal ensemble, preferentially located in IL superficial layer 2/3, was associated with the generalization stimulus. Remarkably, in the identical discrete location, fewer reactivated neurons were associated with the generalization stimulus at the remote timepoint (30 days) following learning. When an IL neuronal ensemble established during learning was selectively chemogenetically silenced, generalization increased. Conversely, IL neuronal ensemble stimulation reduced generalization. Overall, these data identify a crucial role for IL in suppressing generalized responses. Further, we uncover an IL neuronal ensemble, formed during learning, functions to later attenuate the expression of generalization in the presence of ambiguous threat stimuli.

2.
iScience ; 23(8): 101357, 2020 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-32712464

RESUMO

Regular physical activity improves physical and mental health. Here we found that the effect of physical activity extends to the next generation. Voluntary wheel running of dams, from postpartum day 2 to weaning, increased the social dominance and reproductive success, but not the physical/metabolic health, of their otherwise sedentary offspring. The individual's own physical activity did not improve dominance status. Maternal exercise did not disrupt maternal care or the maternal and offspring microbiota. Rather, the development of dominance behavior in the offspring of running mothers could be explained by the reduction of LIF, CXCL1, and CXCL2 cytokines in breast milk. These data reveal a cytokine-mediated lactocrine pathway that responds to the mother's postpartum physical activity and programs offspring social dominance. As dominance behaviors are highly relevant to the individual's survival and reproduction, lactocrine programming could be an evolutionary mechanism by which a mother promotes the social rank of her offspring.

3.
Curr Biol ; 27(24): 3859-3863.e3, 2017 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-29199072

RESUMO

Tumor necrosis factor alpha (TNF-α) is a cytokine that not only coordinates local and systemic immune responses [1, 2] but also regulates neuronal functions. Most prominently, glia-derived TNF-α has been shown to regulate homeostatic synaptic scaling [3-6], but TNF-α-null mice exhibited no apparent cognitive or emotional abnormalities. Instead, we found a TNF-α-dependent intergenerational effect, as mothers with a deficit in TNF-α programmed their offspring to exhibit low innate fear. Cross-fostering and conditional knockout experiments indicated that a TNF-α deficit in the maternal brain, rather than in the hematopoietic system, and during gestation was responsible for the low-fear offspring phenotype. The level of innate fear governs the balance between exploration/foraging and avoidance of predators and is thus fundamentally important in adaptation, fitness, and survival [7]. Because maternal exercise and activity are known to reduce both brain TNF-α [8] and offspring innate fear [9], whereas maternal stress has been reported to increase brain TNF-α [10] and offspring fear and anxiety [11, 12], maternal brain TNF-α may report environmental conditions to promote offspring behavioral adaptation to their anticipated postnatal environment.


Assuntos
Ansiedade/genética , Encéfalo/metabolismo , Medo , Camundongos/fisiologia , Fator de Necrose Tumoral alfa/genética , Animais , Feminino , Masculino , Herança Materna , Camundongos/genética , Camundongos Knockout , Fator de Necrose Tumoral alfa/deficiência
4.
Sci Signal ; 7(315): ra22, 2014 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-24595108

RESUMO

Na(+)-coupled solute transport is crucial for the uptake of nutrients and metabolic precursors, such as myo-inositol, an important osmolyte and precursor for various cell signaling molecules. We found that various solute transporters and potassium channel subunits formed complexes and reciprocally regulated each other in vitro and in vivo. Global metabolite profiling revealed that mice lacking KCNE2, a K(+) channel ß subunit, showed a reduction in myo-inositol concentration in cerebrospinal fluid (CSF) but not in serum. Increased behavioral responsiveness to stress and seizure susceptibility in Kcne2(-/-) mice were alleviated by injections of myo-inositol. Suspecting a defect in myo-inositol transport, we found that KCNE2 and KCNQ1, a voltage-gated potassium channel α subunit, colocalized and coimmunoprecipitated with SMIT1, a Na(+)-coupled myo-inositol transporter, in the choroid plexus epithelium. Heterologous coexpression demonstrated that myo-inositol transport by SMIT1 was augmented by coexpression of KCNQ1 but was inhibited by coexpression of both KCNQ1 and KCNE2, which form a constitutively active, heteromeric K(+) channel. SMIT1 and the related transporter SMIT2 were also inhibited by a constitutively active mutant form of KCNQ1. The activities of KCNQ1 and KCNQ1-KCNE2 were augmented by SMIT1 and the glucose transporter SGLT1 but were suppressed by SMIT2. Channel-transporter signaling complexes may be a widespread mechanism to facilitate solute transport and electrochemical crosstalk.


Assuntos
Canal de Potássio KCNQ1/metabolismo , Complexos Multiproteicos/metabolismo , Neurônios/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Simportadores/metabolismo , Animais , Células CHO , Líquido Cefalorraquidiano/metabolismo , Plexo Corióideo/metabolismo , Cromatografia Líquida de Alta Pressão , Cricetinae , Cricetulus , Imunofluorescência , Imunoprecipitação , Inositol/sangue , Espectrometria de Massas , Metaboloma , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Oócitos/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Transportador 1 de Glucose-Sódio/metabolismo , Xenopus laevis
5.
Nat Neurosci ; 17(1): 97-105, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24292233

RESUMO

Tumor necrosis factor α (TNF) is a proinflammatory cytokine with established roles in host defense and immune system organogenesis. We studied TNF function and found a previously unidentified physiological function that extends its effect beyond the host into the developing offspring. A partial or complete maternal TNF deficit, specifically in hematopoietic cells, resulted in reduced milk levels of the chemokines IP-10, MCP-1, MCP-3, MCP-5 and MIP-1ß, which in turn augmented offspring postnatal hippocampal proliferation, leading to improved adult spatial memory in mice. These effects were reproduced by the postpartum administration of a clinically used anti-TNF agent. Chemokines, fed to suckling pups of TNF-deficient mothers, restored both postnatal proliferation and spatial memory to normal levels. Our results identify a TNF-dependent 'lactrocrine' pathway that programs offspring hippocampal development and memory. The level of ambient TNF is known to be downregulated by physical activity, exercise and adaptive stress. We propose that the maternal TNF-milk chemokine pathway evolved to promote offspring adaptation to post-weaning environmental challenges and competition.


Assuntos
Citocinas/metabolismo , Hipocampo/crescimento & desenvolvimento , Memória/fisiologia , Leite/química , Fator de Necrose Tumoral alfa/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Animais Lactentes , Feminino , Proteína Glial Fibrilar Ácida , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/ultraestrutura , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Nestina/genética , Gravidez , RNA Mensageiro , Fatores de Transcrição SOXB1 , Coloração pela Prata , Proteínas com Domínio T/metabolismo , Fator de Necrose Tumoral alfa/genética
6.
Behav Brain Res ; 226(2): 428-34, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21995942

RESUMO

A nonsense mutation in the human cereblon gene (CRBN) causes a mild type of autosomal recessive non-syndromic intellectual disability (ID). Animal studies show that crbn is a cytosolic protein with abundant expression in the hippocampus (HPC) and neocortex (CTX). Its diverse functions include the developmental regulation of ion channels at the neuronal synapse, the mediation of developmental programs by ubiquitination, and a target for herpes simplex type I virus in HPC neurons. To test the hypothesis that anomalous CRBN expression leads to HPC-mediated memory and learning deficits, we generated germ-line crbn knock-out mice (crbn(-/-)). We also inactivated crbn in forebrain neurons in conditional knock-out mice in which crbn exons 3 and 4 are deleted by cre recombinase under the direction of the Ca(2+)/calmodulin-dependent protein kinase II alpha promoter (CamKII(cre/+), crbn(-/-)). crbn mRNA levels were negligible in the HPC, CTX, and cerebellum (CRBM) of the crbn(-/-) mice. In contrast, crbn mRNA levels were reduced 3- to 4-fold in the HPC, CTX but not in the CRBM in CamKII(cre/+), crbn(-/-) mice as compared to wild type (CamKII(cre/+), crbn(+/+)). Contextual fear conditioning showed a significant decrease in the percentage of freezing time in CamKII(cre/+), crbn(-/-) and crbn(-/-) mice while motor function, exploratory motivation, and anxiety-related behaviors were normal. These findings suggest that CamKII(cre/+), crbn(-/-) mice exhibit selective HPC-dependent deficits in associative learning and supports the use of these mice as in vivo models to study the functional consequences of CRBN aberrations on memory and learning in humans.


Assuntos
Modelos Animais de Doenças , Deficiência Intelectual/genética , Deficiência Intelectual/psicologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Prosencéfalo/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Sequência de Bases/genética , Sequência de Bases/fisiologia , Condicionamento Clássico/fisiologia , Éxons/genética , Medo/fisiologia , Humanos , Deficiência Intelectual/fisiopatologia , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Atividade Motora/genética , Deleção de Sequência/genética , Deleção de Sequência/fisiologia
7.
Results Probl Cell Differ ; 54: 243-53, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22009356

RESUMO

Since fragile X syndrome (FXS) is a typical X-linked mendelian disorder, the protein product associated with the disease (FMRP) is absent or reduced not only in the affected individuals but, in case of full mutation, also in their mothers. Here, by using the mouse model of the disease, we provide evidence that hyperactivity, a typical symptom of FXS, is not wholly induced by the lack of Fmrp in mice but also occurs as a result of its reduced expression in their mother. Genetically wild-type offspring of mutant mothers also had hyperactivity, albeit less pronounced than the mutant offspring. However, other features of FXS reproduced in the mouse model, such as sensory hyperreactivity and seizure susceptibility, were exclusively associated with the absence of Fmrp in the offspring. These data indicate that fmr-1, the gene encoding Fmrp, can be both an offspring genetic and a maternal environmental factor in producing a neurodevelopmental condition.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/fisiopatologia , Exposição Materna , Mutação , Doenças Neurodegenerativas/fisiopatologia , Animais , Comportamento/fisiologia , Modelos Animais de Doenças , Dopamina/metabolismo , Meio Ambiente , Feminino , Síndrome do Cromossomo X Frágil/patologia , Genótipo , Humanos , Camundongos , Transmissão Sináptica/fisiologia
8.
Front Psychiatry ; 2: 25, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21629836

RESUMO

Risk factors for psychiatric disorders have traditionally been classified as genetic or environmental. Risk (candidate) genes, although typically possessing small effects, represent a clear starting point to elucidate downstream cellular/molecular pathways of disease. Environmental effects, especially during development, can also lead to altered behavior and increased risk for disease. An important environmental factor is the mother, demonstrated by the negative effects elicited by maternal gestational stress and altered maternal care. These maternal effects can also have a genetic basis (e.g., maternal genetic variability and mutations). The focus of this review is "maternal genotype effects" that influence the emotional development of the offspring resulting in life-long psychiatric disease-like phenotypes. We have recently found that genetic inactivation of the serotonin 1A receptor (5-HT1AR) and the fmr1 gene (encoding the fragile X mental retardation protein) in mouse dams results in psychiatric disease-like phenotypes in their genetically unaffected offspring. 5-HT1AR deficiency in dams results in anxiety and increased stress responsiveness in their offspring. Offspring of 5-HT1AR deficient dams display altered development of the hippocampus, which could be linked to their anxiety-like phenotype. Maternal inactivation of fmr1, like its inactivation in the offspring, results in a hyperactivity-like condition and is associated with receptor alterations in the striatum. These data indicate a high sensitivity of the offspring to maternal mutations and suggest that maternal genotype effects can increase the impact of genetic risk factors in a population by increasing the risk of the genetically normal offspring as well as by enhancing the effects of offspring mutations.

9.
Neuropsychopharmacology ; 34(10): 2197-207, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19440190

RESUMO

Depression, anxiety, and conduct disorders are common in children and adolescents, and selective serotonin reuptake inhibitors (SSRIs) are often used to treat these conditions. Fluoxetine (Prozac) is the first approved SSRI for the treatment of depression in this population. Although it is believed that overall, fluoxetine is effective in child and adolescent psychiatry, there have been reports of specific adverse drug effects, most prominently, suicidality and psychiatric symptoms such as agitation, worsening of depression, and anxiety. Chronic fluoxetine substantially increases brain extracellular 5-HT concentrations, and the juvenile developing brain may respond to supraphysiological 5-HT levels with specific adverse effects not seen or less prominent in adult brain. Using novelty-induced hypophagia, as well as open-field and elevated plus maze tests, we show that both Swiss Webster and C57Bl/6 mice, receiving fluoxetine in a clinically relevant dose and during their juvenile age corresponding to child-adolescent periods in humans, exhibit a paradoxical anxiogenic response. The adverse effects of juvenile fluoxetine disappeared upon drug discontinuation and no long-term behavioral consequences were apparent. No adverse effect to chronic fluoxetine was seen in adult mice and a dose-dependent anxiolytic effect developed. These data show that the age of the mice, independently of the strains and tests used in this study, is the determining factor of whether the response to chronic fluoxetine is anxiolytic or anxiogenic. Taken together, the response of the juvenile and adult brain to fluoxetine could be fundamentally different and the juvenile fluoxetine administration mouse model described here may help to identify the mechanism underlying this difference.


Assuntos
Comportamento Animal/efeitos dos fármacos , Fluoxetina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Ansiedade/tratamento farmacológico , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Proliferação de Células/efeitos dos fármacos , Giro Denteado/citologia , Giro Denteado/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Comportamento de Ingestão de Líquido/efeitos dos fármacos , Esquema de Medicação , Comportamento Exploratório/efeitos dos fármacos , Fluoxetina/análogos & derivados , Fluoxetina/metabolismo , Bombas de Infusão Implantáveis , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos , Tempo de Reação/efeitos dos fármacos , Natação
10.
J Med Chem ; 52(3): 771-8, 2009 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-19146418

RESUMO

Previous studies with perzinfotel (1), a potent, selective, competitive NMDA receptor antagonist, showed it to be efficacious in inflammatory and neuropathic pain models. To increase the low oral bioavailability of 1 (3-5%), prodrug derivatives (3a-h) were synthesized and evaluated. The oxymethylene-spaced diphenyl analogue 3a demonstrated good stability at acidic and neutral pH, as well as in simulated gastric fluid. In rat plasma, 3a was rapidly converted to 1 via 2a. Pharmacokinetic studies indicated that the amount of systemic exposure of 1 produced by a 10 mg/kg oral dose of 3a was 2.5-fold greater than that produced by a 30 mg/kg oral dose of 1. Consistent with these results, 3a was significantly more potent and had a longer duration of activity than 1 following oral administration in a rodent model of inflammatory pain. Taken together, these results demonstrate that an oxymethylene-spaced prodrug approach increased the bioavailability of 1.


Assuntos
Compostos Azabicíclicos/farmacocinética , Organofosfonatos/farmacocinética , Pró-Fármacos/farmacocinética , Administração Oral , Animais , Compostos Azabicíclicos/administração & dosagem , Bile/metabolismo , Disponibilidade Biológica , Difosfonatos/síntese química , Difosfonatos/farmacocinética , Estabilidade de Medicamentos , Suco Gástrico/metabolismo , Masculino , Organofosfonatos/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores
11.
Neuropsychopharmacology ; 34(6): 1601-8, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19078951

RESUMO

Thyrotropin-releasing hormone (TRH) is a neuropeptide that initiates its effects in mice by interacting with two G-protein-coupled receptors, TRH receptor type 1 (TRH-R1) and TRH receptor type 2 (TRH-R2). Two previous reports described the effects of deleting TRH-R1 in mice. TRH-R1 knockout mice exhibit hypothyroidism, hyperglycemia, and increased depression and anxiety-like behavior. Here we report the generation of TRH-R2 knockout mice. The phenotype of these mice was characterized using gross and histological analyses along with blood hematological assays and chemistries. Standard metabolic tests to assess glucose and insulin tolerance were performed. Behavioral testing included elevated plus maze, open field, tail suspension, forced swim, and novelty-induced hypophagia tests. TRH-R2 knockout mice are euthyroid with normal basal and TRH-stimulated serum levels of thyroid-stimulating hormone (thyrotropin), are normoglycemic, and exhibit normal development and growth. Female, but not male, TRH-R2 knockout mice exhibit moderately increased depression-like and reduced anxiety-like phenotypes. Because the behavioral changes in TRH-R1 knockout mice may have been caused secondarily by their hypothyroidism whereas TRH-R2 knockout mice are euthyroid, these data provide the first evidence for the involvement of the TRH/TRH-R system, specifically extrahypothalamic TRH/TRH-R2, in regulating mood and affect.


Assuntos
Ansiedade/genética , Depressão/genética , Receptores do Hormônio Liberador da Tireotropina/genética , Receptores do Hormônio Liberador da Tireotropina/fisiologia , Animais , Glicemia/genética , Depressão/psicologia , Dieta , Gorduras na Dieta , Feminino , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Prolactina/sangue , Receptores do Hormônio Liberador da Tireotropina/deficiência , Caracteres Sexuais , Estresse Psicológico/genética , Tireotropina/sangue , Hormônio Liberador de Tireotropina/sangue , Tiroxina/sangue
12.
J Pharmacol Exp Ther ; 327(3): 820-6, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18812493

RESUMO

Fragile X syndrome is an X-linked disorder caused by the inactivation of the FMR1 gene, with symptoms ranging from impaired cognitive functions to seizures, anxiety, sensory abnormalities, and hyperactivity. Although fragile X syndrome is considered a typical Mendelian disorder, we have recently reported that the environment, specifically the fmr1(+/-) or fmr1(-/-) [H or knockout (KO)] maternal environment, elicits on its own a partial fragile X-like phenotype and can contribute to the overall phenotype of fmr1(-/0) (KO) male offspring. Genetically fmr1(+/0) (WT) males born to H females (H(maternal) > WT(offspring)), similar to KO male offspring born to H and KO mothers (H > KO and KO > KO), exhibit locomotor hyperactivity. These mice also showed reduced D(2) autoreceptor function, indicating a possible diminished feedback inhibition of dopamine (DA) release in the nigrostriatal and mesolimbic systems. The GABAergic system also regulates DA release, in part via presynaptic GABA(B) receptors (Rs) located on midbrain dopaminergic neurons. Here, we show that the locomotor inhibitory effect of the GABA(B)R agonist baclofen [4-amino-3-(4-chlorophenyl)-butanoic acid] is enhanced in all progeny of mutant mothers (H > WT, H > KO, and KO > KO) compared with WT > WT mice, irrespective of their own genotype. However, increased sensitivity to baclofen was selective and limited to the locomotor response because the muscle-relaxant and sedative effects of the drug were not altered by the maternal environment. These data show that GABA(B)R sensitization, traditionally induced pharmacologically, can also be elicited by the fmr1-deficient maternal environment.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/genética , Padrões de Herança , Receptores de GABA-B/fisiologia , Animais , Baclofeno/farmacologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Mães , Atividade Motora , Fenótipo
13.
Neuropsychopharmacology ; 33(11): 2667-75, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18172434

RESUMO

Fragile X syndrome is an X-linked disorder caused by the inactivation of the FMR-1 gene with symptoms ranging from impaired cognitive functions to seizures, anxiety, sensory abnormalities, and hyperactivity. Males are more severely affected than heterozygote (H) females, who, as carriers, have a 50% chance of transmitting the mutated allele in each pregnancy. fmr-1 knockout (KO) mice reproduce fragile X symptoms, including hyperactivity, seizures, and abnormal sensory processing. In contrast to the expectation that wild-type (WT) males born to H (fmr-1(+/-)) mothers (H>WT) are behaviorally normal and indistinguishable from WT males born to WT mothers (WT>WT); here, we show that H>WT offspring are more active than WT>WT offspring and that their hyperactivity is similar to male KO mice born to H or KO (fmr-1(-/-)) mothers (H>KO/KO>KO). H>WT mice, however, do not exhibit seizures or abnormal sensory processing. Consistent with their hyperactivity, the effect of the D2 agonist quinpirole is reduced in H>WT as well as in H>KO and KO>KO mice compared to WT>WT offspring, suggesting a diminished feedback inhibition of dopamine release. Our data indicate that some aspects of hyperactivity and associated dopaminergic changes in 'fragile X' mice are a maternal fmr-1 genotype rather than an offspring fmr-1 genotype effect.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/patologia , Comportamento Materno/fisiologia , Fenótipo , Animais , Feminino , Triagem de Portadores Genéticos , Masculino , Comportamento Materno/psicologia , Camundongos , Camundongos Knockout , Atividade Motora/genética , Gravidez , Reflexo de Sobressalto/genética
14.
J Neurosci ; 27(27): 7196-207, 2007 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-17611273

RESUMO

Thousands of children receive methylphenidate (MPH; Ritalin) for attention deficit/hyperactivity disorder (ADHD), yet the long-term neurochemical consequences of MPH treatment are unknown. To mimic clinical Ritalin treatment in children, male rats were injected with MPH (5 mg/kg) or vehicle twice daily from postnatal day 7 (PND7)-PND35. At the end of administration (PND35) or in adulthood (PND135), brain sections from littermate pairs were immunocytochemically labeled for neurotransmitters and cytological markers in 16 regions implicated in MPH effects and/or ADHD etiology. At PND35, the medial prefrontal cortex (mPFC) of rats given MPH showed 55% greater immunoreactivity (-ir) for the catecholamine marker tyrosine hydroxylase (TH), 60% more Nissl-stained cells, and 40% less norepinephrine transporter (NET)-ir density. In hippocampal dentate gyrus, MPH-receiving rats showed a 51% decrease in NET-ir density and a 61% expanded distribution of the new-cell marker PSA-NCAM (polysialylated form of neural cell adhesion molecule). In medial striatum, TH-ir decreased by 21%, and in hypothalamus neuropeptide Y-ir increased by 10% in MPH-exposed rats. At PND135, MPH-exposed rats exhibited decreased anxiety in the elevated plus-maze and a trend for decreased TH-ir in the mPFC. Neither PND35 nor PND135 rats showed major structural differences with MPH exposure. These findings suggest that developmental exposure to high therapeutic doses of MPH has short-term effects on select neurotransmitters in brain regions involved in motivated behaviors, cognition, appetite, and stress. Although the observed neuroanatomical changes largely resolve with time, chronic modulation of young brains with MPH may exert effects on brain neurochemistry that modify some behaviors even in adulthood.


Assuntos
Apetite/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Cognição/efeitos dos fármacos , Metilfenidato/administração & dosagem , Motivação , Estresse Fisiológico/prevenção & controle , Fatores Etários , Animais , Apetite/fisiologia , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Cognição/fisiologia , Feminino , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley , Estresse Fisiológico/metabolismo
15.
J Exp Psychol Anim Behav Process ; 31(1): 31-9, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15656725

RESUMO

The peak procedure was used to characterize response timing during acquisition and maintenance of conditioned responding in goldfish. Subjects received light-shock pairings with a 5- or 15-s interstimulus interval. On interspersed peak trials, the conditioned stimulus light was presented for 45 s and no shock was delivered. Peaks in the conditioned response, general activity, occurred at about the time of the expected unconditioned stimulus, and variability in the activity distribution was scalar. Modeling of the changes in the activity distributions over sessions revealed that the temporal features of the conditioned response changed very little during acquisition. The data suggest that times are learned early in training, and, contrary to I. P. Pavlov's (1927/1960) concept of "inhibition of delay," that timing is learning when to respond rather than learning when not to respond.


Assuntos
Aprendizagem por Associação , Condicionamento Clássico , Carpa Dourada , Motivação , Percepção do Tempo , Animais , Atenção , Aprendizagem da Esquiva , Sinais (Psicologia) , Eletrochoque , Generalização Psicológica , Inibição Psicológica , Atividade Motora , Percepção Visual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA