Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Neuroinflammation ; 18(1): 13, 2021 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-33407565

RESUMO

BACKGROUND: Perinatal inflammation is a key factor of brain vulnerability in neonates born preterm or with intra-uterine growth restriction (IUGR), two leading conditions associated with brain injury and responsible for neurocognitive and behavioral disorders. Systemic inflammation is recognized to activate microglia, known to be the critical modulators of brain vulnerability. Although some evidence supports a role for metabotropic glutamate receptor 3 (mGlu3 receptor) in modulation of neuroinflammation, its functions are still unknown in the developing microglia. METHODS: We used a double-hit rat model of perinatal brain injury induced by a gestational low-protein diet combined with interleukin-1ß injections (LPD/IL-1ß), mimicking both IUGR and prematurity-related inflammation. The effect of LPD/IL-1ß on mGlu3 receptor expression and the effect of mGlu3 receptor modulation on microglial reactivity were investigated using a combination of pharmacological, histological, and molecular and genetic approaches. RESULTS: Exposure to LPD/IL-1ß significantly downregulated Grm3 gene expression in the developing microglia. Both transcriptomic analyses and pharmacological modulation of mGlu3 receptor demonstrated its central role in the control of inflammation in resting and activated microglia. Microglia reactivity to inflammatory challenge induced by LPD/IL-1ß exposure was reduced by an mGlu3 receptor agonist. Conversely, both specific pharmacological blockade, siRNA knock-down, and genetic knock-out of mGlu3 receptors mimicked the pro-inflammatory phenotype observed in microglial cells exposed to LPD/IL-1ß. CONCLUSIONS: Overall, these data show that Grm3 plays a central role in the regulation of microglial reactivity in the immature brain. Selective pharmacological activation of mGlu3 receptors may prevent inflammatory-induced perinatal brain injury.


Assuntos
Microglia/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Animais Recém-Nascidos , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Feminino , Perfilação da Expressão Gênica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/patologia , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/antagonistas & inibidores
2.
Glia ; 67(2): 345-359, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30506969

RESUMO

Prematurity and fetal growth restriction (FGR) are frequent conditions associated with adverse neurocognitive outcomes. We have previously identified early deregulation of genes controlling neuroinflammation as a putative mechanism linking FGR and abnormal trajectory of the developing brain. While the oxytocin system was also found to be impaired following adverse perinatal events, its role in the modulation of neuroinflammation in the developing brain is still unknown. We used a double-hit rat model of perinatal brain injury induced by gestational low protein diet (LPD) and potentiated by postnatal injections of subliminal doses of interleukin-1ß (IL1ß) and a zebrafish model of neuroinflammation. Effects of the treatment with carbetocin, a selective, long lasting, and brain diffusible oxytocin receptor agonist, have been assessed using a combination of histological, molecular, and functional tools in vivo and in vitro. In the double-hit model, white matter inflammation, deficient myelination, and behavioral deficits have been observed and the oxytocin system was impaired. Early postnatal supplementation with carbetocin alleviated microglial activation at both transcriptional and cellular levels and provided long-term neuroprotection. The central anti-inflammatory effects of carbetocin have been shown in vivo in rat pups and in a zebrafish model of early-life neuroinflammation and reproduced in vitro on stimulated sorted primary microglial cell cultures from rats subjected to LPD. Carbetocin treatment was associated with beneficial effects on myelination, long-term intrinsic brain connectivity and behavior. Targeting oxytocin signaling in the developing brain may be an effective approach to prevent neuroinflammation - induced brain damage of perinatal origin.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Encéfalo/patologia , Microglia/efeitos dos fármacos , Receptores de Ocitocina/metabolismo , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/patologia , Células Cultivadas , Biologia Computacional , Dieta com Restrição de Proteínas/efeitos adversos , Modelos Animais de Doenças , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interleucina-1beta , Lipopolissacarídeos/toxicidade , Ocitócicos/uso terapêutico , Ocitocina/análogos & derivados , Ocitocina/uso terapêutico , Fragmentos de Peptídeos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , RNA Mensageiro/metabolismo , Peixe-Zebra
3.
Neuroimage ; 185: 851-856, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29649559

RESUMO

The emergence of functional neuroimaging has dramatically accelerated our understanding of the human mind. The advent of functional Magnetic Resonance Imaging paved the way for the next decades' major discoveries in neuroscience and today remains the "gold standard" for deep brain imaging. Recent improvements in imaging technology have been somewhat limited to incremental innovations of mature techniques instead of breakthroughs. Recently, the use of ultrasonic plane waves transmitted at ultrafast frame rates was shown to highly increase Doppler ultrasound sensitivity to blood flows in small vessels in rodents. By identifying regions of brain activation through neurovascular coupling, Ultrafast Doppler was entering into the world of preclinical neuroimaging. The combination of many advantages, including high spatio-temporal resolution, deep penetration, high sensitivity and portability provided unique information about brain function. Recently, Ultrafast Doppler imaging was found able to non-invasively image the spatial and temporal dynamics of microvascular changes during seizures and interictal periods with an unprecedented resolution at bedside. This review summarizes the technical basis, the added value and the clinical perspectives provided by this new brain imaging modality that could create a breakthrough in the knowledge of brain hemodynamics, brain insult, and neuroprotection.


Assuntos
Encéfalo/diagnóstico por imagem , Neuroimagem Funcional/métodos , Ultrassonografia Doppler/métodos , Feminino , Humanos , Processamento de Imagem Assistida por Computador/métodos , Recém-Nascido , Masculino
4.
Glia ; 64(12): 2306-2320, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27687291

RESUMO

Fetal growth restriction (FGR) is a major complication of human pregnancy, frequently resulting from placental vascular diseases and prenatal malnutrition, and is associated with adverse neurocognitive outcomes throughout life. However, the mechanisms linking poor fetal growth and neurocognitive impairment are unclear. Here, we aimed to correlate changes in gene expression induced by FGR in rats and abnormal cerebral white matter maturation, brain microstructure, and cortical connectivity in vivo. We investigated a model of FGR induced by low-protein-diet malnutrition between embryonic day 0 and birth using an interdisciplinary approach combining advanced brain imaging, in vivo connectivity, microarray analysis of sorted oligodendroglial and microglial cells and histology. We show that myelination and brain function are both significantly altered in our model of FGR. These alterations, detected first in the white matter on magnetic resonance imaging significantly reduced cortical connectivity as assessed by ultrafast ultrasound imaging. Fetal growth retardation was found associated with white matter dysmaturation as shown by the immunohistochemical profiles and microarrays analyses. Strikingly, transcriptomic and gene network analyses reveal not only a myelination deficit in growth-restricted pups, but also the extensive deregulation of genes controlling neuroinflammation and the cell cycle in both oligodendrocytes and microglia. Our findings shed new light on the cellular and gene regulatory mechanisms mediating brain structural and functional defects in malnutrition-induced FGR, and suggest, for the first time, a neuroinflammatory basis for the poor neurocognitive outcome observed in growth-restricted human infants. GLIA 2016;64:2306-2320.


Assuntos
Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Retardo do Crescimento Fetal/fisiopatologia , Microglia/metabolismo , Oligodendroglia/metabolismo , Transcriptoma/fisiologia , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Animais Recém-Nascidos , Antígenos/metabolismo , Antígenos CD/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Lesões Encefálicas/diagnóstico por imagem , Citocinas/metabolismo , Feminino , Expressão Gênica/fisiologia , Lipopolissacarídeos/farmacologia , Proteína Básica da Mielina/metabolismo , Vias Neurais/diagnóstico por imagem , Vias Neurais/efeitos dos fármacos , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Gravidez , Proteoglicanas/metabolismo , Ratos , Ratos Sprague-Dawley
5.
Pharmacol Res ; 105: 186-97, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26816085

RESUMO

Intracellular accumulation of hyperphosphorylated tau protein is linked to neuronal degeneration in Alzheimer's disease (AD). Mounting evidence suggests that tau phosphorylation and O-N-acetylglucosamine glycosylation (O-GlcNAcylation) are mutually exclusive post-translational modifications. O-GlcNAcylation depends on 3-5% of intracellular glucose that enters the hexosamine biosynthetic pathway. To our knowledge, the existence of an imbalance between tau phosphorylation and O-GlcNAcylation has not been reported in animal models of AD, as yet. Here, we used triple transgenic (3xTg-AD) mice at 12 months, an age at which hyperphosphorylated tau is already detected and associated with cognitive decline. In these mice, we showed that tau was hyperphosphorylated on both Ser396 and Thr205 in the hippocampus, and to a lower extent and exclusively on Thr205 in the frontal cortex. Tau O-GlcNAcylation, assessed in tau immunoprecipitates, was substantially reduced in the hippocampus of 3xTg-AD mice, with no changes in the frontal cortex or in the cerebellum. No changes in the expression of the three major enzymes involved in O-GlcNAcylation, i.e., glutamine fructose-6-phosphate amidotransferase, O-linked ß-N-acetylglucosamine transferase, and O-GlcNAc hydrolase were found in the hippocampus of 3xTg-AD mice. These data demonstrate that an imbalance between tau phosphorylation and O-GlcNAcylation exists in AD mice, and strengthens the hypothesis that O-GlcNAcylation might be targeted by disease modifying drugs in AD.


Assuntos
Acetilglucosamina/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Proteínas tau/metabolismo , Acilação , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Glicosilação , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Fosforilação
6.
Addict Biol ; 21(6): 1072-1085, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26011513

RESUMO

Palatable food is a strong activator of the reward circuitry and may cause addictive behavior leading to eating disorders. How early life events and sex interact in shaping hedonic sensitivity to palatable food is largely unknown. We used prenatally restraint stressed (PRS) rats, which show abnormalities in the reward system and anxious/depressive-like behavior. Some of the hallmarks of PRS rats are known to be sex-dependent. We report that PRS enhanced and reduced milk chocolate-induced conditioned place preference in males and females, respectively. Male PRS rats also show increases in plasma dihydrotestosterone (DHT) levels and dopamine (DA) levels in the nucleus accumbens (NAc), and reductions in 5-hydroxytryptamine (5-HT) levels in the NAc and prefrontal cortex (PFC). In male rats, systemic treatment with the DHT-lowering drug finasteride reduced both milk chocolate preference and NAc DA levels. Female PRS rats showed lower plasma estradiol (E2 ) levels and lower DA levels in the NAc, and 5-HT levels in the NAc and PFC. E2 supplementation reversed the reduction in milk chocolate preference and PFC 5-HT levels. In the hypothalamus, PRS increased ERα and ERß estrogen receptor and CARTP (cocaine-and-amphetamine receptor transcript peptide) mRNA levels in males, and 5-HT2C receptor mRNA levels in females. Changes were corrected by treatments with finasteride and E2 , respectively. These new findings show that early life stress has a profound impact on hedonic sensitivity to high-palatable food via long-lasting changes in gonadal hormones. This paves the way to the development of hormonal strategies aimed at correcting abnormalities in the response to natural rewards.


Assuntos
Preferências Alimentares/fisiologia , Recompensa , Estresse Psicológico/psicologia , Análise de Variância , Animais , Monoaminas Biogênicas/metabolismo , Encéfalo/metabolismo , Di-Hidrotestosterona/metabolismo , Dopamina/metabolismo , Feminino , Finasterida/farmacologia , Hipotálamo/metabolismo , Masculino , Córtex Pré-Frontal/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/psicologia , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Restrição Física/psicologia , Serotonina/metabolismo , Fatores Sexuais
7.
J Neurosci ; 34(6): 2015-24, 2014 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-24501344

RESUMO

Abnormalities of synaptic transmission in the hippocampus represent an integral part of the altered programming triggered by early life stress, which enhances the vulnerability to stress-related disorders in the adult life. Rats exposed to prenatal restraint stress (PRS) develop enduring biochemical and behavioral changes characteristic of an anxious/depressive-like phenotype. Most neurochemical abnormalities in PRS rats are found in the ventral hippocampus, a region that encodes memories related to stress and emotions. We have recently demonstrated a causal link between the reduction of glutamate release in the ventral hippocampus and anxiety-like behavior in PRS rats. To confer pharmacological validity to the glutamatergic hypothesis of stress-related disorders, we examined whether chronic treatment with two antidepressants with different mechanisms of action could correct the defect in glutamate release and associated behavioral abnormalities in PRS rats. Adult unstressed or PRS rats were treated daily with either agomelatine (40 mg/kg, i.p.) or fluoxetine (5 mg/kg, i.p.) for 21 d. Both treatments reversed the reduction in depolarization-evoked glutamate release and in the expression of synaptic vesicle-associated proteins in the ventral hippocampus of PRS rats. Antidepressant treatment also corrected abnormalities in anxiety-/depression-like behavior and social memory performance in PRS rats. The effect on glutamate release was strongly correlated with the improvement of anxiety-like behavior and social memory. These data offer the pharmacological demonstration that glutamatergic hypofunction in the ventral hippocampus lies at the core of the pathological phenotype caused by early life stress and represents an attractive pharmacological target for novel therapeutic strategies.


Assuntos
Antidepressivos/uso terapêutico , Ácido Glutâmico/metabolismo , Efeitos Tardios da Exposição Pré-Natal/tratamento farmacológico , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/metabolismo , Animais , Ansiedade/tratamento farmacológico , Ansiedade/metabolismo , Ansiedade/psicologia , Depressão/tratamento farmacológico , Depressão/metabolismo , Depressão/psicologia , Feminino , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/psicologia , Ratos , Ratos Sprague-Dawley , Estresse Psicológico/psicologia , Resultado do Tratamento
8.
J Neurosci ; 32(48): 17143-54, 2012 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-23197707

RESUMO

Abnormalities of synaptic transmission and plasticity in the hippocampus represent an integral part of the altered programming triggered by early life stress. Prenatally restraint stressed (PRS) rats develop long-lasting biochemical and behavioral changes, which are the expression of an anxious/depressive-like phenotype. We report here that PRS rats showed a selective impairment of depolarization- or kainate-stimulated glutamate and [(3)H]d-aspartate release in the ventral hippocampus, a region encoding memories related to stress and emotions. GABA release was unaffected in PRS rats. As a consequence of reduced glutamate release, PRS rats were also highly resistant to kainate-induced seizures. Abnormalities of glutamate release were associated with large reductions in the levels of synaptic vesicle-related proteins, such as VAMP (synaptobrevin), syntaxin-1, synaptophysin, synapsin Ia/b and IIa, munc-18, and Rab3A in the ventral hippocampus of PRS rats. Anxiety-like behavior in male PRS (and control) rats was inversely related to the extent of depolarization-evoked glutamate release in the ventral hippocampus. A causal relationship between anxiety-like behavior and reduction in glutamate release was demonstrated using a mixture of the mGlu2/3 receptor antagonist, LY341495, and the GABA(B) receptor antagonist, CGP52432, which was shown to amplify depolarization-evoked [(3)H]d-aspartate release in the ventral hippocampus. Bilateral microinfusion of CGP52432 plus LY341495 in the ventral hippocampus abolished anxiety-like behavior in PRS rats. These findings indicate that an impairment of glutamate release in the ventral hippocampus is a key component of the neuroplastic program induced by PRS, and that strategies aimed at enhancing glutamate release in the ventral hippocampus correct the "anxious phenotype" caused by early life stress.


Assuntos
Ansiedade/metabolismo , Comportamento Animal/fisiologia , Ácido Glutâmico/metabolismo , Hipocampo/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Estresse Psicológico/metabolismo , Aminoácidos/farmacologia , Animais , Benzilaminas/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Antagonistas de Receptores de GABA-A/farmacologia , Hipocampo/efeitos dos fármacos , Ácido Caínico , Masculino , Proteínas Munc18/metabolismo , Ácidos Fosfínicos/farmacologia , Gravidez , Proteínas R-SNARE/metabolismo , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Convulsões/metabolismo , Sinapsinas/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Sinaptofisina/metabolismo , Sintaxina 1/metabolismo , Xantenos/farmacologia , Proteína rab3A de Ligação ao GTP/metabolismo
9.
Mol Pharmacol ; 84(2): 244-51, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23716620

RESUMO

The use of classic antipsychotic drugs is limited by the occurrence of extrapyramidal motor symptoms, which are caused by dopamine (DA) receptor blockade in the neostriatum. We examined the impact of early-life stress on haloperidol-induced catalepsy using the rat model of prenatal restraint stress (PRS). Adult "PRS rats," i.e., the offspring of mothers exposed to restraint stress during pregnancy, were resistant to catalepsy induced by haloperidol (0.5-5 mg/kg i.p.) or raclopride (2 mg/kg s.c.). Resistance to catalepsy in PRS rats did not depend on reductions in blood or striatal levels, as compared with unstressed control rats. PRS rats also showed a greater behavioral response to the DA receptor agonist, apomorphine, suggesting that PRS causes enduring neuroplastic changes in the basal ganglia motor circuit. To examine the activity of this circuit, we performed a stereological counting of c-Fos(+) neurons in the external and internal globus pallidus, subthalamic nucleus, and ventral motor thalamic nuclei. Remarkably, the number of c-Fos(+) neurons in ventral motor thalamic nuclei was higher in PRS rats than in unstressed controls, both under basal conditions and in response to single or repeated injections with haloperidol. Ventral motor thalamic nuclei contain exclusively excitatory projection neurons that convey the basal ganglia motor programming to the cerebral cortex. Hence, an increased activity of ventral motor thalamic nuclei nicely explains the refractoriness of PRS rats to haloperidol-induced catalepsy. Our data raise the interesting possibility that early-life stress is protective against extrapyramidal motor effects of antipsychotic drugs in the adult life.


Assuntos
Catalepsia/induzido quimicamente , Haloperidol/farmacologia , Estresse Fisiológico/fisiologia , Animais , Antipsicóticos/farmacologia , Apomorfina/farmacologia , Catalepsia/sangue , Catecolaminas/sangue , Catecolaminas/farmacologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Agonistas de Dopamina/farmacologia , Feminino , Globo Pálido/efeitos dos fármacos , Globo Pálido/metabolismo , Haloperidol/sangue , Masculino , Exposição Materna , Relações Materno-Fetais/efeitos dos fármacos , Gravidez , Proteínas Proto-Oncogênicas c-fos/metabolismo , Racloprida/farmacologia , Ratos , Receptores Dopaminérgicos/metabolismo , Núcleo Subtalâmico/efeitos dos fármacos , Núcleo Subtalâmico/metabolismo , Núcleos Ventrais do Tálamo/efeitos dos fármacos , Núcleos Ventrais do Tálamo/metabolismo
10.
Int J Neuropsychopharmacol ; 16(2): 323-38, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22310059

RESUMO

Agomelatine is a novel antidepressant acting as an MT1/MT2 melatonin receptor agonist/5-HT2C serotonin receptor antagonist. Because of its peculiar pharmacological profile, this drug caters the potential to correct the abnormalities of circadian rhythms associated with mood disorders, including abnormalities of the sleep/wake cycle. Here, we examined the effect of chronic agomelatine treatment on sleep architecture and circadian rhythms of motor activity using the rat model of prenatal restraint stress (PRS) as a putative 'aetiological' model of depression. PRS was delivered to the mothers during the last 10 d of pregnancy. The adult progeny ('PRS rats') showed a reduced duration of slow wave sleep, an increased duration of rapid eye movement (REM) sleep, an increased number of REM sleep events and an increase in motor activity before the beginning of the dark phase of the light/dark cycle. In addition, adult PRS rats showed an increased expression of the transcript of the primary response gene, c-Fos, in the hippocampus just prior to the beginning of the dark phase. All these changes were reversed by a chronic oral treatment with agomelatine (2000 ppm in the diet). The effect of agomelatine on sleep was largely attenuated by treatment with the MT1/MT2 melatonin receptor antagonist, S22153, which caused PRS-like sleep disturbances on its own. These data provide the first evidence that agomelatine corrects sleep architecture and restores circadian homeostasis in a preclinical model of depression and supports the value of agomelatine as a novel antidepressant that resynchronizes circadian rhythms under pathological conditions.


Assuntos
Acetamidas/uso terapêutico , Transtornos Cronobiológicos/tratamento farmacológico , Hipnóticos e Sedativos/uso terapêutico , Transtornos dos Movimentos/tratamento farmacológico , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Transtornos do Sono-Vigília/tratamento farmacológico , Análise de Variância , Animais , Animais Recém-Nascidos , Nível de Alerta/efeitos dos fármacos , Autorradiografia , Transtornos Cronobiológicos/etiologia , Modelos Animais de Doenças , Esquema de Medicação , Eletroencefalografia , Eletromiografia , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Transtornos dos Movimentos/etiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/tratamento farmacológico , Efeitos Tardios da Exposição Pré-Natal/patologia , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Melatonina/antagonistas & inibidores , Restrição Física/efeitos adversos , Transtornos do Sono-Vigília/etiologia , Tiofenos/farmacologia
11.
J Neurosci ; 30(24): 8096-101, 2010 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-20554860

RESUMO

Oleoylethanolamide (OEA) is a biologically active lipid amide that is released by small-intestinal enterocytes during the absorption of dietary fat and inhibits feeding by engaging the nuclear receptor, peroxisome proliferator-activated receptor-alpha (PPAR-alpha). Previous studies have shown that the anorexic effects of systemically administered OEA require the activation of sensory afferents of the vagus nerve. The central circuits involved in mediating OEA-induced hypophagia remain unknown. In the present study, we report the results of in situ hybridization and immunohistochemistry experiments in rats and mice, which show that systemic injections of OEA (5-10 mg kg(-1), intraperitoneal) enhance expression of the neuropeptide oxytocin in magnocellular neurons of the paraventricular nucleus (PVN) and supraoptic nucleus (SON) of the hypothalamus. No such effect is observed with other hypothalamic neuropeptides, including vasopressin, thyrotropin-releasing hormone and pro-opiomelanocortin. The increase in oxytocin expression elicited by OEA was absent in mutant PPAR-alpha-null mice. Pharmacological blockade of oxytocin receptors in the brain by intracerebroventricular infusion of the selective oxytocin antagonist, L-368,899, prevented the anorexic effects of OEA. The results suggest that OEA suppresses feeding by activating central oxytocin transmission.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Ácidos Oleicos/farmacologia , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Supraóptico/efeitos dos fármacos , Animais , Canfanos/farmacologia , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Endocanabinoides , Regulação da Expressão Gênica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ocitocina/antagonistas & inibidores , Ocitocina/sangue , Ocitocina/genética , PPAR alfa/deficiência , Núcleo Hipotalâmico Paraventricular/metabolismo , Piperazinas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Núcleo Supraóptico/metabolismo
12.
Front Neurol ; 12: 668877, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34220677

RESUMO

Backgroud: Type-3 metabotropic glutamate (mGlu3) receptors are found in both neurons and glial cells and regulate synaptic transmission, astrocyte function, and microglial reactivity. Here we show that the genetic deletion of mGlu3 receptors amplifies ischemic brain damage and associated neuroinflammation in adult mice. An increased infarct size was observed in mGlu3-/- mice of both CD1 and C57Black strains 24 h following a permanent occlusion of the middle cerebral artery (MCA) as compared to their respective wild-type (mGlu3+/+ mice) counterparts. Increases in the expression of selected pro-inflammatory genes including those encoding interleukin-1ß, type-2 cycloxygenase, tumor necrosis factor-α, CD86, and interleukin-6 were more prominent in the peri-infarct region of mGlu3-/- mice. In contrast, the expression of two genes associated with the anti-inflammatory phenotype of microglia (those encoding the mannose-1-phosphate receptor and the α-subunit of interleukin-4 receptor) and the gene encoding the neuroprotective factor, glial cell line-derived neurotrophic factor, was enhanced in the peri-infarct region of wild-type mice, but not mGlu3-/- mice, following MCA occlusion. In C57Black mice, the genetic deletion of mGlu3 receptors worsened the defect in the paw placement test as assessed in the contralateral forepaw at short times (4 h) following MCA occlusion. These findings suggest that mGlu3 receptors are protective against ischemic brain damage and support the way to the use of selective mGlu3 receptor agonists or positive allosteric modulators in experimental animal models of ischemic stroke.

13.
Nat Commun ; 12(1): 1080, 2021 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33597538

RESUMO

Clinicians have long been interested in functional brain monitoring, as reversible functional losses often precedes observable irreversible structural insults. By characterizing neonatal functional cerebral networks, resting-state functional connectivity is envisioned to provide early markers of cognitive impairments. Here we present a pioneering bedside deep brain resting-state functional connectivity imaging at 250-µm resolution on human neonates using functional ultrasound. Signal correlations between cerebral regions unveil interhemispheric connectivity in very preterm newborns. Furthermore, fine-grain correlations between homologous pixels are consistent with white/grey matter organization. Finally, dynamic resting-state connectivity reveals a significant occurrence decrease of thalamo-cortical networks for very preterm neonates as compared to control term newborns. The same method also shows abnormal patterns in a congenital seizure disorder case compared with the control group. These results pave the way to infants' brain continuous monitoring and may enable the identification of abnormal brain development at the bedside.


Assuntos
Encéfalo/diagnóstico por imagem , Córtex Cerebral/diagnóstico por imagem , Epilepsia/diagnóstico por imagem , Substância Cinzenta/diagnóstico por imagem , Substância Branca/diagnóstico por imagem , Algoritmos , Encéfalo/fisiopatologia , Córtex Cerebral/fisiopatologia , Epilepsia/diagnóstico , Epilepsia/fisiopatologia , Feminino , Substância Cinzenta/fisiopatologia , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Imageamento por Ressonância Magnética/métodos , Masculino , Modelos Neurológicos , Rede Nervosa/diagnóstico por imagem , Rede Nervosa/fisiopatologia , Ultrassonografia Doppler/métodos , Substância Branca/fisiopatologia
14.
Nutrients ; 13(11)2021 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-34835975

RESUMO

Microglial activation is a key modulator of brain vulnerability in response to intra-uterine growth restriction (IUGR). However, the consequences of IUGR on microglial development and the microglial proteome are still unknown. We used a model of IUGR induced by a gestational low-protein diet (LPD) in rats. Microglia, isolated from control and growth-restricted animals at P1 and P4, showed significant changes in the proteome between the two groups. The expression of protein sets associated with fetal growth, inflammation, and the immune response were significantly enriched in LPD microglia at P1 and P4. Interestingly, upregulation of protein sets associated with the oxidative stress response and reactive oxygen species production was observed at P4 but not P1. During development, inflammation-associated proteins were upregulated between P1 and P4 in both control and LPD microglia. By contrast, proteins associated with DNA repair and senescence pathways were upregulated in only LPD microglia. Similarly, protein sets involved in protein retrograde transport were significantly downregulated in only LPD microglia. Overall, these data demonstrate significant and multiple effects of LPD-induced IUGR on the developmental program of microglial cells, leading to an abnormal proteome within the first postnatal days.


Assuntos
Retardo do Crescimento Fetal/metabolismo , Microglia/metabolismo , Proteoma/metabolismo , Animais , Animais Recém-Nascidos , Peso Corporal , Análise por Conglomerados , Dieta com Restrição de Proteínas , Inflamação/patologia , Estresse Oxidativo , Ratos Sprague-Dawley
15.
Alcohol Clin Exp Res ; 33(8): 1346-54, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19413649

RESUMO

BACKGROUND: Although it is generally believed that chronic ethanol consumption impairs learning and memory, results obtained in experimental animals are not univocal, and there are conditions in which ethanol paradoxically improves cognitive functions. In the present work, we investigated the effects of prenatal stress and of chronic ethanol exposure during adulthood on spatial memory in rats. METHODS: Rats were subjected to a prenatal stress delivered as 3 daily 45-minute sections of restraint stress to the mothers during the last 10 days of pregnancy (PRS rats). After 7 months of ethanol exposure (ethanol 10%, oral intake), memory performances were evaluated in a spatial discrimination test in control and PRS male rats. Then, the oxidative damages and the expression of metabotropic glutamate (mGlu) receptors were assessed in their hippocampus. RESULTS: Chronic ethanol exposure resulted in a reduced performance in a spatial recognition task in control animals. Unexpectedly, however, the same treatment attenuated spatial memory deficits in rats that had been subjected to prenatal stress. This paradigm of ethanol administration did not produce detectable signs of oxidative damage in the hippocampus in either unstressed or PRS rats. Interestingly, ethanol intake resulted in differential effects in the expression of mGlu receptor subtypes implicated in mechanisms of learning and memory. In control rats, ethanol intake reduced mGlu2/3 and mGlu5 receptor levels in the hippocampus; in PRS rats, which exhibited a constitutive reduction in the levels of these mGlu receptor subtypes, ethanol increased the expression of mGlu1a receptors but did not change the expression of mGlu2/3 or mGlu5 receptors. CONCLUSION: Our findings support the idea that stress-related events occurring before birth have long-lasting effects on brain function and behavior, and suggest that the impact of ethanol on cognition is not only dose- and duration-dependent, but also critically influenced by early life experiences.


Assuntos
Etanol/administração & dosagem , Hipocampo/metabolismo , Transtornos da Memória/prevenção & controle , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Receptores de Glutamato Metabotrópico/biossíntese , Estresse Psicológico/metabolismo , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipocampo/efeitos dos fármacos , Masculino , Transtornos da Memória/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Comportamento Espacial/efeitos dos fármacos , Comportamento Espacial/fisiologia , Regulação para Cima/efeitos dos fármacos
16.
Front Psychol ; 10: 715, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31001173

RESUMO

Preterm infants (PTI) typically experience many painful and stressful procedures or events during their first weeks of life in a neonatal intensive care unit, and these can profoundly impact subsequent brain development and function. Several protective interventions during this sensitive period stimulate the oxytocin system, reduce pain and stress, and improve brain development. This review provides an overview of the environmental risk factors experienced by PTI during hospitalization, with a focus on the effects of pain, and early maternal separation. We also describe the long-term adverse effects of the simultaneous experiences of pain and maternal separation, and the potential beneficial effects of maternal vocalizations, parental contact, and several related processes, which appear to be mediated by the oxytocin system.

17.
Front Mol Neurosci ; 12: 89, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31118884

RESUMO

Stress and the circadian systems play a major role in an organism's adaptation to environmental changes. The adaptive value of the stress system is reactive while that of the circadian system is predictive. Dysfunctions in these two systems may account for many clinically relevant disorders. Despite the evidence that interindividual differences in stress sensitivity and in the functioning of the circadian system are related, there is limited integrated research on these topics. Moreover, sex differences in these systems are poorly investigated. We used the perinatal stress (PRS) rat model, a well-characterized model of maladaptive programming of reactive and predictive adaptation, to monitor the running wheel behavior in male and female adult PRS rats, under a normal light/dark cycle as well as in response to a chronobiological stressor (6-h phase advance/shift). We then analyzed across different time points the expression of genes involved in circadian clocks, stress response, signaling, and glucose metabolism regulation in the suprachiasmatic nucleus (SCN). In the unstressed control group, we found a sex-specific profile that was either enhanced or inverted by PRS. Also, PRS disrupted circadian wheel-running behavior by inducing a phase advance in the activity of males and hypoactivity in females and increased vulnerability to chronobiological stress in both sexes. We also observed oscillations of several genes in the SCN of the unstressed group in both sexes. PRS affected males to greater extent than females, with PRS males displaying a pattern similar to unstressed females. Altogether, our findings provide evidence for a specific profile of dysmasculinization induced by PRS at the behavioral and molecular level, thus advocating the necessity to include sex as a biological variable to study the set-up of circadian system in animal models.

18.
J Neurosci ; 27(26): 7041-6, 2007 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-17596453

RESUMO

A growing body of evidence underscores the importance of early life events as predictors of health in adulthood. Abnormalities in maternal care or other forms of early postnatal stress induce long-term changes in behavior and influence the vulnerability to illnesses throughout life. Some of these changes may be produced by the activation of the hypothalamic-pituitary-adrenal (HPA) axis, which is invariably associated with stress. We used a model in which neonate rats are fed by mothers drinking water supplemented with 0.2 mg/ml corticosterone, the main glucocorticoid hormone in rodents. Plasma corticosterone levels increased in the dams to an extent similar to that induced by a mild stress. Corticosterone-treated dams also showed an increase in maternal care. Remarkably, adult rats that had been nursed by corticosterone-treated mothers were protected against neuronal damage and cognitive impairment produced by transient global brain ischemia. Neuroprotection was associated with a reduced HPA response to ischemia and was primarily decreased when corticosterone was injected at a dose that eliminated any difference in endogenous corticosterone levels between rats raised by mothers supplemented with corticosterone and their matched controls. These data suggest that an increased maternal care protects the offspring against ischemic neuronal damage and that at least a component of neuroprotection is mediated by a reduced response of the HPA axis to ischemia.


Assuntos
Envelhecimento/fisiologia , Isquemia Encefálica/metabolismo , Corticosterona/metabolismo , Lactação/fisiologia , Fármacos Neuroprotetores/metabolismo , Animais , Animais Recém-Nascidos , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/prevenção & controle , Corticosterona/farmacologia , Feminino , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Fármacos Neuroprotetores/farmacologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/metabolismo , Ratos , Ratos Wistar , Estresse Fisiológico/metabolismo , Estresse Fisiológico/fisiopatologia , Tempo
19.
Front Neurol ; 9: 229, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29706926

RESUMO

Oxytocin is a neurohypophysal hormone known for its activity during labor and its role in lactation. However, the function of oxytocin (OTX) goes far beyond the peripheral regulation of reproduction, and the central effects of OTX have been extensively investigated, since it has been recognized to influence the learning and memory processes. OTX has also prominent effects on social behavior, anxiety, and autism. Interaction between glucocorticoids, OTX, and maternal behavior may have long-term effects on the developmental program of the developing brain subjected to adverse events during pre and perinatal periods. OTX treatment in humans improves many aspects of social cognition and behavior. Its effects on the hypothalamic-pituitary-adrenal axis and inflammation appear to be of interest in neonates because these properties may confer benefits when the perinatal brain has been subjected to injury. Indeed, early life inflammation and abnormal adrenal response to stress have been associated with an abnormal white matter development. Recent investigations demonstrated that OTX is involved in the modulation of microglial reactivity in the developing brain. This review recapitulates state-of-the art data supporting the hypothesis that the OTX system could be considered as an innovative candidate for neuroprotection, especially in the immature brain.

20.
Front Neurol ; 9: 605, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30254599

RESUMO

Neuroinflammation has a key role in the pathogenesis of perinatal brain injury. Caffeine, a nonspecific antagonist of adenosine receptors (ARs), is widely used to treat apnea of prematurity and has been linked to a decrease in the incidence of cerebral palsy in premature infants. The mechanisms explaining its neuroprotective effect have not yet been elucidated. The objective of this study was to characterize the expression of adenosine and ARs in two neonatal rat models of neuroinflammation and to determine the effect of A2aR blockade on microglial activation assessed through inflammatory cytokine gene expression. We have used two rat models of microglial activation: the gestational low protein diet (LPD) model, associated with chronic brain injury, and postnatal ibotenate intracerebral injections, responsible for acute excitotoxicity injury. Adenosine blood levels have been measured by Tandem Mass Spectrometry. The expression of ARs in vivo was assessed using qPCR and immunohistochemistry. In vivo models have been replicated in vitro on primary microglial cell cultures exposed to A2aR agonist CGS-21680 or antagonist SCH-58261. The effects of these treatments have been assessed on the M1/M2 cytokine expressions measured by RT-qPCR. LPD during pregnancy was associated with higher adenosine levels in pups at postnatal day 1 and 4. A2aR mRNA expression was significantly increased in both cortex and magnetically sorted microglial cells from LPD animals compared to controls. CD73 expression, responsible for extracellular production of brain adenosine, was significantly increased in LPD cortex and sorted microglia cells. Moreover, CD73 protein level was increased in ibotenate treated animals. In vitro experiments confirmed that LPD or control microglial cells exposed to ibotenate display an increased expression, at both protein and molecular levels, of A2aR and M1 markers (IL-1ß, IL-6, iNOS, TNFα). This pro-inflammatory profile was significantly reduced by SCH-58261, which reduces M1 markers in both LPD and ibotenate-exposed cells, with no effect on control cells. In the same experimental conditions, a partial increased of M1 cytokines was observed in response to A2aR agonist CGS-21680. These results support the involvement of adenosine and particularly of its receptor A2aR in the regulation of microglia in two different animal models of neuroinflammation.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA