Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
Neuropathol Appl Neurobiol ; 47(2): 297-315, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32898926

RESUMO

AIMS: Impairment of blood-brain barrier (BBB) is involved in numerous neurological diseases from developmental to aging stages. Reliable imaging of increased BBB permeability is therefore crucial for basic research and preclinical studies. Today, the analysis of extravasation of exogenous dyes is the principal method to study BBB leakage. However, these procedures are challenging to apply in pups and embryos and may appear difficult to interpret. Here we introduce a novel approach based on agonist-induced internalization of a neuronal G protein-coupled receptor widely distributed in the mammalian brain, the somatostatin receptor type 2 (SST2). METHODS: The clinically approved SST2 agonist octreotide (1 kDa), when injected intraperitoneally does not cross an intact BBB. At sites of BBB permeability, however, OCT extravasates and induces SST2 internalization from the neuronal membrane into perinuclear compartments. This allows an unambiguous localization of increased BBB permeability by classical immunohistochemical procedures using specific antibodies against the receptor. RESULTS: We first validated our approach in sensory circumventricular organs which display permissive vascular permeability. Through SST2 internalization, we next monitored BBB opening induced by magnetic resonance imaging-guided focused ultrasound in murine cerebral cortex. Finally, we proved that after intraperitoneal agonist injection in pregnant mice, SST2 receptor internalization permits analysis of BBB integrity in embryos during brain development. CONCLUSIONS: This approach provides an alternative and simple manner to assess BBB dysfunction and development in different physiological and pathological conditions.


Assuntos
Barreira Hematoencefálica/patologia , Permeabilidade Capilar , Imuno-Histoquímica/métodos , Receptores de Somatostatina/análise , Receptores de Somatostatina/metabolismo , Animais , Anticorpos Monoclonais , Camundongos , Camundongos Endogâmicos C57BL , Octreotida/metabolismo , Ratos , Ratos Wistar
2.
Clin Genet ; 93(6): 1141-1147, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29508392

RESUMO

Ephrin B2, one of the ligand of the EphB receptors, is involved in a complex signaling pathway regulating the development of the nervous system, neuronal migration, erythropoiesis and vasculogenesis. We report a patient with a de novo variant in EFNB2 and a family in which segregates a 610-kb deletion at chromosome 13q33 encompassing only ARGLU1 and EFNB2 genes. The de novo variant was observed in a patient with anal stenosis, hypoplastic left ventricle and mild developmental delay. The deletion was identified in 2 sibs with congenital heart defect and mild developmental delay. One of the affected sibs further had myoclonic epilepsy and bilateral sensorineural hearing loss. The carrier mother was apparently asymptomatic. Because EFNB2 is located in the subtelomeric region of 13q chromosome, we reviewed the previous reports of terminal 13q deletion. We suggest that haploinsufficiency of the EFNB2 could be at the origin of several clinical features reported in 13qter deletions, including intellectual disability, seizures, congenital heart defects, anorectal malformation and hearing loss.


Assuntos
Transtornos Cromossômicos/genética , Efrina-B2/genética , Haploinsuficiência/genética , Transtornos do Neurodesenvolvimento/genética , Pré-Escolar , Deleção Cromossômica , Cromossomos Humanos Par 13/genética , Feminino , Estudos de Associação Genética , Humanos , Lactente , Recém-Nascido , Masculino , Linhagem
3.
Brain Behav Immun ; 73: 427-440, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29879442

RESUMO

Maternal immune activation (MIA) is a common environmental insult on the developing brain and represents a risk factor for neurodevelopmental disorders. Animal models of in utero inflammation further revealed a causal link between maternal inflammatory activation during pregnancy and behavioural impairment relevant to neurodevelopmental disorders in the offspring. Accumulating evidence point out that proinflammatory cytokines produced both in the maternal and fetal compartments are responsible for social, cognitive and emotional behavioral deficits in the offspring. Polyunsaturated fatty acids (PUFAs) are essential fatty acids with potent immunomodulatory activities. PUFAs and their bioactive derivatives can promote or inhibit many aspects of the immune and inflammatory response. PUFAs of the n-3 series ('n-3 PUFAs', also known as omega-3) exhibit anti-inflammatory/pro-resolution properties and promote immune functions, while PUFAs of the n-6 series ('n-6 PUFAs' or omega-6) favor pro-inflammatory responses. The present study aimed at providing insight into the effects of n-3 PUFAs on the consequences of MIA on brain development. We hypothesized that a reduction in n-3 PUFAs exacerbates both maternal and fetal inflammatory responses to MIA and later-life defects in memory in the offspring. Based on a lipopolysaccharide (LPS) model of MIA (LPS injection at embryonic day 17), we showed that n-3 PUFA deficiency 1) alters fatty acid composition of the fetal and adult offspring brain; 2) exacerbates maternal and fetal inflammatory processes with no significant alteration of microglia phenotype, and 3) induces spatial memory deficits in the adult offspring. We also showed a strong negative correlation between brain content in n-3 PUFA and cytokine production in MIA-exposed fetuses. Overall, our study is the first to address the deleterious effects of n-3 PUFA deficiency on brain lipid composition, inflammation and memory performances in MIA-exposed animals and indicates that it should be considered as a potent environmental risk factor for the apparition of neurodevelopmental disorders.


Assuntos
Ácidos Graxos Ômega-3/deficiência , Ácidos Graxos Ômega-3/metabolismo , Memória Espacial/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Citocinas/efeitos dos fármacos , Suplementos Nutricionais , Modelos Animais de Doenças , Ácidos Graxos Ômega-3/farmacologia , Ácidos Graxos Ômega-6/metabolismo , Ácidos Graxos Ômega-6/fisiologia , Feminino , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Transtornos da Memória/metabolismo , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Transtornos do Neurodesenvolvimento/etiologia , Transtornos do Neurodesenvolvimento/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/imunologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Comportamento Social
4.
Acta Anaesthesiol Scand ; 58(6): 733-42, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24724965

RESUMO

BACKGROUND: The highly selective α2 -adrenoreceptor agonist, dexmedetomidine, exerts neuroprotective, analgesic, anti-inflammatory and sympatholytic properties that may be beneficial for perinatal asphyxia. The optimal safe dose for pre-clinical newborn neuroprotection studies is unknown. METHODS: Following cerebral hypoxia-ischaemia, dexmedetomidine was administered to nine newborn piglets in a de-escalation dose study in combination with hypothermia (whole body cooling to 33.5°C). Dexmedetomidine was administered with a loading dose of 1 µg/kg and maintenance infusion at doses from 10 to 0.6 µg/kg/h. One additional piglet was not subjected to hypoxia-ischaemia. Blood for pharmacokinetic analysis was sampled pre-insult and frequently post-insult. A one-compartment linear disposition model was used to fit data. Population parameter estimates were obtained using non-linear mixed effects modelling. RESULTS: All dexmedetomidine infusion regimens led to plasma concentrations above those associated with sedation in neonates and children (0.4-0.8 µg/l). Seven out of the nine piglets with hypoxia-ischaemia experienced periods of bradycardia, hypotension, hypertension and cardiac arrest; all haemodynamic adverse events occurred in piglets with plasma concentrations greater than 1 µg/l. Dexmedetomidine clearance was 0.126 l/kg/h [coefficient of variation (CV) 46.6.%] and volume of distribution was 3.37 l/kg (CV 191%). Dexmedetomidine clearance was reduced by 32.7% at a temperature of 33.5°C. Dexmedetomidine clearance was reduced by 55.8% following hypoxia-ischaemia. CONCLUSIONS: Dexmedetomidine clearance was reduced almost tenfold compared with adult values in the newborn piglet following hypoxic-ischaemic brain injury and subsequent therapeutic hypothermia. Reduced clearance was related to cumulative effects of both hypothermia and exposure to hypoxia. High plasma levels of dexmedetomidine were associated with major cardiovascular complications.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Asfixia Neonatal/complicações , Dexmedetomidina/farmacocinética , Hipotermia Induzida , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/sangue , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Animais , Dexmedetomidina/sangue , Dexmedetomidina/uso terapêutico , Modelos Animais de Doenças , Hipóxia-Isquemia Encefálica/etiologia , Masculino , Taxa de Depuração Metabólica , Fármacos Neuroprotetores/sangue , Fármacos Neuroprotetores/uso terapêutico , Dinâmica não Linear , Sus scrofa , Suínos
5.
Brain Behav Immun Health ; 38: 100791, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38818370

RESUMO

Background: Immune system activation in the neonatal period is associated with white matter injury in preterm infants. In animal studies, neonatal priming of the immune system leads to chronic activation of i.e. microglia cells and altered neuroinflammatory responses potentially years after preterm birth. This may contribute further to brain injury and neurodevelopmental impairment. It is unknown to what extend this also occurs in human. Aim: To identify neuro-inflammatory markers at school age that relate to motor, cognitive and behavioral impairments in preterm born children in a pilot case-control study. Methods: We included n = 20 preterm born children (GA < 28 weeks) in this study, of which n = 10 with motor, cognitive and behavorial impairments and n = 10 preterm born controls next to n = 30 healthy adult controls. In the preterm children, at 8-9 years, 39 inflammatory markers were assessed by Luminex assay in blood serum samples. Firstly, the preterm concentrations of these markers were compared to n = 30 adult controls. Then a univariate analysis was performed to determine differences in values between preterm children with and without impairment at school age. Finally, a principal component analysis and hierarchical clustering was performed to identify protein profiles in preterm born children that relate to impairment at school age. Results: Inflammatory proteins in preterm children at school age differed from values of adult controls. Within the group of preterm children, we found significantly higher levels of GM-CSF in preterms with impairment (p < 0.01) and a trend towards significance for Gal1 and TRAIL (p = 0.06 and p = 0.06 respectively) when compared to preterms without impairment. In addition, differences in clustering of proteins between preterm children was observed, however this variance was not explained by presence of neurodevelopmental impairments. Conclusion: The inflammatory profile at school age in preterm children is different from that of adult controls. The immune modulating cytokines GM-CSF, Gal1 and TRAIL were higher in preterm children with impairment than control preterm children, suggesting that immune responses are altered in these children. No specific cluster of inflammatory markers could be identified. Results indicate that even at school age, neuroinflammatory pathways are activated in preterm born children with neurodevelopmental impairments.

6.
Dev Neurosci ; 35(2-3): 130-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23548575

RESUMO

Toll-like receptors (TLRs) are members of the pattern recognition receptor family that detect components of foreign pathogens or endogenous molecules released in response to injury. Recent studies demonstrate that TLRs also have a functional role in regulating neuronal proliferation in the developing brain. This study investigated cellular expression of TLR3 using immunohistochemistry on human brain tissue. The tissue sections analysed contained anterior and lateral periventricular white matter from the frontal and parietal lobes in post-mortem neonatal cases with a postmenstrual age range of 23.6-31.4 weeks. In addition to preterm brains without overt pathology (control), preterm pathology cases with evidence of white matter injuries (WMI) were also examined. In order to identify TLR-positive cells, we utilized standard double-labelling immunofluorescence co-labelling techniques and confocal microscopy to compare co-expression of TLR3 with a neuronal marker (NeuN) or with glial markers (GFAP for astrocytes, Iba-1 for microglia and Olig2 for oligodendrocytes). We observed an increase in the neuronal (28 vs. 17%) and astroglial (38 vs. 21%) populations in the WMI group compared to controls in the anterior regions of the periventricular white matter in the frontal lobe. The increase in neurons and astrocytes in the WMI cases was associated with an increase in TLR3 immunoreactivity. This expression was significantly increased in the astroglia. The morphology of the TLR3 signal in the control cases was globular and restricted to the perinuclear region of the neurons and astrocytes, whilst in the cases of WMI, both neuronal, axonal and astroglial TLR3 expression was more diffuse (i.e., a different intracellular distribution) and could be detected along the extensions of the processes. This study demonstrates for the first time that neurons and glial cells in human neonatal periventricular white matter express TLR3 during development. The patterns of TLR3 expression were altered in the presence of WMI, which might influence normal developmental processes within the immature brain. Identifying changes in TLR3 expression during fetal development may be key to understanding the reduced volumes of grey matter and impaired cortical development seen in preterm infants.


Assuntos
Encéfalo/metabolismo , Lactente Extremamente Prematuro/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Receptor 3 Toll-Like/biossíntese , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Lesões Encefálicas/metabolismo , Humanos , Lactente Extremamente Prematuro/crescimento & desenvolvimento , Recém-Nascido , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Receptor 3 Toll-Like/análise
7.
Nat Genet ; 17(1): 49-57, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9288097

RESUMO

The cerebro-hepato-renal syndrome of Zellweger is a fatal inherited disease caused by deficient import of peroxisomal matrix proteins. The pathogenic mechanisms leading to extreme hypotonia, severe mental retardation and early death are unknown. We generated a Zellweger animal model through inactivation of the murine Pxr1 gene (formally known as Pex5) that encodes the import receptor for most peroxisomal matrix proteins. Pxr1-/- mice lacked morphologically identifiable peroxisomes and exhibited the typical biochemical abnormalities of Zellweger patients. They displayed intrauterine growth retardation, were severely hypotonic at birth and died within 72 hours. Analysis of the neocortex revealed impaired neuronal migration and maturation and extensive apoptotic death of neurons.


Assuntos
Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/genética , Síndrome de Zellweger/genética , Animais , Animais Recém-Nascidos , Apoptose , Sequência de Bases , Encéfalo/metabolismo , Encéfalo/patologia , Córtex Cerebral/patologia , DNA/biossíntese , Primers do DNA , Morte , Modelos Animais de Doenças , Feminino , Retardo do Crescimento Fetal , Fibroblastos/metabolismo , Humanos , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Neurônios/patologia , Neurônios/fisiologia , Receptor 1 de Sinal de Orientação para Peroxissomos , Reação em Cadeia da Polimerase , Gravidez , Receptores Citoplasmáticos e Nucleares/metabolismo , Recombinação Genética , Síndrome de Zellweger/patologia , Síndrome de Zellweger/fisiopatologia
8.
Nat Genet ; 28(2): 131-8, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11381259

RESUMO

Hypoxia stimulates angiogenesis through the binding of hypoxia-inducible factors to the hypoxia-response element in the vascular endothelial growth factor (Vegf) promotor. Here, we report that deletion of the hypoxia-response element in the Vegf promotor reduced hypoxic Vegf expression in the spinal cord and caused adult-onset progressive motor neuron degeneration, reminiscent of amyotrophic lateral sclerosis. The neurodegeneration seemed to be due to reduced neural vascular perfusion. In addition, Vegf165 promoted survival of motor neurons during hypoxia through binding to Vegf receptor 2 and neuropilin 1. Acute ischemia is known to cause nonselective neuronal death. Our results indicate that chronic vascular insufficiency and, possibly, insufficient Vegf-dependent neuroprotection lead to the select degeneration of motor neurons.


Assuntos
Hipóxia Celular/genética , Fatores de Crescimento Endotelial/genética , Linfocinas/genética , Neurônios Motores/patologia , Degeneração Neural/genética , Elementos de Resposta/genética , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Axônios/fisiologia , Sítios de Ligação , Eletrofisiologia , Fatores de Crescimento Endotelial/metabolismo , Humanos , Linfocinas/metabolismo , Camundongos , Camundongos Knockout , Neurônios Motores/fisiologia , Contração Muscular , Fibras Musculares Esqueléticas/patologia , Atrofia Muscular/genética , Atrofia Muscular/patologia , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuropilina-1 , Nervos Periféricos/patologia , Regiões Promotoras Genéticas , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular , Deleção de Sequência , Medula Espinal/fisiologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
9.
Sci Rep ; 13(1): 2237, 2023 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-36754974

RESUMO

SARS-CoV-2 interacts with cellular cholesterol during many stages of its replication cycle. Pantethine was reported to reduce total cholesterol levels and fatty acid synthesis and potentially alter different processes that might be involved in the SARS-CoV-2 replication cycle. Here, we explored the potential antiviral effects of pantethine in two in vitro experimental models of SARS-CoV-2 infection, in Vero E6 cells and in Calu-3a cells. Pantethine reduced the infection of cells by SARS-CoV-2 in both preinfection and postinfection treatment regimens. Accordingly, cellular expression of the viral spike and nucleocapsid proteins was substantially reduced, and we observed a significant reduction in viral copy numbers in the supernatant of cells treated with pantethine. In addition, pantethine inhibited the infection-induced increase in TMPRSS2 and HECT E3 ligase expression in infected cells as well as the increase in antiviral interferon-beta response and inflammatory gene expression in Calu-3a cells. Our results demonstrate that pantethine, which is well tolerated in humans, was very effective in controlling SARS-CoV-2 infection and might represent a new therapeutic drug that can be repurposed for the prevention or treatment of COVID-19 and long COVID syndrome.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Chlorocebus aethiops , Humanos , Antivirais/farmacologia , Síndrome de COVID-19 Pós-Aguda , Replicação Viral , Células Vero
10.
Neuropsychopharmacology ; 46(3): 579-602, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32781459

RESUMO

Maternal immune activation (MIA) and poor maternal nutritional habits are risk factors for the occurrence of neurodevelopmental disorders (NDD). Human studies show the deleterious impact of prenatal inflammation and low n-3 polyunsaturated fatty acid (PUFA) intake on neurodevelopment with long-lasting consequences on behavior. However, the mechanisms linking maternal nutritional status to MIA are still unclear, despite their relevance to the etiology of NDD. We demonstrate here that low maternal n-3 PUFA intake worsens MIA-induced early gut dysfunction, including modification of gut microbiota composition and higher local inflammatory reactivity. These deficits correlate with alterations of microglia-neuron crosstalk pathways and have long-lasting effects, both at transcriptional and behavioral levels. This work highlights the perinatal period as a critical time window, especially regarding the role of the gut-brain axis in neurodevelopment, elucidating the link between MIA, poor nutritional habits, and NDD.


Assuntos
Ácidos Graxos Ômega-3 , Efeitos Tardios da Exposição Pré-Natal , Animais , Comportamento Animal , Encéfalo , Feminino , Humanos , Inflamação , Microglia , Gravidez
11.
Brain Behav Immun ; 24(5): 776-83, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19903519

RESUMO

Although the role of microglial activation in neural injury remains controversial, there is increasing evidence for a detrimental effect in the immature brain, which may occur in response to release of neurotoxic substances including pro-inflammatory cytokines. However, the signaling mechanisms involved in microglial-induced neuronal cell death are unclear. Microglia isolated from the brains of wild-type (WT) or MyD88 knockout (KO) mice were exposed to PBS or the TLR4-ligand LPS (100 ng/mL) for 2, 6, 14, or 24 h, and the microglia-conditioned medium (MCM) collected. Detection of multiple inflammatory molecules in MCM was performed using a mouse 22-plex cytokine microbead array kit. Primary neuronal cultures were supplemented with the 14 or 24 h MCM, and the degree of neuronal apoptosis examined after exposure for 24 h. Results showed a rapid and sustained elevation in multiple inflammatory mediators in the MCM of WT microglia exposed to LPS, which was largely inhibited in MyD88 KO microglia. There was a significant increase in apoptotic death measured at 24 h in cultured neurons exposed to CM from either 14 or 24 h LPS-stimulated WT microglia (p<.05 vs. WT control). By contrast, there was no increase in apoptotic death in cultured neurons exposed to CM from 14 or 24 h LPS-stimulated MyD88 KO microglia (p=.15 vs. MyD88 KO control). These data suggest that MyD88-dependent activation of microglia by LPS causes release of factors directly toxic to neurons.


Assuntos
Citocinas/metabolismo , Microglia/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Neurônios/metabolismo , Análise de Variância , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Encéfalo/imunologia , Encéfalo/metabolismo , Células Cultivadas , Técnicas de Cocultura , Citocinas/imunologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/imunologia , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
12.
Nat Commun ; 11(1): 6133, 2020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33257673

RESUMO

Omega-3 fatty acids (n-3 PUFAs) are essential for the functional maturation of the brain. Westernization of dietary habits in both developed and developing countries is accompanied by a progressive reduction in dietary intake of n-3 PUFAs. Low maternal intake of n-3 PUFAs has been linked to neurodevelopmental diseases in Humans. However, the n-3 PUFAs deficiency-mediated mechanisms affecting the development of the central nervous system are poorly understood. Active microglial engulfment of synapses regulates brain development. Impaired synaptic pruning is associated with several neurodevelopmental disorders. Here, we identify a molecular mechanism for detrimental effects of low maternal n-3 PUFA intake on hippocampal development in mice. Our results show that maternal dietary n-3 PUFA deficiency increases microglia-mediated phagocytosis of synaptic elements in the rodent developing hippocampus, partly through the activation of 12/15-lipoxygenase (LOX)/12-HETE signaling, altering neuronal morphology and affecting cognitive performance of the offspring. These findings provide a mechanistic insight into neurodevelopmental defects caused by maternal n-3 PUFAs dietary deficiency.


Assuntos
Encéfalo/efeitos dos fármacos , Ácidos Graxos Ômega-3/farmacologia , Microglia/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fagocitose/efeitos dos fármacos , Animais , Encéfalo/crescimento & desenvolvimento , Suplementos Nutricionais , Ácidos Graxos Ômega-3/deficiência , Ácidos Graxos Ômega-3/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Homeostase , Humanos , Lipoxigenase , Masculino , Camundongos , Transtornos do Neurodesenvolvimento
13.
Cell Death Differ ; 15(10): 1542-52, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18551134

RESUMO

In mammals, programmed cell death (PCD) is a central event during brain development. Trophic factors have been shown to prevent PCD in postmitotic neurons. Similarly, cytokines have neurotrophic effects involving regulation of neuronal survival. Nevertheless, neuronal PCD is only partially understood and host determinants are incompletely defined. The present study provides evidence that the cytokine interleukin-9 (IL-9) and its receptor specifically control PCD of neurons in the murine newborn neocortex. IL-9 antiapoptotic action appeared to be time-restricted to early postnatal stages as both ligand and receptor transcripts were mostly expressed in neocortex between postnatal days 0 and 10. This period corresponds to the physiological peak of apoptosis for postmitotic neurons in mouse neocortex. In vivo studies showed that IL-9/IL-9 receptor pathway inhibits apoptosis in the newborn neocortex. Furthermore, in vitro studies demonstrated that IL-9 and its receptor are mainly expressed in neurons. IL-9 effects were mediated by the activation of the JAK/STAT (janus kinase/signal transducer and activator of transcription) pathway, whereas nuclear factor-kappaB (NF-kappaB) or Erk pathways were not involved in mediating IL-9-induced inhibition of cell death. Finally, IL-9 reduced the expression of the mitochondrial pro-apoptotic factor Bax whereas Bcl-2 level was not significantly affected. Together, these data suggest that IL-9/IL-9 receptor signaling pathway represents a novel endogenous antiapoptotic mechanism for cortical neurons by controlling JAK/STAT and Bax levels.


Assuntos
Apoptose/fisiologia , Córtex Cerebral/citologia , Interleucina-9/metabolismo , Neurônios/fisiologia , Receptores de Interleucina-9/metabolismo , Transdução de Sinais/fisiologia , Animais , Animais Recém-Nascidos , Caspase 3/metabolismo , Células Cultivadas , Humanos , Janus Quinases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição STAT/metabolismo , Proteína X Associada a bcl-2/metabolismo
14.
Neuroscience ; 152(4): 859-66, 2008 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-18329816

RESUMO

Several studies using animal models have suggested that the effects of nutritional insult on the developing brain are long-lasting and lead to permanent deficits in learning and behavior. Malnutrition can refer to the availability of all the nutrients but in insufficient quantities or it may imply that one or more of essential nutrients is either missing or is present, but in the wrong proportions in the diet. The hypothesis addressed in this study is that different domains of cognitive functioning can be affected by malnutrition and this can be related to the type of nutritional deficiency that the brain has been exposed to during development. To study the effect of nutritional deprivation during brain development, a paradigm of maternal malnutrition during the period of gestation and lactation was used and its effects were studied on the F1 offspring using Swiss albino mice. Three different types of malnutrition were used, that involve, caloric restriction, inadequate amount of protein in the diet and condition of low iron content. Our results show that the domain of spatial memory affected in the F1 generation depended on the kind of malnutrition that the mother was subjected to. Further our study shows that although hippocampal volume was reduced in all F1 pups, hippocampal subregions of the F1 animals were differentially vulnerable depending on type of malnutrition that the mother was subjected to. These results highlight the importance of qualifying the kind of malnutrition that is suffered by the mother during the period of gestation and lactation as it has consequences for the cognitive domain affected in the offspring. Awareness of this should inform prevention strategies in trying to reverse the effects of adverse maternal nutrition during critical periods in brain development.


Assuntos
Aprendizagem em Labirinto , Memória/fisiologia , Distúrbios Nutricionais/classificação , Distúrbios Nutricionais/fisiopatologia , Comportamento Espacial/fisiologia , Animais , Comportamento Animal , Peso Corporal , Restrição Calórica/métodos , Feminino , Hipocampo/patologia , Deficiências de Ferro , Masculino , Camundongos , Distúrbios Nutricionais/etiologia , Distúrbios Nutricionais/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Deficiência de Proteína/complicações , Tempo de Reação
15.
Arch Pediatr ; 15(2): 157-61, 2008 Feb.
Artigo em Francês | MEDLINE | ID: mdl-18201881

RESUMO

The hypothermia treatment for neonatal hypoxic ischemic encephalopathy is a concept revisited for more than 10 years. With this strategy, animal studies have shown an 80% reduction of brain damage. Conditions for the practice of hypothermia, to obtain neuroprotection, have been described in these studies: rapidity of the onset of cooling after the hypoxic ischemic event, prolonged duration during several hours, ability to obtain neuroprotection with two methods of cooling, selective head cooling or whole body hypothermia. Pilot studies in human newborns have demonstrated the feasibility of these strategies without immediate adverse effects. Two large randomised trials have been conducted in 2005 to test the efficacy. Only with the strategy of whole body cooling, the incidence of poor outcome at 18 months (death or severe disability) was statistically decreased (44% versus 66% in the control group). This reduction seems especially significant in the sub group of intermediate severity (48% versus 66%), whereas severe forms (Grade III in the Sarnat and Sarnat classification) were probably not ameliorated with this treatment. Now, the major problem is to determine the best indications for hypothermia with an early and precise assessment of the grade of the encephalopathy.


Assuntos
Hipotermia Induzida/métodos , Hipóxia-Isquemia Encefálica/terapia , Fatores Etários , Animais , Protocolos Clínicos , Modelos Animais de Doenças , Eletroencefalografia , Estudos de Viabilidade , Seguimentos , Humanos , Hipotermia Induzida/efeitos adversos , Hipóxia-Isquemia Encefálica/diagnóstico , Hipóxia-Isquemia Encefálica/mortalidade , Recém-Nascido , Metanálise como Assunto , Razão de Chances , Projetos Piloto , Ensaios Clínicos Controlados Aleatórios como Assunto , Ratos , Índice de Gravidade de Doença , Ovinos/embriologia , Suínos , Fatores de Tempo , Resultado do Tratamento
16.
Arch Pediatr ; 15(10): 1568-72, 2008 Oct.
Artigo em Francês | MEDLINE | ID: mdl-18804971

RESUMO

In this article, we review the clinical, neuropathological and genetic aspects of distal spinal-muscular atrophy 1 (DSMA1; MIM#604320), formerly designated as autosomal recessive spinal muscular atrophy with respiratory distress type 1 (SMARD1) and also known as distal hereditary-motor neuropathy type 6 (dHMN6 or HMN6).


Assuntos
Atrofia Muscular Espinal/diagnóstico , Diagnóstico Diferencial , Feminino , Humanos , Recém-Nascido , Atrofia Muscular Espinal/genética , Gravidez , Diagnóstico Pré-Natal , Síndrome do Desconforto Respiratório do Recém-Nascido/etiologia
17.
J Clin Invest ; 94(5): 2020-7, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7962548

RESUMO

Vasoactive intestinal peptide (VIP) has potent growth-related actions that influence cell mitosis, neuronal survival, and neurodifferentiation in cell culture. VIP can also produce dramatic growth in postimplantation mouse embryos in vitro, characterized by large increases in cell number. The goal of the present study was to assess the role of VIP on early nervous system development in vivo. Pregnant mice were treated with a specific antagonist to VIP. Prenatal administration of the antagonist early in development (E9-E11) produced severe microcephaly characterized by decreased embryonic brain weight with reduced DNA and protein content. The retardation of growth was disproportionally manifested in the brain compared with the body and was prevented by co-treatment with VIP. Identical treatment with the antagonist later in gestation had no detectable effect on embryonic growth. VIP receptors, which were restricted to the central nervous system during this stage of embryonic development, were increased in the neuroepithelium of antagonist-treated embryos while the number of cells in S-phase was significantly decreased. Thus, VIP regulates brain growth in vivo and inhibition of its action provides new insight into a molecular mechanism for microcephaly.


Assuntos
Microcefalia/etiologia , Peptídeo Intestinal Vasoativo/fisiologia , Animais , Sítios de Ligação , Encéfalo/embriologia , Encéfalo/patologia , Feminino , Camundongos , Neuropeptídeos/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Gravidez , Receptores de Peptídeo Intestinal Vasoativo/análise , Peptídeo Intestinal Vasoativo/antagonistas & inibidores
18.
J Clin Invest ; 100(2): 390-7, 1997 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-9218516

RESUMO

Excitotoxic damage may be a critical factor in the formation of brain lesions associated with cerebral palsy. When injected at birth, the glutamatergic analog ibotenate induces mouse brain lesions that strikingly mimic human microgyria. When ibotenate is injected at postnatal day 5, it produces transcortical necrosis and white matter cysts that mimic human perinatal hypoxic-like lesions. Vasoactive intestinal peptide (VIP) has potent growth-related actions and neuroprotective properties that influence mitosis and neuronal survival in culture. The goal of this study was to assess the protective role of VIP against excitotoxic lesions induced by ibotenate in developing mouse brain. VIP cotreatment reduced ibotenate-induced microgyric-like cortical lesions and white matter cysts by up to 77 and 85%, respectively. VIP protective effects were reproduced by a peptide derived from activity-dependent neurotrophic factor (ADNF), a trophic factor released by VIP-stimulated astrocytes, and by stearyl norleucine VIP, a specific VIP agonist that does not activate adenylate cyclase. Neither forskolin, an adenylate cyclase activator, nor pituitary adenylate cyclase-activating peptide, provided VIP-like protection. VIP and neurotrophic analogs, acting through a cAMP-independent mechanism and inducing ADNF release, could represent new avenues in the understanding and prevention of human cerebral palsy.


Assuntos
Encéfalo/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Ácido Ibotênico/farmacologia , Neurônios/efeitos dos fármacos , Peptídeo Intestinal Vasoativo/farmacologia , Animais , Sítios de Ligação , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Morte Celular , Córtex Cerebral/metabolismo , Colforsina/farmacologia , Histocitoquímica , Ácido Ibotênico/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/farmacologia , Neurônios/citologia , Neuropeptídeos/farmacologia , Fármacos Neuroprotetores/farmacologia , Oligopeptídeos , Fragmentos de Peptídeos/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Transdução de Sinais/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo
19.
J Clin Invest ; 107(4): 457-66, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11181645

RESUMO

Intracerebral administration of the excitotoxin ibotenate to newborn mice induces white-matter lesions, mimicking brain lesions that occur in human preterm infants. Nociceptin (NC), also called orphanin FQ, is the endogenous ligand of the opioid receptor-like 1 (ORL1) receptor and does not bind classical high-affinity opioid receptors. In the present study, administration of NC exacerbated ibotenate-induced white-matter lesions while coadministration of ibotenate with either of two NC antagonists reduced excitotoxic white-matter lesions by up to 64%. Neither ibotenate plus endomorphin I (a selective mu receptor agonist), nor ibotenate plus naloxone (a classical opioid receptor antagonist) modulated the excitotoxic lesion. Pretreatment with antisense oligonucleotides targeting the NC precursor peptide mRNA significantly reduced ibotenate-induced white-matter damage. Finally, high doses of fentanyl, which stimulates both classical mu-opioid receptors and ORL1, exacerbated excitotoxic white-matter lesion. This toxic effect was blocked by inhibiting ORL1 but not classical opioid receptors. Together, these findings show that endogenous or exogenous stimulation of the ORL1 receptor can be neurotoxic and that blocking NC signaling protects the white matter against excitotoxic challenge. These data point to potential new avenues for neuroprotection in human preterm infants at high risk of brain lesions.


Assuntos
Encéfalo/efeitos dos fármacos , Ácido Ibotênico/toxicidade , Peptídeos Opioides/toxicidade , Animais , Animais Recém-Nascidos , Encéfalo/patologia , Fentanila/farmacologia , Humanos , Recém-Nascido , Leucomalácia Periventricular/etiologia , Camundongos , Naloxona/farmacologia , Oligonucleotídeos Antissenso/metabolismo , Oligonucleotídeos Antissenso/uso terapêutico , Receptores de N-Metil-D-Aspartato/fisiologia , Nociceptina
20.
Neuroscience ; 150(3): 712-9, 2007 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-17950543

RESUMO

Perinatal brain injuries often result in irreversible learning disabilities, which manifest in early childhood. These injuries are chiefly ascribable to marked susceptibility of the immature brain to glutamate-induced excitotoxicity. No treatments are available. One well-characterized model of perinatal brain injuries consists in injecting the glutamate analog ibotenate into the brain of 5-day-old mice. The resulting excitotoxic lesions resemble the hypoxic-ischemic gray-matter lesions seen in full-term and near-term newborns, as well as the white-matter lesions of preterm newborns. We previously reported that these lesions disrupted odor preference conditioning in newborn mice. The aim of this study was to assess the effectiveness of the neuroprotector melatonin in preventing learning disabilities in newborn mice with ibotenate-induced brain injury. In postnatal day (P) 6-P7 pups, we tested psychomotor reflexes, spontaneous preference for maternal odors as an index of memory, ultrasonic vocalization responses to stroking as an index of sensitivity to tactile stimuli, and conditioned preference for an odor previously paired with stroking as an index of learning abilities. Without melatonin, conditioning was abolished, whereas spontaneous odor preference, psychomotor reflexes, and sensitivity to tactile stimuli were normal. Thus, abolition of conditioning was not associated with sensorimotor impairments. Histological analysis confirmed the efficacy of melatonin in reducing white-matter lesions induced by ibotenate. Furthermore, treatment with melatonin protected the ability to develop conditioning. Thus, melatonin, which easily crosses the blood-brain barrier and has been proven safe in children, may be effective in preventing learning disabilities caused by perinatal brain injuries in human preterm infants.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Deficiências da Aprendizagem/prevenção & controle , Melatonina/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Animais Recém-Nascidos , Condicionamento Psicológico/efeitos dos fármacos , Modelos Animais de Doenças , Epilepsia Tônico-Clônica/induzido quimicamente , Agonistas de Aminoácidos Excitatórios , Feminino , Ácido Ibotênico , Deficiências da Aprendizagem/induzido quimicamente , Camundongos , Atividade Motora/efeitos dos fármacos , Odorantes , Estimulação Física , Tato , Vocalização Animal/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA