Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Métodos Terapêuticos e Terapias MTCI
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunol Res ; 64(4): 1013-24, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26783030

RESUMO

The Caenorhabditis elegans model can be used to study Candida albicans virulence and host immunity, as well as to identify plant-derived natural products to use against C. albicans. Thymol is a hydrophobic phenol compound from the aromatic plant thyme. In this study, the in vitro data demonstrated concentration-dependent thymol inhibition of both C. albicans growth and biofilm formation during different developmental phases. With the aid of the C. elegans system, we performed in vivo assays, and our results further showed the ability of thymol to increase C. elegans life span during infection, inhibit C. albicans colony formation in the C. elegans intestine, and increase the expression levels of host antimicrobial genes. Moreover, among the genes that encode the p38 MAPK signaling pathway, mutation of the pmk-1 or sek-1 gene decreased the beneficial effects of thymol's antifungal activity against C. albicans and thymol's maintenance of the innate immune response in nematodes. Western blot data showed the level of phosphorylation of pmk-1 was dramatically decreased against C. albicans. In nematodes, treatment with thymol recovered the dysregulation of pmk-1 and sek-1 gene expressions, the phosphorylation level of PMK-1 caused by C. albicans infection. Therefore, thymol may act, at least in part, through the function of the p38 MAPK signaling pathway to protect against C. albicans infection and maintain the host innate immune response to C. albicans. Our results indicate that the p38 MAPK signaling pathway plays a crucial role in regulating the beneficial effects observed after nematodes infected with C. albicans were treated with thymol.


Assuntos
Antifúngicos/uso terapêutico , Proteínas de Caenorhabditis elegans/metabolismo , Candida albicans/imunologia , Candidíase/tratamento farmacológico , Intestinos/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptor EphA4/metabolismo , Timol/uso terapêutico , Animais , Biofilmes/efeitos dos fármacos , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Candidíase/imunologia , Processos de Crescimento Celular/efeitos dos fármacos , Imunidade Inata , Intestinos/imunologia , Intestinos/microbiologia , Proteínas Quinases Ativadas por Mitógeno/genética , Mutação/genética , Fosforilação , Receptor EphA4/genética , Transdução de Sinais , Thymus (Planta)/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
Sleep ; 39(3): 613-24, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26612390

RESUMO

STUDY OBJECTIVES: Optimal sleep is ensured by the interaction of circadian and homeostatic processes. Although synaptic plasticity seems to contribute to both processes, the specific players involved are not well understood. The EphA4 tyrosine kinase receptor is a cell adhesion protein regulating synaptic plasticity. We investigated the role of EphA4 in sleep regulation using electrocorticography in mice lacking EphA4 and gene expression measurements. METHODS: EphA4 knockout (KO) mice, Clock(Δ19/Δ19) mutant mice and littermates, C57BL/6J and CD-1 mice, and Sprague-Dawley rats were studied under a 12 h light: 12 h dark cycle, under undisturbed conditions or 6 h sleep deprivation (SLD), and submitted to a 48 h electrophysiological recording and/or brain sampling at different time of day. RESULTS: EphA4 KO mice showed less rapid eye movement sleep (REMS), enhanced duration of individual bouts of wakefulness and nonrapid eye movement sleep (NREMS) during the light period, and a blunted daily rhythm of NREMS sigma activity. The NREMS delta activity response to SLD was unchanged in EphA4 KO mice. However, SLD increased EphA4 expression in the thalamic/hypothalamic region in C57BL/6J mice. We further show the presence of E-boxes in the promoter region of EphA4, a lower expression of EphA4 in Clock mutant mice, a rhythmic expression of EphA4 ligands in several brain areas, expression of EphA4 in the suprachiasmatic nuclei of the hypothalamus (SCN), and finally an unchanged number of cells expressing Vip, Grp and Avp in the SCN of EphA4 KO mice. CONCLUSIONS: Our results suggest that EphA4 is involved in circadian sleep regulation.


Assuntos
Ritmo Circadiano/fisiologia , Receptor EphA4/metabolismo , Privação do Sono/fisiopatologia , Sono/fisiologia , Animais , Proteínas CLOCK/genética , Ritmo Circadiano/genética , Escuridão , Eletrocorticografia , Fenômenos Eletrofisiológicos , Homeostase , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasticidade Neuronal , Regiões Promotoras Genéticas/genética , Ratos , Ratos Sprague-Dawley , Receptor EphA4/biossíntese , Receptor EphA4/deficiência , Receptor EphA4/genética , Sono/genética , Privação do Sono/genética , Sono REM/genética , Sono REM/fisiologia , Núcleo Supraquiasmático/metabolismo , Tálamo/metabolismo , Fatores de Tempo , Vigília/genética , Vigília/fisiologia
3.
J Comp Neurol ; 524(10): 2080-92, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-26587807

RESUMO

Deciphering the molecular basis for guiding specific aspects of neocortical development remains a challenge because of the complexity of histogenic events and the vast array of protein interactions mediating these events. The Eph family of receptor tyrosine kinases is implicated in a number of neurodevelopmental activities. Eph receptors have been known to be capable of responding to several ephrin ligands within their subgroups, often eliciting similar downstream effects. However, several recent studies have indicated specificity between receptor-ligand pairs within each subfamily, the functional relevance of which is not defined. Here we show that a receptor of the EphA subfamily, EphA4, has effects distinct from those of its close relative, EphA7, in the developing brain. Both EphA4 and EphA7 interact similarly with corresponding ligands expressed in the developing neocortex. However, only EphA7 shows strong interaction with ligands in the somatosensory thalamic nuclei; EphA4 affects only cortical neuronal migration, with no visible effects on the guidance of corticothalamic (CT) axons, whereas EphA7 affects both cortical neuronal migration and CT axon guidance. Our data provide new evidence that Eph receptors in the same subfamily are not simply interchangeable but are functionally specified through selective interactions with distinct ligands in vivo. J. Comp. Neurol. 524:2080-2092, 2016. © 2015 Wiley Periodicals, Inc.


Assuntos
Córtex Cerebral , Vias Neurais/fisiologia , Receptor EphA4/metabolismo , Receptor EphA7/metabolismo , Tálamo , Animais , Animais Recém-Nascidos , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Receptor EphA4/genética , Receptor EphA7/genética , Tálamo/citologia , Tálamo/embriologia , Tálamo/crescimento & desenvolvimento , Tálamo/metabolismo
4.
Physiol Behav ; 135: 232-6, 2014 08.
Artigo em Inglês | MEDLINE | ID: mdl-24949848

RESUMO

Eph-ephrin signaling is known to be important in directing topographic projections in the afferent auditory pathway, including connections to various subdivisions of the inferior colliculus (IC). The acoustic startle-response (ASR) is a reliable reflexive behavioral response in mammals elicited by an unexpected intense acoustic startle-eliciting stimulus (ES). It is mediated by a sub-cortical pathway that includes the IC. The ASR amplitude can be measured with an accelerometer under the subject and can be decreased in amplitude by presenting a less intense, non-startling stimulus 5-300ms before the ES. This reflexive decrement in ASR is called pre-pulse inhibition (PPI) and indicates that the relatively soft pre-pulse was heard. PPI is a general trait among mammals. Mice have been used recently to study this response and to reveal how genetic mutations affect neural circuits and hence the ASR and PPI. In this experiment, we measured the effect of Eph-ephrin mutations using control mice (C57BL/6J), mice with compromised EphA4 signaling (EphA4(lacZ/+), EphA4(lacZ/lacZ)), and knockout ephrin-B3 mice (ephrin-B3 (+/-, -/-)). Control and EphA4(lacZ/+s)trains showed robust PPI (up to 75% decrement in ASR) to an offset of a 70dB SPL background noise at 50ms before the ES. Ephrin-B3 knockout mice and EphA4 homozygous mutants were only marginally significant in PPI (<25% decrement and <33% decrement, respectively) to the same conditions. This decrement in PPI highlights the importance of ephrin-B3 and EphA4 interactions in ordering auditory behavioral circuits. Thus, different mutations in certain members of the signaling family produce a full range of changes in PPI, from minimal to nearly maximal. This technique can be easily adapted to study other aspects of hearing in a wider range of mutations. Along with ongoing neuroanatomical studies, this allows careful quantification of how the auditory anatomical, physiological and now behavioral phenotype is affected by changes in Eph-ephrin expression and functionality.


Assuntos
Efrina-B3/genética , Inibição Pré-Pulso/genética , Receptor EphA4/genética , Reflexo de Sobressalto/genética , Estimulação Acústica , Animais , Efrina-B3/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor EphA4/metabolismo , Transdução de Sinais/genética
5.
PLoS Genet ; 8(4): e1002638, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22511881

RESUMO

The mechanisms of hypoxic injury to the developing human brain are poorly understood, despite being a major cause of chronic neurodevelopmental impairments. Recent work in the invertebrate Caenorhabditis elegans has shown that hypoxia causes discrete axon pathfinding errors in certain interneurons and motorneurons. However, it is unknown whether developmental hypoxia would have similar effects in a vertebrate nervous system. We have found that developmental hypoxic injury disrupts pathfinding of forebrain neurons in zebrafish (Danio rerio), leading to errors in which commissural axons fail to cross the midline. The pathfinding defects result from activation of the hypoxia-inducible transcription factor (hif1) pathway and are mimicked by chemical inducers of the hif1 pathway or by expression of constitutively active hif1α. Further, we found that blocking transcriptional activation by hif1α helped prevent the guidance defects. We identified ephrinB2a as a target of hif1 pathway activation, showed that knock-down of ephrinB2a rescued the guidance errors, and showed that the receptor ephA4a is expressed in a pattern complementary to the misrouting axons. By targeting a constitutively active form of ephrinB2a to specific neurons, we found that ephrinB2a mediates the pathfinding errors via a reverse-signaling mechanism. Finally, magnesium sulfate, used to improve neurodevelopmental outcomes in preterm births, protects against pathfinding errors by preventing upregulation of ephrinB2a. These results demonstrate that evolutionarily conserved genetic pathways regulate connectivity changes in the CNS in response to hypoxia, and they support a potential neuroprotective role for magnesium.


Assuntos
Efrina-B2/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia , Hipóxia , Sulfato de Magnésio/farmacologia , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Axônios/metabolismo , Axônios/fisiologia , Sistema Nervoso Central/metabolismo , Efrina-B2/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Hipóxia/metabolismo , Hipóxia/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neurônios/patologia , Receptor EphA4/genética , Receptor EphA4/metabolismo , Transdução de Sinais , Ativação Transcricional , Peixe-Zebra/genética , Peixe-Zebra/fisiologia
6.
J Comp Neurol ; 505(6): 669-81, 2007 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-17948875

RESUMO

Auditory processing requires proper formation of tonotopically ordered projections. We have evaluated the role of an Eph receptor tyrosine kinase and an ephrin ligand in the development of these frequency maps. We demonstrated expression of EphA4 and ephrin-B2 in auditory nuclei and found expression gradients along the frequency axis in neonates. We tested the roles of EphA4 and ephrin-B2 in development of auditory projections by evaluating whether mutations result in altered patterns of expression of the immediate early gene c-fos after exposure to pure tone stimuli. We evaluated two nuclei, the dorsal cochlear nucleus (DCN) and the medial nucleus of the trapezoid body (MNTB), which project in two distinct auditory pathways. The mean number of c-fos-positive neurons in EphA4(-/-) DCN after 8-kHz pure tone stimulation was 42% lower than in wild-type DCN. Along the dorsoventral, tonotopic axis of DCN, the mean position of c-fos-positive neurons was similar for mutant and wild-type mice, but the spread of these neurons along the tonotopic axis was 35% greater for ephrin-B2(lacZ/+) mice than for wild-type mice. We also examined these parameters in MNTB after exposure to 40-kHz pure tones. Both EphA4(-/-) and ephrin-B2(lacZ/+) mice had significantly fewer c-fos-positive cells than wild-type littermates. The labeled band of cells was narrower and laterally shifted in EphA4(-/-) mice compared with wild-type mice. These differences in cell number and distribution suggest that EphA4 and ephrin-B2 signaling influence auditory activation patterns.


Assuntos
Vias Auditivas/anormalidades , Padronização Corporal/genética , Tronco Encefálico/anormalidades , Efrina-B2/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Receptor EphA4/genética , Estimulação Acústica , Animais , Vias Auditivas/citologia , Vias Auditivas/metabolismo , Percepção Auditiva/genética , Tronco Encefálico/citologia , Tronco Encefálico/metabolismo , Contagem de Células , Núcleo Coclear/anormalidades , Núcleo Coclear/citologia , Núcleo Coclear/metabolismo , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Núcleo Olivar/anormalidades , Núcleo Olivar/citologia , Núcleo Olivar/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética
7.
Proc Natl Acad Sci U S A ; 103(42): 15629-34, 2006 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-17030822

RESUMO

Fine movement in the body is controlled by the motor cortex, which signals in a topographically specific manner to neurons in the spinal cord by means of the corticospinal tract (CST). How the correct topography of the CST is established is unknown. To investigate the possibility that the Eph tyrosine kinase receptor EphA4 is involved in this process, we have traced CST axons in mice in which the EphA4 gene has been deleted. The forelimb subpopulation of CST axons is unaffected in the EphA4-/- mice, but the hindlimb subpopulation branches too early within the cord, both temporally and spatially. EphA4 shows a dynamic expression pattern in the environment of the developing CST in the spinal cord: high at the time of forelimb branching and down-regulated before hindlimb branching. To examine whether the fore- and hindlimb subpopulations of CST axons respond differently to EphA4 in their environment, neurons from fore- and hindlimb motor cortex were cultured on a substrate containing EphA4. Neurons from the hindlimb cortex showed reduced branching on the EphA4 substrate compared with their forelimb counterparts. Neurons from the hindlimb cortex express ephrinA5, a high-affinity ligand for EphA4, at higher levels compared with forelimb cortex neurons, and this expression is down-regulated before hindlimb branching. Together, these findings suggest that EphA4 regulates topographic mapping of the CST by controlling the branching of CST axons in the spinal cord.


Assuntos
Neurônios , Tratos Piramidais , Receptor EphA4/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Membro Anterior/inervação , Membro Posterior/inervação , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Córtex Motor/citologia , Córtex Motor/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Tratos Piramidais/anatomia & histologia , Tratos Piramidais/crescimento & desenvolvimento , Tratos Piramidais/metabolismo , Receptor EphA4/genética , Medula Espinal/anatomia & histologia , Medula Espinal/crescimento & desenvolvimento , Medula Espinal/metabolismo
8.
Neuron ; 39(3): 453-65, 2003 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-12895420

RESUMO

The mechanisms generating precise connections between specific thalamic nuclei and cortical areas remain poorly understood. Using axon tracing analysis of ephrin/Eph mutant mice, we provide in vivo evidence that Eph receptors in the thalamus and ephrins in the cortex control intra-areal topographic mapping of thalamocortical (TC) axons. In addition, we show that the same ephrin/Eph genes unexpectedly control the inter-areal specificity of TC projections through the early topographic sorting of TC axons in an intermediate target, the ventral telencephalon. Our results constitute the first identification of guidance cues involved in inter-areal specificity of TC projections and demonstrate that the same set of mapping labels is used differentially for the generation of topographic specificity of TC projections between and within individual cortical areas.


Assuntos
Córtex Cerebral/metabolismo , Efrina-A4/genética , Efrina-A5/genética , Receptor EphA4/genética , Receptor EphA5/genética , Tálamo/metabolismo , Animais , Mapeamento Encefálico/métodos , Córtex Cerebral/embriologia , Córtex Cerebral/enzimologia , Efrina-A4/biossíntese , Efrina-A4/fisiologia , Efrina-A5/biossíntese , Efrina-A5/fisiologia , Feminino , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vias Neurais/embriologia , Vias Neurais/enzimologia , Vias Neurais/metabolismo , Vias Neurais/fisiologia , Receptor EphA4/biossíntese , Receptor EphA4/fisiologia , Receptor EphA5/biossíntese , Receptor EphA5/fisiologia , Tálamo/embriologia , Tálamo/enzimologia
9.
Eur J Neurosci ; 16(6): 1168-72, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12383247

RESUMO

The role was studied of ephrin-B3, a ligand of the Eph family of tyrosine kinase receptors, in the formation of cortical connectivity. In situ hybridization and immunohistochemistry showed that EphA4, a receptor of ephrin-B3, was expressed in the lateral thalamus (visual and somaotosensory thalamus) of the developing rat brain, but not in the medial thalamic nuclei which project to the limbic cortex. Correspondingly, ephrin-B3 was expressed strongly in the developing limbic cortex including amygdala, entorhinal cortex and hippocampus. To examine the action of ephrin-B3 on thalamic axons, either lateral or medial thalamic explants were cultured on membranes obtained from ephrin-B3-expressing COS cells. Axonal growth was inhibited for cells from the lateral thalamus but not from the medial thalamus. These results suggest that ephrin-B3 contributes to regional specificity by suppressing axonal growth of lateral thalamic neurons.


Assuntos
Axônios/metabolismo , Córtex Cerebral/metabolismo , Efrina-B3/genética , Vias Neurais/metabolismo , Receptor EphA4/genética , Tálamo/metabolismo , Animais , Axônios/ultraestrutura , Células COS , Comunicação Celular/genética , Diferenciação Celular/genética , Membrana Celular/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Técnicas de Cocultura , Feto , Regulação da Expressão Gênica no Desenvolvimento/genética , Vias Neurais/citologia , Vias Neurais/embriologia , Técnicas de Cultura de Órgãos , RNA Mensageiro/metabolismo , Ratos , Tálamo/citologia , Tálamo/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA