Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cereb Cortex ; 29(4): 1644-1658, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29912395

RESUMO

The delicate balance of excitation and inhibition is crucial for proper function of the cerebral cortex, relying on the accurate number and subtype composition of inhibitory gamma-aminobutyric (GABA)-expressing interneurons. Various intrinsic and extrinsic factors precisely orchestrate their multifaceted development including the long-range migration from the basal telencephalon to cortical targets as well as interneuron survival throughout the developmental period. Particularly expressed guidance receptors were described to channel the migration of cortical interneurons deriving from the medial ganglionic eminence (MGE) and the preoptic area (POA) along distinct routes. Hence, unveiling the regulatory genetic networks controlling subtype-specific gene expression profiles is key to understand interneuron-specific developmental programs and to reveal causes for associated disorders. In contrast to MGE-derived interneurons, little is known about the transcriptional networks in interneurons born in the POA. Here, we provide first evidence for the LIM-homeobox transcription factor LHX1 as a crucial key player in the post-mitotic development of POA-derived cortical interneurons. By transcriptional regulation of related genes, LHX1 modulates their survival as well as the subtype-specific expression of guidance receptors of the Eph/ephrin family, thereby affecting directional migration and layer distribution in the adult cortex.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Interneurônios/fisiologia , Proteínas com Homeodomínio LIM/fisiologia , Área Pré-Óptica/crescimento & desenvolvimento , Fatores de Transcrição/fisiologia , Animais , Movimento Celular , Sobrevivência Celular , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Efrina-B3/genética , Efrina-B3/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Interneurônios/citologia , Interneurônios/metabolismo , Proteínas com Homeodomínio LIM/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Área Pré-Óptica/citologia , Área Pré-Óptica/metabolismo , Receptor EphA4/genética , Receptor EphA4/fisiologia , Fatores de Transcrição/genética
2.
Inflammation ; 42(2): 572-585, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30361852

RESUMO

Accumulating evidence indicates that post-injury inflammation characterized by activated microglia contributes much to the neuropathology of ischemic injury. Several studies have demonstrated that microglia exhibit two entirely different functional activation states, referred to as classically activated (M1) and alternatively activated (M2) phenotype. Promoting microglial phenotype to switch from M1 dominant to M2 dominant might be a promising approach for handling ischemic injury. However, the comprehensive mechanism that underlines microglia polarization in ischemic brain remains unclear. Neuronal erythropoietin-producing human hepatocellular carcinoma cell receptor 4 (EphA4), the richest Eph receptor in the central nervous system (CNS), upregulate after ischemia and may have the potential to regulate microglia activation. We hypothesized that modulating EphA4/ephrin signaling could affect ischemic injury through controlling microglia polarization. We therefore knocked down neuronal EphA4 with short hairpin RNA (shRNA) and determined the role of EphA4/ephrin signaling in oxygen-glucose deprivation and reperfusion (OGD/R)-induced injury. We found that EphA4 shRNA treatment attenuated OGD/R-induced apoptosis and microglia proliferation. Neuronal EphA4 knockdown also promoted microglial M2 polarization, which reduced pro-inflammatory mediators and released anti-inflammatory cytokines as well as neurotrophic factors. We further revealed that EphA4 shRNA treatment functioned through RhoA/Rho-associated kinase 2 (ROCK2) signaling, a key mediator of microglia alternative activation. Together, these data suggested that blockage of EphA4/ephrin signaling between neuron and microglia decreased OGD/R-induced injury by promoting alternative activation of microglia via RhoA/ROCK2 signaling.


Assuntos
Apoptose , Microglia/metabolismo , Receptor EphA4/fisiologia , Traumatismo por Reperfusão/patologia , Células Cultivadas , Humanos , Fenótipo , Traumatismo por Reperfusão/etiologia , Transdução de Sinais , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
3.
Genes Brain Behav ; 17(1): 82-92, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28425198

RESUMO

Circadian (∼24 h) rhythms of cellular network plasticity in the central circadian clock, the suprachiasmatic nucleus (SCN), have been described. The neuronal network in the SCN regulates photic resetting of the circadian clock as well as stability of the circadian system during both entrained and constant conditions. EphA4, a cell adhesion molecule regulating synaptic plasticity by controlling connections of neurons and astrocytes, is expressed in the SCN. To address whether EphA4 plays a role in circadian photoreception and influences the neuronal network of the SCN, we have analyzed circadian wheel-running behavior of EphA4 knockout (EphA4-/- ) mice under different light conditions and upon photic resetting, as well as their light-induced protein response in the SCN. EphA4-/- mice exhibited reduced wheel-running activity, longer endogenous periods under constant darkness and shorter periods under constant light conditions, suggesting an effect of EphA4 on SCN function. Moreover, EphA4-/- mice exhibited suppressed phase delays of their wheel-running activity following a light pulse during the beginning of the subjective night (CT15). Accordingly, light-induced c-FOS (FBJ murine osteosarcoma viral oncogene homolog) expression was diminished. Our results suggest a circadian role for EphA4 in the SCN neuronal network, affecting the circadian system and contributing to the circadian response to light.


Assuntos
Relógios Circadianos/fisiologia , Receptor EphA4/fisiologia , Núcleo Supraquiasmático/fisiologia , Animais , Adesão Celular/fisiologia , Relógios Circadianos/genética , Ritmo Circadiano/fisiologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/fisiologia , Neurônios/metabolismo , Estimulação Luminosa , Condicionamento Físico Animal , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptor EphA4/genética , Receptor EphA4/metabolismo , Núcleo Supraquiasmático/metabolismo
4.
Transl Res ; 192: 1-14, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29144959

RESUMO

Innervation is a fundamental basis for function and survival of tissues. In the peripheral tissues, degenerative diseases create a neurotoxic metabolic milieu that either causes neurodegeneration or fails to sustain regenerative growth and reinnervation of injured/diseased tissues. Encapsulation of cells producing neurotrophic factors can augment axon growth and neuron survival; however, sustained innervation in vivo requires a combination of factors promoting axon growth and guidance pathway that are released in a tissue-specific context. Using novel encapsulation techniques and genetic tools, we manipulated retinoic acid-generating enzyme aldehyde dehydrogenase 1a1 (Aldh1a1) in adipocytes that are capable of promoting growth and innervation of white adipose tissue by sympathetic neurons. Aldh1a1-/- adipocytes secrete molecules that regulate axon guidance and markedly stimulate neurite outgrowth in vitro and in vivo. Based on studies with natural and synthetic RAR agonists and antagonists, gene microarray and nanostring arrays, we concluded that ephrin A5/ephrin A4 is a downstream pathway regulated by Aldh1a1. Encapsulation of Aldh1a1-/- adipocytes into alginate poly-L-lysine microcapsules induced functional innervation of adipose tissue in obese wild-type mice. We propose that encapsulated Aldh1a1-/- adipocytes could provide a therapeutic solution for the reinnervation of damaged tissues.


Assuntos
Adipócitos/fisiologia , Tecido Adiposo Branco/inervação , Aldeído Desidrogenase/fisiologia , Sistema Nervoso Simpático/fisiologia , Vitamina A/metabolismo , Células 3T3-L1 , Aldeído Desidrogenase/genética , Família Aldeído Desidrogenase 1 , Animais , Axônios/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neuritos/fisiologia , Receptor EphA4/fisiologia , Retinal Desidrogenase
5.
Development ; 144(2): 321-333, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27993984

RESUMO

The signal-induced proliferation-associated family of proteins comprises four members, SIPA1 and SIPA1L1-3. Mutations of the human SIPA1L3 gene result in congenital cataracts. In Xenopus, loss of Sipa1l3 function led to a severe eye phenotype that was distinguished by smaller eyes and lenses including lens fiber cell maturation defects. We found a direct interaction between Sipa1l3 and Epha4, building a functional platform for proper ocular development. Epha4 deficiency phenocopied loss of Sipa1l3 and rescue experiments demonstrated that Epha4 acts upstream of Sipa1l3 during eye development, with both Sipa1l3 and Epha4 required for early eye specification. The ocular phenotype, upon loss of either Epha4 or Sipa1l3, was partially mediated by rax We demonstrate that canonical Wnt signaling is inhibited downstream of Epha4 and Sipa1l3 during normal eye development. Depletion of either Sipa1l3 or Epha4 resulted in an upregulation of axin2 expression, a direct Wnt/ß-catenin target gene. In line with this, Sipa1l3 or Epha4 depletion could be rescued by blocking Wnt/ß-catenin or activating non-canonical Wnt signaling. We therefore conclude that this pathomechanism prevents proper eye development and maturation of lens fiber cells, resulting in congenital cataracts.


Assuntos
Olho/embriologia , Proteínas Ativadoras de GTPase/fisiologia , Cristalino/embriologia , Cristalino/crescimento & desenvolvimento , Receptor EphA4/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Animais Geneticamente Modificados , Catarata/genética , Diferenciação Celular/genética , Embrião não Mamífero , Olho/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Cristalino/metabolismo , Organogênese/genética , Ligação Proteica , Receptor EphA4/metabolismo , Xenopus/embriologia , Xenopus/genética
6.
PLoS One ; 11(7): e0159930, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27467069

RESUMO

Leptomeningeal anastomoses play a critical role in regulating vascular re-perfusion following obstruction, however, the mechanisms regulating their development remains under investingation. Our current findings indicate that EphA4 receptor is a novel negative regulator of collaterogenesis. We demonstrate that EphA4 is highly expressed on pial arteriole collaterals at post-natal day (P) 1 and 7, then significantly reduced by P21. Endothelial cell (EC)-specific loss of EphA4, EphA4f/f/Tie2::Cre (KO), resulted in an increase in the density but not diameter of pial collaterals compared to WT mice. ECs isolated from KO mice displayed a 3-fold increase in proliferation, enhanced migration, tube formation and elevated levels of phospho(p)-Akt compared to WT ECs. Attenuating p-Akt, using LY294002, reduced the proliferative and migration effects in the KO ECs. RNAseq analysis also revealed altered expression patterns for genes that regulate cell proliferation, vascular development, extracellular matrix and immune-mediate responses, namely MCP-1, MMP2 and angiopoietin-1. Lastly, we show that induction of hindlimb ischemia resulted in accelerated re-perfusion, collateral remodeling and reduced tissue necrosis in the absence of EC-specific EphA4 compared to WT mice. These findings demonstrate a novel role for EphA4 in the early development of the pial collateral network and suggests a role in regulating vascular remodeling after obstruction.


Assuntos
Veias Cerebrais/fisiopatologia , Endotélio Vascular/fisiopatologia , Membro Posterior/irrigação sanguínea , Isquemia/fisiopatologia , Receptor EphA4/fisiologia , Animais , Cromonas , Endotélio Vascular/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfolinas , Perfusão , Receptor EphA4/genética
7.
Tumour Biol ; 37(9): 12411-12422, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27323967

RESUMO

Radiotherapy is widely used for advanced rectal tumors. However, tumor recurrence after this treatment tends to be more aggressive and is associated with a poor prognosis. Uncovering the molecular mechanism that controls this recurrence is essential for developing new therapeutic applications. In the present study, we demonstrated that radiation increases the EphA4 activation level of the survivor progeny of colorectal cancer cells submitted to this treatment and that such activation promoted the internalization of a complex E-cadherin-EphA4, inducing cell-cell adhesion disruption. Moreover, EphA4 knockdown in the progeny of irradiated cells reduced the migratory and invasive potentials and metalloprotease activity induced by irradiation. Finally, we demonstrated that the cell migration and invasion potential were regulated by AKT and ERK1/2 signaling, with the ERK1/2 activity being dependent on EphA4. In summary, our study demonstrates that these signaling pathways could be responsible for the therapeutic failure, thereby promoting local invasion and metastasis in rectal cancer after radiotherapy. We also postulate that EphA4 is a potential therapeutic target for colorectal cancer treatment.


Assuntos
Neoplasias Colorretais/radioterapia , Receptor EphA4/fisiologia , Transdução de Sinais/fisiologia , Antígenos CD , Caderinas/análise , Neoplasias Colorretais/patologia , Doxazossina/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Células HT29 , Humanos , Invasividade Neoplásica , Proteínas Proto-Oncogênicas c-akt/fisiologia
8.
Neurobiol Learn Mem ; 124: 62-70, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26165136

RESUMO

Eph receptors regulate glutamate receptors functions, neuronal morphology and synaptic plasticity, cellular events believed to be involved in memory formation. In this study we aim to explore the roles of Eph receptors in learning and memory. Toward that end, we examined the roles of EphB2 and EphA4 receptors, key regulators of synaptic functions, in fear conditioning memory formation. We show that mice lacking EphB2 (EphB2(-/-)) are impaired in short- and long-term contextual fear conditioning memory. Mice that express a carboxy-terminally truncated form of EphB2 that lacks forward signaling, instead of the full EphB2, are impaired in long-term, but not short-term, contextual fear conditioning memory. Long-term contextual fear conditioning memory is attenuated in CaMKII-cre;EphA4(lx/-) mice where EphA4 is removed from all pyramidal neurons of the forebrain. Mutant mice with targeted kinase-dead EphA4 (EphA4(KD)) exhibit intact long-term contextual fear conditioning memory showing that EphA4 kinase-mediated forward signaling is not needed for contextual fear memory formation. The ability to form long-term conditioned taste aversion (CTA) memory is not impaired in the EphB2(-/-) and CaMKII-cre;EphA4(lx/-) mice. We conclude that EphB2 forward signaling is required for long-term contextual fear conditioning memory formation. In contrast, EphB2 mediates short-term contextual fear conditioning memory formation in a forward signaling-independent manner. EphA4 mediates long-term contextual fear conditioning memory formation in a kinase-independent manner.


Assuntos
Condicionamento Clássico/fisiologia , Medo/fisiologia , Memória de Longo Prazo/fisiologia , Prosencéfalo/fisiologia , Receptor EphA4/fisiologia , Receptor EphB2/fisiologia , Animais , Camundongos , Camundongos Knockout , Receptor EphA4/genética , Receptor EphB2/genética , Transdução de Sinais
9.
PLoS One ; 10(5): e0126942, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25978062

RESUMO

In mouse cerebral corticogenesis, neurons are generated from radial glial cells (RGCs) or from their immediate progeny, intermediate neuronal precursors (INPs). The balance between self-renewal of these neuronal precursors and specification of cell fate is critical for proper cortical development, but the signaling mechanisms that regulate this progression are poorly understood. EphA4, a member of the receptor tyrosine kinase superfamily, is expressed in RGCs during embryogenesis. To illuminate the function of EphA4 in RGC cell fate determination during early corticogenesis, we deleted Epha4 in cortical cells at E11.5 or E13.5. Loss of EphA4 at both stages led to precocious in vivo RGC differentiation toward neurogenesis. Cortical cells isolated at E14.5 and E15.5 from both deletion mutants showed reduced capacity for neurosphere formation with greater differentiation toward neurons. They also exhibited lower phosphorylation of ERK and FRS2α in the presence of FGF. The size of the cerebral cortex at P0 was smaller than that of controls when Epha4 was deleted at E11.5 but not when it was deleted at E13.5, although the cortical layers were formed normally in both mutants. The number of PAX6-positive RGCs decreased at later developmental stages only in the E11.5 Epha4 deletion mutant. These results suggest that EphA4, in cooperation with an FGF signal, contributes to the maintenance of RGC self-renewal and repression of RGC differentiation through the neuronal lineage. This function of EphA4 is especially critical and uncompensated in early stages of corticogenesis, and thus deletion at E11.5 reduces the size of the neonatal cortex.


Assuntos
Diferenciação Celular/fisiologia , Células Ependimogliais/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Regeneração Nervosa/fisiologia , Receptor EphA4/fisiologia , Animais , Apoptose/fisiologia , Western Blotting , Encéfalo/anatomia & histologia , Encéfalo/embriologia , Encéfalo/fisiologia , Linhagem da Célula/fisiologia , Células Cultivadas , Camundongos , Transdução de Sinais/fisiologia
10.
Eur J Neurosci ; 40(7): 3021-31, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25041248

RESUMO

The lack of axonal regeneration in the adult central nervous system is in part attributable to the presence of inhibitory molecules present in the environment of injured axons such as the myelin-associated proteins Nogo-A and MAG and the repulsive guidance molecules Ephrins, Netrins and Semaphorins. In the present study, we hypothesized that EphA4 and one of its potential binding partners EphrinA3 may participate in the inhibition of adult axon regeneration in the model of adult mouse optic nerve injury. Axonal regeneration was analysed in three dimensions after tissue clearing of EphA4 knockout (KO), EphrinA3 KO and wild-type (WT) optic nerves. By immunohistochemistry, EphA4 was highly expressed in Müller glia endfeet in the retina and in astrocytes in the retina and the optic nerve, while EphrinA3 was present in retinal ganglion cells and oligodendrocytes. Optic nerve crush did not cause expression changes. Significantly more axons grew in the crushed optic nerve of EphA4 KO mice than in WT or EphrinA3 KO animals. Single axon analysis revealed that EphA4 KO axons were less prone to form aberrant branching than axons in the other mouse groups. The expression of growth-associated proteins Sprr1a and Gap-43 did not vary between EphA4 KO and WT retinae. However, glial fibrillary acidic protein-expressing astrocytes were withdrawn from the perilesional area in EphA4 KO, suggesting that gliosis down-regulation may locally contribute to improve axonal growth at the injury site. In summary, our three-dimensional analysis of injured mouse optic nerves reveals beneficial effects of EphA4 ablation on the intensity and the pattern of optic nerve axon regeneration.


Assuntos
Axônios/fisiologia , Regeneração Nervosa , Traumatismos do Nervo Óptico/fisiopatologia , Receptor EphA4/fisiologia , Animais , Axônios/patologia , Gliose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/metabolismo , Receptor EphA3/genética , Receptor EphA3/fisiologia , Receptor EphA4/genética , Retina/metabolismo
11.
J Bone Miner Res ; 29(4): 804-19, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23983218

RESUMO

Of the ephrin (Eph) receptors, mature osteoclasts express predominantly EphA4. This study sought to determine if EphA4 has a regulatory role in osteoclasts. Treatment of RAW/C4 cells with Epha4 small interfering RNAs (siRNAs) increased average size, Ctsk mRNA expression level, and bone resorption activity of the derived osteoclast-like cells. Activation of the EphA4 signaling in osteoclast precursors with EfnA4-fc chimeric protein reduced cell size and resorption activity of the derived osteoclasts. Homozygous Epha4 null mice had substantially less trabecular bone in femur and vertebra compared to wild-type controls. The bone loss was due to a decrease in trabecular number and an increase in trabecular spacing, but not to an increase in osteoclast-lined bone surface or an increase in the number of osteoclasts on bone surface. Dynamic histomorphometry and serum biomarker analyses indicate that bone formation in Epha4 null mice was reduced slightly but not significantly. Osteoclasts of Epha4 null mice were also larger, expressed higher levels of Mmp3 and Mmp9 mRNAs, and exhibited greater bone resorption activity than wild-type osteoclasts in vitro. Deficient Epha4 expression had no effects on the total number of osteoclast formed in response to receptor activator of NF-κB ligand nor on apoptosis of osteoclasts in vitro. It also did not affect the protein-tyrosine phosphorylation status of its ligands, EfnB2, EfnA2, and EfnA4, in osteoclasts. Deficient Epha4 expression in Epha4 null osteoclasts activated the ß3 -integrin signaling through reduced phosphorylation of the tyr-747 residue, which led to increased binding of the stimulatory talin and reduced binding of the inhibitory Dok1 to ß3 -integrin. This in turn activated Vav3 and the bone resorption activity of osteoclasts. In conclusion, we demonstrate for the first time that EphA4 is a potent negative regulator of osteoclastic activity, mediated in part through increased Dok1 binding to ß3 -integrin via an increase in EphA4-dependent tyr-747 phosphorylation.


Assuntos
Osteoclastos/citologia , Receptor EphA4/fisiologia , Animais , Remodelação Óssea , Linhagem Celular , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Receptor EphA4/metabolismo , Transdução de Sinais , Microtomografia por Raio-X
12.
Neuropharmacology ; 65: 232-43, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23123677

RESUMO

Neurons within a network have the ability to homeostatically scale-down their excitatory synaptic strength under conditions of persistent neuronal activity elevation, a process pivotal to neural circuit stability. How this homeostatic regulation is achieved at the molecular level in developing neural circuits, which face gradually elevated neuronal activity as part of circuit wiring, is not well-understood. Using dissociated hippocampal neuronal cultures, we identified a critical and cell autonomous role for the receptor tyrosine kinase EphA4 in mediating activity-induced homeostatic down-regulation of excitatory synaptic strength. Reducing the endogenous level of EphA4 in individual neurons by RNAi effectively blocked activity-induced scaling-down of excitatory synaptic strength, while co-transfection of RNAi resistant EphA4 rescued this effect. Furthermore, interfering with EphA4 forward signaling using EphA4-Fc blocked activity-induced homeostatic synaptic scaling-down, while direct activation of EphA4 with its ligand EphrinA1 weakened excitatory synaptic strength. Up- or down-regulating EphA4 function in individual neurons also did not affect the density of excitatory synapses. The kinase activities of EphA4 and its downstream effector Cdk5 were both required for homeostatic synaptic scaling, as overexpression of EphA4 with constitutively active kinase activity reduced excitatory synaptic strength, while interfering with either the kinase activity of EphA4 or Cdk5 blocked activity-induced synaptic scaling. Consistently, the activities of EphA4 and Cdk5 increased significantly during global and persistent activity elevation. Together, our work demonstrated that the kinase activity of EphA4, via activation of downstream Cdk5 activity, mediates the scaling-down of excitatory synaptic strength under conditions of global activity elevation.


Assuntos
Quinase 5 Dependente de Ciclina/metabolismo , Homeostase/fisiologia , Receptor EphA4/metabolismo , Sinapses/enzimologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Quinase 5 Dependente de Ciclina/fisiologia , Ativação Enzimática/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Células HEK293 , Humanos , Ratos , Ratos Sprague-Dawley , Receptor EphA4/fisiologia
13.
J Neurotrauma ; 29(17): 2660-71, 2012 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-22985250

RESUMO

We investigated the role of the axon guidance molecule EphA4 following traumatic brain injury (TBI) in mice. Neutralization of EphA4 improved motor function and axonal regeneration following experimental spinal cord injury (SCI). We hypothesized that genetic absence of EphA4 could improve functional and histological outcome following TBI. Using qRT-PCR in wild-type (WT) mice, we evaluated the EphA4 mRNA levels following controlled cortical impact (CCI) TBI or sham injury and found it to be downregulated in the hippocampus (p<0.05) but not the cortex ipsilateral to the injury at 24 h post-injury. Next, we evaluated the behavioral and histological outcome following CCI using WT mice and Emx1-Cre-driven conditional knockout (cKO) mice. In cKO mice, EphA4 was completely absent in the hippocampus and markedly reduced in the cortical regions from embryonic day 16, which was confirmed using Western blot analysis. EphA4 cKO mice had similar learning and memory abilities at 3 weeks post-TBI compared to WT controls, although brain-injured animals performed worse than sham-injured controls (p<0.05). EphA4 cKO mice performed similarly to WT mice in the rotarod and cylinder tests of motor function up to 29 days post-injury. TBI increased cortical and hippocampal astrocytosis (GFAP immunohistochemistry, p<0.05) and hippocampal sprouting (Timm stain, p<0.05) and induced a marked loss of hemispheric tissue (p<0.05). EphA4 cKO did not alter the histological outcome. Although our results may argue against a beneficial role for EphA4 in the recovery process following TBI, further studies including post-injury pharmacological neutralization of EphA4 are needed to define the role for EphA4 following TBI.


Assuntos
Lesões Encefálicas/patologia , Lesões Encefálicas/psicologia , Receptor EphA4/genética , Animais , Western Blotting , Peso Corporal/fisiologia , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Coxeadura Animal/etiologia , Coxeadura Animal/psicologia , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Equilíbrio Postural/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Receptor EphA4/fisiologia , Caracteres Sexuais
14.
Hum Reprod ; 26(5): 1163-70, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21349856

RESUMO

BACKGROUND: Recently, the Eph-ephrinA system was proposed to contribute to the initial interaction between the maternal endometrial epithelium and embryonic trophectoderm. Since the Eph-ephrin interaction can induce adhesive and/or repulsive forces into the cells, we examined the possible role of this system in functional changes in endometrial epithelial cells using endometrial carcinoma-derived Ishikawa cells. METHODS: The expressions of EphA1, A2 and A4 on Ishikawa cells were examined by RT-PCR and western blotting analyses. The effects of recombinant ephrinA1 on Ishikawa cells were also examined by western blot analysis and cell attachment and aggregation assays. RESULTS: EphA1, A2 and A4 were expressed on Ishikawa cells. Recombinant ephrinA1 bound to the surfaces of Ishikawa cells and induced phosphorylation of EphA2 and A4. In bovine serum albumin-blocked nitrocellulose-coated dishes, Ishikawa cells remained floating and aggregated with each other. Under these conditions, immobilized ephrinA1 promoted Ishikawa cell attachment with increased tyrosine phosphorylation in focal adhesion kinase. In addition, immobilized ephrinA1 reversibly inhibited Ishikawa cell aggregation. Gene-reduction of EphA1, A2 and A4 by siRNAs attenuated the inhibitory effects of ephrinA1 on cell aggregation, confirming that ephrinA1 affects Ishikawa cell functions through Eph-ephrinA interaction. CONCLUSIONS: This study demonstrated that the Eph-ephrinA system can promote cell attachment along with intercellular dissociation in Ishikawa cells. These findings suggest that this system can induce functional changes in endometrial epithelial cells.


Assuntos
Adesão Celular/fisiologia , Agregação Celular/fisiologia , Efrina-A1/fisiologia , Receptores da Família Eph/fisiologia , Animais , Linhagem Celular , Efrina-A1/genética , Efrina-A1/metabolismo , Feminino , Humanos , Camundongos , Fosforilação , Receptor EphA1/metabolismo , Receptor EphA1/fisiologia , Receptor EphA2/metabolismo , Receptor EphA2/fisiologia , Receptor EphA4/metabolismo , Receptor EphA4/fisiologia , Receptores da Família Eph/metabolismo , Proteínas Recombinantes de Fusão , Transdução de Sinais
15.
Dev Biol ; 350(2): 441-50, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21147090

RESUMO

In Xenopus gastrulation, the involuting mesodermal and non-involuting ectodermal cells remain separated from each other, undergoing convergent extension. Here, we show that Eph-ephrin signaling is crucial for the tissue separation and convergence during gastrulation. The loss of EphA4 function results in aberrant gastrulation movements, which are due to selective inhibition of tissue constriction and separation. At the cellular levels, knockdown of EphA4 impairs polarization and migratory activity of gastrulating cells but not specification of their fates. Importantly, rescue experiments demonstrate that EphA4 controls tissue separation via RhoA GTPase in parallel to Fz7 and PAPC signaling. In addition, we show that EphA4 and its putative ligand, ephrin-A1 are expressed in a complementary manner in the involuting mesodermal and non-involuting ectodermal layers of early gastrulae, respectively. Depletion of ephrin-A1 also abrogates tissue separation behaviors. Therefore, these results suggest that Eph receptor and its ephrin ligand might mediate repulsive interaction for tissue separation and convergence during early Xenopus gastrulation movements.


Assuntos
Efrina-A1/fisiologia , Gastrulação , Receptor EphA4/fisiologia , Transdução de Sinais/fisiologia , Xenopus/embriologia , Animais , Caderinas/fisiologia , Movimento Celular , Polaridade Celular , Protocaderinas , Receptores Acoplados a Proteínas G/fisiologia , Proteínas de Xenopus/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia
16.
J Neurosci ; 30(47): 16015-24, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21106840

RESUMO

There is growing evidence that astrocytes play critical roles in neuron-glial interactions at the synapse. Astrocytes are believed to regulate presynaptic and postsynaptic structures and functions, in part, by the release of gliotransmitters such as glutamate, ATP, and d-serine; however, little is known of how neurons and astrocytes communicate to regulate these processes. Here, we investigated a family of transmembrane proteins called ephrinBs and Eph receptors that are expressed in the synapse and are known to regulate synaptic transmission and plasticity. In addition to their presence on CA1 hippocampal neurons, we determined that ephrins and Eph receptors are also expressed on hippocampal astrocytes. Stimulation of hippocampal astrocytes with soluble ephrinB3, known to be expressed on CA1 postsynaptic dendrites, enhanced d-serine synthesis and release in culture. Conversely, ephrinB3 had no effect on d-serine release from astrocytes deficient in EphB3 and EphA4, which are the primary receptors for ephrinB3. Eph receptors mediate this response through interactions with PICK1 (protein interacting with C-kinase) and by dephosphorylating protein kinase C α to activate the conversion of l-serine to d-serine by serine racemase. These findings are supported in vivo, where reduced d-serine levels and synaptic transmissions are observed in the absence of EphB3 and EphA4. These data support a role for ephrins and Eph receptors in regulating astrocyte gliotransmitters, which may have important implications on synaptic transmission and plasticity.


Assuntos
Astrócitos/metabolismo , Efrina-B3/fisiologia , Serina/biossíntese , Serina/metabolismo , Animais , Células Cultivadas , Efrina-B3/deficiência , Hipocampo/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Plasticidade Neuronal/genética , Biossíntese de Proteínas/genética , Receptor EphA4/biossíntese , Receptor EphA4/deficiência , Receptor EphA4/fisiologia , Serina/análogos & derivados , Estereoisomerismo , Transmissão Sináptica/genética
17.
J Neurotrauma ; 27(7): 1321-32, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20486805

RESUMO

Glial scar formation occurs in response to brain injury in mammalian models and inhibits axonal growth. Identification of molecules that may mediate reactivity of astrocytes has become a leading therapeutic goal in the field of neurotrauma. In adult rodent brain and spinal cord, many of the Eph receptors and their ephrin ligands have been demonstrated to be upregulated on reactive astrocytes at the injury site; however, little is known about the expression of these molecules in nonhuman primate injury models. This study examines the role of the tyrosine kinase EphA4 receptor, which predominantly binds most ephrin ligands, after injury in marmoset monkey brain. Following lesioning of the primary visual cortex (V1) in the adult marmoset, EphA4 is strongly upregulated on reactive astrocytes around the lesion site, which secrete extracellular matrix molecules such as chondroitin sulfate proteoglycans, which are known for their inhibitory effect on axonal growth and regeneration. This astrocyte reactivity was also associated with neuronal death in the area adjacent to the lesion site. EphA4 activation induced by clustered ephrin A5-Fc-mediated astrocyte proliferation and glial fibrillary acidic protein expression in vitro, as demonstrated by closure of scratched wound and MTT assays, occurs via two potential signaling pathways, the mitogen-activated protein kinase and Rho pathways. These results in a nonhuman primate model highlight the importance of developing pharmacotherapeutic approaches to block these molecules following brain injury.


Assuntos
Astrócitos/metabolismo , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Córtex Cerebral/metabolismo , Gliose/metabolismo , Gliose/patologia , Receptor EphA4/biossíntese , Regulação para Cima/fisiologia , Animais , Astrócitos/patologia , Biomarcadores/metabolismo , Lesões Encefálicas/complicações , Callithrix , Morte Celular/fisiologia , Córtex Cerebral/lesões , Córtex Cerebral/patologia , Modelos Animais de Doenças , Efrina-A5/metabolismo , Efrinas/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/etiologia , Receptor EphA4/fisiologia
18.
Neuron ; 65(5): 580-2, 2010 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-20223194

RESUMO

Neural circuits of vertebrates often display highly ordered projections between different areas, so-called topographic maps, and are characterized by their ability to undergo structural remodeling. In this issue of Neuron, Galimberti et al. demonstrate that mossy fibers of the rodent hippocampus exhibit prominent structural plasticity according to a novel topographic rule, in a process requiring the EphA4 receptor tyrosine kinase.


Assuntos
Hipocampo/citologia , Fibras Musgosas Hipocampais/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Animais , Camundongos , Receptor EphA4/fisiologia
19.
Proc Natl Acad Sci U S A ; 106(30): 12524-9, 2009 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-19592509

RESUMO

Increasing evidence indicates the importance of neuron-glia communication for synaptic function, but the mechanisms involved are not fully understood. We reported that the EphA4 receptor tyrosine kinase is in dendritic spines of pyramidal neurons of the adult hippocampus and regulates spine morphology. We now show that the ephrin-A3 ligand, which is located in the perisynaptic processes of astrocytes, is essential for maintaining EphA4 activation and normal spine morphology in vivo. Ephrin-A3-knockout mice have spine irregularities similar to those observed in EphA4-knockout mice. Remarkably, loss of ephrin-A3 or EphA4 increases the expression of glial glutamate transporters. Consistent with this, glutamate transport is elevated in ephrin-A3-null hippocampal slices whereas Eph-dependent stimulation of ephrin-A3 signaling inhibits glutamate transport. Furthermore, some forms of hippocampus-dependent learning are impaired in the ephrin-A3-knockout mice. Our results suggest that the interaction between neuronal EphA4 and glial ephrin-A3 bidirectionally controls synapse morphology and glial glutamate transport, ultimately regulating hippocampal function.


Assuntos
Espinhas Dendríticas/fisiologia , Efrina-A3/fisiologia , Glutamatos/metabolismo , Neuroglia/metabolismo , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Análise de Variância , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Transporte Biológico , Espinhas Dendríticas/metabolismo , Efrina-A3/genética , Efrina-A3/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Immunoblotting , Imunoprecipitação , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Camundongos , Camundongos Knockout , Microscopia Confocal , Microscopia de Fluorescência , Atividade Motora/fisiologia , Fosforilação , Células Piramidais/citologia , Células Piramidais/metabolismo , Receptor EphA4/genética , Receptor EphA4/metabolismo , Receptor EphA4/fisiologia
20.
J Cell Biol ; 185(3): 551-64, 2009 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-19414612

RESUMO

Alzheimer's disease is an age-dependent neurodegenerative disorder that is characterized by a progressive decline in cognitive function. gamma-secretase dysfunction is evident in many cases of early onset familial Alzheimer's disease. However, the mechanism by which gamma-secretase dysfunction results in memory loss and neurodegeneration is not fully understood. Here, we demonstrate that gamma-secretase is localized at synapses and regulates spine formation. We identify EphA4, one of the Ephrin receptor family members, as a substrate of gamma-secretase, and find that EphA4 processing is enhanced by synaptic activity. Moreover, overexpression of EphA4 intracellular domain increases the number of dendritic spines by activating the Rac signaling pathway. These findings reveal a function for EphA4-mediated intracellular signaling in the morphogenesis of dendritic spines and suggest that the processing of EphA4 by gamma-secretase affects the pathogenesis of Alzheimer's disease.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Dendritos/enzimologia , Receptor EphA4/metabolismo , Receptor EphA4/fisiologia , Sinapses/fisiologia , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Dimetil Sulfóxido/farmacologia , Hipocampo/enzimologia , Humanos , Camundongos , Emaranhados Neurofibrilares/patologia , Neurônios/fisiologia , Ratos , Coluna Vertebral/citologia , Coluna Vertebral/efeitos dos fármacos , Coluna Vertebral/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...