Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 151(5)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38345254

RESUMO

EphB1 is required for proper guidance of cortical axon projections during brain development, but how EphB1 regulates this process remains unclear. We show here that EphB1 conditional knockout (cKO) in GABAergic cells (Vgat-Cre), but not in cortical excitatory neurons (Emx1-Cre), reproduced the cortical axon guidance defects observed in global EphB1 KO mice. Interestingly, in EphB1 cKOVgat mice, the misguided axon bundles contained co-mingled striatal GABAergic and somatosensory cortical glutamatergic axons. In wild-type mice, somatosensory axons also co-fasciculated with striatal axons, notably in the globus pallidus, suggesting that a subset of glutamatergic cortical axons normally follows long-range GABAergic axons to reach their targets. Surprisingly, the ectopic axons in EphB1 KO mice were juxtaposed to major blood vessels. However, conditional loss of EphB1 in endothelial cells (Tie2-Cre) did not produce the axon guidance defects, suggesting that EphB1 in GABAergic neurons normally promotes avoidance of these ectopic axons from the developing brain vasculature. Together, our data reveal a new role for EphB1 in GABAergic neurons to influence proper cortical glutamatergic axon guidance during brain development.


Assuntos
Orientação de Axônios , Células Endoteliais , Animais , Camundongos , Axônios/fisiologia , Neurônios GABAérgicos , Camundongos Knockout , Receptores Proteína Tirosina Quinases , Receptor EphB1/metabolismo
2.
Cell Rep ; 22(9): 2246-2253, 2018 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-29490264

RESUMO

Altering AMPA receptor (AMPAR) content at synapses is a key mechanism underlying the regulation of synaptic strength during learning and memory. Previous work demonstrated that SynDIG1 (synapse differentiation-induced gene 1) encodes a transmembrane AMPAR-associated protein that regulates excitatory synapse strength and number. Here we show that the related protein SynDIG4 (also known as Prrt1) modifies AMPAR gating properties in a subunit-dependent manner. Young SynDIG4 knockout (KO) mice have weaker excitatory synapses, as evaluated by immunocytochemistry and electrophysiology. Adult SynDIG4 KO mice show complete loss of tetanus-induced long-term potentiation (LTP), while mEPSC amplitude is reduced by only 25%. Furthermore, SynDIG4 KO mice exhibit deficits in two independent cognitive assays. Given that SynDIG4 colocalizes with the AMPAR subunit GluA1 at non-synaptic sites, we propose that SynDIG4 maintains a pool of extrasynaptic AMPARs necessary for synapse development and function underlying higher-order cognitive plasticity.


Assuntos
Cognição , Potenciais Pós-Sinápticos Excitadores , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal , Receptores de AMPA/metabolismo , Sinapses/metabolismo , Animais , Feminino , Genes Reporter , Hipocampo/metabolismo , Cinética , Potenciação de Longa Duração , Memória , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Subunidades Proteicas/metabolismo , Análise e Desempenho de Tarefas , Xenopus laevis
3.
eNeuro ; 3(5)2016.
Artigo em Inglês | MEDLINE | ID: mdl-27800545

RESUMO

Modification of the strength of excitatory synaptic connections is a fundamental mechanism by which neural circuits are refined during development and learning. Synapse Differentiation Induced Gene 1 (SynDIG1) has been shown to play a key role in regulating synaptic strength in vitro. Here, we investigated the role of SynDIG1 in vivo in mice with a disruption of the SynDIG1 gene rather than use an alternate loxP-flanked conditional mutant that we find retains a partial protein product. The gene-trap insertion with a reporter cassette mutant mice shows that the SynDIG1 promoter is active during embryogenesis in the retina with some activity in the brain, and postnatally in the mouse hippocampus, cortex, hindbrain, and spinal cord. Ultrastructural analysis of the hippocampal CA1 region shows a decrease in the average PSD length of synapses and a decrease in the number of synapses with a mature phenotype. Intriguingly, the total synapse number appears to be increased in SynDIG1 mutant mice. Electrophysiological analyses show a decrease in AMPA and NMDA receptor function in SynDIG1-deficient hippocampal neurons. Glutamate stimulation of individual dendritic spines in hippocampal slices from SynDIG1-deficient mice reveals increased short-term structural plasticity. Notably, the overall levels of PSD-95 or glutamate receptors enriched in postsynaptic biochemical fractions remain unaltered; however, activity-dependent synapse development is strongly compromised upon the loss of SynDIG1, supporting its importance for excitatory synapse maturation. Together, these data are consistent with a model in which SynDIG1 regulates the maturation of excitatory synapse structure and function in the mouse hippocampus in vivo.


Assuntos
Região CA1 Hipocampal/crescimento & desenvolvimento , Região CA1 Hipocampal/metabolismo , Proteínas de Transporte/genética , Sinapses/metabolismo , Animais , Região CA1 Hipocampal/ultraestrutura , Células Cultivadas , Proteína 4 Homóloga a Disks-Large , Feminino , Ácido Glutâmico/metabolismo , Guanilato Quinases/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasticidade Neuronal/fisiologia , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/ultraestrutura , Técnicas de Cultura de Tecidos
4.
Dev Neurobiol ; 76(4): 405-20, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26148571

RESUMO

The two cortical hemispheres of the mammalian forebrain are interconnected by major white matter tracts, including the corpus callosum (CC) and the posterior branch of the anterior commissure (ACp), that bridge the telencephalic midline. We show here that the intracellular signaling domains of the EphB1 and EphB2 receptors are critical for formation of both the ACp and CC. We observe partial and complete agenesis of the corpus callosum, as well as highly penetrant ACp misprojection phenotypes in truncated EphB1/2 mice that lack intracellular signaling domains. Consistent with the roles for these receptors in formation of the CC and ACp, we detect expression of these receptors in multiple brain regions associated with the formation of these forebrain structures. Taken together, our findings suggest that a combination of forward and reverse EphB1/2 receptor-mediated signaling contribute to ACp and CC axon guidance.


Assuntos
Comissura Anterior/embriologia , Comissura Anterior/metabolismo , Corpo Caloso/embriologia , Corpo Caloso/metabolismo , Receptor EphB1/metabolismo , Receptor EphB2/metabolismo , Animais , Comissura Anterior/citologia , Axônios/metabolismo , Movimento Celular/fisiologia , Corpo Caloso/citologia , Técnicas de Introdução de Genes , Técnicas de Inativação de Genes , Imuno-Histoquímica , Espaço Intracelular , Camundongos Transgênicos , Técnicas de Rastreamento Neuroanatômico , Domínios Proteicos , Receptor EphB1/genética , Receptor EphB2/genética , Transdução de Sinais
5.
Proc Natl Acad Sci U S A ; 111(6): 2188-93, 2014 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-24453220

RESUMO

In early brain development, ascending thalamocortical axons (TCAs) navigate through the ventral telencephalon (VTel) to reach their target regions in the young cerebral cortex. Descending, deep-layer cortical axons subsequently target appropriate thalamic and subcortical target regions. However, precisely how and when corticothalamic axons (CTAs) identify their appropriate, reciprocal thalamic targets remains unclear. We show here that EphB1 and EphB2 receptors control proper navigation of a subset of TCA and CTA projections through the VTel. We show in vivo that EphB receptor forward signaling and the ephrinB1 ligand are required during the early navigation of L1-CAM(+) thalamic fibers in the VTel, and that the misguided thalamic fibers in EphB1/2 KO mice appear to interact with cortical subregion-specific axon populations during reciprocal cortical axon guidance. As such, our findings suggest that descending cortical axons identify specific TCA subpopulations in the dorsal VTel to coordinate reciprocal cortical-thalamic connectivity in the early developing brain.


Assuntos
Axônios , Córtex Cerebral/metabolismo , Receptores da Família Eph/metabolismo , Transdução de Sinais , Tálamo/metabolismo , Animais , Camundongos , Camundongos Knockout , Receptores da Família Eph/genética
6.
Am J Respir Cell Mol Biol ; 49(4): 680-7, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23742148

RESUMO

Alveolar growth abnormalities and severe respiratory dysfunction are often fatal. Identifying mechanisms that control epithelial proliferation and enlarged, poorly septated airspaces is essential in developing new therapies for lung disease. The membrane-bound ligand ephrin-B2 is strongly expressed in lung epithelium, and yet in contrast to its known requirement for arteriogenesis, considerably less is known regarding the function of this protein in the epithelium. We hypothesize that the vascular mediator ephrin-B2 governs alveolar growth and mechanics beyond the confines of the endothelium. We used the in vivo manipulation of ephrin-B2 reverse signaling to determine the role of this vascular mediator in the pulmonary epithelium and distal lung mechanics. We determined that the ephrin-B2 gene (EfnB2) is strongly expressed in alveolar Type 2 cells throughout development and into adulthood. The role of ephrin-B2 reverse signaling in the lung was assessed in Efnb2(LacZ/6YFΔV) mutants that coexpress the intracellular truncated ephrin-B2-ß-galactosidase fusion and an intracellular point mutant ephrin-B2 protein that is unable to become tyrosine-phosphorylated or to interact with either the SH2 or PDZ domain-containing downstream signaling proteins. In these viable mice, we observed pulmonary hypoplasia and altered pulmonary mechanics, as evidenced by a marked reduction in lung compliance. Associated with the reduction in lung compliance was a significant increase in insoluble fibronectin (FN) basement membrane matrix assembly with FN deposition, and a corresponding increase in the α5 integrin receptor required for FN fibrillogenesis. These experiments indicate that ephrin-B2 reverse signaling mediates distal alveolar formation, fibrillogenesis, and pulmonary compliance.


Assuntos
Efrina-B2/metabolismo , Fibronectinas/metabolismo , Integrina alfa5beta1/metabolismo , Complacência Pulmonar/fisiologia , Transdução de Sinais/fisiologia , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/fisiopatologia , Animais , Citoplasma/genética , Citoplasma/metabolismo , Citoplasma/fisiologia , Efrina-B2/genética , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Fibronectinas/genética , Integrina alfa5beta1/genética , Pulmão/anormalidades , Pulmão/metabolismo , Pulmão/fisiopatologia , Complacência Pulmonar/genética , Pneumopatias/genética , Pneumopatias/metabolismo , Pneumopatias/fisiopatologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Domínios PDZ/genética , Fosforilação/genética , Mutação Puntual/genética , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/fisiologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/fisiologia , Transdução de Sinais/genética , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
7.
Mol Cell Neurosci ; 52: 106-16, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23147113

RESUMO

EphB receptors and their ephrinB ligands transduce bidirectional signals that mediate contact-dependent axon guidance primarily by promoting growth cone repulsion. However, how EphB receptor-mediated forward signaling induces axonal repulsion remains poorly understood. Here, we identify Nck and Pak proteins as essential forward signaling components of EphB2-dependent growth cone collapse in cortical neurons. We show that kinase-active EphB2 binds to Pak and promotes growth cone repulsion via Pak kinase activity, Pak-Nck binding, RhoA signaling and endocytosis. However, Pak's function in this context appears to be independent of Rac/Cdc42-GTP, consistent with the absence of Rac-GTP production after ephrinB treatment of cortical neurons. Taken together, our findings suggest that ephrinB-activated EphB2 receptors recruit a novel Nck/Pak signaling complex to mediate repulsive cortical growth cone guidance, which may be relevant for EphB forward signaling-dependent axon guidance in vivo.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cones de Crescimento/metabolismo , Neurogênese/fisiologia , Proteínas Oncogênicas/metabolismo , Receptor EphB2/metabolismo , Transdução de Sinais/fisiologia , Quinases Ativadas por p21/metabolismo , Animais , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Efrinas/metabolismo , Técnicas de Introdução de Genes , Immunoblotting , Imunoprecipitação , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
8.
Dev Dyn ; 241(12): 1901-10, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23129351

RESUMO

INTRODUCTION: Ephrin-B2 on osteoclasts was reported to promote bone formation as part of homeostasis by activating the EphB4 tyrosine kinase receptor on osteoblasts. Little is known about the role of ephrin-B signaling to EphBs in developmental bone formation. RESULTS: We observed expression of an ephrin-B2 LacZ chimeric allele in the periosteum, sutural bone fronts, and dura mater of embryonic and neonatal mice. Expression in the adult skull was confined to sutures, but was heavily upregulated at sites of bone injury. Culture of embryonic calvariae with soluble recombinant ephrin-B2/Fc doubled their bone content without altering suture width or overall skull morphology. Ephrin-B2/Fc also stimulated osteoblast marker gene expression in cultured MC3T3 preosteoblastic cells without the need for type 1 collagen-induced differentiation. EphB4 was absent in embryonic and adult skulls. However, EphB1 and EphB2, both physiological receptors for ephrin-Bs, were expressed at sites of osteogenesis, and EphB1 knockout mice displayed a reduction in calvarial bone content compared to controls. CONCLUSIONS: These data support a role for ephrin-B2 in the development and healing of bone through activation of osteoblast-specific gene expression. EphB1 and EphB2 are likely candidates receptors for the ephrin-B2 in bone.


Assuntos
Efrina-B2/metabolismo , Osteoblastos/metabolismo , Osteogênese/fisiologia , Crânio/embriologia , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Linhagem Celular , Efrina-B2/genética , Efrina-B2/farmacologia , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/farmacologia , Camundongos , Camundongos Knockout , Técnicas de Cultura de Órgãos , Osteoblastos/citologia , Osteogênese/efeitos dos fármacos , Receptor EphB1/genética , Receptor EphB1/metabolismo , Receptor EphB2/genética , Receptor EphB2/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Crânio/citologia
9.
Eur J Neurosci ; 34(10): 1620-33, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22103419

RESUMO

EphB receptor tyrosine kinases direct axonal pathfinding through interactions with ephrin-B proteins following axon-cell contact. As EphB:ephrin-B binding leads to bidirectional signals, the contributions of signaling into the Eph-expressing cell (forward signaling) or the ephrin-expressing cell (reverse signaling) cannot be assigned using traditional protein null alleles. To determine if EphB1 is functioning solely as a receptor during axon pathfinding, a new knock-in mutant mouse was created, EphB1(T-lacZ), which expresses an intracellular-truncated EphB1-ß-gal fusion protein from the endogenous locus. As in the EphB1(-/-) protein null animals, the EphB1(T-lacZ/T-lacZ) homozygotes fail to form the ipsilateral projecting subpopulation of retinal ganglion cell axons. This indicates that reverse signaling through the extracellular domain of EphB1 is not required for proper axon pathfinding of retinal axons at the optic chiasm. Further analysis of other EphB and ephrin-B mutant mice shows that EphB1 is the preferred receptor of ephrin-B2 and, to a lesser degree, ephrin-B1 in mediating axon guidance at the optic chiasm despite the coexpression of EphB2 in the same ipsilaterally projecting retinal axons.


Assuntos
Comunicação Celular/fisiologia , Efrina-B1/metabolismo , Efrina-B2/metabolismo , Quiasma Óptico/anatomia & histologia , Quiasma Óptico/fisiologia , Receptor EphB2/metabolismo , Animais , Movimento Celular/fisiologia , Efrina-B1/genética , Efrina-B2/genética , Feminino , Corpos Geniculados/citologia , Corpos Geniculados/metabolismo , Humanos , Ligantes , Masculino , Camundongos , Camundongos Knockout , Nervo Óptico/metabolismo , Receptor EphB1/genética , Receptor EphB1/metabolismo , Receptor EphB2/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Retina/metabolismo , Células Ganglionares da Retina/fisiologia , Transdução de Sinais/fisiologia , Vias Visuais/anatomia & histologia , Vias Visuais/embriologia
10.
Nat Commun ; 2: 431, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21847105

RESUMO

Graded expression of EphB and ephrin-B along the dorsoventral axis of the retina indicates a role for these bidirectional signalling molecules in dorsoventral-mediolateral retinocollicular mapping. Although previous studies have implicated EphB2 forward signalling in mice, the intracellular component of EphB2 essential for retinocollicular mapping is unknown as are the roles for EphB1, ephrin-B1, and ephrin-B2. Here we show that EphB2 tyrosine kinase catalytic activity and EphB1 intracellular signalling are key mediators of ventral-temporal retinal ganglion cell axon retinocollicular mapping, by likely interacting with ephrin-B1 in the superior colliculus. We further elucidate roles for the ephrin-B2 intracellular domain in retinocollicular mapping and present the unexpected finding that both dorsal and ventral-temporal retinal ganglion cell axons utilize reverse signalling for topographic mapping. These data demonstrate that both forward and reverse signalling initiated by EphB:ephrin-B interactions have a major role in dorsoventral retinal ganglion cell axon termination along the mediolateral axis of the superior colliculus.


Assuntos
Efrina-B1/metabolismo , Efrina-B2/metabolismo , Receptor EphB1/metabolismo , Receptor EphB2/metabolismo , Células Ganglionares da Retina/metabolismo , Transdução de Sinais , Colículos Superiores/metabolismo , Animais , Axônios/metabolismo , Efrina-B1/genética , Efrina-B2/genética , Feminino , Masculino , Camundongos , Camundongos Knockout , Ligação Proteica , Receptor EphB1/genética , Receptor EphB2/genética , Colículos Superiores/embriologia
11.
J Clin Invest ; 113(6): 856-66, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15067318

RESUMO

The prevailing paradigm is that in human rheumatoid arthritis (RA), the accumulation of monocytes and T cells in the joint, mediated in part by such CC chemokine receptors (CCRs) as CCR2 and CCR5, respectively, plays a central role in disease pathogenesis. To further validate this paradigm, we conducted proof-of-principle studies and tested the hypothesis that gene inactivation of Ccr2 or Ccr5 will ameliorate experimental RA. Contrary to our expectations, we found that in two well-established murine models of experimental RA, CCR2 expression in the hematopoietic cell compartment served as a negative regulator of autoantibody production as well as arthritic disease onset, severity, and resolution. In contrast, the RA phenotype in Ccr5-null mice was similar to that of WT mice. Remarkably, the collagen-induced arthritis phenotype of Ccr2-/- mice mimicked closely that of severe human RA, including production of rheumatoid factor, enhanced T cell production, and monocyte/macrophage accumulation in the joints. Our findings demonstrate an essential protective role of CCR2 expression in RA, indicate the existence of alternative receptors responsible for monocyte/macrophage accumulation to inflamed joints, and emphasize the need to clarify carefully the complex effects of the chemokine system in RA before they can be considered as therapeutic targets.


Assuntos
Artrite Experimental/genética , Artrite Reumatoide/genética , Receptores de Quimiocinas/genética , Animais , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Colágeno Tipo II/imunologia , Humanos , Camundongos , Modelos Animais , Receptores CCR2 , Receptores de Quimiocinas/imunologia , Receptores de Quimiocinas/metabolismo
12.
Atherosclerosis ; 167(1): 25-32, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12618265

RESUMO

The accumulation of macrophages and T lymphocytes in vessel walls is a hallmark of atherogenesis. It has recently been demonstrated in mouse models of atherosclerosis that full disease potential is dependent on several regulators of leukocyte trafficking, including the chemokine monocyte chemotactic protein 1 (MCP-1) and the chemokine receptors CCR2 and CXCR2. A possible role for the chemokine receptor CCR5 in atherogenesis has been suggested by CCR5 expression on macrophages, T cells, coronary endothelial cells and aortic smooth muscle cells and by the presence of CCR5 ligands in atherosclerotic plaques. Moreover, individuals who are naturally deficient in CCR5 were reported to be at reduced risk for severe coronary artery disease (CAD) and early myocardial infarction (MI). To investigate whether CCR5 is pro-atherogenic in mice, we generated CCR5-deficient mice and crossed them with atherosclerosis-prone apoE-deficient mice. Although CCR5-deficient mice exhibit defects in induced macrophage trafficking, mean atherosclerotic lesion area did not differ significantly between apoE-deficient mice and apoE/CCR5-deficient mice after 16 weeks on a diet of normal chow. Ribonuclease protection assays (RPA) on RNA isolated from plaques from both apoE-deficient and apoE/CCR5-deficient animals showed strong signals for the macrophage marker F4/80 but no evidence for expression of prominent markers of T and B lymphocytes. These results indicate that the early stages of plaque formation in this model of lipid-mediated atherogenesis do not depend on CCR5.


Assuntos
Apolipoproteínas E/metabolismo , Arteriosclerose/etiologia , Arteriosclerose/prevenção & controle , Quimiocina CCL2/fisiologia , Macrófagos/fisiologia , Receptores CCR5/deficiência , Análise de Variância , Animais , Northern Blotting , Quimiocina CCL2/genética , Lipoproteínas LDL/análise , Camundongos , Camundongos Knockout , Probabilidade , RNA/análise , Receptores de Quimiocinas/metabolismo , Sensibilidade e Especificidade , Índice de Gravidade de Doença
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA