Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Endocrinol Metab ; 319(1): E81-E90, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32396496

RESUMO

We have previously shown that systemic injection of erythropoietin-producing hepatocellular receptor A7 (EPHA7)-Fc raises serum luteinizing hormone (LH) levels before ovulation in female rats, indicating the induction of EPHA7 in ovulation. In this study, we aimed to identify the mechanism and hypothalamus-pituitary-ovary (HPO) axis level underlying the promotion of LH secretion by EPHA7. Using an ovariectomized (OVX) rat model, in conjunction with low-dose 17ß-estradiol (E2) treatment, we investigated the association between EPHA7-ephrin (EFN)A5 signaling and E2 negative feedback. Various rat models (OVX, E2-treated OVX, and abarelix treated) were injected with the recombinant EPHA7-Fc protein through the caudal vein to investigate the molecular mechanism underlying the promotion of LH secretion by EPHA7. Efna5 was observed strongly expressed in the arcuate nucleus of the female rat by using RNAscope in situ hybridization. Our results indicated that E2, combined with estrogen receptor (ER)α, but not ERß, inhibited Efna5 and gonadotropin-releasing hormone 1 (Gnrh1) expressions in the hypothalamus. In addition, the systemic administration of EPHA7-Fc restrained the inhibition of Efna5 and Gnrh1 by E2, resulting in increased Efna5 and Gnrh1 expressions in the hypothalamus as well as increased serum LH levels. Collectively, our findings demonstrated the involvement of EPHA7-EFNA5 signaling in the regulation of LH and the E2 negative feedback pathway in the hypothalamus, highlighting the functional role of EPHA7 in female reproduction.


Assuntos
Efrina-A5/metabolismo , Receptor alfa de Estrogênio/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hormônio Luteinizante/metabolismo , Precursores de Proteínas/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Efrina-A5/efeitos dos fármacos , Efrina-A5/genética , Estradiol/farmacologia , Receptor beta de Estrogênio/metabolismo , Estrogênios/farmacologia , Retroalimentação Fisiológica/efeitos dos fármacos , Retroalimentação Fisiológica/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/efeitos dos fármacos , Antagonistas de Hormônios/farmacologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/metabolismo , Hipotálamo/efeitos dos fármacos , Hormônio Luteinizante/efeitos dos fármacos , Oligopeptídeos/farmacologia , Ovariectomia , Ovário/efeitos dos fármacos , Ovário/metabolismo , Precursores de Proteínas/efeitos dos fármacos , Ratos , Receptor EphA7/genética , Receptor EphA7/metabolismo , Receptor EphA7/farmacologia , Proteínas Recombinantes
2.
Sci Rep ; 9(1): 263, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30670742

RESUMO

Tumour sequencing identifies highly recurrent point mutations in cancer driver genes, but rare functional mutations are hard to distinguish from large numbers of passengers. We developed a novel computational platform applying a multi-modal approach to filter out passengers and more robustly identify putative driver genes. The primary filter identifies enrichment of cancer mutations in CATH functional families (CATH-FunFams) - structurally and functionally coherent sets of evolutionary related domains. Using structural representatives from CATH-FunFams, we subsequently seek enrichment of mutations in 3D and show that these mutation clusters have a very significant tendency to lie close to known functional sites or conserved sites predicted using CATH-FunFams. Our third filter identifies enrichment of putative driver genes in functionally coherent protein network modules confirmed by literature analysis to be cancer associated. Our approach is complementary to other domain enrichment approaches exploiting Pfam families, but benefits from more functionally coherent groupings of domains. Using a set of mutations from 22 cancers we detect 151 putative cancer drivers, of which 79 are not listed in cancer resources and include recently validated cancer associated genes EPHA7, DCC netrin-1 receptor and zinc-finger protein ZNF479.


Assuntos
Neoplasias/genética , Oncogenes/genética , Mapas de Interação de Proteínas/genética , Biologia Computacional/métodos , Receptor DCC/genética , Receptor DCC/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Bases de Dados Genéticas/estatística & dados numéricos , Conjuntos de Dados como Assunto , Humanos , Mutação Puntual , Mapeamento de Interação de Proteínas/métodos , Receptor EphA7/genética , Receptor EphA7/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
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.
Neuroscience ; 274: 409-18, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-24909897

RESUMO

Within the first two postnatal weeks, corticostriatal axons from the primary somatosensory cortex (S1) form topographic projections that organize into characteristic bands of axon terminals in the dorsolateral striatum. Molecules regulating the development of these topographically organized projections are currently unknown. Thus, the present study investigated whether EphA receptor tyrosine kinases, which regulate axonal guidance in the visual system via axon repulsion, could participate in the formation of corticostriatal connections during development. Prior studies indicate that EphA7-expressing striatal neurons are organized into banded compartments resembling the matrisome innervation pattern formed by cortical afferents from the S1 cortex and that ephrin-A5, a known EphA7 ligand, is expressed in a medial (high) to lateral (low) gradient in S1. Thus, we hypothesized that the organization of EphA7-expressing striatal neurons in banded domains provides a repulsive barrier preventing corticostriatal axons containing EphA7-ligands from innervating inappropriate regions of the striatum. To evaluate this, we injected the anterograde tracer, biotinylated dextran amine (BDA), into two locations in medial areas of S1 (the anterior and posterior whisker fields), which are reported to express high levels of ephrin-A5 during development. Injections were made in mouse pups on postnatal day 9 (P9) and the animals were processed for immunohistochemistry on P12. Our data demonstrate that projections from both the forelimb/anterior whisker field and the posterior whisker field avoid EphA7-expressing neurons and terminate in a banded pattern in regions with very low EphA7-expression. We also determined that corticothalamic projections from medial S1 also exhibit a restricted distribution in the thalamus and avoid neurons expressing EphA7. Thus, our results support the hypothesis that the anatomical organization of striatal and thalamic neurons expressing EphA7 receptors restricts the topographic distribution of cortical afferents from medial regions of S1 which express high levels of ephrin-A5.


Assuntos
Corpo Estriado/anatomia & histologia , Neurônios/citologia , Receptor EphA7/metabolismo , Córtex Somatossensorial/anatomia & histologia , Tálamo/anatomia & histologia , Animais , Biotina/análogos & derivados , Corpo Estriado/metabolismo , Dextranos , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Imuno-Histoquímica , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Vias Neurais/anatomia & histologia , Vias Neurais/metabolismo , Técnicas de Rastreamento Neuroanatômico , Neurônios/metabolismo , Tálamo/metabolismo , Vibrissas
5.
Cereb Cortex ; 23(4): 775-85, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22490549

RESUMO

Auditory stimulus representations are dynamically maintained by ascending and descending projections linking the auditory cortex (Actx), medial geniculate body (MGB), and inferior colliculus. Although the extent and topographic specificity of descending auditory corticofugal projections can equal or surpass that of ascending corticopetal projections, little is known about the molecular mechanisms that guide their development. Here, we used in utero gene electroporation to examine the role of EphA receptor signaling in the development of corticothalamic (CT) and corticocollicular connections. Early in postnatal development, CT axons were restricted to a deep dorsal zone (DDZ) within the MGB that expressed low levels of the ephrin-A ligand. By hearing onset, CT axons had innervated surrounding regions of MGB in control-electroporated mice but remained fixed within the DDZ in mice overexpressing EphA7. In vivo neurophysiological recordings demonstrated a corresponding reduction in spontaneous firing rate, but no changes in sound-evoked responsiveness within MGB regions deprived of CT innervation. Structural and functional CT disruption occurred without gross alterations in thalamocortical connectivity. These data demonstrate a potential role for EphA/ephrin-A signaling in the initial guidance of corticofugal axons and suggest that "genetic rewiring" may represent a useful functional tool to alter cortical feedback without silencing Actx.


Assuntos
Córtex Auditivo , Vias Auditivas/fisiologia , Mapeamento Encefálico , Corpos Geniculados/fisiologia , Receptor EphA7/metabolismo , Transdução de Sinais/fisiologia , Estimulação Acústica , Fatores Etários , Aminoácidos , Animais , Animais Recém-Nascidos , Córtex Auditivo/embriologia , Córtex Auditivo/crescimento & desenvolvimento , Córtex Auditivo/metabolismo , Axônios/fisiologia , Eletroencefalografia , Eletroporação , Embrião de Mamíferos , Potenciais Evocados Auditivos/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/genética , Masculino , Camundongos , Camundongos Transgênicos , RNA Mensageiro/metabolismo , Receptor EphA7/genética , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
6.
J Comp Neurol ; 521(3): 626-37, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22821544

RESUMO

Corticothalamic (CT) feedback outnumbers thalamocortical projections and regulates sensory information processing at the level of the thalamus. It is well established that EphA7, a member of EphA receptor family, is involved in the topographic mapping of CT projections. The aim of the present study was to dissect the precise impact of EphA7 on each step of CT growth. We used in utero electroporation-mediated EphA7 overexpression in developing somatosensory CT axons to dissect EphA7/ephrin-A-dependent mechanisms involved in regulating both initial targeting and postnatal growth of the CT projections. Our data revealed that topographic maps of cortical afferents in the ventrobasal complex and medial part of the posterior complex in the thalamus become discernible shortly after birth and are fully established by the second postnatal week. This process starts with the direct ingrowth of the CT axons to the designated areas within target thalamic nuclei and by progressive increase of axonal processes in the terminal zones. Large-scale overproduction and elimination of exuberant widespread axonal branches outside the target zone was not observed. Each developmental event was coordinated by spatially and temporally different responsiveness of CT axons to the ephrin-A gradient in thalamic nuclei, as well as by the matching levels of EphA7 in CT axons and ephrin-As in thalamic nuclei. These results support the concept that the topographic connections between the maps in the cerebral cortex and corresponding thalamic nuclei are genetically prespecified to a large extent, and established by precise spatiotemporal molecular mechanisms that involve the Eph family of genes.


Assuntos
Efrina-A1/metabolismo , Receptor EphA7/metabolismo , Transdução de Sinais/fisiologia , Córtex Somatossensorial/embriologia , Tálamo/embriologia , Vias Aferentes/citologia , Vias Aferentes/embriologia , Vias Aferentes/metabolismo , Animais , Axônios/metabolismo , Mapeamento Encefálico , Feminino , Camundongos , Camundongos Endogâmicos , Gravidez , Córtex Somatossensorial/citologia , Córtex Somatossensorial/metabolismo , Tálamo/citologia , Tálamo/metabolismo
7.
J Neurosci ; 28(35): 8724-34, 2008 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-18753373

RESUMO

Transcription factor Pax6 exerts a prominent rostrolateral(high) to caudomedial(low) expression gradient in the cortical progenitors and have been implicated in regulation of area identity in the mammalian cortex. Herein, we analyzed the role of Pax6 in molecular arealization and development of thalamocortical connections in the juvenile cortex-specific conditional Pax6 knock-out mice (Pax6cKO). Using a set of molecular markers of positional identity (Id2, Cadherin6, COUP-TF1, RZRbeta, and EphA7), we show that, in the juvenile Pax6cKO, the relative size of caudal cortical areas (putative visual and somatosensory) are mildly enlarged, whereas the rostral domain (putative motor) is severely reduced. Despite the rostral shift of graded expression of areal markers, the distribution of area-specific thalamocortical and corticofugal projections appear normal in the Pax6cKO. This indicates that change of the size of cortical areas is not accompanied by a change in cortical identity. We show furthermore that, despite a severe depletion of supragranular cortical layers and accumulation of cells along the pallial-subpallial boundary, thalamocortical fibers establish a periphery-related pattern of the somatosensory cortex in normal position in Pax6cKO. Our findings indicate that Pax6 expression gradients in cortical progenitors do not directly impart thalamocortical or corticofugal areal identity.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/genética , Morfogênese/genética , Fatores de Transcrição Box Pareados/deficiência , Tálamo/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Fator I de Transcrição COUP/metabolismo , Caderinas/metabolismo , Córtex Cerebral/metabolismo , Proteínas do Olho , Proteínas de Homeodomínio/genética , Proteína 2 Inibidora de Diferenciação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vias Neurais/metabolismo , Membro 2 do Grupo F da Subfamília 1 de Receptores Nucleares , Fator de Transcrição PAX6 , Receptor EphA7/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Repressoras , Tálamo/metabolismo , Fatores de Transcrição/genética
8.
Science ; 313(5792): 1408-13, 2006 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-16902088

RESUMO

In the mouse trigeminal pathway, sensory inputs from distinct facial structures, such as whiskers or lower jaw and lip, are topographically mapped onto the somatosensory cortex through relay stations in the thalamus and hindbrain. In the developing hindbrain, the mechanisms generating such maps remain elusive. We found that in the principal sensory nucleus, the whisker-related map is contributed by rhombomere 3-derived neurons, whereas the rhombomere 2-derived progeny supply the lower jaw and lip representation. Moreover, early Hoxa2 expression in neuroepithelium prevents the trigeminal nerve from ectopically projecting to the cerebellum, whereas late expression in the principal sensory nucleus promotes selective arborization of whisker-related afferents and topographic connectivity to the thalamus. Hoxa2 inactivation further results in the absence of whisker-related maps in the postnatal brain. Thus, Hoxa2- and rhombomere 3-dependent cues determine the whisker area map and are required for the assembly of the whisker-to-barrel somatosensory circuit.


Assuntos
Proteínas de Homeodomínio/fisiologia , Rombencéfalo/embriologia , Córtex Somatossensorial/anatomia & histologia , Nervo Trigêmeo/embriologia , Vibrissas/inervação , Vias Aferentes , Animais , Axônios/ultraestrutura , Face/inervação , Proteínas de Homeodomínio/genética , Lábio/inervação , Mandíbula/embriologia , Mandíbula/inervação , Camundongos , Camundongos Transgênicos , Mutação , Neurônios Aferentes/citologia , Receptor EphA4/metabolismo , Receptor EphA7/metabolismo , Rombencéfalo/citologia , Rombencéfalo/metabolismo , Córtex Somatossensorial/embriologia , Tálamo/embriologia , Tálamo/metabolismo , Gânglio Trigeminal/embriologia , Gânglio Trigeminal/metabolismo , Nervo Trigêmeo/fisiologia , Núcleos Ventrais do Tálamo/embriologia
9.
Neuron ; 48(4): 563-75, 2005 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-16301174

RESUMO

Molecular mechanisms generating the topographic organization of corticothalamic (CT) circuits, which comprise more than three-quarters of the synaptic inputs onto sensory relay neurons, and their interdependence with thalamocortical (TC) axon development are unknown. Using in utero electroporation-mediated gene transfer, we show that EphA7-mediated signaling on neocortical axons controls the within-nucleus topography of CT projections in the thalamus. Notably, CT axons that mis-express EphA7 do not shift the relative positioning of their pathway within the subcortical telencephalon (ST), indicating that they do not depend upon EphA7/ephrin-A signaling in the ST for establishing this topography. Moreover, mis-expression of cortical EphA7 results in disrupted topography of CT projections, but unchanged inter- and intra-areal topography of TC projections. Our results support a model in which EphA/ephrin-A signaling controls independently the precision with which CT and TC projections develop, yet is essential for establishing their topographic reciprocity.


Assuntos
Axônios/fisiologia , Córtex Cerebral/fisiologia , Receptor EphA7/fisiologia , Tálamo/fisiologia , Animais , Axônios/metabolismo , Mapeamento Encefálico , Córtex Cerebral/metabolismo , Camundongos , Camundongos Endogâmicos , Neocórtex/anatomia & histologia , Neocórtex/metabolismo , Vias Neurais/fisiologia , Receptor EphA5/metabolismo , Receptor EphA7/metabolismo , Transmissão Sináptica/fisiologia , Telencéfalo/fisiologia , Tálamo/metabolismo
10.
J Comp Neurol ; 456(3): 203-16, 2003 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-12528186

RESUMO

Parcellation of the mammalian cerebral cortex into distinct areas is essential for proper cortical function; however, the developmental program that results in the genesis of distinct areas is not fully understood. We examined the expression of members of the EphA family-the EphA receptor tyrosine kinases and the ephrin-A ligands-within the developing mouse cerebral cortex, with the aim of characterizing this component of the molecular landscape during cortical parcellation. We found that specific embryonic zones, such as the ventricular, subventricular, intermediate, subplate, and marginal zones, as well as the cortical plate, were positive for particular EphA genes early in corticogenesis (E12-E15). Along with this zone-selective expression, several genes (EphA3, EphA4, EphA5) were evenly expressed along the axes of the developing cortex, whereas one family member (EphA7) was expressed in a distinct anteroposterior pattern. Later in corticogenesis (E16-E18), other EphA family members became selectively expressed, but only within the cortical plate: EphA6 was present posteriorly, and ephrin-A5 was expressed within a middle region. At birth, patterning of EphA gene expression was striking. Thus, we found that the expression of a single EphA gene or a combination of family members can define distinct embryonic zones and anteroposterior regions of the neocortex during development. To examine whether cellular context affects the patterning of EphA expression, we examined gene expression in embryonic cortical cells grown in vitro, such that all cellular contacts are lacking, and in Mash-1 mutant mice, in which thalamocortical connections do not form. We found that the expression patterns of most EphA family members remained stable in these scenarios, whereas the pattern of ephrin-A5 was altered. Taken together, this work provides a comprehensive picture of EphA family expression during mouse corticogenesis and demonstrates that most EphA expression profiles are cell intrinsically based, whereas ephrin-A5 is plastically regulated.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Receptores da Família Eph/metabolismo , Vias Aferentes/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Técnicas de Cultura de Células , Córtex Cerebral/embriologia , Proteínas de Ligação a DNA/genética , Expressão Gênica , Hibridização In Situ , Camundongos , Camundongos Mutantes , Receptor EphA3/metabolismo , Receptor EphA4/metabolismo , Receptor EphA5/metabolismo , Receptor EphA7/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tálamo/metabolismo , Fatores de Tempo , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA