Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Ophthalmol Sci ; 3(4): 100390, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38025164

RESUMO

Purpose: The Retinal Ganglion Cell (RGC) Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) consortium was founded in 2021 to help address the numerous scientific and clinical obstacles that impede development of vision-restorative treatments for patients with optic neuropathies. The goals of the RReSTORe consortium are: (1) to define and prioritize the most critical challenges and questions related to RGC regeneration; (2) to brainstorm innovative tools and experimental approaches to meet these challenges; and (3) to foster opportunities for collaborative scientific research among diverse investigators. Design and Participants: The RReSTORe consortium currently includes > 220 members spanning all career stages worldwide and is directed by an organizing committee comprised of 15 leading scientists and physician-scientists of diverse backgrounds. Methods: Herein, we describe the structure and organization of the RReSTORe consortium, its activities to date, and the perceived impact that the consortium has had on the field based on a survey of participants. Results: In addition to helping propel the field of regenerative medicine as applied to optic neuropathies, the RReSTORe consortium serves as a framework for developing large collaborative groups aimed at tackling audacious goals that may be expanded beyond ophthalmology and vision science. Conclusions: The development of innovative interventions capable of restoring vision for patients suffering from optic neuropathy would be transformative for the ophthalmology field, and may set the stage for functional restoration in other central nervous system disorders. By coordinating large-scale, international collaborations among scientists with diverse and complementary expertise, we are confident that the RReSTORe consortium will help to accelerate the field toward clinical translation. Financial Disclosures: Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.

2.
Mol Neurodegener ; 18(1): 64, 2023 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-37735444

RESUMO

Retinal ganglion cell (RGC) death in glaucoma and other optic neuropathies results in irreversible vision loss due to the mammalian central nervous system's limited regenerative capacity. RGC repopulation is a promising therapeutic approach to reverse vision loss from optic neuropathies if the newly introduced neurons can reestablish functional retinal and thalamic circuits. In theory, RGCs might be repopulated through the transplantation of stem cell-derived neurons or via the induction of endogenous transdifferentiation. The RGC Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) Consortium was established to address the challenges associated with the therapeutic repair of the visual pathway in optic neuropathy. In 2022, the RReSTORe Consortium initiated ongoing international collaborative discussions to advance the RGC repopulation field and has identified five critical areas of focus: (1) RGC development and differentiation, (2) Transplantation methods and models, (3) RGC survival, maturation, and host interactions, (4) Inner retinal wiring, and (5) Eye-to-brain connectivity. Here, we discuss the most pertinent questions and challenges that exist on the path to clinical translation and suggest experimental directions to propel this work going forward. Using these five subtopic discussion groups (SDGs) as a framework, we suggest multidisciplinary approaches to restore the diseased visual pathway by leveraging groundbreaking insights from developmental neuroscience, stem cell biology, molecular biology, optical imaging, animal models of optic neuropathy, immunology & immunotolerance, neuropathology & neuroprotection, materials science & biomedical engineering, and regenerative neuroscience. While significant hurdles remain, the RReSTORe Consortium's efforts provide a comprehensive roadmap for advancing the RGC repopulation field and hold potential for transformative progress in restoring vision in patients suffering from optic neuropathies.


Assuntos
Doenças do Nervo Óptico , Células Ganglionares da Retina , Animais , Humanos , Retina , Encéfalo , Diferenciação Celular , Mamíferos
3.
J Neurosci ; 43(32): 5769-5778, 2023 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-37344233

RESUMO

Semaphorins and Plexins form ligand/receptor pairs that are crucial for a wide range of developmental processes from cell proliferation to axon guidance. The ability of semaphorins to act both as signaling receptors and ligands yields a multitude of responses. Here, we describe a novel role for Semaphorin-6D (Sema6D) and Plexin-A1 in the positioning and targeting of retinogeniculate axons. In Plexin-A1 or Sema6D mutant mice of either sex, the optic tract courses through, rather than along, the border of the dorsal lateral geniculate nucleus (dLGN), and some retinal axons ectopically arborize adjacent and lateral to the optic tract rather than defasciculating and entering the target region. We find that Sema6D and Plexin-A1 act together in a dose-dependent manner, as the number of the ectopic retinal projections is altered in proportion to the level of Sema6D or Plexin-A1 expression. Moreover, using retinal in utero electroporation of Sema6D or Plexin-A1 shRNA, we show that Sema6D and Plexin-A1 are both required in retinal ganglion cells for axon positioning and targeting. Strikingly, nonelectroporated retinal ganglion cell axons also mistarget in the tract region, indicating that Sema6D and Plexin-A1 can act non-cell-autonomously, potentially through axon-axon interactions. These data provide novel evidence for a dose-dependent and non-cell-autonomous role for Sema6D and Plexin-A1 in retinal axon organization in the optic tract and dLGN.SIGNIFICANCE STATEMENT Before innervating their central brain targets, retinal ganglion cell axons fasciculate in the optic tract and then branch and arborize in their target areas. Upon deletion of the guidance molecules Plexin-A1 or Semaphorin-6D, the optic tract becomes disorganized near and extends within the dorsal lateral geniculate nucleus. In addition, some retinal axons form ectopic aggregates within the defasciculated tract. Sema6D and Plexin-A1 act together as a receptor-ligand pair in a dose-dependent manner, and non-cell-autonomously, to produce this developmental aberration. Such a phenotype highlights an underappreciated role for axon guidance molecules in tract cohesion and appropriate defasciculation near, and arborization within, targets.


Assuntos
Células Ganglionares da Retina , Semaforinas , Animais , Camundongos , Axônios/fisiologia , Ligantes , Células Ganglionares da Retina/metabolismo , Semaforinas/genética , Semaforinas/metabolismo
5.
J Comp Neurol ; 527(3): 508-521, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29744881

RESUMO

In the developing mouse optic tract, retinal ganglion cell (RGC) axon position is organized by topography and laterality (i.e., eye-specific or ipsi- and contralateral segregation). Our lab previously showed that ipsilaterally projecting RGCs are segregated to the lateral aspect of the developing optic tract and found that ipsilateral axons self-fasciculate to a greater extent than contralaterally projecting RGC axons in vitro. However, the full complement of axon-intrinsic and -extrinsic factors mediating eye-specific segregation in the tract remain poorly understood. Glia, which are known to express several guidance cues in the visual system and regulate the navigation of ipsilateral and contralateral RGC axons at the optic chiasm, are natural candidates for contributing to eye-specific pre-target axon organization. Here, we investigate the spatiotemporal expression patterns of both putative astrocytes (Aldh1l1+ cells) and microglia (Iba1+ cells) in the embryonic and neonatal optic tract. We quantified the localization of ipsilateral RGC axons to the lateral two-thirds of the optic tract and analyzed glia position and distribution relative to eye-specific axon organization. While our results indicate that glial segregation patterns do not strictly align with eye-specific RGC axon segregation in the tract, we identify distinct spatiotemporal organization of both Aldh1l1+ cells and microglia in and around the developing optic tract. These findings inform future research into molecular mechanisms of glial involvement in RGC axon growth and organization in the developing retinogeniculate pathway.


Assuntos
Família Aldeído Desidrogenase 1/metabolismo , Neuroglia/metabolismo , Trato Óptico/embriologia , Trato Óptico/metabolismo , Retinal Desidrogenase/metabolismo , Células Ganglionares da Retina/metabolismo , Fatores Etários , Família Aldeído Desidrogenase 1/análise , Animais , Axônios/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Trato Óptico/citologia , Retinal Desidrogenase/análise , Vias Visuais/citologia , Vias Visuais/embriologia , Vias Visuais/metabolismo
6.
J Comp Neurol ; 526(7): 1077-1096, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29322522

RESUMO

Prior to forming and refining synaptic connections, axons of projection neurons navigate long distances to their targets. While much is known about guidance cues for axon navigation through intermediate choice points, whether and how axons are organized within tracts is less clear. Here we analyze the organization of retinal ganglion cell (RGC) axons in the developing mouse retinogeniculate pathway. RGC axons are organized by both eye-specificity and topography in the optic nerve and tract: ipsilateral RGC axons are segregated from contralateral axons and are offset laterally in the tract relative to contralateral axon topographic position. To identify potential cell-autonomous factors contributing to the segregation of ipsilateral and contralateral RGC axons in the visual pathway, we assessed their fasciculation behavior in a retinal explant assay. Ipsilateral RGC neurites self-fasciculate more than contralateral neurites in vitro and maintain this difference in the presence of extrinsic chiasm cues. To further probe the role of axon self-association in circuit formation in vivo, we examined RGC axon organization and fasciculation in an EphB1-/- mutant, in which a subset of ipsilateral RGC axons aberrantly crosses the midline but targets the ipsilateral zone in the dorsal lateral geniculate nucleus on the opposite side. Aberrantly crossing axons retain their association with ipsilateral axons in the contralateral tract, indicating that cohort-specific axon affinity is maintained independently of guidance signals present at the midline. Our results provide a comprehensive assessment of RGC axon organization in the retinogeniculate pathway and suggest that axon self-association contributes to pre-target axon organization.


Assuntos
Axônios/fisiologia , Nervo Óptico/fisiologia , Células Ganglionares da Retina/citologia , Vias Visuais , Aminoácidos/metabolismo , Animais , Animais Recém-Nascidos , Embrião de Mamíferos , Olho/citologia , Olho/inervação , Fasciculação , Lateralidade Funcional , Técnicas In Vitro , Filamentos Intermediários/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Nervo Óptico/embriologia , Nervo Óptico/crescimento & desenvolvimento , Receptor EphB1/genética , Receptor EphB1/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Vias Visuais/anatomia & histologia , Vias Visuais/embriologia , Vias Visuais/crescimento & desenvolvimento
7.
J Neurosci ; 36(42): 10707-10722, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27798125

RESUMO

Although much is known about the regenerative capacity of retinal ganglion cells, very significant barriers remain in our ability to restore visual function following traumatic injury or disease-induced degeneration. Here we summarize our current understanding of the factors regulating axon guidance and target engagement in regenerating axons, and review the state of the field of neural regeneration, focusing on the visual system and highlighting studies using other model systems that can inform analysis of visual system regeneration. This overview is motivated by a Society for Neuroscience Satellite meeting, "Reconnecting Neurons in the Visual System," held in October 2015 sponsored by the National Eye Institute as part of their "Audacious Goals Initiative" and co-organized by Carol Mason (Columbia University) and Michael Crair (Yale University). The collective wisdom of the conference participants pointed to important gaps in our knowledge and barriers to progress in promoting the restoration of visual system function. This article is thus a summary of our existing understanding of visual system regeneration and provides a blueprint for future progress in the field.


Assuntos
Encéfalo/fisiologia , Fenômenos Fisiológicos Oculares , Vias Visuais/fisiologia , Animais , Axônios/fisiologia , Humanos , Nervo Óptico/fisiologia , Células Ganglionares da Retina/fisiologia , Vias Visuais/citologia
8.
Elife ; 52016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27648578

RESUMO

The regulation of protein degradation is essential for maintaining the appropriate environment to coordinate complex cell signaling events and to promote cellular remodeling. The Autophagy linked FYVE protein (Alfy), previously identified as a molecular scaffold between the ubiquitinated cargo and the autophagic machinery, is highly expressed in the developing central nervous system, indicating that this pathway may have yet unexplored roles in neurodevelopment. To examine this possibility, we used mouse genetics to eliminate Alfy expression. We report that this evolutionarily conserved protein is required for the formation of axonal tracts throughout the brain and spinal cord, including the formation of the major forebrain commissures. Consistent with a phenotype reflecting a failure in axon guidance, the loss of Alfy in mice disrupts localization of glial guidepost cells, and attenuates axon outgrowth in response to Netrin-1. These findings further support the growing indication that macroautophagy plays a key role in the developing CNS.


Assuntos
Encéfalo/embriologia , Vias Neurais/embriologia , Neurônios/fisiologia , Proteínas de Transporte Vesicular/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Relacionadas à Autofagia , Técnicas de Inativação de Genes , Camundongos Endogâmicos C57BL
9.
Neural Dev ; 10: 23, 2015 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-26492970

RESUMO

BACKGROUND: In the visual system of most binocular vertebrates, the axons of retinal ganglion cells (RGCs) diverge at the diencephalic midline and extend to targets on both ipsi- and contralateral sides of the brain. While a molecular mechanism explaining ipsilateral guidance decisions has been characterized, less is known of how RGC axons cross the midline. RESULTS: Here, we took advantage of the zebrafish, in which all RGC axons project contralaterally at the optic chiasm, to characterize Islr2 as an RGC receptor required for complete retinal axon midline crossing. We used a systematic extracellular protein-protein interaction screening assay to identify two Vasorin paralogs, Vasna and Vasnb, as specific Islr2 ligands. Antibodies against Vasna and Vasnb reveal cellular populations surrounding the retinal axon pathway, suggesting the involvement of these proteins in guidance decisions made by axons of the optic nerve. Specifically, Vasnb marks the membranes of a cellular barricade located anteriorly to the optic chiasm, a structure termed the "glial knot" in higher vertebrates. Loss of function mutations in either vasorin paralog, individually or combined, however, do not exhibit an overt retinal axon projection phenotype, suggesting that additional midline factors, acting either independently or redundantly, compensate for their loss. Analysis of Islr2 knockout mice supports a scenario in which Islr2 controls the coherence of RGC axons through the ventral midline and optic tract. CONCLUSIONS: Although stereotypic guidance of RGC axons at the vertebrate optic chiasm is controlled by multiple, redundant mechanisms, and despite the differences in ventral diencephalic tissue architecture, we identify a novel role for the LRR receptor Islr2 in ensuring proper axon navigation at the optic chiasm of both zebrafish and mouse.


Assuntos
Axônios/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/fisiologia , Quiasma Óptico/embriologia , Retina/embriologia , Animais , Padronização Corporal/fisiologia , Processamento de Imagem Assistida por Computador , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Vias Visuais/embriologia , Peixe-Zebra
10.
Dev Neurobiol ; 75(12): 1385-401, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25788284

RESUMO

During development of the mammalian eye, the first retinal ganglion cells (RGCs) that extend to the brain are located in the dorsocentral (DC) retina. These RGCs extend to either ipsilateral or contralateral targets, but the ipsilateral projections do not survive into postnatal periods. The function and means of disappearance of the transient ipsilateral projection are not known. We have followed the course of this transient early ipsilateral cohort of RGCs, paying attention to how far they extend, whether they enter targets and if so, which ones, and the time course of their disappearance. The DC ipsilateral RGC axons were traced using DiI labeling at E13.5 and E15.5 to compare the proportion of ipsi- versus contralateral projections during the first period of growth. In utero electroporation of E12.5 retina with GFP constructs was used to label axons that could be visualized at succeeding time points into postnatal ages. Our results show that the earliest ipsilateral axons grow along the cellular border of the brain, and are segregated from the laterally positioned contralateral axons from the same retinal origin. In agreement with previous reports, although many early RGCs extend ipsilaterally, after E16 their number rapidly declines. Nonetheless, some ipsilateral axons from the DC retina enter the superior colliculus and arborize minimally, but very few enter the dorsal lateral geniculate nucleus and those that do extend only short branches. While the mechanism of selective axonal disappearance remains elusive, these data give further insight into establishment of the visual pathways.


Assuntos
Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Células Ganglionares da Retina/citologia , Vias Visuais/citologia , Vias Visuais/crescimento & desenvolvimento , Animais , Axônios , Estudos de Coortes , Eletroporação , Lateralidade Funcional , Imuno-Histoquímica , Camundongos Endogâmicos C57BL , Técnicas de Rastreamento Neuroanatômico
11.
J Neurosci ; 34(20): 6746-58, 2014 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-24828630

RESUMO

Thalamus is a potent driver of cortical activity even though cortical synapses onto excitatory layer 4 neurons outnumber thalamic synapses 10 to 1. Previous in vitro studies have proposed that thalamocortical (TC) synapses are stronger than corticocortical (CC) synapses. Here, we investigated possible anatomical and physiological differences between these inputs in the rat in vivo. We developed a high-throughput light microscopy method, validated by electron microscopy, to completely map the locations of synapses across an entire dendritic tree. This demonstrated that TC synapses are slightly more proximal to the soma than CC synapses, but detailed compartmental modeling predicted that dendritic filtering does not appreciably favor one synaptic class over another. Measurements of synaptic strength in intact animals confirmed that both TC and CC synapses are weak and approximately equivalent. We conclude that thalamic effectiveness does not rely on enhanced TC strength, but rather on coincident activation of converging inputs.


Assuntos
Córtex Cerebral/fisiologia , Dendritos/fisiologia , Neurônios/fisiologia , Sinapses/fisiologia , Tálamo/fisiologia , Potenciais de Ação/fisiologia , Animais , Espinhas Dendríticas/fisiologia , Modelos Neurológicos , Vias Neurais/fisiologia , Ratos , Ratos Wistar , Transmissão Sináptica/fisiologia
12.
PLoS One ; 8(3): e57184, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23526936

RESUMO

BACKGROUND: The absence or deficiency of melanin as in albinos, has detrimental effects on retinal development that include aberrant axonal projections from eye to brain and impaired vision. In pigmented retinal pigment epithelium (RPE), dihydroxyphenalanine (L-Dopa), an intermediate in the synthetic path for melanin, has been hypothesized to regulate the tempo of neurogenesis. The time course of expression of retinal L-Dopa, whether it is harbored exclusively in the RPE, the extent of deficiency in albinos compared to isogenic controls, and whether L-Dopa can be restored if exogenously delivered to the albino have been unknown. METHODOLOGY/ PRINCIPAL FINDINGS: L-Dopa and catecholamines including dopamine extracted from retinas of pigmented (C57BL/6J) and congenic albino (C57BL/6J-tyr(c2j) ) mice, were measured throughout development beginning at E10.5 and at maturity. L-Dopa, but not dopamine nor any other catecholamine, appears in pigmented retina as soon as tyrosinase is expressed in RPE at E10.5. In pigmented retina, L-Dopa content increases throughout pre- and postnatal development until the end of the first postnatal month after which it declines sharply. This time course reflects the onset and completion of retinal development. L-Dopa is absent from embryonic albino retina and is greatly reduced in postnatal albino retina compared to pigmented retina. Dopamine is undetectable in both albino and pigmented retinas until after the postnatal expression of the neuronal enzyme tyrosine hydroxylase. If provided to pregnant albino mothers, L-Dopa accumulates in the RPE of the fetuses. CONCLUSIONS: L-Dopa in pigmented RPE is most abundant during development after which content declines. This L-Dopa is not converted to dopamine. L-Dopa is absent or at low levels in albino retina and can be restored to the RPE by administration in utero. These findings further implicate L-Dopa as a factor in the RPE that could influence development, and demonstrate that administration of L-Dopa could be a means to rescue developmental abnormalities characteristic of albinos.


Assuntos
Albinismo/embriologia , Albinismo/metabolismo , Levodopa/metabolismo , Retina/embriologia , Retina/metabolismo , Albinismo/genética , Animais , Dopamina/metabolismo , Feminino , Levodopa/administração & dosagem , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Monofenol Mono-Oxigenase/genética , Monofenol Mono-Oxigenase/metabolismo , Neurogênese , Gravidez , Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo
14.
J Neurosci ; 32(14): 4821-6, 2012 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-22492037

RESUMO

The divergence of retinal ganglion cell (RGC) axons into ipsilateral and contralateral projections at the optic chiasm and the subsequent segregation of retinal inputs into eye-specific domains in their target, the dorsal lateral geniculate nucleus (dLGN), are crucial for binocular vision. In albinism, affected individuals exhibit a lack or reduction of pigmentation in the eye and skin, a concomitant reduced ipsilateral projection, and diverse visual defects. Here we investigate how such altered decussation affects eye-specific retinogeniculate targeting in albino mice using the C57BL/6 Tyr(c-2J/c-2J) strain, in which tyrosinase, necessary for melanogenesis, is mutated. In albino mice, fewer RGCs from the ventrotemporal (VT) retina project ipsilaterally, reflected in a decrease in cells expressing ipsilateral markers. In addition, a population of RGCs from the VT retina projects contralaterally and, within the dLGN, their axons cluster into a patch separated from the contralateral termination area. Furthermore, eye-specific segregation is not complete in the albino dLGN and, upon perturbing postnatal retinal activity with epibatidine, the ipsilateral projection fragments and the aberrant contralateral patch disappears. These results suggest that the defects in afferent targeting and activity-dependent refinement in the albino dLGN arise from RGC misspecification together with potential perturbations of early activity patterns in the albino retina.


Assuntos
Albinismo/patologia , Axônios/patologia , Olho/patologia , Corpos Geniculados/patologia , Vias Visuais/patologia , Albinismo/genética , Animais , Animais Recém-Nascidos , Olho/crescimento & desenvolvimento , Olho/inervação , Feminino , Corpos Geniculados/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Neurogênese/genética , Células Ganglionares da Retina/patologia , Vias Visuais/crescimento & desenvolvimento
15.
Neuron ; 73(1): 79-91, 2012 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-22243748

RESUMO

Dendrites achieve characteristic spacing patterns during development to ensure appropriate coverage of territories. Mechanisms of dendrite positioning via repulsive dendrite-dendrite interactions are beginning to be elucidated, but the control, and importance, of dendrite positioning relative to their substrate is poorly understood. We found that dendritic branches of Drosophila dendritic arborization sensory neurons can be positioned either at the basal surface of epidermal cells, or enclosed within epidermal invaginations. We show that integrins control dendrite positioning on or within the epidermis in a cell autonomous manner by promoting dendritic retention on the basal surface. Loss of integrin function in neurons resulted in excessive self-crossing and dendrite maintenance defects, the former indicating a role for substrate interactions in self-avoidance. In contrast to a contact-mediated mechanism, we find that integrins prevent crossings that are noncontacting between dendrites in different three-dimensional positions, revealing a requirement for combined dendrite-dendrite and dendrite-substrate interactions in self-avoidance.


Assuntos
Padronização Corporal/fisiologia , Dendritos/fisiologia , Integrinas/metabolismo , Células Receptoras Sensoriais/citologia , Animais , Animais Geneticamente Modificados , Padronização Corporal/genética , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Dendritos/ultraestrutura , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Células Epidérmicas , Epiderme/fisiologia , Epiderme/ultraestrutura , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/genética , Peroxidase do Rábano Silvestre/metabolismo , Integrinas/genética , Larva , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Modelos Biológicos , Morfogênese , Órgãos dos Sentidos/citologia , Células Receptoras Sensoriais/metabolismo
16.
Dev Neurobiol ; 72(6): 789-804, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22095825

RESUMO

Precise regulation of neuroprogenitor cell proliferation and differentiation is required for successful brain development, but the factors that contribute to this are only incompletely understood. The transcription factor ATF5 promotes proliferation of cerebral cortical neuroprogenitor cells and its down regulation permits their differentiation. Here, we examine the expression and regulation of ATF5 in cerebellar granule neuron progenitor cells (CGNPs) as well as the role of ATF5 in the transition of CGNPs to postmitotic cerebellar granule neurons (GCNs). We find that ATF5 is expressed by proliferating CGNPs in both the embryonic and postnatal cerebellar external granule layer (EGL) and in the rhombic lip, the embryonic structure from which the EGL arises. In contrast, ATF5 is undetectable in postmitotic GCNs. In highly enriched dissociated cultures of CGNPs and CGNs, ATF5 is expressed only in CGNPs. Constitutive ATF5 expression in CGNPs does not affect their proliferation or exit from the cell cycle. In contrast, in presence of sonic hedgehog (Shh), a mitogen for CGNPs, constitutively expressed ATF5 promotes CGNP proliferation and delays their cell cycle exit and differentiation. Conversely, ATF5 loss-of-function conferred by a dominant-negative form of ATF5 significantly diminishes Shh-stimulated CGNP proliferation and promotes differentiation. In parallel with its stimulation of CGNP proliferation, Shh enhances ATF5 expression by what appeared to be a posttranscriptional mechanism involving protein stabilization. These findings indicate a reciprocal interaction between ATF5 and Shh in which Shh stimulates ATF5 expression and in which ATF5 contributes to Shh-stimulated CGNP expansion.


Assuntos
Fatores Ativadores da Transcrição/metabolismo , Proliferação de Células , Cerebelo/citologia , Proteínas Hedgehog/metabolismo , Células-Tronco Neurais/citologia , Neurônios/citologia , Fatores Ativadores da Transcrição/genética , Animais , Células Cultivadas , Cerebelo/metabolismo , Regulação da Expressão Gênica , Proteínas Hedgehog/genética , Camundongos , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Transdução de Sinais
17.
Proc Natl Acad Sci U S A ; 108(49): E1349-58, 2011 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-22049344

RESUMO

Carefully designed animal models of genetic risk factors are likely to aid our understanding of the pathogenesis of schizophrenia. Here, we study a mouse strain with a truncating lesion in the endogenous Disc1 ortholog designed to model the effects of a schizophrenia-predisposing mutation and offer a detailed account of the consequences that this mutation has on the development and function of a hippocampal circuit. We uncover widespread and cumulative cytoarchitectural alterations in the dentate gyrus during neonatal and adult neurogenesis, which include errors in axonal targeting and are accompanied by changes in short-term plasticity at the mossy fiber/CA3 circuit. We also provide evidence that cAMP levels are elevated as a result of the Disc1 mutation, leading to altered axonal targeting and dendritic growth. The identified structural alterations are, for the most part, not consistent with the growth-promoting and premature maturation effects inferred from previous RNAi-based Disc1 knockdown. Our results provide support to the notion that modest disturbances of neuronal connectivity and accompanying deficits in short-term synaptic dynamics is a general feature of schizophrenia-predisposing mutations.


Assuntos
Axônios/metabolismo , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal , Potenciais de Ação , Animais , Animais Recém-Nascidos , Proliferação de Células , Células Cultivadas , AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Dendritos/metabolismo , Dendritos/fisiologia , Giro Denteado/citologia , Giro Denteado/crescimento & desenvolvimento , Giro Denteado/metabolismo , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Imuno-Histoquímica , Potenciação de Longa Duração , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fibras Musgosas Hipocampais/metabolismo , Proteínas do Tecido Nervoso/genética , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Neurônios/fisiologia , Técnicas de Patch-Clamp
18.
Neuron ; 71(4): 566-8, 2011 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-21867873

RESUMO

Cadherins implement afferent-target matching in invertebrates, but proof for this concept in mammalian circuits has remained elusive. Two new studies in this issue of Neuron show that cadherin-6 mediates retinal ganglion cell target selection and that cadherin-9 promotes synapse specificity in the hippocampus.

19.
PLoS Biol ; 9(2): e1001013, 2011 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-21346800

RESUMO

The function of neuronal networks relies on selective assembly of synaptic connections during development. We examined how synaptic specificity emerges in the pontocerebellar projection. Analysis of axon-target interactions with correlated light-electron microscopy revealed that developing pontine mossy fibers elaborate extensive cell-cell contacts and synaptic connections with Purkinje cells, an inappropriate target. Subsequently, mossy fiber-Purkinje cell connections are eliminated resulting in granule cell-specific mossy fiber connectivity as observed in mature cerebellar circuits. Formation of mossy fiber-Purkinje cell contacts is negatively regulated by Purkinje cell-derived BMP4. BMP4 limits mossy fiber growth in vitro and Purkinje cell-specific ablation of BMP4 in mice results in exuberant mossy fiber-Purkinje cell interactions. These findings demonstrate that synaptic specificity in the pontocerebellar projection is achieved through a stepwise mechanism that entails transient innervation of Purkinje cells, followed by synapse elimination. Moreover, this work establishes BMP4 as a retrograde signal that regulates the axon-target interactions during development.


Assuntos
Axônios/fisiologia , Comunicação Celular/fisiologia , Rede Nervosa/fisiologia , Animais , Axônios/ultraestrutura , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Proteína Morfogenética Óssea 4/fisiologia , Cerebelo/embriologia , Cerebelo/fisiologia , Cerebelo/ultraestrutura , Camundongos , Rede Nervosa/embriologia , Células de Purkinje/fisiologia , Células de Purkinje/ultraestrutura , Transmissão Sináptica/fisiologia
20.
J Neurosci ; 29(47): 14855-63, 2009 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-19940181

RESUMO

Partial decussation of sensory pathways allows neural inputs from both sides of the body to project to the same target region where these signals will be integrated. Here, to better understand mechanisms of eye-specific targeting, we studied how retinal ganglion cell (RGC) axons terminate in their thalamic target, the dorsal lateral geniculate nucleus (dLGN), when crossing at the optic chiasm midline is altered. In models with gain- and loss-of-function of EphB1, the receptor that directs the ipsilateral projection at the optic chiasm, misrouted RGCs target the appropriate retinotopic zone in the opposite dLGN. However, in EphB1(-/-) mice, the misrouted axons do not intermingle with normally projecting RGC axons and segregate instead into a distinct patch. We also revisited the role of retinal activity on eye-specific targeting by blocking correlated waves of activity with epibatidine into both eyes. We show that, in wild-type mice, retinal waves are necessary during the first postnatal week for both proper distribution and eye-specific segregation of ipsilateral axons in the mature dLGN. Moreover, in EphB1(-/-) mice, refinement of ipsilateral axons is perturbed in control conditions and is further impaired after epibatidine treatment. Finally, retinal waves are required for the formation of the segregated patch of misrouted axons in EphB1(-/-) mice. These findings implicate molecular determinants for targeting of eye-specific zones that are independent of midline guidance cues and that function in concert with correlated retinal activity to sculpt retinogeniculate projections.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Corpos Geniculados/anormalidades , Cones de Crescimento/ultraestrutura , Receptor EphB1/genética , Retina/anormalidades , Vias Visuais/anormalidades , Potenciais de Ação/fisiologia , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Sinais (Psicologia) , Lateralidade Funcional/genética , Corpos Geniculados/metabolismo , Cones de Crescimento/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurogênese/genética , Agonistas Nicotínicos/farmacologia , Piridinas/farmacologia , Retina/metabolismo , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/metabolismo , Sinapses/metabolismo , Sinapses/ultraestrutura , Transmissão Sináptica/genética , Visão Ocular/fisiologia , Vias Visuais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA