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










Base de dados
Intervalo de ano de publicação
1.
Invest Ophthalmol Vis Sci ; 64(13): 32, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-37862028

RESUMO

Purpose: We aimed to generate and phenotype a mouse model of foveal hypoplasia, optic nerve decussation defects, and anterior segment dysgenesis (FHONDA), a rare disease associated with mutations in Slc38a8 that causes severe visual alterations similar to albinism without affecting pigmentation. Methods: The FHONDA mouse model was generated with clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 technology using an RNA guide targeting the Scl38a8 murine locus. The resulting mice were backcrossed to C57BL/6J. Melanin content was measured using spectrophotometry. Retinal cell architecture was analyzed through light and electron microscopy. Retinal projections to the brain were evaluated with anterograde labelling in embryos and adults. Visual function was assessed by electroretinography (ERG) and the optomotor test (OT). Results: From numerous Slc38a8 mouse mutant alleles generated, we selected one that encodes a truncated protein (p.196Pro*, equivalent to p.199Pro* in the human protein) closely resembling a mutant allele described in patients (p.200Gln*). Slc38a8 mutant mice exhibit wild-type eye and coat pigmentation with comparable melanin content. Subcellular abnormalities were observed in retinal pigment epithelium cells of Slc38a8 mutant mice. Anterograde labeling experiments of retinal projections in embryos and adults showed a reduction of ipsilateral fibers. Functional visual analyses revealed a decreased ERG response in scotopic conditions and a reduction of visual acuity in mutant mice measured by OT. Conclusions: Slc38a8 mutant mice recapitulate the phenotype of patients with FHONDA concerning their normal pigmentation and their abnormal visual system, in the latter being a hallmark of all types of albinism. These mice will be helpful in better understanding the pathophysiology of this genetic condition.


Assuntos
Albinismo , Sistemas de Transporte de Aminoácidos Neutros , Anormalidades do Olho , Adulto , Humanos , Camundongos , Animais , Melaninas , Camundongos Endogâmicos C57BL , Pigmentação , Sistemas de Transporte de Aminoácidos Neutros/genética
2.
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
3.
Mol Psychiatry ; 28(6): 2328-2342, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37217677

RESUMO

The proper maturation of emotional and sensory circuits requires fine-tuning of serotonin (5-HT) level during early postnatal development. Consistently, dysfunctions of the serotonergic system have been associated with neurodevelopmental psychiatric diseases, including autism spectrum disorders (ASD). However, the mechanisms underlying the developmental effects of 5-HT remain partially unknown, one obstacle being the action of 5-HT on different cell types. Here, we focused on microglia, which play a role in brain wiring refinement, and we investigated whether the control of these cells by 5-HT is relevant for neurodevelopment and spontaneous behaviors in mice. Since the main 5-HT sensor in microglia is the 5-HT2B receptor subtype, we prevented 5-HT signaling specifically in microglia by conditional invalidation of the Htr2b gene in these cells. We observed that abrogating the serotonergic control of microglia during early postnatal development affects the phagolysosomal compartment of these cells and their proximity to dendritic spines and perturbs neuronal circuits maturation. Furthermore, this early ablation of microglial 5-HT2B receptors leads to adult hyperactivity in a novel environment and behavioral defects in sociability and flexibility. Importantly, we show that these behavioral alterations result from a developmental effect, since they are not observed when microglial Htr2b invalidation is induced later, at P30 onward. Thus, a primary alteration of 5-HT sensing in microglia, during a critical time window between birth and P30, is sufficient to impair social and flexibility skills. This link between 5-HT and microglia may explain the association between serotonergic dysfunctions and behavioral traits like impaired sociability and inadaptability to novelty, which are prominent in psychiatric disorders such as ASD.


Assuntos
Microglia , Serotonina , Humanos , Camundongos , Animais , Serotonina/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Encéfalo/metabolismo , Transdução de Sinais
4.
Neuron ; 107(4): 617-630.e6, 2020 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-32559415

RESUMO

Stable genomic integration of exogenous transgenes is essential in neurodevelopmental and stem cell studies. Despite tools driving increasingly efficient genomic insertion with DNA vectors, transgenesis remains fundamentally hindered by the impossibility of distinguishing integrated from episomal transgenes. Here, we introduce an integration-coupled On genetic switch, iOn, which triggers gene expression upon incorporation into the host genome through transposition, thus enabling rapid and accurate identification of integration events following transfection with naked plasmids. In vitro, iOn permits rapid drug-free stable transgenesis of mouse and human pluripotent stem cells with multiple vectors. In vivo, we demonstrate faithful cell lineage tracing, assessment of regulatory elements, and mosaic analysis of gene function in somatic transgenesis experiments that reveal neural progenitor potentialities and interaction. These results establish iOn as a universally applicable strategy to accelerate and simplify genetic engineering in cultured systems and model organisms by conditioning transgene activation to genomic integration.


Assuntos
Expressão Gênica , Técnicas de Transferência de Genes , Células-Tronco Neurais , Transgenes , Animais , Linhagem da Célula , Vetores Genéticos , Humanos , Camundongos
5.
Int J Mol Sci ; 21(2)2020 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-31936811

RESUMO

Across all species, retinal ganglion cells (RGCs) are the first retinal neurons generated during development, followed by the other retinal cell types. How are retinal progenitor cells (RPCs) able to produce these cell types in a specific and timely order? Here, we will review the different models of retinal neurogenesis proposed over the last decades as well as the extrinsic and intrinsic factors controlling it. We will then focus on the molecular mechanisms, especially the cascade of transcription factors that regulate, more specifically, RGC fate. We will also comment on the recent discovery that the ciliary marginal zone is a new stem cell niche in mice contributing to retinal neurogenesis, especially to the generation of ipsilateral RGCs. Furthermore, RGCs are composed of many different subtypes that are anatomically, physiologically, functionally, and molecularly defined. We will summarize the different classifications of RGC subtypes and will recapitulate the specification of some of them and describe how a genetic disease such as albinism affects neurogenesis, resulting in profound visual deficits.


Assuntos
Neurogênese/fisiologia , Células Ganglionares da Retina/metabolismo , Albinismo , Animais , Fatores de Crescimento de Fibroblastos , Proteínas Hedgehog , Humanos , Retina/crescimento & desenvolvimento , Retina/metabolismo , Células Ganglionares da Retina/classificação , Fatores de Transcrição/metabolismo
6.
Sci Rep ; 7(1): 3236, 2017 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-28607399

RESUMO

Neural activity is crucial for the refinement of neuronal connections during development, but the contribution of synaptic release mechanisms is not known. In the mammalian retina, spontaneous neural activity controls the refinement of retinal projections to the dorsal lateral geniculate nucleus (dLGN) and the superior colliculus (SC) to form appropriate topographic and eye-specific maps. To evaluate the role of synaptic release, the rab-interacting molecules (RIMs), a family of active zone proteins that play a central role in calcium-triggered release, were conditionally ablated in a subset of retinal ganglion cells (RGCs). We found that this deletion is sufficient to reduce presynaptic release probability onto dLGN neurons. Furthermore, eye-specific segregation in the dLGN and topographic refinement of ipsilateral axons in the SC and the dLGN, are impaired in RIM1/2 conditional knock-out (Rim-cDKO) mice. These defects are similar to those found when retinal activity is globally disturbed. However, reduction in synaptic release had no effect on eye-specific lamination in the SC nor on the retinotopic refinement of contralateral axons in the SC. This study highlights a potential distinction between synaptic and non-synaptic roles of neuronal activity for different mapping rules operating in visual system development.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Retina/crescimento & desenvolvimento , Células Ganglionares da Retina/fisiologia , Vias Visuais/crescimento & desenvolvimento , Animais , Axônios/fisiologia , Corpos Geniculados/metabolismo , Camundongos Knockout , Técnicas de Patch-Clamp , Retina/citologia , Retina/fisiologia , Células Ganglionares da Retina/metabolismo , Colículos Superiores/crescimento & desenvolvimento , Colículos Superiores/metabolismo , Vias Visuais/metabolismo
7.
Dev Neurobiol ; 77(7): 844-860, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27907266

RESUMO

The visual representation of the outside world relies on the appropriate connectivity between the eyes and the brain. Retinal ganglion cells are the sole neurons that send an axon from the retina to the brain, and thus the guidance decisions of retinal axons en route to their targets in the brain shape the neural circuitry that forms the basis of vision. Here, we focus on the choice made by retinal axons to cross or avoid the midline at the optic chiasm. This decision allows each brain hemisphere to receive inputs from both eyes corresponding to the same visual hemifield, and is thus crucial for binocular vision. In achiasmatic conditions, all retinal axons from one eye project to the ipsilateral brain hemisphere. In albinism, abnormal guidance of retinal axons at the optic chiasm leads to a change in the ratio of contralateral and ipsilateral projections with the consequence that each brain hemisphere receives inputs primarily from the contralateral eye instead of an almost equal distribution from both eyes in humans. In both cases, this misrouting of retinal axons leads to reduced visual acuity and poor depth perception. While this defect has been known for decades, mouse genetics have led to a better understanding of the molecular mechanisms at play in retinal axon guidance and at the origin of the guidance defect in albinism. In addition, fMRI studies on humans have now confirmed the anatomical and functional consequences of axonal misrouting at the chiasm that were previously only assumed from animal models. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 844-860, 2017.


Assuntos
Orientação de Axônios/fisiologia , Axônios/fisiologia , Encéfalo/fisiologia , Retina/fisiologia , Vias Visuais/fisiologia , Animais , Humanos , Quiasma Óptico/fisiologia
8.
Nat Commun ; 7: 12896, 2016 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-27694812

RESUMO

The development of neuronal circuits is controlled by guidance molecules that are hypothesized to interact with the cholesterol-enriched domains of the plasma membrane termed lipid rafts. Whether such domains enable local intracellular signalling at the submicrometre scale in developing neurons and are required for shaping the nervous system connectivity in vivo remains controversial. Here, we report a role for lipid rafts in generating domains of local cAMP signalling in axonal growth cones downstream of ephrin-A repulsive guidance cues. Ephrin-A-dependent retraction of retinal ganglion cell axons involves cAMP signalling restricted to the vicinity of lipid rafts and is independent of cAMP modulation outside of this microdomain. cAMP modulation near lipid rafts controls the pruning of ectopic axonal branches of retinal ganglion cells in vivo, a process requiring intact ephrin-A signalling. Together, our findings indicate that lipid rafts structure the subcellular organization of intracellular cAMP signalling shaping axonal arbors during the nervous system development.


Assuntos
Axônios/metabolismo , AMP Cíclico/metabolismo , Efrina-A1/metabolismo , Microdomínios da Membrana/química , Retina/citologia , Retina/embriologia , Animais , Feminino , Regulação da Expressão Gênica , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Células Ganglionares da Retina/citologia , Transdução de Sinais
9.
Biol Aujourdhui ; 209(1): 87-95, 2015.
Artigo em Francês | MEDLINE | ID: mdl-26115714

RESUMO

Numerous neurotransmitters have been implicated in neurodevelopmental processes. In addition, developing neurons show an abundance of vesicles in the growth cones, and express proteins of the SNARE complex early on. This has led to propose a role for vesicular fusion machinery in axonal growth and synapse formation. However, as the molecular machinery of vesicular fusion started to unveil, and knockouts for the major proteins of this complex were generated, it came as a surprise that none of these proteins was essential for the construction of brain architecture, although they were crucial for vital functions of the organism, leading to early mortality of exocytosis mutants. Because of this early death, conditional ablation of these genes in well-defined neuronal populations was necessary to study their role at later stages of neural circuit development, when activity-dependent mechanisms are best defined. Early studies showed that mutants of Munc18-1, a gene essential for both constitutive and calcium triggered release, were required for target dependent cell survival but not for axon growth or early refinement of topographic targeting, at least in the retinotectal system. Conditional knockout of the Rim1 and Rim2 genes allowed to interrogate more specifically the role of calcium-triggered release. Rims (rab interacting molecules) play a key role in the assembly of calcium channels and their coupling to the SNARE complex alters calcium-triggered release with little effect on constitutive release. When Rim1/Rim2 genes were ablated in the thalamus, layer IV neurons failed to organize into barrel structures, and to form the characteristic asymmetric distribution of their dendrites. More surprisingly, thalamocortical axons still organized in precise topographic maps and formed well differentiated synapses despite considerable reduction of calcium-induced synaptic release. However, this reduction in release probability altered axon targeting in the visual system where axons from both eyes compete for the same target. Thus, genetic tools targeting the exocytosis machinery are allowing to dissect more precisely the contribution of synaptic and non-synaptic mechanisms to activity-dependent circuit wiring.


Assuntos
Sistema Nervoso/crescimento & desenvolvimento , Neurotransmissores/fisiologia , Sinapses/fisiologia , Animais , Axônios/fisiologia , Exocitose/genética , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/fisiologia , Técnicas de Inativação de Genes , Camundongos , Camundongos Knockout , Proteínas Munc18/genética , Proteínas Munc18/fisiologia , Mutação , Neurônios/fisiologia , Retina/ultraestrutura , Proteínas SNARE/genética , Proteínas SNARE/fisiologia , Sensação , Vesículas Sinápticas/fisiologia , Tálamo , Visão Ocular
10.
ACS Chem Neurosci ; 6(7): 1219-30, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25857335

RESUMO

Maturation of functional neuronal circuits during central nervous system development relies on sophisticated mechanisms. First, axonal and dendritic growth should reach appropriate targets for correct synapse elaboration. Second, pruning and neuronal death are required to eliminate redundant or inappropriate neuronal connections. Serotonin, in addition to its role as a neurotransmitter, actively participates in postnatal establishment and refinement of brain wiring in mammals. Brain resident macrophages, that is, microglia, also play an important role in developmentally regulated neuronal death as well as in synaptic maturation and elimination. Here, we tested the hypothesis of cross-regulation between microglia and serotonin during postnatal brain development in a mouse model of synaptic refinement. We found expression of the serotonin 5-HT2B receptor on postnatal microglia, suggesting that serotonin could participate in temporal and spatial synchronization of microglial functions. Using two-photon microscopy, acute brain slices, and local delivery of serotonin, we observed that microglial processes moved rapidly toward the source of serotonin in Htr2B(+/+) mice, but not in Htr2B(-/-) mice lacking the 5-HT2B receptor. We then investigated whether some developmental steps known to be controlled by serotonin could potentially result from microglia sensitivity to serotonin. Using an in vivo model of synaptic refinement during early brain development, we investigated the maturation of the retinal projections to the thalamus and observed that Htr2B(-/-) mice present anatomical alterations of the ipsilateral projecting area of retinal axons into the thalamus. In addition, activation markers were upregulated in microglia from Htr2B(-/-) compared to control neonates, in the absence of apparent morphological modifications. These results support the hypothesis that serotonin interacts with microglial cells and these interactions participate in brain maturation.


Assuntos
Corpos Geniculados/crescimento & desenvolvimento , Microglia/fisiologia , Receptor 5-HT2A de Serotonina/metabolismo , Retina/crescimento & desenvolvimento , Serotonina/metabolismo , Sinapses/fisiologia , Animais , Receptor 1 de Quimiocina CX3C , Células Cultivadas , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/fisiologia , Corpos Geniculados/fisiologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/fisiologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptor 5-HT2A de Serotonina/genética , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Retina/fisiologia , Técnicas de Cultura de Tecidos , Vias Visuais/crescimento & desenvolvimento , Vias Visuais/fisiologia
11.
Semin Cell Dev Biol ; 35: 136-46, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25152335

RESUMO

The refinement of neural connections requires activity-dependent mechanisms in addition to the genetic program initially establishing wiring diagrams. The well-understood organization of the visual system makes it an accessible model for analyzing the contribution of activity in the formation of connectivity. Prior to visual experience, patterned spontaneous activity in the form of retinal waves has an important role for the establishment of eye-specific and retinotopic maps by acting on the refinement of axon arborization. In the present review, which focuses on experimental data obtained in mice and ferrets, we highlight the features of retinal activity that are important for visual map formation and question whether synaptic release and Hebbian based competition rules apply to this system. Recent evidence using genetic tools that allowed the manipulation of different features of neural activity have clarified the controversy on whether activity is instructive or permissive for visual map formation. Furthermore, current evidence strongly suggests that different mechanisms are at play for different types of axons (ipsilateral vs. contralateral), maps (eye-specific vs. retinotopic) or targets. Many molecules that either modulate activity or are modulated by activity are important in the formation of the visual map, such as adenylate cyclase 1, serotonin, or molecules from the immune system. Finally, new players in the game include retrograde messengers signaling from the target cell to the retinal axons as well as microglia that could help to eliminate inappropriate synapses.


Assuntos
Modelos Neurológicos , Retina/fisiologia , Células Ganglionares da Retina/fisiologia , Vias Visuais/fisiologia , Animais , Mapeamento Encefálico/métodos , Corpos Geniculados/anatomia & histologia , Corpos Geniculados/fisiologia , Retina/anatomia & histologia , Células Ganglionares da Retina/citologia , Colículos Superiores/anatomia & histologia , Colículos Superiores/fisiologia , Vias Visuais/anatomia & histologia
12.
Neural Dev ; 9: 11, 2014 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-24885435

RESUMO

BACKGROUND: Proper binocular vision depends on the routing at the optic chiasm of the correct proportion of retinal ganglion cell (RGC) axons that project to the same (ipsilateral) and opposite (contralateral) side of the brain. The ipsilateral RGC projection is reduced in mammals with albinism, a congenital disorder characterized by deficient pigmentation in the skin, hair, and eyes. Compared to the pigmented embryonic mouse retina, the albino embryonic mouse retina has fewer RGCs that express the zinc-finger transcription factor, Zic2, which is transiently expressed by RGCs fated to project ipsilaterally. Here, using Zic2 as a marker of ipsilateral RGCs, Islet2 as a marker of contralateral RGCs, and birthdating, we investigate spatiotemporal dynamics of RGC production as they relate to the phenotype of diminished ipsilateral RGC number in the albino retina. RESULTS: At embryonic day (E)15.5, fewer Zic2-positive (Zic2+) RGCs are found in the albino ventrotemporal (VT) retina compared with the pigmented VT retina, as we previously reported. However, the reduction in Zic2+ RGCs in the albino is not accompanied by a compensatory increase in Zic2-negative (Zic2-) RGCs, resulting in fewer RGCs in the VT retina at this time point. At E17.5, however, the number of RGCs in the VT region is similar in pigmented and albino retinae, implicating a shift in the timing of RGC production in the albino. Short-term birthdating assays reveal a delay in RGC production in the albino VT retina between E13 and E15. Specifically, fewer Zic2+ RGCs are born at E13 and more Zic2- RGCs are born at E15. Consistent with an increase in the production of Zic2- RGCs born at later ages, more RGCs at E17.5 express the contralateral marker, Islet2, in the albino VT retina compared with the pigmented retina. CONCLUSIONS: A delay in neurogenesis in the albino retina is linked to the alteration of RGC subtype specification and consequently leads to the reduced ipsilateral projection that characterizes albinism.


Assuntos
Neurogênese , Retina/embriologia , Células Ganglionares da Retina/metabolismo , Animais , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Retina/metabolismo , Fatores de Transcrição/metabolismo
13.
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
14.
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.

15.
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
16.
J Vis Exp ; (31)2009 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-19779401

RESUMO

The retina and its sole output neuron, the retinal ganglion cell (RGC), comprise an excellent model in which to examine biological questions such as cell differentiation, axon guidance, retinotopic organization and synapse formation. One drawback is the inability to efficiently and reliably manipulate gene expression in RGCs in vivo, especially in the otherwise accessible murine visual pathways. Transgenic mice can be used to manipulate gene expression, but this approach is often expensive, time consuming, and can produce unwanted side effects. In chick, in ovo electroporation is used to manipulate gene expression in RGCs for examining retina and RGC development. Although similar electroporation techniques have been developed in neonatal mouse pups, adult rats, and embryonic murine retinae in vitro, none of these strategies allow full characterization of RGC development and axon projections in vivo. To this end, we have developed two applications of electroporation, one in utero and the other ex vivo, to specifically target embryonic murine RGCs. With in utero retinal electroporation, we can misexpress or downregulate specific genes in RGCs and follow their axon projections through the visual pathways in vivo, allowing examination of guidance decisions at intermediate targets, such as the optic chiasm, or at target regions, such as the lateral geniculate nucleus. Perturbing gene expression in a subset of RGCs in an otherwise wild-type background facilitates an understanding of gene function throughout the retinal pathway. Additionally, we have developed a companion technique for analyzing RGC axon growth in vitro. We electroporate embryonic heads ex vivo, collect and incubate the whole retina, then prepare explants from these retinae several days later. Retinal explants can be used in a variety of in vitro assays in order to examine the response of electroporated RGC axons to guidance cues or other factors. In sum, this set of techniques enhances our ability to misexpress or downregulate genes in RGCs and should greatly aid studies examining RGC development and axon projections.


Assuntos
Eletroporação/métodos , Regulação da Expressão Gênica no Desenvolvimento , Células Ganglionares da Retina/fisiologia , Animais , Feminino , Camundongos , Gravidez , Ratos , Retina/embriologia , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/metabolismo
17.
Cell ; 134(3): 386-7, 2008 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-18692461

RESUMO

A surprising new mechanism that regulates the plasticity of postnatal neurons is reported in this issue by Sugiyama et al. (2008). These authors show in mice that visual experience triggers cell-to-cell transfer of the homeoprotein Otx2 to cortical interneurons, where it promotes maturation of inhibitory neural circuitry and opens the critical period for plasticity in the visual cortex.


Assuntos
Plasticidade Neuronal , Fatores de Transcrição Otx/metabolismo , Córtex Visual/fisiologia , Animais , Humanos , Interneurônios/fisiologia , Camundongos , Privação Sensorial
18.
Annu Rev Neurosci ; 31: 295-315, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18558857

RESUMO

At the optic chiasm, retinal ganglion cell axons from each eye converge and segregate into crossed and uncrossed projections, a pattern critical for binocular vision. Here, we review recent findings on optic chiasm development, highlighting the specific transcription factors and guidance cues that implement retinal axon divergence into crossed and uncrossed pathways. Although mechanisms underlying the formation of the uncrossed projection have been identified, the means by which retinal axons are guided across the midline are still unclear. In addition to directives provided by transcription factors and receptors in the retina, gene expression in the ventral diencephalon influences chiasm formation. Throughout this review, we compare guidance mechanisms at the optic chiasm with those in other midline models and highlight unanswered questions both for retinal axon growth and axon guidance in general.


Assuntos
Encéfalo/embriologia , Cones de Crescimento/ultraestrutura , Quiasma Óptico/embriologia , Retina/embriologia , Células Ganglionares da Retina/citologia , Visão Binocular/fisiologia , Animais , Encéfalo/fisiologia , Lateralidade Funcional/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Cones de Crescimento/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Quiasma Óptico/fisiologia , Retina/fisiologia , Células Ganglionares da Retina/fisiologia , Vias Visuais/embriologia , Vias Visuais/fisiologia
19.
J Neurosci ; 25(3): 706-10, 2005 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-15659608

RESUMO

In the mouse somatosensory cortex, thalamocortical axons (TCAs) corresponding to individual whiskers cluster into restricted barrel domains during the first days of life. If whiskers are lesioned before that time, the cortical space devoted to the afferents from the damaged whisker shrinks and becomes occupied by thalamocortical afferents from neighboring unlesioned whiskers. This plasticity ends by postnatal day 3 (P3) to P4 when barrels emerge. To test whether TCA development and lesion-induced plasticity are linked, we used monoamine oxidase A knock-out (MAOA-KO) mice in which normal TCA development is halted by an excess of serotonin. Normal TCA development can be restored when serotonin levels are lowered by parachlorophenylalanine (PCPA). By varying the time of PCPA administration, we found that barrel development can be reinitiated until P11, although the emergence of TCA clusters becomes gradually slower and less complete. In mice in which barrels emerge 3 d later than the normal schedule, at P6 instead of P3, we examined lesion-induced plasticity. We find a progressive decline of the lesion-induced plasticity and a closure at P3, similar to normal mice, showing that this plasticity is not influenced by an excess of serotonin levels. Thus, in MAOA-KO mice, the emergence of barrel patterning can be delayed without a concomitant delay in lesion-induced plasticity, and the cortical space devoted to one whisker representation cannot be modified by the periphery once patterning is imprinted in the subcortical relays. We conclude that the closure of the lesion-induced plasticity period in the barrelfield is probably not determined at the cortical level.


Assuntos
Plasticidade Neuronal/fisiologia , Córtex Somatossensorial/crescimento & desenvolvimento , Tálamo/crescimento & desenvolvimento , Vibrissas/inervação , Animais , Axônios/fisiologia , Padronização Corporal , Mapeamento Encefálico , Fenclonina/farmacologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Monoaminoxidase/genética , Vias Neurais/crescimento & desenvolvimento , Serotonina/metabolismo , Fatores de Tempo , Triptofano Hidroxilase/antagonistas & inibidores , Vibrissas/lesões
20.
J Neurosci ; 22(19): 8541-52, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12351728

RESUMO

In the rodent primary somatosensory cortex, the thalamocortical axons (TCAs) are organized into clusters that correspond to functional units in the periphery. Around these axons, neurons in layer IV aggregate as barrels. To understand how this organization emerges, we analyzed TCA development in mice that do not form barrels, the monoamine oxidase A knock-out (MAOA-KO), and in MAOA/5-HT(1B) receptor double-KO mice, which have a restored barrel field. We show that TCAs already attain cortical layer IV on the day of birth. They are uniformly distributed in this layer from postnatal day 0 (P0) to P2 and secondarily coalesce into barrel domains in layer IV, over a 3 d period (P3-P5), with no prepatterning in the deeper layers. In MAOA-KO mice, the uniform distribution of the TC projection is maintained, and no axon clusters emerge. Individual TCA arbors were traced after carbocyanine injections. At P1, TCAs were poorly branched and covered variable tangential widths, encompassing one to two prospective barrels. At P7 the number of TCA branches increased 10-fold in layer IV and became restricted to one barrel. In MAOA-KO mice, there was a 50% reduction of the TCA terminal branches in layer IV, with a 40% increase in their tangential extent. These defects were corrected in the MAOA/5-HT(1B) double knock-out mice, indicating an effect of the presynaptic 5-HT(1B) receptor on axon branching. Our results indicate that the barrel-deficient phenotype of MAOA-KO mice results from an altered refinement of the TCA arbors in their target layer IV, involving branch elaboration and collateral retraction during early postnatal life.


Assuntos
Axônios/ultraestrutura , Proteínas de Membrana Transportadoras , Monoaminoxidase/deficiência , Proteínas do Tecido Nervoso , Córtex Somatossensorial/citologia , Tálamo/citologia , Animais , Axônios/metabolismo , Proteínas de Transporte/biossíntese , Corantes , Imuno-Histoquímica , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Monoaminoxidase/biossíntese , Monoaminoxidase/genética , Neurônios/metabolismo , Neurônios/ultraestrutura , Fenótipo , Receptor 5-HT1B de Serotonina , Receptores de Serotonina/biossíntese , Receptores de Serotonina/deficiência , Receptores de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina , Córtex Somatossensorial/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...