Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 641
Filtrar
1.
EMBO J ; 40(21): e107532, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34549820

RESUMO

Astrocytes regulate brain-wide functions and also show region-specific differences, but little is known about how general and region-specific functions are aligned at the single-cell level. To explore this, we isolated adult mouse diencephalic astrocytes by ACSA-2-mediated magnetic-activated cell sorting (MACS). Single-cell RNA-seq revealed 7 gene expression clusters of astrocytes, with 4 forming a supercluster. Within the supercluster, cells differed by gene expression related to ion homeostasis or metabolism, with the former sharing gene expression with other regions and the latter being restricted to specific regions. All clusters showed expression of proliferation-related genes, and proliferation of diencephalic astrocytes was confirmed by immunostaining. Clonal analysis demonstrated low level of astrogenesis in the adult diencephalon, but not in cerebral cortex grey matter. This led to the identification of Smad4 as a key regulator of diencephalic astrocyte in vivo proliferation and in vitro neurosphere formation. Thus, astrocytes show diverse gene expression states related to distinct functions with some subsets being more widespread while others are more regionally restricted. However, all share low-level proliferation revealing the novel concept of adult astrogenesis in the diencephalon.


Assuntos
Astrócitos/metabolismo , Linhagem da Célula/genética , Diencéfalo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Neurogênese/genética , Proteína Smad4/genética , Animais , Astrócitos/classificação , Astrócitos/citologia , Ciclo Celular/genética , Diferenciação Celular , Proliferação de Células , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Diencéfalo/citologia , Diencéfalo/crescimento & desenvolvimento , Ontologia Genética , Redes Reguladoras de Genes , Substância Cinzenta/citologia , Substância Cinzenta/crescimento & desenvolvimento , Substância Cinzenta/metabolismo , Redes e Vias Metabólicas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Anotação de Sequência Molecular , Família Multigênica , Transdução de Sinais , Proteína Smad4/metabolismo
2.
Neuron ; 109(10): 1721-1738.e4, 2021 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-33823137

RESUMO

Basal ganglia play a central role in regulating behavior, but the organization of their outputs to other brain areas is incompletely understood. We investigate the largest output nucleus, the substantia nigra pars reticulata (SNr), and delineate the organization and physiology of its projection populations in mice. Using genetically targeted viral tracing and whole-brain anatomical analysis, we identify over 40 SNr targets that encompass a roughly 50-fold range of axonal densities. Retrograde tracing from the volumetrically largest targets indicates that the SNr contains segregated subpopulations that differentially project to functionally distinct brain stem regions. These subpopulations are electrophysiologically specialized and topographically organized and collateralize to common diencephalon targets, including the motor and intralaminar thalamus as well as the pedunculopontine nucleus and the midbrain reticular formation. These findings establish that SNr signaling is organized as dense, parallel outputs to specific brain stem targets concurrent with extensive collateral branches that encompass the majority of SNr axonal boutons.


Assuntos
Gânglios da Base/citologia , Tronco Encefálico/citologia , Diencéfalo/citologia , Neurônios/fisiologia , Animais , Gânglios da Base/fisiologia , Tronco Encefálico/fisiologia , Diencéfalo/fisiologia , Potenciais Evocados , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais/citologia , Vias Neurais/fisiologia
3.
J Comp Neurol ; 529(1): 52-86, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32964417

RESUMO

This study provides an analysis of the cytoarchitecture, myeloarchitecture, and chemoarchitecture of the diencephalon (dorsal thalamus, ventral thalamus, and epithalamus) of the banded mongoose (Mungos mungo) and domestic ferret (Mustela putorius furo). Using architectural and immunohistochemical stains, we observe that the nuclear organization of the diencephalon is very similar in the two species, and similar to that reported in other carnivores, such as the domestic cat and dog. The same complement of putatively homologous nuclei were identified in both species, with only one variance, that being the presence of the perireticular nucleus in the domestic ferret, that was not observed in the banded mongoose. The chemoarchitecture was also mostly consistent between species, although there were a number of minor variations across a range of nuclei in the density of structures expressing the calcium-binding proteins parvalbumin, calbindin, and calretinin. Thus, despite almost 53 million years since these two species of carnivores shared a common ancestor, strong phylogenetic constraints appear to limit the potential for adaptive evolutionary plasticity within the carnivore order. Apart from the presence of the perireticular nucleus, the most notable difference between the species studied was the physical inversion of the dorsal lateral geniculate nucleus, as well as the lateral posterior and pulvinar nuclei in the domestic ferret compared to the banded mongoose and other carnivores, although this inversion appears to be a feature of the Mustelidae family. While no functional sequelae are suggested, this inversion is likely to result from the altricial birth of Mustelidae species.


Assuntos
Diencéfalo/anatomia & histologia , Furões/anatomia & histologia , Herpestidae/anatomia & histologia , Animais , Carnívoros/anatomia & histologia , Carnívoros/genética , Diencéfalo/citologia , Furões/genética , Herpestidae/genética , Masculino , Especificidade da Espécie
4.
Sci Rep ; 10(1): 8814, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32483163

RESUMO

Promoting the regeneration or survival of retinal ganglion cells (RGCs) is one focus of regenerative medicine. Homeobox Barhl transcription factors might be instrumental in these processes. In mammals, only barhl2 is expressed in the retina and is required for both subtype identity acquisition of amacrine cells and for the survival of RGCs downstream of Atoh7, a transcription factor necessary for RGC genesis. The underlying mechanisms of this dual role of Barhl2 in mammals have remained elusive. Whole genome duplication in the teleost lineage generated the barhl1a and barhl2 paralogues. In the Zebrafish retina, Barhl2 functions as a determinant of subsets of amacrine cells lineally related to RGCs independently of Atoh7. In contrast, barhl1a expression depends on Atoh7 but its expression dynamics and function have not been studied. Here we describe for the first time a Barhl1a reporter line in vivo showing that barhl1a turns on exclusively in subsets of RGCs and their post-mitotic precursors. We also show transient expression of barhl1a:GFP in diencephalic neurons extending their axonal projections as part of the post-optic commissure, at the time of optic chiasm formation. This work sets the ground for future studies on RGC subtype identity, axonal projections and genetic specification of Barhl1a-positive RGCs and commissural neurons.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Quiasma Óptico/embriologia , Células Ganglionares da Retina/metabolismo , Proteínas de Peixe-Zebra/biossíntese , Peixe-Zebra/genética , Células Amácrinas/metabolismo , Animais , Axônios/ultraestrutura , Linhagem da Célula , Proteínas de Ligação a DNA/fisiologia , Diencéfalo/citologia , Diencéfalo/embriologia , Duplicação Gênica , Genes Reporter , Proteínas de Homeodomínio/genética , Microscopia Intravital , Microscopia de Fluorescência , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Quiasma Óptico/citologia , Células Ganglionares da Retina/classificação , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia
5.
Sci Rep ; 10(1): 8315, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32433594

RESUMO

Neuronal migration is essential for constructing functional neural networks. Two posterior septal (PS) nuclei, the triangular septal nucleus and bed nuclei of the anterior commissure, are involved in fear and anxiety. During development, glutamatergic PS neurons undergo long-distance rostrodorsal migration from the thalamic eminence (TE) of the diencephalon, then settle in the caudalmost telencephalon. However, the developmental behavior of PS neurons and the guidance structures facilitating their migration remain unknown. We previously demonstrated the migration of PS neurons along the fornix, a major efferent pathway from the hippocampal formation. Here, we show that the postcommissural fornix is essential for PS neuron migration which is largely confined to its axonal tract, which grows in the opposite direction as PS neuron migration. Fornical axons reach the TE prior to initiation of PS neuron rostrodorsal migration. Ectopic expression of Semaphorin 3 A in the dorsomedial cortex resulted in defective fornix formation. Furthermore, loss of the postcommissural fornix stalled PS neuron migration resulting in abnormal accumulation near their origin. This suggests that PS neurons utilize the postcommissural fornix as a permissive corridor during migration beyond the diencephalic-telencephalic boundary. This axonal support is essential for the functional organization of the heterogeneous septal nuclear complex.


Assuntos
Movimento Celular , Diencéfalo/citologia , Hipocampo/fisiologia , Neurônios/citologia , Telencéfalo/citologia , Animais , Diencéfalo/embriologia , Eletroporação , Feminino , Hipocampo/citologia , Hipocampo/embriologia , Camundongos , Gravidez , Semaforina-3A/metabolismo , Telencéfalo/embriologia
6.
Development ; 147(1)2020 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-31908317

RESUMO

Zebrafish display widespread and pronounced adult neurogenesis, which is fundamental for their regeneration capability after central nervous system injury. However, the cellular identity and the biological properties of adult newborn neurons are elusive for most brain areas. Here, we have used short-term lineage tracing of radial glia progeny to prospectively isolate newborn neurons from the her4.1+ radial glia lineage in the homeostatic adult forebrain. Transcriptome analysis of radial glia, newborn neurons and mature neurons using single cell sequencing identified distinct transcriptional profiles, including novel markers for each population. Specifically, we detected two separate newborn neuron types, which showed diversity of cell fate commitment and location. Further analyses showed that these cell types are homologous to neurogenic cells in the mammalian brain, identified neurogenic commitment in proliferating radial glia and indicated that glutamatergic projection neurons are generated in the adult zebrafish telencephalon. Thus, we prospectively isolated adult newborn neurons from the adult zebrafish forebrain, identified markers for newborn and mature neurons in the adult brain, and revealed intrinsic heterogeneity among adult newborn neurons and their homology with mammalian adult neurogenic cell types.


Assuntos
Encéfalo/citologia , Linhagem da Célula , Células Ependimogliais/citologia , Neurogênese , Neurônios/citologia , Peixe-Zebra/anatomia & histologia , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos/anatomia & histologia , Diencéfalo/citologia , Perfilação da Expressão Gênica , Camundongos , Análise de Sequência de RNA , Análise de Célula Única , Telencéfalo/citologia , Peixe-Zebra/crescimento & desenvolvimento
7.
J Neurosci ; 39(24): 4694-4713, 2019 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-30948475

RESUMO

Adult zebrafish, in contrast to mammals, regenerate neurons in their brain, but the extent and variability of this capacity is unclear. Here we ask whether the loss of various dopaminergic neuron populations is sufficient to trigger their functional regeneration. Both sexes of zebrafish were analyzed. Genetic lineage tracing shows that specific diencephalic ependymo-radial glial (ERG) progenitor cells give rise to new dopaminergic [tyrosine hydroxylase-positive (TH+)] neurons. Ablation elicits an immune response, increased proliferation of ERG progenitor cells, and increased addition of new TH+ neurons in populations that constitutively add new neurons (e.g., diencephalic population 5/6). Inhibiting the immune response attenuates neurogenesis to control levels. Boosting the immune response enhances ERG proliferation, but not addition of TH+ neurons. In contrast, in populations in which constitutive neurogenesis is undetectable (e.g., the posterior tuberculum and locus ceruleus), cell replacement and tissue integration are incomplete and transient. This is associated with a loss of spinal TH+ axons, as well as permanent deficits in shoaling and reproductive behavior. Hence, dopaminergic neuron populations in the adult zebrafish brain show vast differences in regenerative capacity that correlate with constitutive addition of neurons and depend on immune system activation.SIGNIFICANCE STATEMENT Despite the fact that zebrafish show a high propensity to regenerate neurons in the brain, this study reveals that not all types of dopaminergic neurons are functionally regenerated after specific ablation. Hence, in the same adult vertebrate brain, mechanisms of successful and incomplete regeneration can be studied. We identify progenitor cells for dopaminergic neurons and show that activating the immune system promotes the proliferation of these cells. However, in some areas of the brain this only leads to insufficient replacement of functionally important dopaminergic neurons that later disappear. Understanding the mechanisms of regeneration in zebrafish may inform interventions targeting the regeneration of functionally important neurons, such as dopaminergic neurons, from endogenous progenitor cells in nonregenerating mammals.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Fenômenos do Sistema Imunitário/fisiologia , Regeneração Nervosa/fisiologia , Peixe-Zebra/fisiologia , Envelhecimento , Animais , Axônios/fisiologia , Linhagem da Célula/genética , Proliferação de Células , Diencéfalo/citologia , Diencéfalo/fisiologia , Feminino , Masculino , Microglia/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/genética , Neurogênese/fisiologia , Comportamento Sexual Animal/fisiologia
8.
Sci Rep ; 8(1): 11728, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-30082833

RESUMO

Septal nuclei are telencephalic structures associated with a variety of brain functions as part of the limbic system. The two posterior septal nuclei, the triangular septal nucleus (TS) and the bed nuclei of the anterior commissure (BAC), are involved in fear and anxiety through their projections to the medial habenular nucleus. However, the development of both the TS and BAC remains unclear. Here, we found a novel caudal origin and putative migratory stream of mouse posterior septal neurons arising from the thalamic eminence (TE), a transient developmental structure at the rostral end of the rodent diencephalon. TE-derived cells, which have glutamatergic identity, migrated rostrally and entered the telencephalic territory by passing beneath the third ventricle. Subsequently, they turned dorsally toward the posterior septum. We also observed that TS and BAC neurons in the postnatal septum were labeled with GFP by in utero electroporation into the TE, suggesting a shared origin. Furthermore, TE-derived septal neurons migrated along the fornix, an efferent pathway from the hippocampus. These results demonstrate that posterior septal neurons have a distinct extratelencephalic origin from other septal nuclei. This heterogeneous origin may contribute to neuronal diversity of the septal nuclear complex.


Assuntos
Diencéfalo/citologia , Diencéfalo/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Animais , Axônios/metabolismo , Feminino , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Neurônios/citologia , Neurônios/metabolismo , Gravidez , Núcleos Septais/metabolismo , Tálamo/citologia , Tálamo/metabolismo
9.
J Physiol ; 596(16): 3775-3791, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29874406

RESUMO

KEY POINTS: Perioral tactile signals are transmitted via the infraorbital nerve (ION) to trigeminal nuclei. Each cerebellar Purkinje cell (PC) receives this signal as complex spikes (CSs) via a climbing fibre (CF) emerging from the inferior olive (IO). The anatomical pathway from trigeminal nuclei to the IO is not clearly identified. In the present study, we examined candidate anatomical pathways for perioral sensory signalling by analysing CSs recorded from PCs in male mice by single unit recording. CS generation by ION stimulation was inhibited by injection of a GABAA receptor agonist, muscimol, into the contralateral mesodiencephalic junction, which is referred to as the area parafascicularis prerubralis (PfPr). The number of CSs evoked by mechanical whisker stimulation was also decreased by contralateral PfPr inhibition. These results suggest the existence of a sensory signalling pathway to the IO via the PfPr in mice. ABSTRACT: Perioral tactile signals are transmitted via the infraorbital nerve (ION) to trigeminal nuclei. Each cerebellar Purkinje cell receives this signal as complex spikes (CSs) via a climbing fibre emerging from the inferior olive (IO). However, the anatomical pathway from the trigeminal nuclei to the IO is not clearly identified. In the present study, we recorded CSs from Purkinje cells in male mice by single unit recording, and examined the signal transduction pathway. CSs were evoked by electrical stimulation of the ipsilateral or contralateral ION with a latency of 20-70 ms. CS generation by ipsilateral ION stimulation was inhibited by injection of a GABAA receptor agonist, muscimol, into the contralateral mesodiencephalic junction, ranging from around the fasciculus retroflexus to the interstitial nucleus of Cajal, which is referred to as the area parafascicularis prerubralis (PfPr). CSs evoked by contralateral ION stimulation were also suppressed by muscimol injection into the PfPr, although the effective area was more restricted. Furthermore, CSs evoked by mechanical stimulation around the whisker region were suppressed by PfPr inhibition. We also found that the primary motor cortex plays a role to suppress this signalling pathway. These results indicate the existence of an anatomical pathway for conducting perioral sensory signals to the IO via the PfPr.


Assuntos
Cerebelo/fisiologia , Diencéfalo/fisiologia , Mesencéfalo/fisiologia , Boca/fisiologia , Núcleo Olivar/fisiologia , Células de Purkinje/fisiologia , Células Receptoras Sensoriais/fisiologia , Animais , Células Cultivadas , Cerebelo/citologia , Cerebelo/efeitos dos fármacos , Diencéfalo/citologia , Diencéfalo/efeitos dos fármacos , Agonistas de Receptores de GABA-A/farmacologia , Masculino , Mesencéfalo/citologia , Mesencéfalo/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Boca/citologia , Boca/efeitos dos fármacos , Muscimol/farmacologia , Núcleo Olivar/citologia , Núcleo Olivar/efeitos dos fármacos , Células de Purkinje/citologia , Células de Purkinje/efeitos dos fármacos , Receptores de GABA-A/química , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/efeitos dos fármacos
10.
Front Neural Circuits ; 11: 44, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28676745

RESUMO

There has been controversy regarding the precise mechanisms of anesthetic-induced unconsciousness, with two salient approaches that have emerged within systems neuroscience. One prominent approach is the "bottom up" paradigm, which argues that anesthetics suppress consciousness by modulating sleep-wake nuclei and neural circuits in the brainstem and diencephalon that have evolved to control arousal states. Another approach is the "top-down" paradigm, which argues that anesthetics suppress consciousness by modulating the cortical and thalamocortical circuits involved in the integration of neural information. In this article, we synthesize these approaches by mapping bottom-up and top-down mechanisms of general anesthetics to two distinct but inter-related dimensions of consciousness: level and content. We show how this explains certain empirical observations regarding the diversity of anesthetic drug effects. We conclude with a more nuanced discussion of how levels and contents of consciousness interact to generate subjective experience and what this implies for the mechanisms of anesthetic-induced unconsciousness.


Assuntos
Anestésicos/farmacologia , Mapeamento Encefálico , Estado de Consciência/efeitos dos fármacos , Estado de Consciência/fisiologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Animais , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/fisiologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiologia , Diencéfalo/citologia , Diencéfalo/fisiologia , Humanos , Tálamo/efeitos dos fármacos , Tálamo/fisiologia
11.
J Exp Biol ; 220(Pt 13): 2328-2334, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28679791

RESUMO

The brain structure of many animals is influenced by their predators, but the cellular processes underlying this brain plasticity are not well understood. Previous studies showed that electric fish (Brachyhypopomus occidentalis) naturally exposed to high predator (Rhamdia quelen) density and tail injury had reduced brain cell proliferation compared with individuals facing few predators and those with intact tails. However, these field studies described only correlations between predator exposure and cell proliferation. Here, we used a congener Brachyhypopomus gauderio and another electric fish Apteronotus leptorhynchus to experimentally test the hypothesis that exposure to a predator stimulus and tail injury causes alterations in brain cell proliferation. To simulate predator exposure, we either amputated the tail followed by short-term (1 day) or long-term (17-18 days) recovery or repeatedly chased intact fish with a plastic rod over a 7 day period. We measured cell proliferation (PCNA+ cell density) in the telencephalon and diencephalon, and plasma cortisol, which commonly mediates stress-induced changes in brain cell proliferation. In both species, either tail amputation or simulated predator chase decreased cell proliferation in the telencephalon in a manner resembling the effect of predators in the field. In A. leptorhynchus, cell proliferation decreased drastically in the short term after tail amputation and partially rebounded after long-term recovery. In B. gauderio, tail amputation elevated cortisol levels, but repeated chasing had no effect. In A. leptorhynchus, tail amputation elevated cortisol levels in the short term but not in the long term. Thus, predator stimuli can cause reductions in brain cell proliferation, but the role of cortisol is not clear.


Assuntos
Diencéfalo/fisiologia , Gimnotiformes/fisiologia , Estimulação Luminosa , Comportamento Predatório , Cauda/lesões , Telencéfalo/fisiologia , Animais , Proliferação de Células , Diencéfalo/citologia , Cadeia Alimentar
12.
BMC Dev Biol ; 17(1): 5, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28407732

RESUMO

BACKGROUND: Cranial neural crest cells (NCCs) are a unique embryonic cell type which give rise to a diverse array of derivatives extending from neurons and glia through to bone and cartilage. Depending on their point of origin along the antero-posterior axis cranial NCCs are rapidly sorted into distinct migratory streams that give rise to axial specific structures. These migratory streams mirror the underlying segmentation of the brain with NCCs exiting the diencephalon and midbrain following distinct paths compared to those exiting the hindbrain rhombomeres (r). The genetic landscape of cranial NCCs arising at different axial levels remains unknown. RESULTS: Here we have used RNA sequencing to uncover the transcriptional profiles of mouse cranial NCCs arising at different axial levels. Whole transcriptome analysis identified over 120 transcripts differentially expressed between NCCs arising anterior to r3 (referred to as r1-r2 migratory stream for simplicity) and the r4 migratory stream. Eight of the genes differentially expressed between these populations were validated by RT-PCR with 2 being further validated by in situ hybridisation. We also explored the expression of the Neuropilins (Nrp1 and Nrp2) and their co-receptors and show that the A-type Plexins are differentially expressed in different cranial NCC streams. CONCLUSIONS: Our analyses identify a large number of genes differentially regulated between cranial NCCs arising at different axial levels. This data provides a comprehensive description of the genetic landscape driving diversity of distinct cranial NCC streams and provides novel insight into the regulatory networks controlling the formation of specific skeletal elements and the mechanisms promoting migration along different paths.


Assuntos
Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Crista Neural/citologia , Crista Neural/crescimento & desenvolvimento , Análise de Sequência de RNA/métodos , Animais , Movimento Celular , Diencéfalo/citologia , Diencéfalo/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Mesencéfalo/citologia , Mesencéfalo/crescimento & desenvolvimento , Camundongos , Proteínas do Tecido Nervoso/genética , Neuropilina-1/genética , Neuropilina-2/genética , Rombencéfalo/citologia , Rombencéfalo/crescimento & desenvolvimento
13.
Curr Biol ; 27(2): 270-278, 2017 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-28065605

RESUMO

Most neuronal populations form on both the left and right sides of the brain. Their efferent axons appear to grow synchronously along similar pathways on each side, although the neurons or their environment often differ between the two hemispheres [1-4]. How this coordination is controlled has received little attention. Frequently, neurons establish interhemispheric connections, which can function to integrate information between brain hemispheres (e.g., [5]). Such commissures form very early, suggesting their potential developmental role in coordinating ipsilateral axon navigation during embryonic development [4]. To address the temporal-spatial control of bilateral axon growth, we applied long-term time-lapse imaging to visualize the formation of the conserved left-right asymmetric habenular neural circuit in the developing zebrafish embryo [6]. Although habenular neurons are born at different times across brain hemispheres [7], we found that elongation of habenular axons occurs synchronously. The initiation of axon extension is not controlled within the habenular network itself but through an early developing proximal diencephalic network. The commissural neurons of this network influence habenular axons both ipsilaterally and contralaterally. Their unilateral absence impairs commissure formation and coordinated habenular axon elongation and causes their subsequent arrest on both sides of the brain. Thus, habenular neural circuit formation depends on a second intersecting commissural network, which facilitates the exchange of information between hemispheres required for ipsilaterally projecting habenular axons. This mechanism of network formation may well apply to other circuits, and has only remained undiscovered due to technical limitations.


Assuntos
Axônios/fisiologia , Diencéfalo/fisiologia , Neurônios/fisiologia , Peixe-Zebra/fisiologia , Animais , Padronização Corporal , Diencéfalo/citologia , Embrião não Mamífero/citologia , Embrião não Mamífero/fisiologia , Neurogênese , Neurônios/citologia , Imagem com Lapso de Tempo , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/metabolismo
14.
Curr Biol ; 27(3): 318-333, 2017 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-28089511

RESUMO

The vertebrate diencephalic A11 system provides the sole dopaminergic innervation of hindbrain and spinal cord and has been implicated in modulation of locomotion and sensory processes. However, the exact contributions of sensory stimuli and motor behavior to A11 dopaminergic activity remain unclear. We recorded cellular calcium activity in four anatomically distinct posterior tubercular A11-type dopaminergic subgroups and two adjacent hypothalamic dopaminergic groups in GCaMP7a-transgenic, semi-restrained zebrafish larvae. Our analyses reveal the contributions of different sensory modalities and motor states to dopaminergic activity. Each posterior tubercular and hypothalamic subgroup showed distinct activity patterns, while activity was synchronous within individual subgroups. Caudal and dorsomedial hypothalamic dopaminergic neurons are activated following vigorous tail movements and stay active for about 10 s, revealing predominantly post-motor activity. In contrast, posterior tubercular dopaminergic neurons are predominantly sensory driven, with subgroups differentially responding to different tactile or visual sensory modalities. In the anterior subgroups, neuronal response magnitudes are tuned to tactile stimulus intensities, revealing features similar to sensory systems. We identify the lateral line system as source for this tactile tuning. In contrast, the posterior subgroup is responsive to distinct moving visual stimuli. Specifically, translational forward stimuli, which may indicate insufficient rheotaxis and drift, induce dopaminergic activity, but backward or rotational stimuli not. The activation of posterior tubercular dopaminergic neurons by sensory stimuli, and their projections onto peripheral mechanosensory systems, suggests a participation of A11-type neurons in the feedback regulation of sensory systems. Together with the adjacent hypothalamic neurons, they may serve to set basic behavioral states.


Assuntos
Diencéfalo/fisiologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/fisiologia , Atividade Motora/fisiologia , Células Receptoras Sensoriais/metabolismo , Peixe-Zebra/fisiologia , Animais , Diencéfalo/citologia , Neurônios Dopaminérgicos/citologia , Mecanotransdução Celular , Reconhecimento Visual de Modelos , Estimulação Luminosa , Vias Visuais
15.
Neuroscience ; 334: 245-258, 2016 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-27531857

RESUMO

Using an immunohistochemical technique, we mapped the immunoreactive structures containing methionine-enkephalin-Arg6-Gly7-Leu8 (Met-8) (a marker for the pro-enkephalin system) in the human diencephalon. Compared with previous studies, we observed a more widespread distribution of Met-8 in the human diencephalon. Met-8-immunoreactive cell bodies and fibers exhibited a more widespread distribution in the hypothalamus than in the thalamus. We observed six populations of Met-8-immunoreactive cell bodies. These perikarya were observed in the paratenial thalamic nucleus, ventromedial and dorsomedial hypothalamic nuclei, lateral hypothalamic area, pallidohypothalamic nucleus and in the paraventricular hypothalamic nucleus (posterior part). In the thalamus, Met-8-immunoreactive fibers were primarily observed in the midline region, whereas in the hypothalamus, these fibers were widely distributed. In general, a moderate/low density of Met-8-immunoreactive fibers was observed in the diencephalic nuclei. A moderate density was observed in the paraventricular thalamic nucleus, reuniens thalamic nucleus, lateral and medial geniculate nuclei, dorsomedial hypothalamic nucleus, paraventricular hypothalamic nucleus (posterior part) and ventromedial hypothalamic nucleus. The present study is the first to demonstrate the presence of clusters of Met-8-immunoreactive cell bodies in the human thalamus and hypothalamus, the distribution of fibers containing neuropeptides in the hypothalamus and the presence of these fibers in several thalamic nuclei. This neuroanatomical study will serve to elucidate the physiological roles of Met-8 in future studies of the human diencephalon.


Assuntos
Diencéfalo/citologia , Diencéfalo/metabolismo , Encefalina Metionina/análogos & derivados , Idoso de 80 Anos ou mais , Encefalina Metionina/metabolismo , Encefalinas/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Precursores de Proteínas/metabolismo
16.
Physiol Behav ; 164(Pt A): 268-76, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27317836

RESUMO

Recently, it was found that the avian central vasotocin receptor (V1aR) is associated with the regulation of food intake. To identify V1aR-containing brain structures regulating food intake, a selective V1aR antagonist SR-49059 that induced food intake was administrated intracerebroventricularly in male chickens followed by detection of brain structures using FOS immunoreactivity. Particularly, the hypothalamic core region of the paraventricular nucleus, lateral hypothalamic area, dorsomedial hypothalamic nucleus, a subnucleus of the central extended amygdalar complex [dorsolateral bed nucleus of the stria terminalis], medial septal nucleus and caudal brainstem [nucleus of the solitary tract] showed significantly increased FOS-ir cells. On the other hand, the supraoptic nucleus of the preoptic area and the nucleus of the hippocampal commissure of the septum showed suppressed FOS immunoreactivity in the V1aR antagonist treatment group. Further investigation revealed that neuronal activity of arginine vasotocin (AVT-ir) magnocellular neurons in the supraoptic nucleus, preoptic periventricular nucleus, paraventricular nucleus and ventral periventricular hypothalamic nucleus and most likely corticotropin releasing hormone (CRH-ir) neurons in the nucleus of the hippocampal commissure were reduced following the antagonist treatment. Dual immunofluorescence labeling results showed that perikarya of AVT-ir magnocellular neurons in the preoptic area and hypothalamus were colabeled with V1aR. Within the nucleus of the hippocampal commissure, CRH-ir neurons were shown in close contact with V1aR-ir glial cells. Results of the present study suggest that the V1aR plays a role in the regulation of food intake by modulating neurons that synthesize and release anorectic neuropeptides in the avian brain.


Assuntos
Regulação do Apetite/fisiologia , Proteínas Aviárias/metabolismo , Diencéfalo/metabolismo , Ingestão de Alimentos/fisiologia , Receptores de Vasopressinas/metabolismo , Septo do Cérebro/metabolismo , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Regulação do Apetite/efeitos dos fármacos , Comportamento Apetitivo/efeitos dos fármacos , Comportamento Apetitivo/fisiologia , Proteínas Aviárias/antagonistas & inibidores , Fármacos do Sistema Nervoso Central/administração & dosagem , Galinhas , Diencéfalo/citologia , Diencéfalo/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Indóis/farmacologia , Masculino , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeo Y/administração & dosagem , Proteínas Proto-Oncogênicas c-fos/metabolismo , Pirrolidinas/farmacologia , Distribuição Aleatória , Septo do Cérebro/citologia , Septo do Cérebro/efeitos dos fármacos
17.
Proc Natl Acad Sci U S A ; 113(21): 6053-8, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27166423

RESUMO

Nonvisual photoreceptors are widely distributed in the retina and brain, but their roles in animal behavior remain poorly understood. Here we document a previously unidentified form of deep-brain photoreception in Xenopus laevis frog tadpoles. The isolated nervous system retains sensitivity to light even when devoid of input from classical eye and pineal photoreceptors. These preparations produce regular bouts of rhythmic swimming activity in ambient light but fall silent in the dark. This sensitivity is tuned to short-wavelength UV light; illumination at 400 nm initiates motor activity over a broad range of intensities, whereas longer wavelengths do not cause a response. The photosensitive tissue is located in a small region of caudal diencephalon-this region is necessary to retain responses to illumination, whereas its focal illumination is sufficient to drive them. We present evidence for photoreception via the light-sensitive proteins opsin (OPN)5 and/or cryptochrome 1, because populations of OPN5-positive and cryptochrome-positive cells reside within the caudal diencephalon. This discovery represents a hitherto undescribed vertebrate pathway that links luminance detection to motor output. The pathway provides a simple mechanism for light avoidance and/or may reinforce classical circadian systems.


Assuntos
Criptocromos/metabolismo , Diencéfalo/metabolismo , Opsinas/metabolismo , Células Fotorreceptoras/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Diencéfalo/citologia , Larva , Células Fotorreceptoras/citologia , Xenopus laevis
18.
Dev Dyn ; 245(6): 667-77, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27005305

RESUMO

BACKGROUND: Antioxidants such as the green tea polyphenol epigallocatechin gallate (EGCG) are neuroprotective under many conditions in mature nervous systems; however, their impact has rarely been explored in developing nervous systems, in which a critical step is the formation of connections between neurons. Axons emerge from newly formed neurons and are led by a dynamic structure found at their tip called a growth cone. Here we explore the impact of EGCG on the development of retinal ganglion cell (RGC) axons, which connect the eye to the brain. RESULTS: EGCG acts directly on RGC axons to increase the number of growth cone filopodia, fingerlike projections that respond to extrinsic signals, in vitro and in vivo. Furthermore, EGCG exposure leads to a dramatic defect in the guided growth of RGC axons where the axons fail to make a key turn in the mid-diencephalon required to reach their target. Intriguingly, at guidance points where RGCs do not show a change in direction, EGCG has no influence on RGC axon behavior. CONCLUSIONS: We propose that EGCG stabilizes filopodia and prevents normal filopodial dynamics required for axons to change their direction of outgrowth at guidance decision points. Developmental Dynamics 245:667-677, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Axônios/efeitos dos fármacos , Axônios/metabolismo , Catequina/análogos & derivados , Cones de Crescimento/efeitos dos fármacos , Pseudópodes/efeitos dos fármacos , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/efeitos dos fármacos , Animais , Orientação de Axônios/efeitos dos fármacos , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Catequina/farmacologia , Gonadotropina Coriônica/farmacologia , Diencéfalo/citologia , Diencéfalo/efeitos dos fármacos , Embrião não Mamífero/citologia , Embrião não Mamífero/efeitos dos fármacos , Feminino , Humanos , Hibridização In Situ , Neurogênese/efeitos dos fármacos , Retina/citologia , Retina/efeitos dos fármacos , Xenopus
19.
Development ; 143(4): 691-702, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26755703

RESUMO

Mesodiencephalic dopaminergic (mdDA) neurons are located in the ventral mesencephalon and caudal diencephalon of all tetrapod species studied so far. They are the most prominent DA neuronal population and are implicated in control and modulation of motor, cognitive and rewarding/affective behaviors. Their degeneration or dysfunction is intimately linked to several neurological and neuropsychiatric human diseases. To gain further insights into their generation, we studied spatiotemporal expression patterns and epistatic interactions in chick embryos of selected marker genes and signaling pathways associated with mdDA neuron development in mouse. We detected striking differences in the expression patterns of the chick orthologs of the mouse mdDA marker genes Pitx3 and Aldh1a1, which suggests important differences between the species in the generation/generating of these cells. We also discovered that the sonic hedgehog signaling pathway is both necessary and sufficient for the induction of ectopic PITX3 expression in chick mesencephalon downstream of WNT9A-induced LMX1a transcription. These aspects of early chicken development resemble the ontogeny of zebrafish diencephalic DA neuronal populations, and suggest a divergence between birds and mammals during evolution.


Assuntos
Galinhas/genética , Diencéfalo/citologia , Neurônios Dopaminérgicos/citologia , Epistasia Genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Mesencéfalo/citologia , Fatores de Transcrição/genética , Animais , Biomarcadores/metabolismo , Proliferação de Células , Embrião de Galinha , Diencéfalo/metabolismo , Neurônios Dopaminérgicos/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Mesencéfalo/metabolismo , Camundongos , Mitose , Modelos Biológicos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Transdução de Sinais/genética , Análise Espaço-Temporal , Fatores de Transcrição/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
20.
J Comp Neurol ; 524(4): 896-913, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26287569

RESUMO

Teleosts possess two or three paralogs of gonadotropin-releasing hormone (GnRH) genes: gnrh1, gnrh2, and gnrh3. Some species have lost the gnrh1 and/or gnrh3 genes, whereas gnrh2 has been completely conserved in the teleost species analyzed to date. In most teleosts that possess gnrh1, GnRH1 peptide is the authentic GnRH that stimulates gonadotropin release, whereas GnRH2 and GnRH3, if present, are neuromodulatory. Progenitors of GnRH1 and GnRH3 neurons originate from olfactory placodes and migrate to their destination during early development. However, because of the relatively low affinity/specificity of generally available antibodies that recognize GnRH1 or GnRH3, labeling of these neurons has only been possible using genetic manipulation. We used a model teleost, medaka, which possesses all three paralogous gnrh genes, to analyze development of forebrain GnRH neurons composed of GnRH1 and GnRH3 neurons. Here, we newly generated transgenic medaka lines that express enhanced green fluorescent protein under the control of promoters for gnrh1 or gnrh3, to detect GnRH neurons and facilitate immunohistochemical analysis of the neuronal morphology. We used a combination of immunohistochemistry and three-dimensional confocal microscopy image reconstructions to improve identification of neurites from GnRH1 or GnRH3 neuronal populations with greater precision. This led us to clearly identify the hypophysiotropic innervation of GnRH1 neurons residing in the ventral preoptic area (vPOA) from as early as 10 days post hatching. Furthermore, these analyses also revealed retinopetal projections of nonhypophysiotropic GnRH1 neurons in vPOA, prominent during early developmental stages, and multiple populations of GnRH3 neurons with different origins and migratory pathways.


Assuntos
Diencéfalo/crescimento & desenvolvimento , Proteínas de Peixes/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/citologia , Oryzias/crescimento & desenvolvimento , Telencéfalo/crescimento & desenvolvimento , Animais , Animais Geneticamente Modificados , Movimento Celular/fisiologia , Diencéfalo/citologia , Diencéfalo/metabolismo , Proteínas de Peixes/genética , Hormônio Liberador de Gonadotropina/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Imageamento Tridimensional/métodos , Imuno-Histoquímica , Hibridização In Situ , Microscopia Confocal/métodos , Neurônios/metabolismo , Oryzias/anatomia & histologia , Oryzias/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Telencéfalo/citologia , Telencéfalo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...