Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros













Base de dados
Intervalo de ano de publicação
1.
Neuron ; 112(1): 41-55.e3, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-37898123

RESUMO

Primary cilia act as antenna receivers of environmental signals and enable effective neuronal or glial responses. Disruption of their function is associated with circuit disorders. To understand the signals these cilia receive, we comprehensively mapped cilia's contacts within the human cortical connectome using serial-section EM reconstruction of a 1 mm3 cortical volume, spanning the entire cortical thickness. We mapped the "contactome" of cilia emerging from neurons and astrocytes in every cortical layer. Depending on the layer and cell type, cilia make distinct patterns of contact. Primary cilia display cell-type- and layer-specific variations in size, shape, and microtubule axoneme core, which may affect their signaling competencies. Neuronal cilia are intrinsic components of a subset of cortical synapses and thus a part of the connectome. This diversity in the structure, contactome, and connectome of primary cilia endows each neuron or glial cell with a unique barcode of access to the surrounding neural circuitry.


Assuntos
Cílios , Conectoma , Humanos , Neurônios/fisiologia , Córtex Cerebral , Neuroglia/fisiologia
2.
bioRxiv ; 2023 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-37790363

RESUMO

Microglia diversity emerges from interactions between intrinsic genetic programs and environment-derived signals, but how these processes unfold and interact in the developing brain remains unclear. Here, we show that radial glia-expressed integrin beta 8 (ITGB8) expressed in radial glia progenitors activates microglia-expressed TGFß1, permitting microglial development. Domain-restricted deletion of Itgb8 in these progenitors establishes complementary regions with developmentally arrested "dysmature" microglia that persist into adulthood. In the absence of autocrine TGFß1 signaling, we find that microglia adopt a similar dysmature phenotype, leading to neuromotor symptoms almost identical to Itgb8 mutant mice. In contrast, microglia lacking the TGFß signal transducers Smad2 and Smad3 have a less polarized dysmature phenotype and correspondingly less severe neuromotor dysfunction. Finally, we show that non-canonical (Smad-independent) signaling partially suppresses disease and development associated gene expression, providing compelling evidence for the adoption of microglial developmental signaling pathways in the context of injury or disease.

3.
EBioMedicine ; 78: 103931, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35306341

RESUMO

Congenital hydrocephalus occurs in one in 500-1000 babies born in the United States and acquired hydrocephalus may occur as the consequence of stroke, intraventricular and subarachnoid hemorrhage, traumatic brain injuries, brain tumors, craniectomy or may be idiopathic, as in the case of normal pressure hydrocephalus. Irrespective of its prevalence and significant impact on quality of life, neurosurgeons still rely on invasive cerebrospinal fluid shunt systems for the treatment of hydrocephalus that are exceptionally prone to failure and/or infection. Further understanding of this process at a molecular level, therefore, may have profound implications for improving treatment and quality of life for millions of individuals worldwide. The purpose of this article is to review the current research landscape on hydrocephalus with a focus on recent advances in our understanding of cerebrospinal fluid pathways from an evolutionary, genetics and molecular perspective.


Assuntos
Hidrocefalia , Hemorragia Subaracnóidea , Ventrículos do Coração , Humanos , Hidrocefalia/etiologia , Hidrocefalia/cirurgia , Lactente , Resolução de Problemas , Qualidade de Vida
4.
Neuron ; 110(7): 1100-1115, 2022 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-35216663

RESUMO

Radial progenitor development and function lay the foundation for the construction of the cerebral cortex. Radial glial scaffold, through its functions as a source of neurogenic progenitors and neuronal migration guide, is thought to provide a template for the formation of the cerebral cortex. Emerging evidence is challenging this limited view. Intriguingly, radial glial scaffold may also play a role in axonal growth, guidance, and neuronal connectivity. Radial glial cells not only facilitate the generation, placement, and allocation of neurons in the cortex but also regulate how they wire up. The organization and function of radial glial cells may thus be a unifying feature of the developing cortex that helps to precisely coordinate the right patterns of neurogenesis, neuronal placement, and connectivity necessary for the emergence of a functional cerebral cortex. This perspective critically explores this emerging view and its impact in the context of human brain development and disorders.


Assuntos
Córtex Cerebral , Neuroglia , Movimento Celular/fisiologia , Córtex Cerebral/fisiologia , Células Ependimogliais , Humanos , Neurogênese/fisiologia , Neuroglia/fisiologia , Neurônios/fisiologia
5.
Elife ; 102021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34812142

RESUMO

Variants in the high confident autism spectrum disorder (ASD) gene ANK2 target both ubiquitously expressed 220 kDa ankyrin-B and neurospecific 440 kDa ankyrin-B (AnkB440) isoforms. Previous work showed that knock-in mice expressing an ASD-linked Ank2 variant yielding a truncated AnkB440 product exhibit ectopic brain connectivity and behavioral abnormalities. Expression of this variant or loss of AnkB440 caused axonal hyperbranching in vitro, which implicated AnkB440 microtubule bundling activity in suppressing collateral branch formation. Leveraging multiple mouse models, cellular assays, and live microscopy, we show that AnkB440 also modulates axon collateral branching stochastically by reducing the number of F-actin-rich branch initiation points. Additionally, we show that AnkB440 enables growth cone (GC) collapse in response to chemorepellent factor semaphorin 3 A (Sema 3 A) by stabilizing its receptor complex L1 cell adhesion molecule/neuropilin-1. ASD-linked ANK2 variants failed to rescue Sema 3A-induced GC collapse. We propose that impaired response to repellent cues due to AnkB440 deficits leads to axonal targeting and branch pruning defects and may contribute to the pathogenicity of ANK2 variants.


Assuntos
Anquirinas/genética , Orientação de Axônios/genética , Axônios/fisiologia , Semaforina-3A/genética , Transdução de Sinais/genética , Animais , Anquirinas/metabolismo , Camundongos , Semaforina-3A/metabolismo
6.
Cell Rep ; 35(1): 108946, 2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33826889

RESUMO

Although embryonic brain development and neurodegeneration have received considerable attention, the events that govern postnatal brain maturation are less understood. Here, we identify the miR-29 family to be strikingly induced during the late stages of brain maturation. Brain maturation is associated with a transient, postnatal period of de novo non-CG (CH) DNA methylation mediated by DNMT3A. We examine whether an important function of miR-29 during brain maturation is to restrict the period of CH methylation via its targeting of Dnmt3a. Deletion of miR-29 in the brain, or knockin mutations preventing miR-29 to specifically target Dnmt3a, result in increased DNMT3A expression, higher CH methylation, and repression of genes associated with neuronal activity and neuropsychiatric disorders. These mouse models also develop neurological deficits and premature lethality. Our results identify an essential role for miR-29 in restricting CH methylation in the brain and illustrate the importance of CH methylation regulation for normal brain maturation.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Metilação de DNA/genética , MicroRNAs/metabolismo , Regiões 3' não Traduzidas/genética , Animais , Animais Recém-Nascidos , Sequência de Bases , Comportamento Animal , DNA (Citosina-5-)-Metiltransferases/metabolismo , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/patologia , Neurônios/metabolismo , Neurônios/patologia , Convulsões/genética , Convulsões/patologia , Transdução de Sinais , Sinapses/metabolismo , Regulação para Cima/genética
7.
Curr Top Dev Biol ; 142: 99-146, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33706927

RESUMO

Primary cilium, first described in the 19th century in different cell types and organisms by Alexander Ecker, Albert Kolliker, Aleksandr Kowalevsky, Paul Langerhans, and Karl Zimmermann (Ecker, 1844; Kolliker, 1854; Kowalevsky, 1867; Langerhans, 1876; Zimmermann, 1898), play an essential modulatory role in diverse aspects of nervous system development and function. The primary cilium, sometimes referred to as the cell's 'antennae', can receive wide ranging inputs from cellular milieu, including morphogens, growth factors, neuromodulators, and neurotransmitters. Its unique structural and functional organization bequeaths it the capacity to hyper-concentrate signaling machinery in a restricted cellular domain approximately one-thousandth the volume of cell soma. Thus enabling it to act as a signaling hub that integrates diverse developmental and homestatic information from cellular milieu to regulate the development and function of neural cells. Dysfunction of primary cilia contributes to the pathophysiology of several brain malformations, intellectual disabilities, epilepsy, and psychiatric disorders. This review focuses on the most essential contributions of primary cilia to cerebral cortical development and function, in the context of neurodevelopmental disorders and malformations. It highlights the recent progress made in identifying the mechanisms underlying primary cilia's role in cortical progenitors, neurons and glia, in health and disease. A future challenge will be to translate these insights and advances into effective clinical treatments for ciliopathies.


Assuntos
Cílios , Neurogênese , Humanos , Neurônios , Transdução de Sinais
8.
BMC Res Notes ; 13(1): 542, 2020 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-33213515

RESUMO

OBJECTIVE: Hardness of water in the form of CaCO3 affects reproductive potential in various fish species, differently. This study evaluates the effect of water hardness on growth and reproduction of two aquarium fishes, Poecilia reticulata (Ovo-viviparous sp.) and Betta splendens (Oviparous sp.) by growing them under 150 (control), 320, 540 and 900 ppm CaCO3 levels in semi natural aquaria. RESULTS: Growth increased with increasing water hardness, reporting a significant progress of P. reticulata (p = 0.005) at 900 ppm. Similarly, the reproductive potential of P. reticulata was improved significantly, recording the highest fecundity (16.22 ± 3.90) and Gonadosomatic Index (GSI-2.48 ± 0.6) at 900 ppm. However, in B. splendens water hardness adversely affected the reproduction by resulting a significantly low hatchability and disturbed bubble nests at 900 ppm, compared to the largest bubble nest formed at the control condition (108.58 ± 16.19 cm2). Thus, the study revealed differential effects of water hardness on reproductive potential of the test species, by increasing the potential of P. reticulata while decreasing that of B. splendens. Though larval survival was affected in both species, larval growth was improved significantly in P. reticulata at 900 ppm level. Understanding reproductive potential of aquarium fishes in natural waters is crucial for their management purposes.


Assuntos
Poecilia , Animais , Água Doce , Dureza , Reprodução , Água
9.
Dev Cell ; 51(6): 759-774.e5, 2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31846650

RESUMO

Appropriate axonal growth and connectivity are essential for functional wiring of the brain. Joubert syndrome-related disorders (JSRD), a group of ciliopathies in which mutations disrupt primary cilia function, are characterized by axonal tract malformations. However, little is known about how cilia-driven signaling regulates axonal growth and connectivity. We demonstrate that the deletion of related JSRD genes, Arl13b and Inpp5e, in projection neurons leads to de-fasciculated and misoriented axonal tracts. Arl13b deletion disrupts the function of its downstream effector, Inpp5e, and deregulates ciliary-PI3K/AKT signaling. Chemogenetic activation of ciliary GPCR signaling and cilia-specific optogenetic modulation of downstream second messenger cascades (PI3K, AKT, and AC3) commonly regulated by ciliary signaling receptors induce rapid changes in axonal dynamics. Further, Arl13b deletion leads to changes in transcriptional landscape associated with dysregulated PI3K/AKT signaling. These data suggest that ciliary signaling acts to modulate axonal connectivity and that impaired primary cilia signaling underlies axonal tract defects in JSRD.


Assuntos
Anormalidades Múltiplas/metabolismo , Axônios/metabolismo , Cerebelo/anormalidades , Cílios/metabolismo , Anormalidades do Olho/genética , Doenças Renais Císticas/metabolismo , Retina/anormalidades , Anormalidades Múltiplas/genética , Animais , Cerebelo/metabolismo , Modelos Animais de Doenças , Anormalidades do Olho/metabolismo , Doenças Renais Císticas/genética , Camundongos , Mutação/genética , Neurogênese/fisiologia , Retina/metabolismo
10.
J Anat ; 235(3): 432-451, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31373394

RESUMO

The cerebral cortex constitutes more than half the volume of the human brain and is presumed to be responsible for the neuronal computations underlying complex phenomena, such as perception, thought, language, attention, episodic memory and voluntary movement. Rodent models are extremely valuable for the investigation of brain development, but cannot provide insight into aspects that are unique or highly derived in humans. Many human psychiatric and neurological conditions have developmental origins but cannot be studied adequately in animal models. The human cerebral cortex has some unique genetic, molecular, cellular and anatomical features, which need to be further explored. The Anatomical Society devoted its summer meeting to the topic of Human Brain Development in June 2018 to tackle these important issues. The meeting was organized by Gavin Clowry (Newcastle University) and Zoltán Molnár (University of Oxford), and held at St John's College, Oxford. The participants provided a broad overview of the structure of the human brain in the context of scaling relationships across the brains of mammals, conserved principles and recent changes in the human lineage. Speakers considered how neuronal progenitors diversified in human to generate an increasing variety of cortical neurons. The formation of the earliest cortical circuits of the earliest generated neurons in the subplate was discussed together with their involvement in neurodevelopmental pathologies. Gene expression networks and susceptibility genes associated to neurodevelopmental diseases were discussed and compared with the networks that can be identified in organoids developed from induced pluripotent stem cells that recapitulate some aspects of in vivo development. New views were discussed on the specification of glutamatergic pyramidal and γ-aminobutyric acid (GABA)ergic interneurons. With the advancement of various in vivo imaging methods, the histopathological observations can be now linked to in vivo normal conditions and to various diseases. Our review gives a general evaluation of the exciting new developments in these areas. The human cortex has a much enlarged association cortex with greater interconnectivity of cortical areas with each other and with an expanded thalamus. The human cortex has relative enlargement of the upper layers, enhanced diversity and function of inhibitory interneurons and a highly expanded transient subplate layer during development. Here we highlight recent studies that address how these differences emerge during development focusing on diverse facets of our evolution.


Assuntos
Córtex Cerebral/embriologia , Animais , Redes Reguladoras de Genes , Humanos , Interneurônios , Transtornos do Neurodesenvolvimento/genética , Neurogênese , Células Piramidais
11.
Neuron ; 103(5): 836-852.e5, 2019 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-31277925

RESUMO

Polarized, non-overlapping, regularly spaced, tiled organization of radial glial cells (RGCs) serves as a framework to generate and organize cortical neuronal columns, layers, and circuitry. Here, we show that mediator of cell motility 1 (Memo1) is a critical determinant of radial glial tiling during neocortical development. Memo1 deletion or knockdown leads to hyperbranching of RGC basal processes and disrupted RGC tiling, resulting in aberrant radial unit assembly and neuronal layering. Memo1 regulates microtubule (MT) stability necessary for RGC tiling. Memo1 deficiency leads to disrupted MT minus-end CAMSAP2 distribution, initiation of aberrant MT branching, and altered polarized trafficking of key basal domain proteins such as GPR56, and thus aberrant RGC tiling. These findings identify Memo1 as a mediator of RGC scaffold tiling, necessary to generate and organize neurons into functional ensembles in the developing cerebral cortex.


Assuntos
Células Ependimogliais/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neocórtex/embriologia , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Animais , Transtorno Autístico/genética , Movimento Celular/genética , Polaridade Celular , Cerebelo/embriologia , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Células Ependimogliais/citologia , Técnicas de Silenciamento de Genes , Células HEK293 , Hipocampo/embriologia , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Neocórtex/citologia , Neocórtex/metabolismo , Células-Tronco Neurais/citologia , Neurônios/citologia , Transporte Proteico , Receptores Acoplados a Proteínas G/metabolismo
12.
Neuron ; 103(3): 423-431.e4, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31196673

RESUMO

The paraventricular thalamus (PVT) is an interface for brain reward circuits, with input signals arising from structures, such as prefrontal cortex and hypothalamus, that are broadcast to downstream limbic targets. However, the precise synaptic connectivity, activity, and function of PVT circuitry for reward processing are unclear. Here, using in vivo two-photon calcium imaging, we find that PVT neurons projecting to the nucleus accumbens (PVT-NAc) develop inhibitory responses to reward-predictive cues coding for both cue-reward associative information and behavior. The multiplexed activity in PVT-NAc neurons is directed by opposing activity patterns in prefrontal and lateral hypothalamic afferent axons. Further, we find that prefrontal cue encoding may maintain accurate cue-reward processing, as optogenetic disruption of this encoding induced long-lasting effects on downstream PVT-NAc cue responses and behavioral cue discrimination. Together, these data reveal that PVT-NAc neurons act as an interface for reward processing by integrating relevant inputs to accurately inform reward-seeking behavior.


Assuntos
Aprendizagem por Associação/fisiologia , Região Hipotalâmica Lateral/fisiologia , Núcleos da Linha Média do Tálamo/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Animais , Condicionamento Clássico , Fissura/fisiologia , Sinais (Psicologia) , Ácido Glutâmico/fisiologia , Região Hipotalâmica Lateral/citologia , Camundongos , Núcleos da Linha Média do Tálamo/citologia , Vias Neurais/fisiologia , Optogenética , Técnicas de Patch-Clamp , Córtex Pré-Frontal/citologia , Recompensa , Ácido gama-Aminobutírico/fisiologia
13.
Nat Commun ; 10(1): 134, 2019 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-30635555

RESUMO

The development of the mammalian cerebral cortex depends on careful orchestration of proliferation, maturation, and migration events, ultimately giving rise to a wide variety of neuronal and non-neuronal cell types. To better understand cellular and molecular processes that unfold during late corticogenesis, we perform single-cell RNA-seq on the mouse cerebral cortex at a progenitor driven phase (embryonic day 14.5) and at birth-after neurons from all six cortical layers are born. We identify numerous classes of neurons, progenitors, and glia, their proliferative, migratory, and activation states, and their relatedness within and across age. Using the cell-type-specific expression patterns of genes mutated in neurological and psychiatric diseases, we identify putative disease subtypes that associate with clinical phenotypes. Our study reveals the cellular template of a complex neurodevelopmental process, and provides a window into the cellular origins of brain diseases.


Assuntos
Linhagem da Célula/genética , Desenvolvimento Embrionário/genética , Perfilação da Expressão Gênica , Neocórtex/embriologia , Animais , Sequência de Bases , Linhagem Celular , Ciliopatias/genética , Feminino , Células HEK293 , Humanos , Masculino , Transtornos Mentais/genética , Camundongos , Camundongos Endogâmicos C57BL , Transtornos do Neurodesenvolvimento/genética , Neurônios/citologia , Neurônios/metabolismo , Análise de Sequência de RNA , Análise de Célula Única , Células-Tronco/citologia , Transcrição Gênica/genética
14.
Genes Dev ; 31(16): 1679-1692, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28916710

RESUMO

Adenomatous polyposis coli (APC) regulates the activity of ß-catenin, an integral component of Wnt signaling. However, the selective role of the APC-ß-catenin pathway in cerebral cortical development is unknown. Here we genetically dissected the relative contributions of APC-regulated ß-catenin signaling in cortical progenitor development, a necessary early step in cerebral cortical formation. Radial progenitor-specific inactivation of the APC-ß-catenin pathway indicates that the maintenance of appropriate ß-catenin-mediated Wnt tone is necessary for the orderly differentiation of cortical progenitors and the resultant formation of the cerebral cortex. APC deletion deregulates ß-catenin, leads to high Wnt tone, and disrupts Notch1 signaling and primary cilium maintenance necessary for radial progenitor functions. ß-Catenin deregulation directly disrupts cilium maintenance and signaling via Tulp3, essential for intraflagellar transport of ciliary signaling receptors. Surprisingly, deletion of ß-catenin or inhibition of ß-catenin activity in APC-null progenitors rescues the APC-null phenotype. These results reveal that APC-regulated ß-catenin activity in cortical progenitors sets the appropriate Wnt tone necessary for normal cerebral cortical development.


Assuntos
Proteína da Polipose Adenomatosa do Colo/fisiologia , Córtex Cerebral/embriologia , Células-Tronco Neurais/metabolismo , Neurogênese , Via de Sinalização Wnt , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Proliferação de Células , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Cílios/metabolismo , Proteínas Hedgehog/metabolismo , Camundongos , Camundongos Knockout , Células-Tronco Neurais/citologia , Receptor Notch1/metabolismo , beta Catenina/fisiologia
15.
Dev Cell ; 42(3): 286-300.e4, 2017 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-28787594

RESUMO

Appropriate growth and synaptic integration of GABAergic inhibitory interneurons are essential for functional neural circuits in the brain. Here, we demonstrate that disruption of primary cilia function following the selective loss of ciliary GTPase Arl13b in interneurons impairs interneuronal morphology and synaptic connectivity, leading to altered excitatory/inhibitory activity balance. The altered morphology and connectivity of cilia mutant interneurons and the functional deficits are rescued by either chemogenetic activation of ciliary G-protein-coupled receptor (GPCR) signaling or the selective induction of Sstr3, a ciliary GPCR, in Arl13b-deficient cilia. Our results thus define a specific requirement for primary cilia-mediated GPCR signaling in interneuronal connectivity and inhibitory circuit formation.


Assuntos
Interneurônios/metabolismo , Transdução de Sinais , Sinapses/metabolismo , Potenciais Sinápticos , Fatores de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/metabolismo , Animais , Células Cultivadas , Cílios/metabolismo , Interneurônios/citologia , Interneurônios/fisiologia , Camundongos , Neurogênese , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Sinapses/fisiologia
16.
Cell Rep ; 16(10): 2576-2592, 2016 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-27568284

RESUMO

The mechanisms underlying Zika virus (ZIKV)-related microcephaly and other neurodevelopment defects remain poorly understood. Here, we describe the derivation and characterization, including single-cell RNA-seq, of neocortical and spinal cord neuroepithelial stem (NES) cells to model early human neurodevelopment and ZIKV-related neuropathogenesis. By analyzing human NES cells, organotypic fetal brain slices, and a ZIKV-infected micrencephalic brain, we show that ZIKV infects both neocortical and spinal NES cells as well as their fetal homolog, radial glial cells (RGCs), causing disrupted mitoses, supernumerary centrosomes, structural disorganization, and cell death. ZIKV infection of NES cells and RGCs causes centrosomal depletion and mitochondrial sequestration of phospho-TBK1 during mitosis. We also found that nucleoside analogs inhibit ZIKV replication in NES cells, protecting them from ZIKV-induced pTBK1 relocalization and cell death. We established a model system of human neural stem cells to reveal cellular and molecular mechanisms underlying neurodevelopmental defects associated with ZIKV infection and its potential treatment.


Assuntos
Mitose , Células-Tronco Neurais/enzimologia , Células-Tronco Neurais/virologia , Células Neuroepiteliais/virologia , Neuroglia/virologia , Proteínas Serina-Treonina Quinases/metabolismo , Zika virus/patogenicidade , Encéfalo/embriologia , Encéfalo/patologia , Encéfalo/virologia , Morte Celular/efeitos dos fármacos , Centrossomo/efeitos dos fármacos , Centrossomo/metabolismo , Feto/virologia , Perfilação da Expressão Gênica , Humanos , Imunidade Inata/efeitos dos fármacos , Microcefalia/patologia , Microcefalia/virologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitose/efeitos dos fármacos , Neocórtex/patologia , Células-Tronco Neurais/imunologia , Células-Tronco Neurais/ultraestrutura , Células Neuroepiteliais/efeitos dos fármacos , Células Neuroepiteliais/imunologia , Células Neuroepiteliais/ultraestrutura , Neuroglia/patologia , Neuroglia/ultraestrutura , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neurônios/virologia , Fármacos Neuroprotetores/farmacologia , Nucleosídeos/farmacologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Medula Espinal/patologia , Transcrição Gênica/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Zika virus/efeitos dos fármacos , Zika virus/fisiologia , Zika virus/ultraestrutura , Infecção por Zika virus/patologia , Infecção por Zika virus/virologia , Receptor Tirosina Quinase Axl
17.
J Neurosci ; 36(20): 5448-61, 2016 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-27194326

RESUMO

UNLABELLED: Apoptosis plays an essential role during brain development, yet the precise mechanism by which this pathway is regulated in the brain remains unknown. In particular, mammalian cells are known to express multiple anti-apoptotic Bcl-2 family proteins. However, the cells of the developing brain could also exist in a primed state in which the loss of a single anti-apoptotic Bcl-2 family protein is sufficient to trigger apoptosis. Here, we examined the critical role of Bcl-xL, an anti-apoptotic protein, during brain development. Using conditional knock-out mice in which Bcl-xL is deleted in neural progenitor cells (Bcl-xL(Emx1-Cre)), we show that the loss of Bcl-xL is not sufficient to trigger apoptosis in these proliferating progenitors. In contrast, specific populations of postmitotic neurons derived from these progenitors, including upper layer cortical neurons and the CA1-CA3 regions of the hippocampus, were acutely dependent on Bcl-xL. Consistent with this finding, deletion of Bcl-xL selectively in the postmitotic neurons in the brain (Bcl-xL(Nex-Cre)) also resulted in similar patterns of apoptosis. This Bcl-xL deficiency-induced neuronal death was a consequence of activation of the apoptotic pathway, because the cell death was rescued with codeletion of the proapoptotic proteins Bax and Bak. Importantly, the loss of these Bcl-xL-dependent neurons led to severe neurobehavioral abnormalities, including deficits in motor learning, hyperactivity, and increased risk-taking and self-injurious behaviors. Together, our results identify a population of neurons in the developing brain that are acutely dependent on Bcl-xL during the peak period of synaptic connectivity that are important for the establishment of higher-order complex behaviors. SIGNIFICANCE STATEMENT: Although Bcl-xL is known to inhibit apoptosis, exactly which cells in the brain are dependent on Bcl-xL has remained unclear because of the embryonic lethality of mice globally deleted for Bcl-xL. Here, we conditionally deleted Bcl-xL in the brain and found that this did not result in widespread apoptosis in the proliferating progenitors. Instead, Bcl-xL deficiency induced apoptosis in a select population of differentiated neurons predominantly in the early postnatal stages. Importantly, these Bcl-xL-dependent neurons are not essential for survival of the organism but instead regulate complex behaviors. Our results show that the selective loss of these Bcl-xL-dependent neurons results in mice exhibiting severe neurobehavioral abnormalities, including self-injurious and risk-taking behaviors, hyperactivity, and learning and memory defects.


Assuntos
Apoptose , Hipocampo/metabolismo , Aprendizagem , Atividade Motora , Neurônios/metabolismo , Proteína bcl-X/metabolismo , Animais , Feminino , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/fisiologia , Masculino , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Neurogênese , Neurônios/citologia , Neurônios/fisiologia , Proteína bcl-X/genética
18.
Development ; 143(13): 2356-66, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27226321

RESUMO

The tumor suppressor protein adenomatous polyposis coli (APC) is multifunctional - it participates in the canonical Wnt/ß-catenin signal transduction pathway as well as modulating cytoskeleton function. Although APC is expressed by Schwann cells, the role that it plays in these cells and in the myelination of the peripheral nervous system (PNS) is unknown. Therefore, we used the Cre-lox approach to generate a mouse model in which APC expression is specifically eliminated from Schwann cells. These mice display hindlimb weakness and impaired axonal conduction in sciatic nerves. Detailed morphological analyses revealed that APC loss delays radial axonal sorting and PNS myelination. Furthermore, APC loss delays Schwann cell differentiation in vivo, which correlates with persistent activation of the Wnt signaling pathway and results in perturbed extension of Schwann cell processes and disrupted lamellipodia formation. In addition, APC-deficient Schwann cells display a transient diminution of proliferative capacity. Our data indicate that APC is required by Schwann cells for their timely differentiation to mature, myelinating cells and plays a crucial role in radial axonal sorting and PNS myelination.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Axônios/metabolismo , Bainha de Mielina/metabolismo , Sistema Nervoso Periférico/metabolismo , Animais , Diferenciação Celular/genética , Membro Posterior/patologia , Integrases/metabolismo , Camundongos , Pseudópodes/metabolismo , Células de Schwann/citologia , Células de Schwann/metabolismo , Nervo Isquiático/metabolismo , Via de Sinalização Wnt/genética
19.
Nat Commun ; 6: 10118, 2015 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-26656849

RESUMO

Neuregulin-1 (NRG1) and its receptor ErbB4 influence several processes of neurodevelopment, but the mechanisms regulating this signalling in the mature brain are not well known. DISC1 is a multifunctional scaffold protein that mediates many cellular processes. Here we present a functional relationship between DISC1 and NRG1-ErbB4 signalling in mature cortical interneurons. By cell type-specific gene modulation in vitro and in vivo including in a mutant DISC1 mouse model, we demonstrate that DISC1 inhibits NRG1-induced ErbB4 activation and signalling. This effect is likely mediated by competitive inhibition of binding of ErbB4 to PSD95. Finally, we show that interneuronal DISC1 affects NRG1-ErbB4-mediated phenotypes in the fast spiking interneuron-pyramidal neuron circuit. Post-mortem brain analyses and some genetic studies have reported interneuronal deficits and involvement of the DISC1, NRG1 and ErbB4 genes in schizophrenia, respectively. Our results suggest a mechanism by which cross-talk between DISC1 and NRG1-ErbB4 signalling may contribute to these deficits.


Assuntos
Interneurônios/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neuregulina-1/metabolismo , Receptor ErbB-4/metabolismo , Sinapses/fisiologia , Animais , Células Cultivadas , Córtex Cerebral , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Neuregulina-1/genética , Ratos , Ratos Sprague-Dawley , Receptor ErbB-4/genética , Transdução de Sinais/fisiologia
20.
Nat Commun ; 6: 7857, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26206566

RESUMO

Primary cilia are essential conveyors of signals underlying major cell functions. Cerebral cortical progenitors and neurons have a primary cilium. The significance of cilia function for brain development and function is evident in the plethora of developmental brain disorders associated with human ciliopathies. Nevertheless, the role of primary cilia function in corticogenesis remains largely unknown. Here we delineate the functions of primary cilia in the construction of cerebral cortex and their relevance to ciliopathies, using an shRNA library targeting ciliopathy genes known to cause brain disorders, but whose roles in brain development are unclear. We used the library to query how ciliopathy genes affect distinct stages of mouse cortical development, in particular neural progenitor development, neuronal migration, neuronal differentiation and early neuronal connectivity. Our results define the developmental functions of ciliopathy genes and delineate disrupted developmental events that are integrally related to the emergence of brain abnormalities in ciliopathies.


Assuntos
Encefalopatias/genética , Córtex Cerebral/embriologia , Cílios/genética , Animais , Feminino , Biblioteca Gênica , Humanos , Camundongos Endogâmicos C57BL , Neurogênese , Gravidez , RNA Interferente Pequeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA