Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(39): e2404395121, 2024 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-39292743

RESUMO

Adult central nervous system (CNS) neurons down-regulate growth programs after injury, leading to persistent regeneration failure. Coordinated lipids metabolism is required to synthesize membrane components during axon regeneration. However, lipids also function as cell signaling molecules. Whether lipid signaling contributes to axon regeneration remains unclear. In this study, we showed that lipin1 orchestrates mechanistic target of rapamycin (mTOR) and STAT3 signaling pathways to determine axon regeneration. We established an mTOR-lipin1-phosphatidic acid/lysophosphatidic acid-mTOR loop that acts as a positive feedback inhibitory signaling, contributing to the persistent suppression of CNS axon regeneration following injury. In addition, lipin1 knockdown (KD) enhances corticospinal tract (CST) sprouting after unilateral pyramidotomy and promotes CST regeneration following complete spinal cord injury (SCI). Furthermore, lipin1 KD enhances sensory axon regeneration after SCI. Overall, our research reveals that lipin1 functions as a central regulator to coordinate mTOR and STAT3 signaling pathways in the CNS neurons and highlights the potential of lipin1 as a promising therapeutic target for promoting the regeneration of motor and sensory axons after SCI.


Assuntos
Axônios , Neurônios Motores , Regeneração Nervosa , Fosfatidato Fosfatase , Fator de Transcrição STAT3 , Transdução de Sinais , Traumatismos da Medula Espinal , Serina-Treonina Quinases TOR , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/genética , Animais , Axônios/metabolismo , Axônios/fisiologia , Regeneração Nervosa/fisiologia , Fator de Transcrição STAT3/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Fosfatidato Fosfatase/metabolismo , Fosfatidato Fosfatase/genética , Neurônios Motores/metabolismo , Neurônios Motores/fisiologia , Camundongos , Ácidos Fosfatídicos/metabolismo , Células Receptoras Sensoriais/metabolismo , Feminino , Tratos Piramidais/metabolismo , Tratos Piramidais/patologia
2.
Sci Data ; 11(1): 987, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39256463

RESUMO

Astrocytes, the predominant glial cells in the central nervous system, play essential roles in maintaining brain function. Reprogramming induced pluripotent stem cells (iPSCs) to become astrocytes through overexpression of the transcription factors, NFIB and SOX9, is a rapid and efficient approach for studying human neurological diseases and identifying therapeutic targets. However, the precise differentiation path and molecular signatures of induced astrocytes remain incompletely understood. Accordingly, we performed single-cell RNA sequencing analysis on 64,736 cells to establish a comprehensive atlas of NFIB/SOX9-directed astrocyte differentiation from human iPSCs. Our dataset provides detailed information about the path of astrocyte differentiation, highlighting the stepwise molecular changes that occur throughout the differentiation process. This dataset serves as a valuable reference for dissecting uncharacterized transcriptomic features of NFIB/SOX9-induced astrocytes and investigating lineage progression during astrocyte differentiation. Moreover, these findings pave the way for future studies on neurological diseases using the NFIB/SOX9-induced astrocyte model.


Assuntos
Astrócitos , Diferenciação Celular , Células-Tronco Pluripotentes Induzidas , Fatores de Transcrição NFI , Fatores de Transcrição SOX9 , Transcriptoma , Fatores de Transcrição SOX9/genética , Astrócitos/metabolismo , Astrócitos/citologia , Humanos , Fatores de Transcrição NFI/genética , Fatores de Transcrição NFI/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Análise de Célula Única
3.
bioRxiv ; 2024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39091722

RESUMO

Circular RNAs (circRNAs) are noncoding RNAs abundant in brain tissue, and many are derived from activity-dependent, linear mRNAs encoding for synaptic proteins, suggesting that circRNAs may directly or indirectly play a role in regulating synaptic development, plasticity, and function. However, it is unclear if the circular forms of these RNAs are similarly regulated by activity and what role these circRNAs play in developmental plasticity. Here, we employed transcriptome-wide analysis comparing differential expression of both mRNAs and circRNAs in juvenile mouse primary visual cortex (V1) following monocular deprivation (MD), a model of developmental plasticity. Among the differentially expressed mRNAs and circRNAs following 3-day MD, the circular and the activity-dependent linear forms of the Homer1 gene, circHomer1 and Homer1a respectively, were of interest as their expression changed in opposite directions: circHomer1 expression increased while the expression of Homer1a decreased following MD. Knockdown of circHomer1 prevented the depression of closed-eye responses normally observed after 3-day MD. circHomer1-knockdown led to a reduction in average dendritic spine size prior to MD, but critically there was no further reduction after 3-day MD, consistent with impaired structural plasticity. circHomer1-knockdown also prevented the reduction of surface AMPA receptors after 3-day MD. Synapse-localized puncta of the AMPA receptor endocytic protein Arc increased in volume after MD but were smaller in circHomer1-knockdown neurons, suggesting that circHomer1 regulates plasticity through mechanisms of activity-dependent AMPA receptor endocytosis. Thus, activity-dependent circRNAs regulate developmental synaptic plasticity, and our findings highlight the essential role of circHomer1 in V1 plasticity induced by short-term MD.

4.
Nat Neurosci ; 27(2): 373-383, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38212587

RESUMO

Rabies-virus-based monosynaptic tracing is a widely used technique for mapping neural circuitry, but its cytotoxicity has confined it primarily to anatomical applications. Here we present a second-generation system for labeling direct inputs to targeted neuronal populations with minimal toxicity, using double-deletion-mutant rabies viruses. Viral spread requires expression of both deleted viral genes in trans in postsynaptic source cells. Suppressing this expression with doxycycline following an initial period of viral replication reduces toxicity to postsynaptic cells. Longitudinal two-photon imaging in vivo indicated that over 90% of both presynaptic and source cells survived for the full 12-week course of imaging. Ex vivo whole-cell recordings at 5 weeks postinfection showed that the second-generation system perturbs input and source cells much less than the first-generation system. Finally, two-photon calcium imaging of labeled networks of visual cortex neurons showed that their visual response properties appeared normal for 10 weeks, the longest we followed them.


Assuntos
Vírus da Raiva , Vírus da Raiva/genética , Neurônios/fisiologia , Replicação Viral
6.
IBRO Neurosci Rep ; 15: 218-224, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37822516

RESUMO

Rett Syndrome (RTT) is a neurodevelopmental disorder caused by pathogenic variants in the MECP2 gene. While the majority of RTT-causing variants are clustered in the methyl-CpG binding domain and NCoR/SMRT interaction domain, we report a female patient with a functionally uncharacterized MECP2 variant in the C-terminal domain, c.1030C>T (R344W). We functionally characterized MECP2-R344W in terms of protein stability, NCoR/SMRT complex interaction, and protein nuclear localization in vitro. MECP2-R344W cells showed an increased protein degradation rate without significant change in NCoR/SMRT complex interaction and nuclear localization pattern, suggesting that enhanced MECP2 degradation is sufficient to cause a Rett Syndrome-like phenotype. This study highlights the pathogenicity of the C-terminal domain in Rett Syndrome, and demonstrates the potential of targeting MECP2 protein stability as a therapeutic approach.

7.
Cell Rep Methods ; 3(5): 100462, 2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-37323579

RESUMO

Calcium imaging provides advantages in monitoring large populations of neuronal activities simultaneously. However, it lacks the signal quality provided by neural spike recording in traditional electrophysiology. To address this issue, we developed a supervised data-driven approach to extract spike information from calcium signals. We propose the ENS2 (effective and efficient neural networks for spike inference from calcium signals) system for spike-rate and spike-event predictions using ΔF/F0 calcium inputs based on a U-Net deep neural network. When testing on a large, ground-truth public database, it consistently outperformed state-of-the-art algorithms in both spike-rate and spike-event predictions with reduced computational load. We further demonstrated that ENS2 can be applied to analyses of orientation selectivity in primary visual cortex neurons. We conclude that it would be a versatile inference system that may benefit diverse neuroscience studies.


Assuntos
Modelos Neurológicos , Redes Neurais de Computação , Potenciais de Ação/fisiologia , Algoritmos , Cálcio da Dieta
8.
Stem Cell Res ; 66: 103000, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36521383

RESUMO

ABCA7 encodes an ATP-binding cassette transporter, and its loss-of-function variants are associated with Alzheimer's disease. To investigate the role of ABCA7 deficiency in the pathogenesis of Alzheimer's disease, we generated a homozygous ABCA7-knockout induced pluripotent stem cell (iPSC) line using CRISPR/Cas9-mediated gene editing. This ABCA7-deficient iPSC line maintains a normal karyotype, expression of pluripotency markers, and trilineage differentiation capacity.


Assuntos
Doença de Alzheimer , Células-Tronco Pluripotentes Induzidas , Humanos , Doença de Alzheimer/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Sistemas CRISPR-Cas/genética , Homozigoto , Transportadores de Cassetes de Ligação de ATP/genética
9.
Biomolecules ; 12(10)2022 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-36291714

RESUMO

Neurons interact with astrocytes, microglia, and vascular cells. These interactions become unbalanced in disease states, resulting in damage to neurons and synapses, and contributing to cognitive impairment. Importantly, synaptic loss and synaptic dysfunction have been considered for years as a main pathological factor of cognitive impairment in Alzheimer's disease (AD). Recently, miRNAs have emerged as essential regulators of physiological and pathological processes in the brain. Focusing on the role of miRNAs in regulating synaptic functions, as well as different cell types in the brain, offers opportunities for the early prevention, diagnosis, and potential treatment of AD-related cognitive impairment. Here, we review the recent research conducted on miRNAs regulating astrocytes, microglia, cerebrovasculature, and synaptic functions in the context of AD-related cognitive impairment. We also review potential miRNA-related biomarkers and therapeutics, as well as emerging imaging technologies relevant for AD research.


Assuntos
Doença de Alzheimer , MicroRNAs , Humanos , Doença de Alzheimer/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Sinapses/genética , Sinapses/metabolismo , Neurônios/metabolismo , Biomarcadores/metabolismo
10.
Elife ; 112022 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-35904330

RESUMO

Human cerebral organoids are unique in their development of progenitor-rich zones akin to ventricular zones from which neuronal progenitors differentiate and migrate radially. Analyses of cerebral organoids thus far have been performed in sectioned tissue or in superficial layers due to their high scattering properties. Here, we demonstrate label-free three-photon imaging of whole, uncleared intact organoids (~2 mm depth) to assess early events of early human brain development. Optimizing a custom-made three-photon microscope to image intact cerebral organoids generated from Rett Syndrome patients, we show defects in the ventricular zone volumetric structure of mutant organoids compared to isogenic control organoids. Long-term imaging live organoids reveals that shorter migration distances and slower migration speeds of mutant radially migrating neurons are associated with more tortuous trajectories. Our label-free imaging system constitutes a particularly useful platform for tracking normal and abnormal development in individual organoids, as well as for screening therapeutic molecules via intact organoid imaging.


Assuntos
Organoides , Síndrome de Rett , Encéfalo/diagnóstico por imagem , Humanos , Neurônios , Organoides/fisiologia , Síndrome de Rett/diagnóstico por imagem , Síndrome de Rett/genética
11.
Nat Metab ; 4(2): 203-212, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35177851

RESUMO

GDNF-family receptor a-like (GFRAL) has been identified as the cognate receptor of growth/differentiation factor 15 (GDF15/MIC-1), considered a key signaling axis in energy homeostasis and body weight regulation. Currently, little is known about the physiological regulation of the GDF15-GFRAL signaling pathway. Here we show that membrane-bound matrix metalloproteinase 14 (MT1-MMP/MMP14) is an endogenous negative regulator of GFRAL in the context of obesity. Overnutrition-induced obesity increased MT1-MMP activation, which proteolytically inactivated GFRAL to suppress GDF15-GFRAL signaling, thus modulating the anorectic effects of the GDF15-GFRAL axis in vivo. Genetic ablation of MT1-MMP specifically in GFRAL+ neurons restored GFRAL expression, resulting in reduced weight gain, along with decreased food intake in obese mice. Conversely, depletion of GFRAL abolished the anti-obesity effects of MT1-MMP inhibition. MT1-MMP inhibition also potentiated GDF15 activity specifically in obese phenotypes. Our findings identify a negative regulator of GFRAL for the control of non-homeostatic body weight regulation, provide mechanistic insights into the regulation of GDF15 sensitivity, highlight negative regulators of the GDF15-GFRAL pathway as a therapeutic avenue against obesity and identify MT1-MMP as a promising target.


Assuntos
Metaloproteinase 14 da Matriz , Obesidade , Animais , Anorexia/metabolismo , Peso Corporal , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Metaloproteinase 14 da Matriz/uso terapêutico , Camundongos , Obesidade/metabolismo
12.
Front Neural Circuits ; 15: 803401, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34949992

RESUMO

Neurons remodel the structure and strength of their synapses during critical periods of development in order to optimize both perception and cognition. Many of these developmental synaptic changes are thought to occur through synapse-specific homosynaptic forms of experience-dependent plasticity. However, homosynaptic plasticity can also induce or contribute to the plasticity of neighboring synapses through heterosynaptic interactions. Decades of research in vitro have uncovered many of the molecular mechanisms of heterosynaptic plasticity that mediate local compensation for homosynaptic plasticity, facilitation of further bouts of plasticity in nearby synapses, and cooperative induction of plasticity by neighboring synapses acting in concert. These discoveries greatly benefited from new tools and technologies that permitted single synapse imaging and manipulation of structure, function, and protein dynamics in living neurons. With the recent advent and application of similar tools for in vivo research, it is now feasible to explore how heterosynaptic plasticity contribute to critical periods and the development of neuronal circuits. In this review, we will first define the forms heterosynaptic plasticity can take and describe our current understanding of their molecular mechanisms. Then, we will outline how heterosynaptic plasticity may lead to meaningful refinement of neuronal responses and observations that suggest such mechanisms are indeed at work in vivo. Finally, we will use a well-studied model of cortical plasticity-ocular dominance plasticity during a critical period of visual cortex development-to highlight the molecular overlap between heterosynaptic and developmental forms of plasticity, and suggest potential avenues of future research.


Assuntos
Plasticidade Neuronal , Córtex Visual , Neurônios , Sinapses
13.
Proc Natl Acad Sci U S A ; 116(27): 13651-13660, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31209021

RESUMO

Adult hippocampal neurogenesis involves the lifelong generation of neurons. The process depends on the homeostasis of the production of neurons and maintenance of the adult neural stem cell (NSC) pool. Here, we report that α2-chimaerin, a Rho GTPase-activating protein, is essential for NSC homeostasis in adult hippocampal neurogenesis. Conditional deletion of α2-chimaerin in adult NSCs resulted in the premature differentiation of NSCs into intermediate progenitor cells (IPCs), which ultimately depleted the NSC pool and impaired neuron generation. Single-cell RNA sequencing and pseudotime analyses revealed that α2-chimaerin-conditional knockout (α2-CKO) mice lacked a unique NSC subpopulation, termed Klotho-expressing NSCs, during the transition of NSCs to IPCs. Furthermore, α2-CKO led to defects in hippocampal synaptic plasticity and anxiety/depression-like behaviors in mice. Our findings collectively demonstrate that α2-chimaerin plays an essential role in adult hippocampal NSC homeostasis to maintain proper brain function.


Assuntos
Proteínas Quimerinas/fisiologia , Ativadores de GTP Fosfo-Hidrolase/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Animais , Diferenciação Celular , Técnicas de Silenciamento de Genes , Hipocampo/fisiologia , Homeostase , Camundongos , Camundongos Knockout , Células-Tronco Neurais/fisiologia , Células-Tronco/fisiologia
14.
Science ; 360(6395): 1349-1354, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29930137

RESUMO

Plasticity of cortical responses in vivo involves activity-dependent changes at synapses, but the manner in which different forms of synaptic plasticity act together to create functional changes in neurons remains unknown. We found that spike timing-induced receptive field plasticity of visual cortex neurons in mice is anchored by increases in the synaptic strength of identified spines. This is accompanied by a decrease in the strength of adjacent spines on a slower time scale. The locally coordinated potentiation and depression of spines involves prominent AMPA receptor redistribution via targeted expression of the immediate early gene product Arc. Hebbian strengthening of activated synapses and heterosynaptic weakening of adjacent synapses thus cooperatively orchestrate cell-wide plasticity of functional neuronal responses.


Assuntos
Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Córtex Visual/fisiologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteínas do Citoesqueleto/genética , Espinhas Dendríticas/fisiologia , Eletroporação , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Potenciação de Longa Duração/fisiologia , Depressão Sináptica de Longo Prazo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Receptores de AMPA/genética , Receptores de AMPA/metabolismo , Transmissão Sináptica , Córtex Visual/citologia , Córtex Visual/metabolismo
15.
Nat Rev Neurosci ; 19(6): 368-382, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29740174

RESUMO

Rett syndrome (RTT) is a severe neurological disorder caused by mutations in the gene encoding methyl-CpG-binding protein 2 (MeCP2). Almost two decades of research into RTT have greatly advanced our understanding of the function and regulation of the multifunctional protein MeCP2. Here, we review recent advances in understanding how loss of MeCP2 impacts different stages of brain development, discuss recent findings demonstrating the molecular role of MeCP2 as a transcriptional repressor, assess primary and secondary effects of MeCP2 loss and examine how loss of MeCP2 can result in an imbalance of neuronal excitation and inhibition at the circuit level along with dysregulation of activity-dependent mechanisms. These factors present challenges to the search for mechanism-based therapeutics for RTT and suggest specific approaches that may be more effective than others.


Assuntos
Encéfalo/fisiopatologia , Proteína 2 de Ligação a Metil-CpG/genética , Síndrome de Rett/genética , Síndrome de Rett/fisiopatologia , Animais , Encéfalo/crescimento & desenvolvimento , Diferenciação Celular , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , MicroRNAs/genética , Mutação , Neurônios/fisiologia
16.
J Neurosci ; 38(16): 3890-3900, 2018 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-29540554

RESUMO

Microdeletion of a region in chromosome 16p11.2 increases susceptibility to autism. Although this region contains exons of 29 genes, disrupting only a small segment of the region, which spans five genes, is sufficient to cause autistic traits. One candidate gene in this critical segment is MVP, which encodes for the major vault protein (MVP) that has been implicated in regulation of cellular transport mechanisms. MVP expression levels in MVP+/- mice closely phenocopy those of 16p11.2 mutant mice, suggesting that MVP+/- mice may serve as a model of MVP function in 16p11.2 microdeletion. Here we show that MVP regulates the homeostatic component of ocular dominance (OD) plasticity in primary visual cortex. MVP+/- mice of both sexes show impairment in strengthening of open-eye responses after several days of monocular deprivation (MD), whereas closed-eye responses are weakened as normal, resulting in reduced overall OD plasticity. The frequency of miniature EPSCs (mEPSCs) in pyramidal neurons is decreased in MVP+/- mice after extended MD, suggesting a reduction of functional synapses. Correspondingly, upregulation of surface GluA1 AMPA receptors is reduced in MVP+/- mice after extended MD, and is accompanied by altered expression of STAT1 and phosphorylated ERK, which have been previously implicated in OD plasticity. Normalization of STAT1 levels by introducing STAT1 shRNA rescues surface GluA1 and open-eye responses, implicating STAT1 as a downstream effector of MVP. These findings demonstrate a specific role for MVP as a key molecule influencing the homeostatic component of activity-dependent synaptic plasticity, and potentially the corresponding phenotypes of 16p11.2 microdeletion syndrome.SIGNIFICANCE STATEMENT Major vault protein (MVP), a candidate gene in 16p11.2 microdeletion syndrome, has been implicated in the regulation of several cellular processes including transport mechanisms and scaffold signaling. However, its role in brain function and plasticity remains unknown. In this study, we identified MVP as an important regulator of the homeostatic component of experience-dependent plasticity, via regulation of STAT1 and ERK signaling. This study helps reveal a new mechanism for an autism-related gene in brain function, and suggests a broader role for neuro-immune interactions in circuit level plasticity. Importantly, our findings might explain specific components of the pathophysiology of 16p11.2 microdeletion syndrome.


Assuntos
Transtorno Autístico/genética , Transtornos Cromossômicos/genética , Deficiência Intelectual/genética , Plasticidade Neuronal , Partículas de Ribonucleoproteínas em Forma de Abóbada/metabolismo , Córtex Visual/fisiologia , Animais , Deleção Cromossômica , Cromossomos Humanos Par 16/genética , Dominância Ocular , Potenciais Pós-Sinápticos Excitadores , Feminino , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Potenciais Pós-Sinápticos em Miniatura , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Células Piramidais/metabolismo , Células Piramidais/fisiologia , Receptores de AMPA/metabolismo , Fator de Transcrição STAT1/metabolismo , Partículas de Ribonucleoproteínas em Forma de Abóbada/genética , Córtex Visual/citologia , Córtex Visual/metabolismo
17.
Nat Commun ; 5: 4826, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25189171

RESUMO

During cerebral cortex development, pyramidal neurons migrate through the intermediate zone and integrate into the cortical plate. These neurons undergo the multipolar-bipolar transition to initiate radial migration. While perturbation of this polarity acquisition leads to cortical malformations, how this process is initiated and regulated is largely unknown. Here we report that the specific upregulation of the Rap1 guanine nucleotide exchange factor, RapGEF2, in migrating neurons corresponds to the timing of this polarity transition. In utero electroporation and live-imaging studies reveal that RapGEF2 acts on the multipolar-bipolar transition during neuronal migration via a Rap1/N-cadherin pathway. Importantly, activation of RapGEF2 is controlled via phosphorylation by a serine/threonine kinase Cdk5, whose activity is largely restricted to the radial migration zone. Thus, the specific expression and Cdk5-dependent phosphorylation of RapGEF2 during multipolar-bipolar transition within the intermediate zone are essential for proper neuronal migration and wiring of the cerebral cortex.


Assuntos
Movimento Celular/fisiologia , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Quinase 5 Dependente de Ciclina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Eletroporação , Imunofluorescência , Humanos , Imuno-Histoquímica , Fosforilação , RNA Interferente Pequeno/genética
18.
J Neurosci ; 34(22): 7425-36, 2014 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-24872548

RESUMO

The radial migration of newborn neurons is critical for the lamination of the cerebral cortex. Proper neuronal migration requires precise and rapid reorganization of the actin and microtubule cytoskeleton. However, the underlying signaling mechanisms controlling cytoskeletal reorganization are not well understood. Here, we show that Mst3, a serine/threonine kinase highly expressed in the developing mouse brain, is essential for radial neuronal migration and final neuronal positioning in the developing mouse neocortex. Mst3 silencing by in utero electroporation perturbed the multipolar-to-bipolar transition of migrating neurons and significantly retards radial migration. Although the kinase activity of Mst3 is essential for its functions in neuronal morphogenesis and migration, it is regulated via its phosphorylation at Ser79 by a serine/threonine kinase, cyclin-dependent kinase 5 (Cdk5). Our results show that Mst3 regulates neuronal migration through modulating the activity of RhoA, a Rho-GTPase critical for actin cytoskeletal reorganization. Mst3 phosphorylates RhoA at Ser26, thereby negatively regulating the GTPase activity of RhoA. Importantly, RhoA knockdown successfully rescues neuronal migration defect in Mst3-knockdown cortices. Our findings collectively suggest that Cdk5-Mst3 signaling regulates neuronal migration via RhoA-dependent actin dynamics.


Assuntos
Movimento Celular/fisiologia , Quinase 5 Dependente de Ciclina/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/fisiologia , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Células Cultivadas , Ativação Enzimática/fisiologia , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Dados de Sequência Molecular , Neocórtex/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Fosforilação/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Ratos , Proteína rhoA de Ligação ao GTP
19.
Neuroscientist ; 20(6): 589-98, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24402611

RESUMO

Cytoskeletal restructuring is essential for nearly all cellular processes in the developing brain. After cell fate determination, newborn cortical neurons must migrate to their final positions while establishing proper axon-dendrite polarity. Significant progress has recently been made towards understanding the cellular and molecular mechanisms underlying neuronal polarization in vivo. Collapsin response mediator protein 2 (CRMP2) has long been identified as a microtubule-binding protein that regulates neuronal polarity in vitro. Recent studies provide new insights into the roles of CRMP2 in neuronal migration and subsequent neuronal differentiation. Both the expression and activity of CRMP2 are tightly regulated during cortex development. CRMP2 is suggested to be important in the multipolar-bipolar transition in radial migration. The increasing number of known interaction partners indicates that CRMP2 has functions beyond cytoskeletal regulation, including axonal transport, vesicle trafficking, and neurotransmitter release. This review discusses the current knowledge about CRMP2 in the context of neuronal development and highlights a recent emerging theme regarding its potential therapeutic applications.


Assuntos
Encefalopatias/terapia , Encéfalo/crescimento & desenvolvimento , Movimento Celular , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Animais , Encefalopatias/fisiopatologia , Polaridade Celular , Cones de Crescimento/fisiologia , Humanos
20.
J Virol ; 88(1): 679-89, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24173226

RESUMO

Lipocalin 2 (Lcn2) is a bacteriostatic factor produced during the innate immune response to bacterial infection. Whether Lcn2 has a function in viral infection is unknown. We investigated the regulation and function of Lcn2 in the central nervous system (CNS) of mice during West Nile virus (WNV) encephalitis. Lcn2 mRNA and protein were induced in the brain by day 5, and this induction increased further by day 7 postinfection but was delayed compared with the induction of the toll-like receptor 3 (TLR3) gene, retinoic acid-inducible gene 1 (RIG-I), and melanoma differentiation-associated protein 5 (MDA5) gene. The Lcn2 mRNA and protein were both found at high levels in the choroid plexus, vascular endothelium, macrophage/microglia, and astrocytes. However, some neuronal subsets contained Lcn2 protein but no detectable mRNA. In Lcn2 knockout (KO) mice, with the exception of CXC motif chemokine 5 (CXCL5), which was significantly more downregulated than in wild-type (WT) mice, expression levels of a number of other host response genes were similar in the two genotypes. The brain from Lcn2 and WT mice with WNV encephalitis contained similar numbers of infiltrating macrophages, granulocytes, and T cells. Lcn2 KO and WT mice had no significant difference in tissue viral loads or survival after infection with different doses of WNV. We conclude that Lcn2 gene expression is induced to high levels in a time-dependent fashion in a variety of cells and regions of the CNS of mice with WNV encephalitis. The function of Lcn2 in the host response to WNV infection remains largely unknown, but our data indicate that it is dispensable as an antiviral or immunoregulatory factor in WNV encephalitis.


Assuntos
Proteínas de Fase Aguda/metabolismo , Sistema Nervoso Central/metabolismo , Lipocalinas/metabolismo , Proteínas Oncogênicas/metabolismo , Febre do Nilo Ocidental/metabolismo , Animais , Hibridização In Situ , Lipocalina-2 , Camundongos , Camundongos Endogâmicos C57BL , Regulação para Cima , Febre do Nilo Ocidental/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA