Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Cereb Cortex ; 29(3): 1280-1290, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29462287

RESUMO

Neural progenitor proliferation and cell fate decision from self-renewal to differentiation are crucial factors in determining brain size and morphology. The cytoskeletal dependent regulation of these processes is not entirely known. The actin-binding filamin A (FlnA) was shown to regulate proliferation of progenitors by directing changes in cell cycles proteins such as Cdk1 during G2/M phase. Here we report that functional loss of FlnA not only affects the rate of proliferation by altering cell cycle length but also causes a defect in early differentiation through changes in cell fate specification. FlnA interacts with Rho GTPase RhoA, and FlnA loss impairs RhoA activation. Disruption of either of these cytoskeletal associated proteins delays neurogenesis and promotes neural progenitors to remain in proliferative states. Aurora kinase B (Aurkb) has been implicated in cytokinesis, and peaks in expression during the G2/M phase. Inhibition of FlnA or RhoA impairs Aurkb degradation and alters its localization during mitosis. Overexpression of Aurkb replicates the same delay in neurogenesis seen with loss of FlnA or RhoA. Our findings suggest that shared cytoskeletal processes can direct neural progenitor proliferation by regulating the expression and localization of proteins that are implicated in the cell cycle progression and cell fate specification.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Citoesqueleto/fisiologia , Filaminas/fisiologia , Mitose/fisiologia , Células-Tronco Neurais/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Animais , Aurora Quinase B/fisiologia , Diferenciação Celular , Proliferação de Células , Camundongos , Neurogênese
2.
Cereb Cortex ; 29(5): 1938-1952, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29659741

RESUMO

Although neural progenitor proliferation along the ventricular zone is regulated by ß-catenin through Wnt signaling, the cytoskeletal mechanisms that regulate expression and localization of these proteins are not well understood. Our prior studies have shown that loss of the actin-binding Filamin A (FlnA) and actin-nucleating protein Formin 2 (Fmn2) impairs endocytosis of low-density-lipoprotein-receptor-related protein 6 (Lrp6), thereby disrupting ß-catenin activation, resulting in decreased brain size. Here, we report that activated RhoA-GTPase disengages Fmn2 N- to C-terminal binding to promote Fmn2 activation and redistribution into lysosomal vesicles. Fmn2 colocalizes with ß-catenin in lysosomes and promotes its degradation. Further, Fmn2 binds the E3 ligase Smurf2, enhances Smurf2-dependent ubiquitination, and degradation of Dishevelled-2 (Dvl2), thereby initiates ß-catenin degradation. Finally, Fmn2 overexpression disrupts neuroepithelial integrity, neuronal migration, and proliferation-phenotypes in E13 mouse embryos, as seen with loss of Fmn2+FlnA function. Conversely, co-expression of Dvl2 with Fmn2 rescues the proliferation defect due to Fmn2 overexpression in mouse embryos. These findings suggest that there is a homeostatic feedback mechanism in the cytoskeletal-dependent regulation of neural proliferation within the cerebral cortex. Upstream, Fmn2 promotes proliferation by stabilizing the Lrp6 receptor, leading to ß-catenin activation. Downstream, RhoA-activated Fmn2 promotes lysosomal degradation of Dvl2, leading to ß-catenin degradation.


Assuntos
Forminas/metabolismo , Lisossomos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Proteólise , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Caderinas/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Camundongos , Ubiquitina-Proteína Ligases/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
3.
Hum Mol Genet ; 26(7): 1294-1304, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28175289

RESUMO

Filamins are a family of actin-binding proteins responsible for diverse biological functions in the context of regulating actin dynamics and vesicle trafficking. Disruption of these proteins has been implicated in multiple human developmental disorders. To investigate the roles of different filamin isoforms, we focused on FlnA and FlnB interactions in the cartilage growth plate, since mutations in both molecules cause chondrodysplasias. Current studies show that FlnA and FlnB share a common function in stabilizing the actin cytoskeleton, they physically interact in the cytoplasm of chondrocytes, and loss of FlnA enhances FlnB expression of chondrocytes in the growth plate (and vice versa), suggesting compensation. Prolonged FlnB loss, however, promotes actin-stress fiber formation following plating onto an integrin activating substrate whereas FlnA inhibition leads to decreased actin formation. FlnA more strongly binds RhoA, although both filamins overlap with RhoA expression in the cell cytoplasm. FlnA promotes RhoA activation whereas FlnB indirectly inhibits this pathway. Moreover, FlnA loss leads to diminished expression of ß1-integrin, whereas FlnB loss promotes integrin expression. Finally, fibronectin mediated integrin activation has been shown to activate RhoA and activated RhoA leads to stress fiber formation and cell spreading. Fibronectin stimulation in null FlnA cells impairs enhanced spreading whereas FlnB inhibited cells show enhanced spreading. While filamins serve a primary static function in stabilization of the actin cytoskeleton, these studies are the first to demonstrate a dynamic and antagonistic relationship between different filamin isoforms in the dynamic regulation of integrin expression, RhoGTPase activity and actin stress fiber remodeling.


Assuntos
Filaminas/genética , Fibras de Estresse/genética , Proteína rhoA de Ligação ao GTP/genética , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Condrócitos/metabolismo , Fibronectinas/metabolismo , Filaminas/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Lâmina de Crescimento/metabolismo , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Ligação Proteica , Fibras de Estresse/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
4.
Development ; 143(23): 4509-4520, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27789627

RESUMO

Actin-associated proteins regulate multiple cellular processes, including proliferation and differentiation, but the molecular mechanisms underlying these processes are unclear. Here, we report that the actin-binding protein filamin A (FlnA) physically interacts with the actin-nucleating protein formin 2 (Fmn2). Loss of FlnA and Fmn2 impairs proliferation, thereby generating multiple embryonic phenotypes, including microcephaly. FlnA interacts with the Wnt co-receptor Lrp6. Loss of FlnA and Fmn2 impairs Lrp6 endocytosis, downstream Gsk3ß activity, and ß-catenin accumulation in the nucleus. The proliferative defect in Flna and Fmn2 null neural progenitors is rescued by inhibiting Gsk3ß activity. Our findings thus reveal a novel mechanism whereby actin-associated proteins regulate proliferation by mediating the endocytosis and transportation of components in the canonical Wnt pathway. Moreover, the Fmn2-dependent signaling in this pathway parallels that seen in the non-canonical Wnt-dependent regulation of planar cell polarity through the Formin homology protein Daam. These studies provide evidence for integration of actin-associated processes in directing neuroepithelial proliferation.


Assuntos
Proliferação de Células/fisiologia , Endocitose/fisiologia , Filaminas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Nucleares/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Diferenciação Celular , Linhagem Celular , Membrana Celular/fisiologia , Proliferação de Células/genética , Filaminas/genética , Forminas , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta/metabolismo , Células HEK293 , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Camundongos Knockout , Microcefalia/genética , Proteínas dos Microfilamentos/genética , Proteínas do Tecido Nervoso , Proteínas Nucleares/genética , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
5.
Hum Mol Genet ; 25(9): 1714-27, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26911678

RESUMO

Down syndrome (DS) is caused by a triplication of chromosome 21 (HSA21). Increased oxidative stress, decreased neurogenesis and synaptic dysfunction from HSA21 gene overexpression are thought to cause mental retardation, dementia and seizure in this disorder. Recent epigenetic studies have raised the possibility that DNA methylation has significant effects on DS neurodevelopment. Here, we performed methylome profiling in normal and DS fetal cortices and observed a significant hypermethylation in ∼4% of probes in the DS samples compared with age-matched normals. The probes with differential methylation were distributed across all chromosomes, with no enrichment on HSA21. Functional annotation and pathway analyses showed that genes in the ubiquitination pathway were significantly altered, including: BRCA1, TSPYL5 and PEX10 HSA21 located DNMT3L was overexpressed in DS neuroprogenitors, and this overexpression increased the promoter methylation of TSPYL5 potentially through DNMT3B, and decreased its mRNA expression. DNMT3L overexpression also increased mRNA levels for TP53 and APP, effectors of TSPYL5 Furthermore, DNMT3L overexpression increased APP and PSD95 expression in differentiating neurons, whereas DNMT3LshRNA could partially rescue the APP and PSD95 up-regulation in DS cells. These results provide some of the first mechanistic insights into causes for epigenetic changes in DS, leading to modification of genes relevant for the DS neural endophenotype.


Assuntos
Córtex Cerebral/metabolismo , Cromossomos Humanos Par 21/genética , DNA (Citosina-5-)-Metiltransferases/genética , Síndrome de Down/genética , Epigênese Genética/genética , Feto/metabolismo , Biomarcadores/metabolismo , Proteínas Sanguíneas/genética , Estudos de Casos e Controles , Células Cultivadas , Córtex Cerebral/patologia , Metilação de DNA , Proteína 4 Homóloga a Disks-Large , Feto/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Proteínas Nucleares/genética , Estresse Oxidativo , Proteínas de Ligação a Poli(A)/genética , Regiões Promotoras Genéticas , Ubiquitinação
6.
Hum Mol Genet ; 23(17): 4663-73, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24760772

RESUMO

Filamin B (FlnB) is an actin-binding protein thought to transduce signals from various membrane receptors and intracellular proteins onto the actin cytoskeleton. Formin1 (Fmn1) is an actin-nucleating protein, implicated in actin assembly and intracellular signaling. Human mutations in FLNB cause several skeletal disorders associated with dwarfism and early bone fusion. Mouse mutations in Fmn1 cause aberrant fusion of carpal digits. We report here that FlnB and Fmn1 physically interact, are co-expressed in chondrocytes in the growth plate and share overlapping expression in the cell cytoplasm and nucleus. Loss of FlnB leads to a dramatic decrease in Fmn1 expression at the hypertrophic-to-ossification border. Loss of Fmn1-FlnB in mice leads to a more severe reduction in body size, weight and growth plate length, than observed in mice following knockout of either gene alone. Shortening of the long bone is associated with a decrease in chondrocyte proliferation and an overall delay in ossification in the double-knockout mice. In contrast to FlnB null, Fmn1 loss results in a decrease in the width of the prehypertrophic zone. Loss of both proteins, however, causes an overall decrease in the width of the proliferation zone and an increase in the differentiated hypertrophic zone. The current findings suggest that Fmn1 and FlnB have shared and independent functions. FlnB loss promotes prehypertrophic differentiation whereas Fmn1 leads to a delay. Both proteins, however, regulate chondrocyte proliferation, and FlnB may regulate Fmn1 function at the hypertrophic-to-ossification border, thereby explaining the overall delay in ossification.


Assuntos
Diferenciação Celular , Condrócitos/metabolismo , Condrócitos/patologia , Proteínas Fetais/metabolismo , Filaminas/metabolismo , Lâmina de Crescimento/metabolismo , Lâmina de Crescimento/patologia , Proteínas dos Microfilamentos/metabolismo , Proteínas Nucleares/metabolismo , Animais , Calcificação Fisiológica , Proliferação de Células , Proteínas Fetais/deficiência , Filaminas/deficiência , Forminas , Humanos , Hipertrofia , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Proteínas Nucleares/deficiência , Ligação Proteica , Transporte Proteico , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo
7.
Neurobiol Dis ; 77: 106-16, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25747816

RESUMO

The basic helix-loop-helix (bHLH) transcription factor Olig2 is crucial for mammalian central nervous system development. Human ortholog OLIG2 is located in the Down syndrome critical region in trisomy 21. To investigate the effect of Olig2 misexpression on brain development, we generated a developmentally regulated Olig2-overexpressing transgenic line with a Cre/loxP system. The transgenic mice with Olig2 misexpression in cortical neural stem/progenitor cells exhibited microcephaly, cortical dyslamination, hippocampus malformation, and profound motor deficits. Ectopic misexpression of Olig2 impaired cortical progenitor proliferation and caused precocious cell cycle exit. Massive neuronal cell death was detected in the developing cortex of Olig2-misexpressing mice. In addition, Olig2 misexpression led to a significant downregulation of neuronal specification factors including Ngn1, Ngn2 and Pax6, and a defect in cortical neurogenesis. Chromatin-immunoprecipitation and sequencing (ChIP-Seq) analysis indicates that Olig2 directly targets the promoter and/or enhancer regions of Nfatc4, Dscr1/Rcan1 and Dyrk1a, the critical neurogenic genes that contribute to Down syndrome phenotypes, and inhibits their expression. Together, our study suggests that Olig2 misexpression in neural stem cells elicits neurogenesis defects and neuronal cell death, which may contribute to developmental disorders including Down syndrome, where OLIG2 is triplicated on chromosomal 21.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Córtex Cerebral , Síndrome de Down/genética , Síndrome de Down/patologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas do Tecido Nervoso/genética , Neurogênese/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Calbindinas/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Morte Celular/genética , Córtex Cerebral/embriologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/patologia , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Embrião de Mamíferos , Proteínas de Homeodomínio/metabolismo , Interneurônios/metabolismo , Interneurônios/patologia , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Fatores do Domínio POU/metabolismo , Parvalbuminas/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Repetições de Trinucleotídeos/genética
8.
J Neurosci ; 33(40): 15735-46, 2013 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-24089482

RESUMO

Periventricular heterotopias is a malformation of cortical development, characterized by ectopic neuronal nodules around ventricle lining and caused by an initial migration defect during early brain development. Human mutations in the Filamin A (FLNA) and ADP-ribosylation factor guanine exchange factor 2 [ARFGEF2; encoding brefeldin-A-inhibited guanine exchange factor-2 (BIG2)] genes give rise to this disorder. Previously, we have reported that Big2 inhibition impairs neuronal migration and binds to FlnA, and its loss promotes FlnA phosphorylation. FlnA phosphorylation dictates FlnA-actin binding affinity and consequently alters focal adhesion size and number to effect neuronal migration. Here we show that FlnA loss similarly impairs migration, reciprocally enhances Big2 expression, but also alters Big2 subcellular localization in both null and conditional FlnA mice. FlnA phosphorylation promotes relocalization of Big2 from the Golgi toward the lipid ruffles, thereby activating Big2-dependent Arf1 at the cell membrane. Loss of FlnA phosphorylation or Big2 function impairs Arf1-dependent vesicle trafficking at the periphery, and Arf1 is required for maintenance of cell-cell junction connectivity and focal adhesion assembly. Loss of Arf1 activity disrupts neuronal migration and cell adhesion. Collectively, these studies demonstrate a potential mechanism whereby coordinated interactions between actin (through FlnA) and vesicle trafficking (through Big2-Arf) direct the assembly and disassembly of membrane protein complexes required for neuronal migration and neuroependymal integrity.


Assuntos
Fator 1 de Ribosilação do ADP/metabolismo , Movimento Celular/fisiologia , Filaminas/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Neurônios/metabolismo , Fator 1 de Ribosilação do ADP/genética , Animais , Adesão Celular/fisiologia , Filaminas/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Fosforilação
9.
Hum Mol Genet ; 21(10): 2330-40, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22343408

RESUMO

Mental retardation and early Alzheimer's disease (AD) have generally been attributed to progressive neuronal loss in the developing and mature Down syndrome (DS) brain. However, reduced neuronal production during development could also contribute to the smaller brain size and simplified gyral patterning seen in this disorder. Here, we show impairments in proliferation within the ventricular zone (VZ) of early DS fetal cortex and in cultured early passage DS human neural progenitors (HNPs). We find that the reduced proliferative rates correspond temporally with increased expression of the chromosome 21 (HSA21) associated, oligodendrocyte transcription factor OLIG2 at 14-18 weeks gestational age (GA) (period of neurogenesis). Moreover, the DS HNPs adopt more oligodendrocyte-specific features including increased oligodendrocyte marker expression, as well as a reduction in KCNA3 potassium channel expression and function. We further show that OLIG2 inhibition or over-expression regulates potassium channel expression levels and that activation or inhibition of these channels influences the rate of progenitor proliferation. Finally, neural progenitors from Olig2 over-expressing transgenic mice exhibit these same impairments in proliferation and potassium channel expression. These findings suggest that OLIG2 over-expression inhibits neural progenitor proliferation through changes in potassium channel activity, thereby contributing to the reduced neuronal numbers and brain size in DS.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proliferação de Células , Síndrome de Down/patologia , Expressão Gênica , Proteínas do Tecido Nervoso/genética , Células-Tronco Neurais/patologia , Neurônios/patologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Síndrome de Down/genética , Síndrome de Down/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Neurônios/metabolismo , Fator de Transcrição 2 de Oligodendrócitos
10.
J Neurosci ; 32(36): 12619-29, 2012 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-22956851

RESUMO

Periventricular heterotopia (PH) is a human malformation of cortical development associated with gene mutations in ADP-ribosylation factor guanine exchange factor 2 (ARFGEF2 encodes for Big2 protein) and Filamin A (FLNA). PH is thought to derive from neuroependymal disruption, but the extent to which neuronal migration contributes to this phenotype is unknown. Here, we show that Arfgef2 null mice develop PH and exhibit impaired neural migration with increased protein expression for both FlnA and phosphoFlnA at Ser2152. Big2 physically interacts with FlnA and overexpression of phosphomimetic Ser2512 FLNA impairs neuronal migration. FlnA phosphorylation directs FlnA localization toward the cell cytoplasm, diminishes its binding affinity to actin skeleton, and alters the number and size of paxillin focal adhesions. Collectively, our results demonstrate a molecular mechanism whereby Big2 inhibition promotes phosphoFlnA (Ser2152) expression, and increased phosphoFlnA impairs its actin binding affinity and the distribution of focal adhesions, thereby disrupting cell intrinsic neuronal migration.


Assuntos
Movimento Celular/fisiologia , Proteínas Contráteis/metabolismo , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Proteínas dos Microfilamentos/metabolismo , Neurônios/fisiologia , Animais , Feminino , Filaminas , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/fisiologia
11.
J Neurosci ; 32(22): 7672-84, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22649246

RESUMO

Cytoskeleton-associated proteins play key roles not only in regulating cell morphology and migration but also in proliferation. Mutations in the cytoskeleton-associated gene filamin A (FlnA) cause the human disorder periventricular heterotopia (PH). PH is a disorder of neural stem cell development that is characterized by disruption of progenitors along the ventricular epithelium and subsequent formation of ectopic neuronal nodules. FlnA-dependent regulation of cytoskeletal dynamics is thought to direct neural progenitor migration and proliferation. Here we show that embryonic FlnA-null mice exhibited a reduction in brain size and decline in neural progenitor numbers over time. The drop in the progenitor population was not attributable to cell death or changes in premature differentiation, but to prolonged cell cycle duration. Suppression of FlnA led to prolongation of the entire cell cycle length, principally in M phase. FlnA loss impaired degradation of cyclin B1-related proteins, thereby delaying the onset and progression through mitosis. We found that the cdk1 kinase Wee1 bound FlnA, demonstrated increased expression levels after loss of FlnA function, and was associated with increased phosphorylation of cdk1. Phosphorylation of cdk1 inhibited activation of the anaphase promoting complex degradation system, which was responsible for cyclin B1 degradation and progression through mitosis. Collectively, our results demonstrate a molecular mechanism whereby FlnA loss impaired G2 to M phase entry, leading to cell cycle prolongation, compromised neural progenitor proliferation, and reduced brain size.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Córtex Cerebral/fisiologia , Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Células-Tronco Neurais/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fatores Etários , Animais , Bromodesoxiuridina/metabolismo , Proteína Quinase CDC2/genética , Ciclo Celular/genética , Diferenciação Celular/genética , Células Cultivadas , Córtex Cerebral/citologia , Proteínas Contráteis/deficiência , Ciclina B1/metabolismo , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos , Filaminas , Citometria de Fluxo , Regulação Enzimológica da Expressão Gênica/genética , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Antígeno Ki-67 , Camundongos , Camundongos Transgênicos , Microcefalia/genética , Proteínas dos Microfilamentos/deficiência , Heterotopia Nodular Periventricular/genética , Heterotopia Nodular Periventricular/patologia , Fosforilação/genética , Proteínas com Domínio T/metabolismo , Tirosina/metabolismo
12.
Nat Genet ; 36(1): 69-76, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14647276

RESUMO

Disruption of human neural precursor proliferation can give rise to a small brain (microcephaly), and failure of neurons to migrate properly can lead to an abnormal arrest of cerebral cortical neurons in proliferative zones near the lateral ventricles (periventricular heterotopia). Here we show that an autosomal recessive condition characterized by microcephaly and periventricular heterotopia maps to chromosome 20 and is caused by mutations in the gene ADP-ribosylation factor guanine nucleotide-exchange factor-2 (ARFGEF2). By northern-blot analysis, we found that mouse Arfgef2 mRNA levels are highest during embryonic periods of ongoing neuronal proliferation and migration, and by in situ hybridization, we found that the mRNA is widely distributed throughout the embryonic central nervous system (CNS). ARFGEF2 encodes the large (>200 kDa) brefeldin A (BFA)-inhibited GEF2 protein (BIG2), which is required for vesicle and membrane trafficking from the trans-Golgi network (TGN). Inhibition of BIG2 by BFA, or by a dominant negative ARFGEF2 cDNA, decreases cell proliferation in vitro, suggesting a cell-autonomous regulation of neural expansion. Inhibition of BIG2 also disturbed the intracellular localization of such molecules as E-cadherin and beta-catenin by preventing their transport from the Golgi apparatus to the cell surface. Our findings show that vesicle trafficking is an important regulator of proliferation and migration during human cerebral cortical development.


Assuntos
Fatores de Ribosilação do ADP/genética , Córtex Cerebral/fisiologia , Fatores de Troca do Nucleotídeo Guanina/genética , Proteínas de Saccharomyces cerevisiae , Adolescente , Sequência de Aminoácidos , Animais , Divisão Celular , Movimento Celular , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Imageamento por Ressonância Magnética , Camundongos , Dados de Sequência Molecular , Mutação , Neurônios/fisiologia
13.
Biomacromolecules ; 13(2): 358-68, 2012 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-22206477

RESUMO

We present a method of tuning surface chemistry and nerve cell behavior by photo-cross-linking methoxy poly(ethylene glycol) monoacrylate (mPEGA) with hydrophobic, semicrystalline poly(ε-caprolactone) diacrylate (PCLDA) at various weight compositions of mPEGA (ø(m)) from 2 to 30%. Improved surface wettability is achieved with corresponding decreases in friction, water contact angle, and capability of adsorbing proteins from cell culture media because of repulsive PEG chains tethered in the network. The responses of rat Schwann cell precursor line (SpL201), rat pheochromocytoma (PC12), and E14 mouse neural progenitor cells (NPCs) to the modified surfaces are evaluated. Nonmonotonic or parabolic dependence of cell attachment, spreading, proliferation, and differentiation on ø(m) is identified for these cell types with maximal values at ø(m) of 5-7%. In addition, NPCs demonstrate enhanced neuronal differentiated lineages on the mPEGA/PCLDA network at ø(m) of 5% with intermediate wettability and surface energy. This approach lays the foundation for fabricating heterogeneous nerve conduits with a compositional gradient along the wall thickness, which are able to promote nerve cell functions within the conduit while inhibiting cell attachment on the outer wall to prevent potential fibrous tissue formation following implantation.


Assuntos
Acrilatos/síntese química , Materiais Biocompatíveis/síntese química , Células-Tronco Neurais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Células de Schwann/efeitos dos fármacos , Acrilatos/farmacologia , Animais , Materiais Biocompatíveis/farmacologia , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Luz , Lubrificantes/química , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Células PC12 , Processos Fotoquímicos , Polimerização , Ratos , Células de Schwann/citologia , Células de Schwann/fisiologia , Espectroscopia de Infravermelho com Transformada de Fourier , Alicerces Teciduais , Molhabilidade
14.
Biomacromolecules ; 13(2): 342-9, 2012 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-22251248

RESUMO

We present a novel photopolymerizable poly(L-lysine) (PLL) and use it to modify polyethylene glycol diacrylate (PEGDA) hydrogels for creating a better, permissive nerve cell niche. Compared with their neutral counterparts, these PLL-grafted hydrogels greatly enhance pheochromocytoma (PC12) cell survival in encapsulation, proliferation, and neurite growth and also promote neural progenitor cell proliferation and differentiation capacity, represented by percentages of both differentiated neurons and astrocytes. The role of efficiently controlled substrate stiffness in regulating nerve cell behavior is also investigated and a polymerizable cationic small molecule, [2-(methacryloyloxy)ethyl]-trimethylammonium chloride (MTAC), is used to compare with this newly developed PLL. The results indicate that these PLL-grafted hydrogels are promising biomaterials for nerve repair and regeneration.


Assuntos
Astrócitos/efeitos dos fármacos , Materiais Biocompatíveis/síntese química , Neurônios/efeitos dos fármacos , Polietilenoglicóis/química , Polilisina/síntese química , Animais , Astrócitos/citologia , Astrócitos/fisiologia , Materiais Biocompatíveis/farmacologia , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Hidrogéis , Luz , Espectroscopia de Ressonância Magnética , Neurônios/citologia , Neurônios/fisiologia , Células PC12 , Processos Fotoquímicos , Polilisina/farmacologia , Polimerização , Ratos , Alicerces Teciduais
15.
Biomacromolecules ; 13(5): 1663-74, 2012 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-22533450

RESUMO

Recently, we have developed a photopolymerizable poly(L-lysine) (PLL) that can be covalently incorporated into poly(ethylene glycol) diacrylate (PEGDA) hydrogels to improve their bioactivity by providing positive charges. To explore the potential of these PLL-grafted PEGDA hydrogels as a cell delivery vehicle and luminal filler in nerve guidance conduits for peripheral and central nerve regeneration, we varied the number of pendent PLL chains in the hydrogels by photo-cross-linking PEGDA with weight compositions of PLL (φ(PLL)) of 0, 1, 2, 3, and 5%. We further investigated the effect of PLL grafting density on E14 mouse neural progenitor cell (NPC) behavior including cell viability, attachment, proliferation, differentiation, and gene expression. The amount of actually grafted PLL and charge densities were characterized, showing a proportional increase with the feed composition φ(PLL). NPC viability in 3D hydrogels was significantly improved in a PLL grafting density-dependent manner at days 7 and 14 postencapsulation. Similarly, NPC attachment and proliferation were promoted on the PLL-grafted hydrogels with increasing φ(PLL) up to 2%. More intriguingly, NPC lineage commitment was dramatically altered by the amount of grafted PLL chains in the hydrogels. NPC differentiation demonstrated a parabolic or nonmonotonic dependence on φ(PLL), resulting in cells mostly differentiated toward mature neurons with extensive neurite formation and astrocytes rather than oligodendrocytes on the PLL-grafted hydrogels with φ(PLL) of 2%, whereas the neutral hydrogels and PLL-grafted hydrogels with higher φ(PLL) of 5% support NPC differentiation less. Gene expression of lineage markers further illustrated this trend, indicating that PLL-grafted hydrogels with an optimal φ(PLL) of 2% could be a promising cell carrier that promoted NPC functions for treatment of nerve injuries.


Assuntos
Hidrogéis/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Polilisina/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Hidrogéis/química , Camundongos , Modelos Moleculares , Estrutura Molecular , Células-Tronco Neurais/citologia , Polietilenoglicóis/química , Polilisina/química , Relação Estrutura-Atividade
16.
Hum Mol Genet ; 18(3): 497-516, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18996916

RESUMO

Periventricular heterotopia (PH) is a disorder characterized by neuronal nodules, ectopically positioned along the lateral ventricles of the cerebral cortex. Mutations in either of two human genes, Filamin A (FLNA) or ADP-ribosylation factor guanine exchange factor 2 (ARFGEF2), cause PH (Fox et al. in 'Mutations in filamin 1 prevent migration of cerebral cortical neurons in human periventricular heterotopia'. Neuron, 21, 1315-1325, 1998; Sheen et al. in 'Mutations in ARFGEF2 implicate vesicle trafficking in neural progenitor proliferation and migration in the human cerebral cortex'. Nat. Genet., 36, 69-76, 2004). Recent studies have shown that mutations in mitogen-activated protein kinase kinase kinase-4 (Mekk4), an indirect interactor with FlnA, also lead to periventricular nodule formation in mice (Sarkisian et al. in 'MEKK4 signaling regulates filamin expression and neuronal migration'. Neuron, 52, 789-801, 2006). Here we show that neurons in post-mortem human PH brains migrated appropriately into the cortex, that periventricular nodules were primarily composed of later-born neurons, and that the neuroependyma was disrupted in all PH cases. As studied in the mouse, loss of FlnA or Big2 function in neural precursors impaired neuronal migration from the germinal zone, disrupted cell adhesion and compromised neuroepithelial integrity. Finally, the hydrocephalus with hop gait (hyh) mouse, which harbors a mutation in Napa [encoding N-ethylmaleimide-sensitive factor attachment protein alpha (alpha-SNAP)], also develops a progressive denudation of the neuroepithelium, leading to periventricular nodule formation. Previous studies have shown that Arfgef2 and Napa direct vesicle trafficking and fusion, whereas FlnA associates dynamically with the Golgi membranes during budding and trafficking of transport vesicles. Our current findings suggest that PH formation arises from a final common pathway involving disruption of vesicle trafficking, leading to impaired cell adhesion and loss of neuroependymal integrity.


Assuntos
Ventrículos Cerebrais/citologia , Heterotopia Nodular Periventricular/patologia , Células-Tronco/citologia , Adulto , Idoso de 80 Anos ou mais , Animais , Adesão Celular , Movimento Celular , Ventrículos Cerebrais/fisiopatologia , Proteínas Contráteis/genética , Proteínas Contráteis/metabolismo , Feminino , Filaminas , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Recém-Nascido , Masculino , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Neurônios/fisiologia , Heterotopia Nodular Periventricular/fisiopatologia , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/genética , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/metabolismo
17.
Prog Neurobiol ; 201: 102028, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33636226

RESUMO

Previously, we reported global hypermethylation in DS might be attributed to the overexpression of HSA21 gene DNMT3L, which can enhance DNMT3A and DNMT3B activities in DNA methylation. To test this hypothesis, we compared the DNA methylation and RNA expression profiles of early-differentiated human neuroprogenitors with and without DNMT3L overexpression. We found DNMT3L overexpression only moderately increased the DNA methylation of limited genes, yet significantly altered global RNA expression of genes involved in neural differentiation. We further found that DNMT3L bound STAT1 or STAT3, and increased its phosphorylation and nuclear translocation, which in turn activated the expression of transcription factors including HES3, ASCL1, NEUROD2 and NEUROG2 and CDK inhibitor CDKN1A, which promoted cell cycle exit and neural differentiation. This phenomenon was also confirmed in Dnmt3l conditional knockin mice, which could be rescued by STAT1 and STAT3 phosphorylation inhibitors (Fludarabine and SH-4-54) but not DNA methylation inhibitor (Decitabine). These results suggest that DNMT3L play an important role during neurodevelopment independent of DNA methylation, which may contribute to the abnormal phenotypes observed in Down syndrome cortex.


Assuntos
Metilação de DNA , RNA , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Diferenciação Celular , DNA (Citosina-5-)-Metiltransferases/genética , Camundongos , Proteínas do Tecido Nervoso , Fosforilação , Fator de Transcrição STAT1
18.
Hum Mol Genet ; 17(3): 440-57, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17984171

RESUMO

Down syndrome (DS) is caused by trisomy of chromosome 21 and is characterized by mental retardation, seizures and premature Alzheimer's disease. To examine neuropathological mechanisms giving rise to this disorder, we generated multiple human DS neural progenitor cell (NPC) lines from the 19-21 week frontal cortex and characterized their genomic and functional properties. Microarray profiling of DS progenitors indicated that increased levels of gene expression were not limited to chromosome 21, suggesting that increased expression of genes on chromosome 21 altered transcriptional regulation of a subset of genes throughout the entire genome. Moreover, many transcriptionally dysregulated genes were involved in cell death and oxidative stress. Network analyses suggested that upregulated expression of chromosome 21 genes such as S100B and amyloid precursor protein activated the stress response kinase pathways, and furthermore, could be linked to upregulation of the water channel aquaporin 4 (AQP4). We further demonstrate in DS NPCs that S100B is constitutively overexpressed, that overexpression leads to increased reactive oxygen species (ROS) formation and activation of stress response kinases, and that activation of this pathway results in compensatory AQP4 expression. In addition, AQP4 expression could be induced by direct exposure to ROS, and siRNA inhibition of AQP4 resulted in elevated levels of ROS following S100B exposure. Finally, elevated levels of S100B-induced ROS and loss of AQP4 expression led to increased programmed cell death. These findings suggest that dysregulation of chromosome 21 genes in DS neural progenitors leads to increased ROS and thereby alters transcriptional regulation of cytoprotective, non-chromosome 21 genes in response to ongoing cellular insults.


Assuntos
Aquaporina 4/genética , Aquaporina 4/metabolismo , Síndrome de Down/genética , Síndrome de Down/metabolismo , Células-Tronco Embrionárias/metabolismo , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Neurônios/metabolismo , Proteínas S100/genética , Proteínas S100/metabolismo , Apoptose , Sequência de Bases , Estudos de Casos e Controles , Linhagem Celular , Cromossomos Humanos Par 21/genética , Primers do DNA/genética , Síndrome de Down/embriologia , Síndrome de Down/patologia , Células-Tronco Embrionárias/patologia , Perfilação da Expressão Gênica , Humanos , Neurônios/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/patologia , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo , Subunidade beta da Proteína Ligante de Cálcio S100 , Transfecção
19.
Neurologist ; 14(3): 181-91, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18469675

RESUMO

BACKGROUND: Malformations of cortical development (MCD) are increasingly recognized as an important cause of epilepsy and developmental delay. MCD encompass a wide spectrum of disorders with various underlying genetic etiologies and clinical manifestations. High resolution imaging has dramatically improved our recognition of MCD. REVIEW SUMMARY: This review will provide a brief overview of the stages of normal cortical development, including neuronal proliferation, neuroblast migration, and neuronal organization. Disruptions at various stages lead to characteristic MCD. Disorders of neurogenesis give rise to microcephaly (small brain) or macrocephaly (large brain). Disorders of early neuroblast migration give rise to periventricular heterotopia (neurons located along the ventricles), whereas abnormalities later in migration lead to lissencephaly (smooth brain) or subcortical band heterotopia (smooth brain with a band of heterotopic neurons under the cortex). Abnormal neuronal migration arrest give rise to over migration of neurons in cobblestone lissencephaly. Lastly, disorders of neuronal organization cause polymicrogyria (abnormally small gyri and sulci). This review will also discuss the known genetic mutations and potential mechanisms that contribute to these syndromes. CONCLUSION: Identification of various gene mutations has not only given us greater insight into some of the pathophysiologic basis of MCD, but also an understanding of the processes involved in normal cortical development.


Assuntos
Córtex Cerebral/anormalidades , Córtex Cerebral/patologia , Predisposição Genética para Doença/genética , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/patologia , Diferenciação Celular/genética , Movimento Celular/genética , Córtex Cerebral/fisiopatologia , Humanos , Mutação , Malformações do Sistema Nervoso/fisiopatologia
20.
J Neurosci Methods ; 294: 1-6, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29061345

RESUMO

BACKGROUND: Morphometric analyses of biological features have become increasingly common in recent years with such analyses being subject to a large degree of observer bias, variability, and time consumption. While commercial software packages exist to perform these analyses, they are expensive, require extensive user training, and are usually dependent on the observer tracing the morphology. NEW METHOD: To address these issues, we have developed a broadly applicable, no-cost ImageJ plugin we call 'BranchAnalysis2D/3D', to perform morphometric analyses of structures with branching morphologies, such as neuronal dendritic spines, vascular morphology, and primary cilia. RESULTS: Our BranchAnalysis2D/3D algorithm allows for rapid quantification of the length and thickness of branching morphologies, independent of user tracing, in both 2D and 3D data sets. COMPARISON WITH EXISTING METHODS: We validated the performance of BranchAnalysis2D/3D against pre-existing software packages using trained human observers and images from brain and retina. We found that the BranchAnalysis2D/3D algorithm outputs results similar to available software (i.e., Metamorph, AngioTool, Neurolucida), while allowing faster analysis times and unbiased quantification. CONCLUSIONS: BranchAnalysis2D/3D allows inexperienced observers to output results like a trained observer but more efficiently, thereby increasing the consistency, speed, and reliability of morphometric analyses.


Assuntos
Encéfalo/citologia , Imageamento Tridimensional/métodos , Microscopia Confocal/métodos , Neurônios/citologia , Software , Algoritmos , Animais , Camundongos , Variações Dependentes do Observador , Reprodutibilidade dos Testes , Retina/anatomia & histologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA