Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Hum Genet ; 110(5): 826-845, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37098352

RESUMO

Alterations in cortical neurogenesis are implicated in neurodevelopmental disorders including autism spectrum disorders (ASDs). The contribution of genetic backgrounds, in addition to ASD risk genes, on cortical neurogenesis remains understudied. Here, using isogenic induced pluripotent stem cell (iPSC)-derived neural progenitor cells (NPCs) and cortical organoid models, we report that a heterozygous PTEN c.403A>C (p.Ile135Leu) variant found in an ASD-affected individual with macrocephaly dysregulates cortical neurogenesis in an ASD-genetic-background-dependent fashion. Transcriptome analysis at both bulk and single-cell level revealed that the PTEN c.403A>C variant and ASD genetic background affected genes involved in neurogenesis, neural development, and synapse signaling. We also found that this PTEN p.Ile135Leu variant led to overproduction of NPC subtypes as well as neuronal subtypes including both deep and upper layer neurons in its ASD background, but not when introduced into a control genetic background. These findings provide experimental evidence that both the PTEN p.Ile135Leu variant and ASD genetic background contribute to cellular features consistent with ASD associated with macrocephaly.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Células-Tronco Pluripotentes Induzidas , Megalencefalia , Células-Tronco Neurais , Humanos , Transtorno do Espectro Autista/genética , Transtorno Autístico/genética , Megalencefalia/genética , Neurogênese/genética , Neurônios , PTEN Fosfo-Hidrolase/genética
2.
Hum Mol Genet ; 32(18): 2832-2841, 2023 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-37387247

RESUMO

Neurons within the cerebellum form temporal-spatial connections through the cerebellum, and the entire brain. Organoid models provide an opportunity to model the early differentiation of the developing human cerebellum, which is difficult to study in vivo, and affords the opportunity to study neurodegenerative and neurodevelopmental diseases of the cerebellum. Previous cerebellar organoid models focused on early neuron generation and single cell activity. Here, we modify previous protocols to generate more mature cerebellar organoids that allow for the establishment of several classes of mature neurons during cerebellar differentiation and development, including the establishment of neural networks during whole-organoid maturation. This will provide a means to study the generation of several more mature cerebellar cell types, including Purkinje cells, granule cells and interneurons expression as well as neuronal communication for biomedical, clinical and pharmaceutical applications.


Assuntos
Cerebelo , Neurônios , Humanos , Neurônios/metabolismo , Células de Purkinje/metabolismo , Neurogênese , Organoides
3.
Hum Mol Genet ; 32(10): 1589-1606, 2023 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-36519762

RESUMO

Autism spectrum disorders (ASD) display both phenotypic and genetic heterogeneity, impeding the understanding of ASD and development of effective means of diagnosis and potential treatments. Genes affected by genomic variations for ASD converge in dozens of gene ontologies (GOs), but the relationship between the variations at the GO level have not been well elucidated. In the current study, multiple types of genomic variations were mapped to GOs and correlations among GOs were measured in ASD and control samples. Several ASD-unique GO correlations were found, suggesting the importance of co-occurrence of genomic variations in genes from different functional categories in ASD etiology. Combined with experimental data, several variations related to WNT signaling, neuron development, synapse morphology/function and organ morphogenesis were found to be important for ASD with macrocephaly, and novel co-occurrence patterns of them in ASD patients were found. Furthermore, we applied this gene ontology correlation analysis method to find genomic variations that contribute to ASD etiology in combination with changes in gene expression and transcription factor binding, providing novel insights into ASD with macrocephaly and a new methodology for the analysis of genomic variation.


Assuntos
Transtorno do Espectro Autista , Megalencefalia , Humanos , Transtorno do Espectro Autista/genética , Genômica , Megalencefalia/genética
4.
Mol Cell ; 68(5): 885-900.e6, 2017 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-29220654

RESUMO

The integrated stress response (ISR) is a homeostatic mechanism induced by endoplasmic reticulum (ER) stress. In acute/transient ER stress, decreased global protein synthesis and increased uORF mRNA translation are followed by normalization of protein synthesis. Here, we report a dramatically different response during chronic ER stress. This chronic ISR program is characterized by persistently elevated uORF mRNA translation and concurrent gene expression reprogramming, which permits simultaneous stress sensing and proteostasis. The program includes PERK-dependent switching to an eIF3-dependent translation initiation mechanism, resulting in partial, but not complete, translational recovery, which, together with transcriptional reprogramming, selectively bolsters expression of proteins with ER functions. Coordination of transcriptional and translational reprogramming prevents ER dysfunction and inhibits "foamy cell" development, thus establishing a molecular basis for understanding human diseases associated with ER dysfunction.


Assuntos
Estresse do Retículo Endoplasmático , Fator de Iniciação 3 em Eucariotos/metabolismo , Fibroblastos/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/biossíntese , Transcrição Gênica , eIF-2 Quinase/metabolismo , Animais , Reprogramação Celular , Fator de Iniciação 3 em Eucariotos/genética , Fibroblastos/patologia , Células HEK293 , Humanos , Camundongos , Fases de Leitura Aberta , Fenótipo , Proteostase , Interferência de RNA , RNA Mensageiro/genética , Transdução de Sinais , Fatores de Tempo , Transfecção , eIF-2 Quinase/genética
5.
Am J Hum Genet ; 108(3): 375-382, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33667389

RESUMO

This article is based on the address given by the author at the 2020 virtual meeting of the American Society of Human Genetics (ASHG) on October 26, 2020. The video of the original address can be found at the ASHG website.


Assuntos
Genética Médica/tendências , Genômica/tendências , Genética Humana/tendências , Humanos , Estados Unidos
6.
Cell ; 136(3): 393-4, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19203572

RESUMO

Neuronal cell migration and maturation are crucial for the development of the mammalian cerebral cortex. In this issue, Creppe et al. (2009) now show that the multisubunit histone acetyltransferase Elongator may regulate migration and branch formation of cortical neurons in the developing mouse brain by acetylating alpha-tubulin.


Assuntos
Histona Acetiltransferases/metabolismo , Neurogênese , Neurônios/metabolismo , Tubulina (Proteína)/metabolismo , Acetilação , Animais , Movimento Celular , Camundongos
7.
Cell ; 132(3): 474-86, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18267077

RESUMO

Mitotic spindle orientation and plane of cleavage in mammals is a determinant of whether division yields progenitor expansion and/or birth of new neurons during radial glial progenitor cell (RGPC) neurogenesis, but its role earlier in neuroepithelial stem cells is poorly understood. Here we report that Lis1 is essential for precise control of mitotic spindle orientation in both neuroepithelial stem cells and radial glial progenitor cells. Controlled gene deletion of Lis1 in vivo in neuroepithelial stem cells, where cleavage is uniformly vertical and symmetrical, provokes rapid apoptosis of those cells, while radial glial progenitors are less affected. Impaired cortical microtubule capture via loss of cortical dynein causes astral and cortical microtubules to be greatly reduced in Lis1-deficient cells. Increased expression of the LIS/dynein binding partner NDEL1 restores cortical microtubule and dynein localization in Lis1-deficient cells. Thus, control of symmetric division, essential for neuroepithelial stem cell proliferation, is mediated through spindle orientation determined via LIS1/NDEL1/dynein-mediated cortical microtubule capture.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Embrião de Mamíferos/citologia , Proteínas Associadas aos Microtúbulos/metabolismo , Células Neuroepiteliais/citologia , Fuso Acromático/metabolismo , Células-Tronco/citologia , 1-Alquil-2-acetilglicerofosfocolina Esterase/genética , Animais , Encéfalo/citologia , Encéfalo/embriologia , Ciclo Celular , Movimento Celular , Proliferação de Células , Dineínas/metabolismo , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/metabolismo , Modelos Biológicos , Neurônios/citologia
8.
Dev Biol ; 464(2): 161-175, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32579954

RESUMO

The Dishevelled proteins transduce both canonical Wnt/ß-catenin and non-canonical Wnt/planar cell polarity (PCP) signaling pathways to regulate many key developmental processes during embryogenesis. Here, we disrupt both canonical and non-canonical Wnt pathways by targeting the entire Dishevelled family of genes (Dvl1, Dvl2, and Dvl3) to investigate their functional roles in the early embryo. We identified several defects in anterior-posterior axis specification and mesoderm patterning in Dvl1+/-; Dvl2-/-; Dvl3-/- embryos. Homozygous deletions in all three Dvl genes (Dvl TKO) resulted in defects in distal visceral endoderm migration and a complete failure to induce mesoderm formation. To identify potential mechanisms that lead to the defects in the developmental processes preceding gastrulation, we generated Dvl TKO mouse embryonic stem cells (mESCs) and compared the transcriptional profile of these cells with wild-type (WT) mESCs during germ lineage differentiation into 3D embryoid bodies (EBs). While the Dvl TKO mESCs displayed similar morphology, self-renewal properties, and minor transcriptional variation from WT mESCs, we identified major transcriptional dysregulation in the Dvl TKO EBs during differentiation in a number of genes involved in anterior-posterior pattern specification, gastrulation induction, mesenchyme morphogenesis, and mesoderm-derived tissue development. The absence of the Dvls leads to specific down-regulation of BMP signaling genes. Furthermore, exogenous activation of canonical Wnt, BMP, and Nodal signaling all fail to rescue the mesodermal defects in the Dvl TKO EBs. Moreover, endoderm differentiation was promoted in the absence of mesoderm in the Dvl TKO EBs, while the suppression of ectoderm differentiation was delayed. Overall, we demonstrate that the Dvls are dispensable for maintaining self-renewal in mESCs but are critical during differentiation to regulate key developmental signaling pathways to promote proper axis specification and mesoderm formation.


Assuntos
Diferenciação Celular , Proteínas Desgrenhadas/deficiência , Embrião de Mamíferos , Deleção de Genes , Mesoderma/embriologia , Transdução de Sinais , Animais , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Desgrenhadas/metabolismo , Camundongos , Camundongos Knockout
9.
Proc Natl Acad Sci U S A ; 115(20): E4661-E4669, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29712858

RESUMO

Nonischemic cardiomyopathy (NICM) resulting from long-standing hypertension, valvular disease, and genetic mutations is a major cause of heart failure worldwide. Recent observations suggest that myeloid cells can impact cardiac function, but the role of tissue-intrinsic vs. tissue-extrinsic myeloid cells in NICM remains poorly understood. Here, we show that cardiac resident macrophage proliferation occurs within the first week following pressure overload hypertrophy (POH; a model of heart failure) and is requisite for the heart's adaptive response. Mechanistically, we identify Kruppel-like factor 4 (KLF4) as a key transcription factor that regulates cardiac resident macrophage proliferation and angiogenic activities. Finally, we show that blood-borne macrophages recruited in late-phase POH are detrimental, and that blockade of their infiltration improves myocardial angiogenesis and preserves cardiac function. These observations demonstrate previously unappreciated temporal and spatial roles for resident and nonresident macrophages in the development of heart failure.


Assuntos
Cardiomegalia/patologia , Cardiomiopatias/patologia , Insuficiência Cardíaca/patologia , Fatores de Transcrição Kruppel-Like/metabolismo , Macrófagos/patologia , Miocárdio/patologia , Animais , Cardiomegalia/imunologia , Cardiomegalia/metabolismo , Cardiomiopatias/imunologia , Cardiomiopatias/metabolismo , Células Cultivadas , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/metabolismo , Fator 4 Semelhante a Kruppel , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Miocárdio/imunologia , Miocárdio/metabolismo , Pressão
10.
Nature ; 507(7490): 99-103, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24413397

RESUMO

Ring chromosomes are structural aberrations commonly associated with birth defects, mental disabilities and growth retardation. Rings form after fusion of the long and short arms of a chromosome, and are sometimes associated with large terminal deletions. Owing to the severity of these large aberrations that can affect multiple contiguous genes, no possible therapeutic strategies for ring chromosome disorders have been proposed. During cell division, ring chromosomes can exhibit unstable behaviour leading to continuous production of aneuploid progeny with low viability and high cellular death rate. The overall consequences of this chromosomal instability have been largely unexplored in experimental model systems. Here we generated human induced pluripotent stem cells (iPSCs) from patient fibroblasts containing ring chromosomes with large deletions and found that reprogrammed cells lost the abnormal chromosome and duplicated the wild-type homologue through the compensatory uniparental disomy (UPD) mechanism. The karyotypically normal iPSCs with isodisomy for the corrected chromosome outgrew co-existing aneuploid populations, enabling rapid and efficient isolation of patient-derived iPSCs devoid of the original chromosomal aberration. Our results suggest a fundamentally different function for cellular reprogramming as a means of 'chromosome therapy' to reverse combined loss-of-function across many genes in cells with large-scale aberrations involving ring structures. In addition, our work provides an experimentally tractable human cellular system for studying mechanisms of chromosomal number control, which is of critical relevance to human development and disease.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Cromossomos em Anel , Aneuploidia , Animais , Reprogramação Celular/genética , Instabilidade Cromossômica/genética , Deleção Cromossômica , Transtornos Cromossômicos/genética , Transtornos Cromossômicos/patologia , Cromossomos Humanos Par 13/genética , Cromossomos Humanos Par 17/genética , Células Clonais/citologia , Células Clonais/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Cariótipo , Cariotipagem , Masculino , Camundongos , Modelos Genéticos , Dissomia Uniparental/genética
11.
Chromosoma ; 126(4): 457-463, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27882407

RESUMO

Approximately 1 in 500 newborns are born with chromosomal abnormalities that include trisomies, translocations, large deletions, and duplications. There is currently no therapeutic approach for correcting such chromosomal aberrations in vivo or in vitro. When we attempted to produce induced pluripotent stem cell (iPSC) models from patient-derived fibroblasts that contained ring chromosomes, we found that the ring chromosomes were eliminated and replaced by duplicated normal copies of chromosomes through a mechanism of uniparental isodisomy (Bershteyn et al. 2014, Nature 507:99). The discovery of this previously unforeseen system for aberrant chromosome correction during reprogramming enables us for the first time to model and understand this process of cell-autonomous correction of ring chromosomes during human patient somatic cell reprograming to iPSCs. This knowledge could lead to a potential therapeutic strategy to correct common large-scale chromosomal aberrations, termed "chromosome therapy".


Assuntos
Reprogramação Celular/genética , Terapia Genética/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Cromossomos em Anel , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Dissomia Uniparental/genética
13.
Hum Mol Genet ; 25(20): 4376-4388, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28172828

RESUMO

A defined set of genetic instructions encodes functionality in complex organisms. Delineating these unique genetic signatures is essential to understanding the formation and functionality of specialized tissues. Vision, one of the five central senses of perception, is initiated by the retina and has evolved over time to produce rod and cone photoreceptors that vary in a species-specific manner, and in some cases by geographical region resulting in higher order visual acuity in humans. RNA-sequencing and use of existing and de novo transcriptome assemblies allowed ocular transcriptome mapping from a diverse set of rodent and primate species. Global genomic refinements along with systems-based comparative and co-expression analyses of these transcriptome maps identified gene modules that correlated with specific features of rod versus cone retinal cellular composition. Organization of the ocular transcriptome demonstrated herein defines the molecular basis of photoreceptor architecture and functionality, providing a new paradigm for neurogenetic analyses of the mammalian retina in health and disease.


Assuntos
Mamíferos/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Transcriptoma , Animais , Perfilação da Expressão Gênica , Humanos , Macaca fascicularis/genética , Macaca fascicularis/metabolismo , Mamíferos/genética , Camundongos , Ratos , Análise de Sequência de RNA
14.
Mol Psychiatry ; 22(6): 820-835, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27378147

RESUMO

Autism spectrum disorders (ASD) are common, complex and heterogeneous neurodevelopmental disorders. Cellular and molecular mechanisms responsible for ASD pathogenesis have been proposed based on genetic studies, brain pathology and imaging, but a major impediment to testing ASD hypotheses is the lack of human cell models. Here, we reprogrammed fibroblasts to generate induced pluripotent stem cells, neural progenitor cells (NPCs) and neurons from ASD individuals with early brain overgrowth and non-ASD controls with normal brain size. ASD-derived NPCs display increased cell proliferation because of dysregulation of a ß-catenin/BRN2 transcriptional cascade. ASD-derived neurons display abnormal neurogenesis and reduced synaptogenesis leading to functional defects in neuronal networks. Interestingly, defects in neuronal networks could be rescued by insulin growth factor 1 (IGF-1), a drug that is currently in clinical trials for ASD. This work demonstrates that selection of ASD subjects based on endophenotypes unraveled biologically relevant pathway disruption and revealed a potential cellular mechanism for the therapeutic effect of IGF-1.


Assuntos
Transtorno Autístico/metabolismo , Transtorno Autístico/patologia , Técnicas de Cultura de Tecidos/métodos , Adolescente , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/fisiopatologia , Encéfalo/metabolismo , Proliferação de Células/genética , Células Cultivadas , Criança , Pré-Escolar , Feminino , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/uso terapêutico , Masculino , Células-Tronco Neurais/metabolismo , Neurogênese , Neurônios/metabolismo , Neurônios/fisiologia , beta Catenina/metabolismo
15.
Cereb Cortex ; 27(8): 3918-3929, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27371763

RESUMO

Neuronal nitric oxide synthase is involved in diverse signaling cascades that regulate neuronal development and functions via S-Nitrosylation-mediated mechanism or the soluble guanylate cyclase (sGC)/cyclic guanosine monophosphate (cGMP) pathway activated by nitric oxide. Although it has been studied extensively in vitro and in invertebrate animals, effects on mammalian brain development and underlying mechanisms remain poorly understood. Here we report that genetic deletion of "Nos1" disrupts dendritic development, whereas pharmacological inhibition of the sGC/cGMP pathway does not alter dendritic growth during cerebral cortex development. Instead, nuclear distribution element-like (NDEL1), a protein that regulates dendritic development, is specifically S-nitrosylated at cysteine 203, thereby accelerating dendritic arborization. This post-translational modification is enhanced by N-methyl-D-aspartate receptor-mediated neuronal activity, the main regulator of dendritic formation. Notably, we found that disruption of S-Nitrosylation of NDEL1 mediates impaired dendritic maturation caused by developmental alcohol exposure, a model of developmental brain abnormalities resulting from maternal alcohol use. These results highlight S-Nitrosylation as a key activity-dependent mechanism underlying neonatal brain maturation and suggest that reduction of S-Nitrosylation of NDEL1 acts as a pathological factor mediating neurodevelopmental abnormalities caused by maternal alcohol exposure.


Assuntos
Proteínas de Transporte/metabolismo , Dendritos/metabolismo , Transtornos do Espectro Alcoólico Fetal/metabolismo , Córtex Pré-Frontal/metabolismo , Células Piramidais/metabolismo , Transmissão Sináptica/fisiologia , Animais , Proteínas de Transporte/genética , Dendritos/efeitos dos fármacos , Dendritos/patologia , Modelos Animais de Doenças , Transtornos do Espectro Alcoólico Fetal/patologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico Sintase Tipo I/genética , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/crescimento & desenvolvimento , Córtex Pré-Frontal/patologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/patologia
16.
N Engl J Med ; 370(13): 1209-1219, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24670167

RESUMO

BACKGROUND: Autism involves early brain overgrowth and dysfunction, which is most strongly evident in the prefrontal cortex. As assessed on pathological analysis, an excess of neurons in the prefrontal cortex among children with autism signals a disturbance in prenatal development and may be concomitant with abnormal cell type and laminar development. METHODS: To systematically examine neocortical architecture during the early years after the onset of autism, we used RNA in situ hybridization with a panel of layer- and cell-type-specific molecular markers to phenotype cortical microstructure. We assayed markers for neurons and glia, along with genes that have been implicated in the risk of autism, in prefrontal, temporal, and occipital neocortical tissue from postmortem samples obtained from children with autism and unaffected children between the ages of 2 and 15 years. RESULTS: We observed focal patches of abnormal laminar cytoarchitecture and cortical disorganization of neurons, but not glia, in prefrontal and temporal cortical tissue from 10 of 11 children with autism and from 1 of 11 unaffected children. We observed heterogeneity between cases with respect to cell types that were most abnormal in the patches and the layers that were most affected by the pathological features. No cortical layer was uniformly spared, with the clearest signs of abnormal expression in layers 4 and 5. Three-dimensional reconstruction of layer markers confirmed the focal geometry and size of patches. CONCLUSIONS: In this small, explorative study, we found focal disruption of cortical laminar architecture in the cortexes of a majority of young children with autism. Our data support a probable dysregulation of layer formation and layer-specific neuronal differentiation at prenatal developmental stages. (Funded by the Simons Foundation and others.).


Assuntos
Transtorno Autístico/patologia , Neocórtex/ultraestrutura , Adolescente , Transtorno Autístico/genética , Biomarcadores/análise , Biomarcadores/metabolismo , Calbindina 1/genética , Contagem de Células , Criança , Pré-Escolar , Crioultramicrotomia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 1/genética , Expressão Gênica , Humanos , Imageamento Tridimensional , Hibridização In Situ , Neocórtex/crescimento & desenvolvimento , Proteínas do Tecido Nervoso/genética , Proteínas de Neurofilamentos/genética , Neurogênese , Neurônios/patologia , Membro 2 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , RNA/genética
17.
Dev Biol ; 398(2): 177-92, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25448697

RESUMO

Planar cell polarity (PCP) signaling is an evolutionarily conserved mechanism that coordinates polarized cell behavior to regulate tissue morphogenesis during vertebrate gastrulation, neurulation and organogenesis. In Xenopus and zebrafish, PCP signaling is activated by non-canonical Wnts such as Wnt11, and detailed understanding of Wnt11 expression has provided important clues on when, where and how PCP may be activated to regulate tissue morphogenesis. To explore the role of Wnt11 in mammalian development, we established a Wnt11 expression and lineage map with high spatial and temporal resolution by creating and analyzing a tamoxifen-inducible Wnt11-CreER BAC (bacterial artificial chromosome) transgenic mouse line. Our short- and long-term lineage tracing experiments indicated that Wnt11-CreER could faithfully recapitulate endogenous Wnt11 expression, and revealed for the first time that cells transiently expressing Wnt11 at early gastrulation were fated to become specifically the progenitors of the entire endoderm. During mid-gastrulation, Wnt11-CreER expressing cells also contribute extensively to the endothelium in both embryonic and extraembryonic compartments, and the endocardium in all chambers of the developing heart. In contrast, Wnt11-CreER expression in the myocardium starts from late-gastrulation, and occurs in three transient, sequential waves: first in the precursors of the left ventricular (LV) myocardium from E7.0 to 8.0; subsequently in the right ventricular (RV) myocardium from E8.0 to 9.0; and finally in the superior wall of the outflow tract (OFT) myocardium from E8.5 to 10.5. These results provide formal genetic proof that the majority of the endocardium and myocardium diverge by mid-gastrulation in the mouse, and suggest a tight spatial and temporal control of Wnt11 expression in the myocardial lineage to coordinate with myocardial differentiation in the first and second heart field progenitors to form the LV, RV and OFT. The insights gained from this study will also guide future investigations to decipher the role of non-canonical Wnt/PCP signaling in endoderm development, vasculogenesis and heart formation.


Assuntos
Linhagem da Célula , Desenvolvimento Embrionário , Proteínas Wnt/metabolismo , Animais , Cromossomos Artificiais Bacterianos/metabolismo , Embrião de Mamíferos/irrigação sanguínea , Embrião de Mamíferos/metabolismo , Endoderma/embriologia , Endoderma/metabolismo , Gastrulação , Integrases/metabolismo , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos Transgênicos , Miocárdio/metabolismo , Linha Primitiva/embriologia , Linha Primitiva/metabolismo , Transgenes , Proteínas Wnt/genética
18.
Am J Med Genet C Semin Med Genet ; 172(4): 422-430, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27813255

RESUMO

Large chromosomal aberrations occur commonly during development, resulting in complex and multisystem diseases. In spite of this high frequency, there are currently no means for correcting these disorders due to their complexity and involvement of multiple genes. Recently, several new approaches have been devised that target whole chromosomes in vitro, which are collectively referred to as "Chromosome Therapies." These include silencing and selection for loss of the extra chromosome in trisomies, promotion of euploidy in an aneuploid culture, and forced loss and replacement of a chromosome. Here, we provide a review of Chromosome Therapy, and discuss potential directions for these methods clinically, as well as research applications and cellular models that can be made using these technologies. © 2016 Wiley Periodicals, Inc.


Assuntos
Aberrações Cromossômicas , Transtornos Cromossômicos/terapia , Terapia Genética/métodos , Cromossomos/genética , Terapia Genética/tendências , Humanos
19.
Hum Mol Genet ; 23(2): 449-66, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24030547

RESUMO

Heterozygous LIS1 mutations are responsible for the human neuronal migration disorder lissencephaly. Mitotic functions of LIS1 have been suggested from many organisms throughout evolution. However, the cellular functions of LIS1 at distinct intracellular compartments such as the centrosome and the cell cortex have not been well defined especially during mitotic cell division. Here, we used detailed cellular approaches and time-lapse live cell imaging of mitosis from Lis1 mutant mouse embryonic fibroblasts to reveal critical roles of LIS1 in mitotic spindle regulation. We found that LIS1 is required for the tight control of chromosome congression and segregation to dictate kinetochore-microtubule (MT) interactions and anaphase progression. In addition, LIS1 is essential for the establishment of mitotic spindle pole integrity by maintaining normal centrosome number. Moreover, LIS1 plays crucial roles in mitotic spindle orientation by increasing the density of astral MT plus-end movements toward the cell cortex, which enhances cortical targeting of LIS1-dynein complex. Overexpression of NDEL1-dynein and MT stabilization rescues spindle orientation defects in Lis1 mutants, demonstrating that mouse LIS1 acts via the LIS1-NDEL1-dynein complex to regulate astral MT plus-ends dynamics and establish proper contacts of MTs with the cell cortex to ensure precise cell division.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Proteínas de Transporte/metabolismo , Dineínas/metabolismo , Lisencefalia/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Mitose , Fuso Acromático/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterase/genética , Animais , Células Cultivadas , Centrossomo , Córtex Cerebral , Segregação de Cromossomos , Células HEK293 , Humanos , Lisencefalia/genética , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Mutação , Neurônios/metabolismo , Estabilidade Proteica , Fuso Acromático/genética
20.
J Neurosci ; 34(36): 12168-81, 2014 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-25186760

RESUMO

During brain development, neural progenitor cells proliferate and differentiate into neural precursors. These neural precursors migrate along the radial glial processes and localize at their final destination in the cortex. Numerous reports have revealed that 14-3-3 proteins are involved in many neuronal activities, although their functions in neurogenesis remain unclear. Here, using 14-3-3ε/ζ double knock-out mice, we found that 14-3-3 proteins are important for proliferation and differentiation of neural progenitor cells in the cortex, resulting in neuronal migration defects and seizures. 14-3-3 deficiency resulted in the increase of δ-catenin and the decrease of ß-catenin and αN-catenin. 14-3-3 proteins regulated neuronal differentiation into neurons via direct interactions with phosphorylated δ-catenin to promote F-actin formation through a catenin/Rho GTPase/Limk1/cofilin signaling pathway. Conversely, neuronal migration defects seen in the double knock-out mice were restored by phosphomimic Ndel1 mutants, but not δ-catenin. Our findings provide new evidence that 14-3-3 proteins play important roles in neurogenesis and neuronal migration via the regulation of distinct signaling cascades.


Assuntos
Proteínas 14-3-3/metabolismo , Córtex Cerebral/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Proteínas 14-3-3/genética , Actinas/metabolismo , Animais , Cateninas/metabolismo , Movimento Celular , Proliferação de Células , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Neurônios/fisiologia , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa