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










Base de dados
Intervalo de ano de publicação
1.
Hum Mol Genet ; 32(15): 2511-2522, 2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37216650

RESUMO

FOXG1 is a critical transcription factor in human brain where loss-of-function mutations cause a severe neurodevelopmental disorder, while increased FOXG1 expression is frequently observed in glioblastoma. FOXG1 is an inhibitor of cell patterning and an activator of cell proliferation in chordate model organisms but different mechanisms have been proposed as to how this occurs. To identify genomic targets of FOXG1 in human neural progenitor cells (NPCs), we engineered a cleavable reporter construct in endogenous FOXG1 and performed chromatin immunoprecipitation (ChIP) sequencing. We also performed deep RNA sequencing of NPCs from two females with loss-of-function mutations in FOXG1 and their healthy biological mothers. Integrative analyses of RNA and ChIP sequencing data showed that cell cycle regulation and Bone Morphogenic Protein (BMP) repression gene ontology categories were over-represented as FOXG1 targets. Using engineered brain cell lines, we show that FOXG1 specifically activates SMAD7 and represses CDKN1B. Activation of SMAD7 which inhibits BMP signaling may be one way that FOXG1 patterns the forebrain, while repression of cell cycle regulators such as CDKN1B may be one way that FOXG1 expands the NPC pool to ensure proper brain size. Our data reveal novel mechanisms on how FOXG1 may control forebrain patterning and cell proliferation in human brain development.


Assuntos
Fatores de Transcrição Forkhead , Células-Tronco Neurais , Feminino , Humanos , Fatores de Transcrição Forkhead/metabolismo , Ciclo Celular/genética , Células-Tronco Neurais/metabolismo , Divisão Celular , Regulação da Expressão Gênica , Proteínas do Tecido Nervoso/metabolismo
2.
Hum Mol Genet ; 31(21): 3715-3728, 2022 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-35640156

RESUMO

Kabuki syndrome is frequently caused by loss-of-function mutations in one allele of histone 3 lysine 4 (H3K4) methyltransferase KMT2D and is associated with problems in neurological, immunological and skeletal system development. We generated heterozygous KMT2D knockout and Kabuki patient-derived cell models to investigate the role of reduced dosage of KMT2D in stem cells. We discovered chromosomal locus-specific alterations in gene expression, specifically a 110 Kb region containing Synaptotagmin 3 (SYT3), C-Type Lectin Domain Containing 11A (CLEC11A), Chromosome 19 Open Reading Frame 81 (C19ORF81) and SH3 And Multiple Ankyrin Repeat Domains 1 (SHANK1), suggesting locus-specific targeting of KMT2D. Using whole genome histone methylation mapping, we confirmed locus-specific changes in H3K4 methylation patterning coincident with regional decreases in gene expression in Kabuki cell models. Significantly reduced H3K4 peaks aligned with regions of stem cell maps of H3K27 and H3K4 methylation suggesting KMT2D haploinsufficiency impact bivalent enhancers in stem cells. Preparing the genome for subsequent differentiation cues may be of significant importance for Kabuki-related genes. This work provides a new insight into the mechanism of action of an important gene in bone and brain development and may increase our understanding of a specific function of a human disease-relevant H3K4 methyltransferase family member.


Assuntos
Histona-Lisina N-Metiltransferase , Histonas , Doenças Vestibulares , Humanos , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Células-Tronco/metabolismo , Doenças Vestibulares/genética
3.
Stem Cell Reports ; 17(3): 475-488, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35148845

RESUMO

Heterozygous loss-of-function mutations in Forkhead box G1 (FOXG1), a uniquely brain-expressed gene, cause microcephaly, seizures, and severe intellectual disability, whereas increased FOXG1 expression is frequently observed in glioblastoma. To investigate the role of FOXG1 in forebrain cell proliferation, we modeled FOXG1 syndrome using cells from three clinically diagnosed cases with two sex-matched healthy parents and one unrelated sex-matched control. Cells with heterozygous FOXG1 loss showed significant reduction in cell proliferation, increased ratio of cells in G0/G1 stage of the cell cycle, and increased frequency of primary cilia. Engineered loss of FOXG1 recapitulated this effect, while isogenic repair of a patient mutation reverted output markers to wild type. An engineered inducible FOXG1 cell line derived from a FOXG1 syndrome case demonstrated that FOXG1 dose-dependently affects all cell proliferation outputs measured. These findings provide strong support for the critical importance of FOXG1 levels in controlling human brain cell growth in health and disease.


Assuntos
Fatores de Transcrição Forkhead , Proteínas do Tecido Nervoso , Proliferação de Células , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Prosencéfalo/metabolismo , Células-Tronco/metabolismo , Síndrome
4.
Cells ; 10(11)2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34831347

RESUMO

Cardiovascular disease (CVD) is one of the contributing factors to more than one-third of human mortality and the leading cause of death worldwide. The death of cardiac myocyte is a fundamental pathological process in cardiac pathologies caused by various heart diseases, including myocardial infarction. Thus, strategies for replacing fibrotic tissue in the infarcted region with functional myocardium have long been a goal of cardiovascular research. This review begins by briefly discussing a variety of somatic stem- and progenitor-cell populations that were frequently studied in early investigations of regenerative myocardial therapy and then focuses primarily on pluripotent stem cells (PSCs), especially induced-pluripotent stem cells (iPSCs), which have emerged as perhaps the most promising source of cardiomyocytes for both therapeutic applications and drug testing. We also describe attempts to generate cardiomyocytes directly from cardiac fibroblasts (i.e., transdifferentiation), which, if successful, may enable the pool of endogenous cardiac fibroblasts to be used as an in-situ source of cardiomyocytes for myocardial repair.


Assuntos
Insuficiência Cardíaca/terapia , Miocárdio/patologia , Regeneração/fisiologia , Transplante de Células-Tronco , Animais , Ensaios Clínicos como Assunto , Humanos , Células-Tronco Pluripotentes Induzidas/citologia
5.
Stem Cell Reports ; 16(7): 1749-1762, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34214487

RESUMO

Mutations in HPRT1, a gene encoding a rate-limiting enzyme for purine salvage, cause Lesch-Nyhan disease which is characterized by self-injury and motor impairments. We leveraged stem cell and genetic engineering technologies to model the disease in isogenic and patient-derived forebrain and midbrain cell types. Dopaminergic progenitor cells deficient in HPRT showed decreased intensity of all developmental cell-fate markers measured. Metabolic analyses revealed significant loss of all purine derivatives, except hypoxanthine, and impaired glycolysis and oxidative phosphorylation. real-time glucose tracing demonstrated increased shunting to the pentose phosphate pathway for de novo purine synthesis at the expense of ATP production. Purine depletion in dopaminergic progenitor cells resulted in loss of RHEB, impairing mTORC1 activation. These data demonstrate dopaminergic-specific effects of purine salvage deficiency and unexpectedly reveal that dopaminergic progenitor cells are programmed to a high-energy state prior to higher energy demands of terminally differentiated cells.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Metabolismo Energético , Síndrome de Lesch-Nyhan/metabolismo , Síndrome de Lesch-Nyhan/patologia , Mesencéfalo/patologia , Biomarcadores/metabolismo , Linhagem da Célula , Córtex Cerebral/patologia , Glucose/metabolismo , Glicólise , Humanos , Hipoxantina Fosforribosiltransferase/deficiência , Síndrome de Lesch-Nyhan/enzimologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Células-Tronco Neurais/metabolismo , Fosforilação Oxidativa , Via de Pentose Fosfato , Purinas/metabolismo
6.
Stem Cells Transl Med ; 9(6): 697-712, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32154672

RESUMO

Making high-quality dopamine (DA)-producing cells for basic biological or small molecule screening studies is critical for the development of novel therapeutics for disorders of the ventral midbrain. Currently, many ventral midbrain assays have low signal-to-noise ratio due to low levels of cellular DA and the rate-limiting enzyme of DA synthesis, tyrosine hydroxylase (TH), hampering discovery efforts. Using intensively characterized ventral midbrain cells derived from human skin, which demonstrate calcium pacemaking activity and classical electrophysiological properties, we show that an L-type calcium agonist can significantly increase TH protein levels and DA content and release. Live calcium imaging suggests that it is the immediate influx of calcium occurring simultaneously in all cells that drives this effect. Genome-wide expression profiling suggests that L-type calcium channel stimulation has a significant effect on specific genes related to DA synthesis and affects expression of L-type calcium receptor subunits from the CACNA1 and CACNA2D families. Together, our findings provide an advance in the ability to increase DA and TH levels to improve the accuracy of disease modeling and small molecule screening for disorders of the ventral midbrain, including Parkinson's disease.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Dopamina/metabolismo , Mesencéfalo/citologia , Tirosina 3-Mono-Oxigenase/metabolismo , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Cálcio/metabolismo , Diferenciação Celular , Linhagem Celular , Forma Celular/efeitos dos fármacos , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Fenômenos Eletrofisiológicos , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Células-Tronco Neurais/citologia , Transcriptoma/genética
7.
Hum Mol Genet ; 29(5): 785-802, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-31943018

RESUMO

Down syndrome (DS), caused by the triplication of human chromosome 21, leads to significant alterations in brain development and is a major genetic cause of intellectual disability. While much is known about changes to neurons in DS, the effects of trisomy 21 on non-neuronal cells such as astrocytes are poorly understood. Astrocytes are critical for brain development and function, and their alteration may contribute to DS pathophysiology. To better understand the impact of trisomy 21 on astrocytes, we performed RNA-sequencing on astrocytes from newly produced DS human induced pluripotent stem cells (hiPSCs). While chromosome 21 genes were upregulated in DS astrocytes, we found consistent up- and down-regulation of genes across the genome with a strong dysregulation of neurodevelopmental, cell adhesion and extracellular matrix molecules. ATAC (assay for transposase-accessible chromatin)-seq also revealed a global alteration in chromatin state in DS astrocytes, showing modified chromatin accessibility at promoters of cell adhesion and extracellular matrix genes. Along with these transcriptomic and epigenomic changes, DS astrocytes displayed perturbations in cell size and cell spreading as well as modifications to cell-cell and cell-substrate recognition/adhesion, and increases in cellular motility and dynamics. Thus, triplication of chromosome 21 is associated with genome-wide transcriptional, epigenomic and functional alterations in astrocytes that may contribute to altered brain development and function in DS.


Assuntos
Astrócitos/patologia , Adesão Celular , Síndrome de Down/patologia , Regulação da Expressão Gênica , Genoma Humano , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Neurais/patologia , Astrócitos/metabolismo , Diferenciação Celular , Movimento Celular , Síndrome de Down/genética , Síndrome de Down/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Neurais/metabolismo , Transcriptoma
8.
Am J Hum Genet ; 104(5): 815-834, 2019 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-31031012

RESUMO

We identified individuals with variations in ACTL6B, a component of the chromatin remodeling machinery including the BAF complex. Ten individuals harbored bi-allelic mutations and presented with global developmental delay, epileptic encephalopathy, and spasticity, and ten individuals with de novo heterozygous mutations displayed intellectual disability, ambulation deficits, severe language impairment, hypotonia, Rett-like stereotypies, and minor facial dysmorphisms (wide mouth, diastema, bulbous nose). Nine of these ten unrelated individuals had the identical de novo c.1027G>A (p.Gly343Arg) mutation. Human-derived neurons were generated that recaptured ACTL6B expression patterns in development from progenitor cell to post-mitotic neuron, validating the use of this model. Engineered knock-out of ACTL6B in wild-type human neurons resulted in profound deficits in dendrite development, a result recapitulated in two individuals with different bi-allelic mutations, and reversed on clonal genetic repair or exogenous expression of ACTL6B. Whole-transcriptome analyses and whole-genomic profiling of the BAF complex in wild-type and bi-allelic mutant ACTL6B neural progenitor cells and neurons revealed increased genomic binding of the BAF complex in ACTL6B mutants, with corresponding transcriptional changes in several genes including TPPP and FSCN1, suggesting that altered regulation of some cytoskeletal genes contribute to altered dendrite development. Assessment of bi-alleic and heterozygous ACTL6B mutations on an ACTL6B knock-out human background demonstrated that bi-allelic mutations mimic engineered deletion deficits while heterozygous mutations do not, suggesting that the former are loss of function and the latter are gain of function. These results reveal a role for ACTL6B in neurodevelopment and implicate another component of chromatin remodeling machinery in brain disease.


Assuntos
Actinas/genética , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Dendritos/patologia , Epilepsia/etiologia , Células-Tronco Pluripotentes Induzidas/patologia , Mutação , Transtornos do Neurodesenvolvimento/etiologia , Neurônios/patologia , Adulto , Criança , Pré-Escolar , Cromatina/genética , Cromatina/metabolismo , Dendritos/metabolismo , Epilepsia/patologia , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lactente , Masculino , Transtornos do Neurodesenvolvimento/patologia , Neurônios/metabolismo , Adulto Jovem
9.
Bio Protoc ; 9(5): e3188, 2019 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-33654990

RESUMO

Induced Pluripotent Stem Cells (iPSCs) are pluripotent stem cells that can be generated from somatic cells, and provide a way to model the development of neural tissues in vitro. One particularly interesting application of iPSCs is the development of neurons analogous to those found in the human forebrain. Forebrain neurons play a central role in cognition and sensory processing, and deficits in forebrain neuronal activity contributes to a host of conditions, including epilepsy, Alzheimer's disease, and schizophrenia. Here, we present our protocol for differentiating iPSCs into forebrain neural progenitor cells (NPCs) and neurons, whereby neural rosettes are generated from stem cells without dissociation and NPCs purified from rosettes based on their adhesion, resulting in a more rapid generation of pure NPC cultures. Neural progenitor cells can be maintained as long-term cultures, or differentiated into forebrain neurons. This protocol provides a simplified and fast methodology of generating forebrain NPCs and neurons, and enables researchers to generate effective in vitro models to study forebrain disease and neurodevelopment. This protocol can also be easily adapted to generate other neural lineages.

10.
Stem Cell Reports ; 11(1): 183-196, 2018 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-29937144

RESUMO

Heterozygous loss-of-function mutations in GRIN2B, a subunit of the NMDA receptor, cause intellectual disability and language impairment. We developed clonal models of GRIN2B deletion and loss-of-function mutations in a region coding for the glutamate binding domain in human cells and generated neurons from a patient harboring a missense mutation in the same domain. Transcriptome analysis revealed extensive increases in genes associated with cell proliferation and decreases in genes associated with neuron differentiation, a result supported by extensive protein analyses. Using electrophysiology and calcium imaging, we demonstrate that NMDA receptors are present on neural progenitor cells and that human mutations in GRIN2B can impair calcium influx and membrane depolarization even in a presumed undifferentiated cell state, highlighting an important role for non-synaptic NMDA receptors. It may be this function, in part, which underlies the neurological disease observed in patients with GRIN2B mutations.


Assuntos
Diferenciação Celular , Mutação , Neurônios/citologia , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Biomarcadores , Diferenciação Celular/genética , Análise Mutacional de DNA , Reparo do DNA , Dosagem de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutação com Perda de Função , Modelos Moleculares , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Conformação Proteica , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/metabolismo
11.
Front Aging Neurosci ; 9: 258, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28824419

RESUMO

Synaptic transmission requires intricate coordination of the components involved in processing of incoming signals, formation and stabilization of synaptic machinery, neurotransmission and in all related signaling pathways. Changes to any of these components cause synaptic imbalance and disruption of neuronal circuitry. Extensive studies at the neuromuscular junction (NMJ) have greatly aided in the current understanding of synapses and served to elucidate the underlying physiology as well as associated adaptive and homeostatic processes. The heparan sulfate proteoglycan agrin is a vital component of the NMJ, mediating synaptic formation and maintenance in both brain and muscle, but very little is known about direct control of its expression. Here, we investigated the relationship between agrin and transcription factor early growth response-1 (Egr-1), as Egr-1 regulates the expression of many genes involved in synaptic homeostasis and plasticity. Using chromatin immunoprecipitation (ChIP), cell culture with cell lines derived from brain and muscle, and animal models, we show that Egr-1 binds to the AGRN gene locus and suppresses its expression. When compared with wild type (WT), mice deficient in Egr-1 (Egr-1-/-) display a marked increase in AGRN mRNA and agrin full-length and cleavage fragment protein levels, including the 22 kDa, C-terminal fragment in brain and muscle tissue homogenate. Because agrin is a crucial component of the NMJ, we explored possible physiological implications of the Egr-1-agrin relationship. In the diaphragm, Egr-1-/- mice display increased NMJ motor endplate density, individual area and area of innervation. In addition to increased density, soleus NMJs also display an increase in fragmented and faint endplates in Egr-1-/- vs. WT mice. Moreover, the soleus NMJ electrophysiology of Egr-1-/- mice revealed increased quantal content and motor testing showed decreased movement and limb muscle strength compared with WT. This study provides evidence for the potential involvement of a novel Egr-1-agrin pathway in synaptic homeostatic and compensatory mechanisms at the NMJ. Synaptic homeostasis is greatly affected by the process of aging. These and other data suggest that changes in Egr-1 expression may directly or indirectly promote age-related pathologies.

12.
Stem Cells Transl Med ; 6(3): 886-896, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28170165

RESUMO

The development of targeted therapeutics for rare neurodevelopmental disorders (NDDs) faces significant challenges due to the scarcity of subjects and the difficulty of obtaining human neural cells. Here, we illustrate a rapid, simple protocol by which patient derived cells can be reprogrammed to induced pluripotent stem cells (iPSCs) using an episomal vector and differentiated into neurons. Using this platform enables patient somatic cells to be converted to physiologically active neurons in less than two months with minimal labor. This platform includes a method to combine somatic cell reprogramming with CRISPR/Cas9 gene editing at single cell resolution, which enables the concurrent development of clonal knockout or knock-in models that can be used as isogenic control lines. This platform reduces the logistical barrier for using iPSC technology, allows for the development of appropriate control lines for use in rare neurodevelopmental disease research, and establishes a fundamental component to targeted therapeutics and precision medicine. Stem Cells Translational Medicine 2017;6:886-896.


Assuntos
Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Edição de Genes , Modelos Biológicos , Transtornos do Neurodesenvolvimento/patologia , Sequência de Bases , Diferenciação Celular , Fibroblastos/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Mesencéfalo/patologia , Neurônios/patologia , Prosencéfalo/patologia
13.
Am J Med Genet A ; 170A(5): 1225-35, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26789910

RESUMO

Mutations in chromodomain helicase DNA-binding domain 8 (CHD8) have been identified in independent genotyping studies of autism spectrum disorder. To better understand the phenotype associated with CHD8 mutations, we genotyped all CHD8 exons in carefully assessed cohorts of autism (n = 142), schizophrenia (SCZ; n = 143), and intellectual disability (ID; n = 94). We identified one frameshift mutation, seven non-synonymous variants, and six synonymous variants. The frameshift mutation, p.Asn2092Lysfs*2, which creates a premature stop codon leading to the loss of 212 amino acids of the protein, was from an autism case on whom we present multiple clinical assessments and pharmacological treatments spanning more than 10 years. RNA and protein analysis support a model where the transcript generated from the mutant allele results in haploinsufficiency of CHD8. This case report supports the association of CHD8 mutations with classical autism, macrocephaly, infantile hypotonia, speech delay, lack of major ID, and psychopathology in late adolescence caused by insufficient dosage of CHD8. Review of 16 other CHD8 mutation cases suggests that clinical features and their severity vary considerably across individuals; however, these data support a CHD8 mutation syndrome, further highlighting the importance of genomic medicine to guide clinical assessment and treatment.


Assuntos
Transtorno do Espectro Autista/genética , Proteínas de Ligação a DNA/genética , Esquizofrenia/genética , Fatores de Transcrição/genética , Adolescente , Transtorno do Espectro Autista/fisiopatologia , Criança , Pré-Escolar , Hibridização Genômica Comparativa , Éxons/genética , Feminino , Mutação da Fase de Leitura , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Humanos , Masculino , Esquizofrenia/fisiopatologia
14.
Proc Natl Acad Sci U S A ; 111(18): 6810-5, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24753587

RESUMO

Dystroglycan (DG), a cell adhesion molecule well known to be essential for skeletal muscle integrity and formation of neuromuscular synapses, is also present at inhibitory synapses in the central nervous system. Mutations that affect DG function not only result in muscular dystrophies, but also in severe cognitive deficits and epilepsy. Here we demonstrate a role of DG during activity-dependent homeostatic regulation of hippocampal inhibitory synapses. Prolonged elevation of neuronal activity up-regulates DG expression and glycosylation, and its localization to inhibitory synapses. Inhibition of protein synthesis prevents the activity-dependent increase in synaptic DG and GABAA receptors (GABAARs), as well as the homeostatic scaling up of GABAergic synaptic transmission. RNAi-mediated knockdown of DG blocks homeostatic scaling up of inhibitory synaptic strength, as does knockdown of like-acetylglucosaminyltransferase (LARGE)--a glycosyltransferase critical for DG function. In contrast, DG is not required for the bicuculline-induced scaling down of excitatory synaptic strength or the tetrodotoxin-induced scaling down of inhibitory synaptic strength. The DG ligand agrin increases GABAergic synaptic strength in a DG-dependent manner that mimics homeostatic scaling up induced by increased activity, indicating that activation of this pathway alone is sufficient to regulate GABAAR trafficking. These data demonstrate that DG is regulated in a physiologically relevant manner in neurons and that DG and its glycosylation are essential for homeostatic plasticity at inhibitory synapses.


Assuntos
Distroglicanas/metabolismo , Neurônios GABAérgicos/metabolismo , Plasticidade Neuronal/fisiologia , Agrina/metabolismo , Animais , Distroglicanas/antagonistas & inibidores , Distroglicanas/genética , Feminino , Glicosilação , Hipocampo/metabolismo , Homeostase , Humanos , Masculino , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Mutação , N-Acetilglucosaminiltransferases/antagonistas & inibidores , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/metabolismo , Sinapses/metabolismo
15.
Dev Neurobiol ; 73(5): 333-53, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22949126

RESUMO

In response to a wound, astrocytes in culture extend microtubule-rich processes and polarize, orienting their centrosomes and Golgi apparatus woundside. ß1 Integrin null astrocytes fail to extend processes toward the wound, and are disoriented, and often migrate away orthogonal, to the wound. The centrosome is unusually fragmented in ß1 integrin null astrocytes. Expression of a ß1 integrin cDNA in the null background yields cells with intact centrosomes that polarize and extend processes normally. Fragmented centrosomes rapidly assemble following integrin ligation and cell attachment. However, several experiments indicated that cell adhesion is not necessary. For example, astrocytes in suspension expressing a chimeric ß1 subunit that can be activated by an antibody assemble centrosomes suggesting that ß1 activation is sufficient to cause centrosome assembly in the absence of cell adhesion. siRNA knockdown of PCM1, a major centrosomal protein, inhibits cell polarization, consistent with the notion that centrosomes are necessary for polarity and that integrins regulate polarity via centrosome integrity. Screening inhibitors of molecules downstream of integrins indicate that neither FAK nor ILK is involved in regulation of centrosome integrity. In contrast, blebbistatin, a specific inhibitor of non-muscle myosin II (NMII), mimics the response of ß1 integrin null astrocytes by disrupting centrosome integrity and cell polarization. Blebbistatin also inhibits integrin-mediated centrosome assembly in astrocytes attaching to fibronectin, consistent with the hypothesis that NMII functions downstream of integrins in regulating centrosome integrity.


Assuntos
Astrócitos/ultraestrutura , Centrossomo/ultraestrutura , Integrina beta1/fisiologia , Cicatrização/fisiologia , Animais , Adesão Celular , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Polaridade Celular , Células Cultivadas/fisiologia , Embrião de Galinha , DNA Complementar/genética , Matriz Extracelular/fisiologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Integrina beta1/biossíntese , Integrina beta1/genética , Camundongos , Miosina não Muscular Tipo IIB/antagonistas & inibidores , Miosina não Muscular Tipo IIB/fisiologia , Proteínas Tirosina Quinases/metabolismo , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/fisiologia , Retina/citologia , Retina/embriologia , Suspensões
16.
Proc Natl Acad Sci U S A ; 107(17): 7863-8, 2010 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-20385823

RESUMO

Schizophrenia likely results from poorly understood genetic and environmental factors. We studied the gene encoding the synaptic protein SHANK3 in 285 controls and 185 schizophrenia patients with unaffected parents. Two de novo mutations (R1117X and R536W) were identified in two families, one being found in three affected brothers, suggesting germline mosaicism. Zebrafish and rat hippocampal neuron assays revealed behavior and differentiation defects resulting from the R1117X mutant. As mutations in SHANK3 were previously reported in autism, the occurrence of SHANK3 mutations in subjects with a schizophrenia phenotype suggests a molecular genetic link between these two neurodevelopmental disorders.


Assuntos
Proteínas de Transporte/genética , Mutação de Sentido Incorreto/genética , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Esquizofrenia/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Biologia Computacional , Primers do DNA/genética , Feminino , Humanos , Masculino , Repetições de Microssatélites/genética , Dados de Sequência Molecular , Linhagem , Ratos , Análise de Sequência de DNA , Peixe-Zebra
17.
Hum Mol Genet ; 17(24): 3965-74, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18801879

RESUMO

In a systematic sequencing screen of synaptic genes on the X chromosome, we have identified an autistic female without mental retardation (MR) who carries a de novo frameshift Ile367SerfsX6 mutation in Interleukin-1 Receptor Accessory Protein-Like 1 (IL1RAPL1), a gene implicated in calcium-regulated vesicle release and dendrite differentiation. We showed that the function of the resulting truncated IL1RAPL1 protein is severely altered in hippocampal neurons, by measuring its effect on neurite outgrowth activity. We also sequenced the coding region of the close related member IL1RAPL2 and of NCS-1/FREQ, which physically interacts with IL1RAPL1, in a cohort of subjects with autism. The screening failed to identify non-synonymous variant in IL1RAPL2, whereas a rare missense (R102Q) in NCS-1/FREQ was identified in one autistic patient. Furthermore, we identified by comparative genomic hybridization a large intragenic deletion of exons 3-7 of IL1RAPL1 in three brothers with autism and/or MR. This deletion causes a frameshift and the introduction of a premature stop codon, Ala28GlufsX15, at the very beginning of the protein. All together, our results indicate that mutations in IL1RAPL1 cause a spectrum of neurological impairments ranging from MR to high functioning autism.


Assuntos
Transtorno Autístico/genética , Cálcio/fisiologia , Proteína Acessória do Receptor de Interleucina-1/genética , Deleção de Sequência/genética , Animais , Síndrome de Asperger/genética , Síndrome de Asperger/patologia , Transtorno Autístico/patologia , Sequência de Bases , Diferenciação Celular/genética , Linhagem Celular , Criança , Códon sem Sentido/genética , Feminino , Mutação da Fase de Leitura , Triagem de Portadores Genéticos , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Proteína Acessória do Receptor de Interleucina-1/fisiologia , Masculino , Neuritos/metabolismo , Neuritos/patologia , Linhagem , Ratos
18.
Dev Neurobiol ; 68(5): 559-74, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18188865

RESUMO

Monolayers of astrocytes in culture respond to a scrape wound by orienting towards the wound and extending processes that will repair it. We show here that they also upregulate the expression of extracellular matrix (ECM) proteins, laminin, and chondroitin sulfated proteoglycan, that are deposited in astrocytic scars in vivo. We have previously shown that the major functional ECM receptors on astrocytes are dystroglycan (DG) plus integrins alpha1beta1, alpha5beta1, alpha6beta1, and alphavbeta3. Consistent with this, laminin fragments that activate alpha1beta1 integrin, alpha6beta1 integrin, and DG all contribute to attachment. During astrocyte attachment, or process extension, integrins and DG are found at the leading edge of the lammelipodium, though they change in distribution with the extent of attachment and the alpha and beta subunits of DG can be spatially uncoupled. Functionally, inhibitory antibodies to DG and integrin alpha1beta1 or the RGD peptide all inhibit process extension, showing that ligand engagement of integrins and DG contribute to process extension. Astrocytes differentiated from DG or beta1 null ES cells respond very differently to wounding. The former fail to extend process and cell polarization is disrupted partially. However, beta1 null astrocytes not only fail to extend processes perpendicular to the wound, but cell polarization is completely disrupted and cells migrate randomly into the wound. We conclude that integrins are essential for astrocyte polarity.


Assuntos
Astrócitos/fisiologia , Distroglicanas/fisiologia , Integrina beta1/fisiologia , Integrinas/fisiologia , Microtúbulos/fisiologia , Cicatrização/fisiologia , Animais , Astrócitos/ultraestrutura , Adesão Celular/fisiologia , Diferenciação Celular/fisiologia , Polaridade Celular/fisiologia , Células Cultivadas , Células-Tronco Embrionárias/fisiologia , Matriz Extracelular/fisiologia , Imunofluorescência , Integrina alfa1beta1/metabolismo , Ratos , Ratos Sprague-Dawley , Frações Subcelulares/fisiologia
19.
J Gen Virol ; 87(Pt 3): 673-678, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16476990

RESUMO

Dystroglycan (DG) is an extracellular matrix receptor necessary for the development of metazoans from flies to humans and is also an entry route for various pathogens. Lymphocytic choriomeningitis virus (LCMV), a member of the family Arenaviridae, infects by binding to alpha-DG. Here, the role of cholesterol lipid rafts in infection by LCMV via alpha-DG was investigated. The cholesterol-sequestering drugs methyl-beta-cyclodextrin (MbetaCD), filipin and nystatin inhibited the infectivity of LCMV selectively, but did not affect infection by vesicular stomatitis virus. Cholesterol loading after depletion with MbetaCD restored infectivity to control levels. DG was not found in lipid rafts identified with the raft marker ganglioside GM1. Treatment with MbetaCD, however, enhanced the solubility of DG. This may reflect the association of DG with cholesterol outside lipid rafts and suggests that association of DG with non-raft cholesterol is critical for infection by LCMV through alpha-DG.


Assuntos
Infecções por Arenaviridae/virologia , Colesterol/fisiologia , Distroglicanas/metabolismo , Vírus da Coriomeningite Linfocítica/fisiologia , Animais , Linhagem Celular , Colesterol/metabolismo , Distroglicanas/química , Camundongos , Solubilidade , Replicação Viral
20.
Life Sci ; 76(3): 263-80, 2004 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-15531379

RESUMO

Though the nicotinic acetylcholine receptor (nAChR) subunits alpha9 and alpha 10 have been thoroughly characterized within hair cells of the organ of Corti in the inner ear, prior studies have shown that they are also expressed in lymphocytes. In this report, we sought to more definitively characterize the nAChR subunits alpha9 and alpha10 within various populations of human lymphocytes. Using a combination of techniques, including RT-PCR, single-cell RT-PCR, Northern and western blot analysis, and immunofluorescence, expression of both alpha9 and alpha 10 was demonstrated in purified populations of T-cells (CD3+, CD4+, CD8+ and the Jurkat, MT2 and CEM T-cell lines) and B-cells (CD19+, CD80+ and EBV-immortalized B-cells). Single-lymphocyte recording techniques failed to identify an ionic current in response to applied acetylcholine in either T-cells or B-cells. These results clearly demonstrate the presence of these nicotinic receptor subunits within several populations of human lymphocytes, implicating their role in the immune response. However, a lack of demonstrated response to applied acetylcholine using standard single-cell recording techniques suggests a physiology different than that seen in hair cells of the inner ear.


Assuntos
Subpopulações de Linfócitos B/metabolismo , Receptores Nicotínicos/metabolismo , Subpopulações de Linfócitos T/metabolismo , Acetilcolina/farmacologia , Subpopulações de Linfócitos B/efeitos dos fármacos , Separação Celular , Citometria de Fluxo , Humanos , Células Jurkat , RNA Mensageiro/metabolismo , Receptores Nicotínicos/análise , Receptores Nicotínicos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Subpopulações de Linfócitos T/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...