Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Genet Med ; 26(4): 101057, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38158856

RESUMO

PURPOSE: We established the genetic etiology of a syndromic neurodevelopmental condition characterized by variable cognitive impairment, recognizable facial dysmorphism, and a constellation of extra-neurological manifestations. METHODS: We performed phenotypic characterization of 6 participants from 4 unrelated families presenting with a neurodevelopmental syndrome and used exome sequencing to investigate the underlying genetic cause. To probe relevance to the neurodevelopmental phenotype and craniofacial dysmorphism, we established two- and three-dimensional human stem cell-derived neural models and generated a stable cachd1 zebrafish mutant on a transgenic cartilage reporter line. RESULTS: Affected individuals showed mild cognitive impairment, dysmorphism featuring oculo-auriculo abnormalities, and developmental defects involving genitourinary and digestive tracts. Exome sequencing revealed biallelic putative loss-of-function variants in CACHD1 segregating with disease in all pedigrees. RNA sequencing in CACHD1-depleted neural progenitors revealed abnormal expression of genes with key roles in Wnt signaling, neurodevelopment, and organ morphogenesis. CACHD1 depletion in neural progenitors resulted in reduced percentages of post-mitotic neurons and enlargement of 3D neurospheres. Homozygous cachd1 mutant larvae showed mandibular patterning defects mimicking human facial dysmorphism. CONCLUSION: Our findings support the role of loss-of-function variants in CACHD1 as the cause of a rare neurodevelopmental syndrome with facial dysmorphism and multisystem abnormalities.


Assuntos
Anormalidades Múltiplas , Anormalidades Craniofaciais , Anormalidades Musculoesqueléticas , Transtornos do Neurodesenvolvimento , Animais , Humanos , Anormalidades Múltiplas/genética , Anormalidades Craniofaciais/genética , Deficiência Intelectual/genética , Anormalidades Musculoesqueléticas/genética , Transtornos do Neurodesenvolvimento/genética , Fenótipo , Síndrome , Peixe-Zebra/genética
2.
Semin Cell Dev Biol ; 111: 67-73, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32654970

RESUMO

Until the discovery of human embryonic stem cells and human induced pluripotent stem cells, biotechnology companies were severely limited in the number of human tissues that they could model in large-scale in vitro studies. Until this point, companies have been limited to immortalized cancer lines or a small number of primary cell types that could be extracted and expanded. Nowadays, protocols continue to be developed in the stem cell field, enabling researchers to model an ever-growing library of cell types in controlled, large-scale screens. One differentiation method in particular- cerebral organoids- shows substantial potential in the field of neuroscience and developmental neurobiology. Cerebral organoid technology is still in an early phase of development, and there are several challenges that are currently being addressed by academic and industrial researchers alike. Here we briefly describe some of the early adopters of cerebral organoids, several of the challenges that they are likely facing, and various technologies that are currently being implemented to overcome them.


Assuntos
Descoberta de Drogas/métodos , Drogas em Investigação/farmacologia , Modelos Biológicos , Doenças Neurodegenerativas/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Organoides/efeitos dos fármacos , Sistemas CRISPR-Cas , Diferenciação Celular , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Drogas em Investigação/química , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Aprendizado de Máquina , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/química , Organoides/metabolismo , Organoides/patologia , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos
3.
Nucleic Acids Res ; 43(10): e65, 2015 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-25765640

RESUMO

Isogenic pluripotent stem cells are critical tools for studying human neurological diseases by allowing one to study the effects of a mutation in a fixed genetic background. Of particular interest are the spectrum of autism disorders, some of which are monogenic such as Timothy syndrome (TS); others are multigenic such as the microdeletion and microduplication syndromes of the 16p11.2 chromosomal locus. Here, we report engineered human embryonic stem cell (hESC) lines for modeling these two disorders using locus-specific endonucleases to increase the efficiency of homology-directed repair (HDR). We developed a system to: (1) computationally identify unique transcription activator-like effector nuclease (TALEN) binding sites in the genome using a new software program, TALENSeek, (2) assemble the TALEN genes by combining golden gate cloning with modified constructs from the FLASH protocol, and (3) test the TALEN pairs in an amplification-based HDR assay that is more sensitive than the typical non-homologous end joining assay. We applied these methods to identify, construct, and test TALENs that were used with HDR donors in hESCs to generate an isogenic TS cell line in a scarless manner and to model the 16p11.2 copy number disorder without modifying genomic loci with high sequence similarity.


Assuntos
Engenharia Celular , Transtornos Globais do Desenvolvimento Infantil/genética , Células-Tronco Embrionárias , Modelos Genéticos , Transtorno Autístico , Sítios de Ligação , Linhagem Celular , Desoxirribonucleases/metabolismo , Marcação de Genes , Genoma Humano , Humanos , Síndrome do QT Longo/genética , Reparo de DNA por Recombinação , Software , Sindactilia/genética
4.
BMC Genomics ; 15: 154, 2014 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-24564186

RESUMO

BACKGROUND: High-throughput sequencing is gradually replacing microarrays as the preferred method for studying mRNA expression levels, providing nucleotide resolution and accurately measuring absolute expression levels of almost any transcript, known or novel. However, existing microarray data from clinical, pharmaceutical, and academic settings represent valuable and often underappreciated resources, and methods for assessing and improving the quality of these data are lacking. RESULTS: To quantitatively assess the quality of microarray probes, we directly compare RNA-Seq to Agilent microarrays by processing 231 unique samples from the Allen Human Brain Atlas using RNA-Seq. Both techniques provide highly consistent, highly reproducible gene expression measurements in adult human brain, with RNA-Seq slightly outperforming microarray results overall. We show that RNA-Seq can be used as ground truth to assess the reliability of most microarray probes, remove probes with off-target effects, and scale probe intensities to match the expression levels identified by RNA-Seq. These sequencing scaled microarray intensities (SSMIs) provide more reliable, quantitative estimates of absolute expression levels for many genes when compared with unscaled intensities. Finally, we validate this result in two human cell lines, showing that linear scaling factors can be applied across experiments using the same microarray platform. CONCLUSIONS: Microarrays provide consistent, reproducible gene expression measurements, which are improved using RNA-Seq as ground truth. We expect that our strategy could be used to improve probe quality for many data sets from major existing repositories.


Assuntos
Encéfalo/metabolismo , Perfilação da Expressão Gênica/métodos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Análise de Sequência de RNA/métodos , Análise por Conglomerados , Biologia Computacional/métodos , Expressão Gênica , Perfilação da Expressão Gênica/normas , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Neocórtex/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/normas , Reprodutibilidade dos Testes , Análise de Sequência de RNA/normas , Transcriptoma
5.
Nat Genet ; 32(2): 326-30, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12172548

RESUMO

Familial exudative vitreoretinopathy (FEVR) is a hereditary ocular disorder characterized by a failure of peripheral retinal vascularization. Loci associated with FEVR map to 11q13-q23 (EVR1; OMIM 133780, ref. 1), Xp11.4 (EVR2; OMIM 305390, ref. 2) and 11p13-12 (EVR3; OMIM 605750, ref. 3). Here we have confirmed linkage to the 11q13-23 locus for autosomal dominant FEVR in one large multigenerational family and refined the disease locus to a genomic region spanning 1.55 Mb. Mutations in FZD4, encoding the putative Wnt receptor frizzled-4, segregated completely with affected individuals in the family and were detected in affected individuals from an additional unrelated family, but not in normal controls. FZD genes encode Wnt receptors, which are implicated in development and carcinogenesis. Injection of wildtype and mutated FZD4 into Xenopus laevis embryos revealed that wildtype, but not mutant, frizzled-4 activated calcium/calmodulin-dependent protein kinase II (CAMKII) and protein kinase C (PKC), components of the Wnt/Ca(2+) signaling pathway. In one of the mutants, altered subcellular trafficking led to defective signaling. These findings support a function for frizzled-4 in retinal angiogenesis and establish the first association between a Wnt receptor and human disease.


Assuntos
Neovascularização Patológica/genética , Proteínas/genética , Doenças Retinianas/genética , Vasos Retinianos/patologia , Sequência de Aminoácidos , Pré-Escolar , Feminino , Receptores Frizzled , Marcadores Genéticos , Haplótipos , Humanos , Masculino , Dados de Sequência Molecular , Mutação , Linhagem , Polimorfismo Genético , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G , Retina/patologia , Doenças Retinianas/patologia , Alinhamento de Sequência , Transdução de Sinais
6.
Cell Stem Cell ; 30(3): 312-332.e13, 2023 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-36796362

RESUMO

Human genome variation contributes to diversity in neurodevelopmental outcomes and vulnerabilities; recognizing the underlying molecular and cellular mechanisms will require scalable approaches. Here, we describe a "cell village" experimental platform we used to analyze genetic, molecular, and phenotypic heterogeneity across neural progenitor cells from 44 human donors cultured in a shared in vitro environment using algorithms (Dropulation and Census-seq) to assign cells and phenotypes to individual donors. Through rapid induction of human stem cell-derived neural progenitor cells, measurements of natural genetic variation, and CRISPR-Cas9 genetic perturbations, we identified a common variant that regulates antiviral IFITM3 expression and explains most inter-individual variation in susceptibility to the Zika virus. We also detected expression QTLs corresponding to GWAS loci for brain traits and discovered novel disease-relevant regulators of progenitor proliferation and differentiation such as CACHD1. This approach provides scalable ways to elucidate the effects of genes and genetic variation on cellular phenotypes.


Assuntos
Células-Tronco Neurais , Infecção por Zika virus , Zika virus , Humanos , Células-Tronco Neurais/metabolismo , Diferenciação Celular/genética , Encéfalo/metabolismo , Zika virus/metabolismo , Expressão Gênica , Proteínas de Membrana/metabolismo , Proteínas de Ligação a RNA/metabolismo
7.
Cell Death Dis ; 13(3): 262, 2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35322011

RESUMO

Mutations in N-glycanase 1 (NGLY1), which deglycosylates misfolded glycoproteins for degradation, can cause NGLY1 deficiency in patients and their abnormal fetal development in multiple organs, including microcephaly and other neurological disorders. Using cerebral organoids (COs) developed from human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs), we investigate how NGLY1 dysfunction disturbs early brain development. While NGLY1 loss had limited impact on the undifferentiated cells, COs developed from NGLY1-deficient hESCs showed defective formation of SATB2-positive upper-layer neurons, and attenuation of STAT3 and HES1 signaling critical for sustaining radial glia. Bulk and single-cell transcriptomic analysis revealed premature neuronal differentiation accompanied by downregulation of secreted and transcription factors, including TTR, IGFBP2, and ID4 in NGLY1-deficient COs. NGLY1 malfunction also dysregulated ID4 and enhanced neuronal differentiation in CO transplants developed in vivo. NGLY1-deficient CO cells were more vulnerable to multiple stressors; treating the deficient cells with recombinant TTR reduced their susceptibility to stress from proteasome inactivation, likely through LRP2-mediated activation of MAPK signaling. Expressing NGLY1 led to IGFBP2 and ID4 upregulation in CO cells developed from NGLY1-deficiency patient's hiPSCs. In addition, treatment with recombinant IGFBP2 enhanced ID4 expression, STAT3 signaling, and proliferation of NGLY1-deficient CO cells. Overall, our discoveries suggest that dysregulation of stress responses and neural precursor differentiation underlies the brain abnormalities observed in NGLY1-deficient individuals.


Assuntos
Organoides , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/metabolismo , Complexo de Endopeptidases do Proteassoma , Glicoproteínas/metabolismo , Humanos , Neurogênese , Organoides/metabolismo , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/genética , Complexo de Endopeptidases do Proteassoma/metabolismo
8.
Expert Opin Ther Targets ; 26(9): 811-822, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36424892

RESUMO

INTRODUCTION: The Helping to End Addiction Long-termSM Initiative supports a wide range of programs to develop new or improved prevention and opioid addiction treatment strategies. An essential component of this effort is to accelerate development of non-opioid pain therapeutics. In all fields of medicine, therapeutics development is an arduous process and late-stage translational efforts such as clinical trials to validate targets are particularly complex and costly. While there are plentiful novel targets for pain treatment, successful clinical validation is rare. It is therefore crucial to develop processes whereby therapeutic targets can be reasonably 'de-risked' prior to substantial late-stage validation efforts. Such rigorous validation of novel therapeutic targets in the preclinical space will give potential private sector partners the confidence to pursue clinical validation of promising therapeutic concepts and compounds. AREAS COVERED: In 2020, the National Institutes of Health (NIH) held the Target Validation for Non-Addictive Therapeutics Development for Pain workshop to gather insights from key opinion leaders in academia, industry, and venture-financing. EXPERT OPINION: The result was a roadmap for pain target validation focusing on three modalities: 1) human evidence; 2) assay development in vitro; 3) assay development in vivo.


Assuntos
Transtornos Relacionados ao Uso de Opioides , Dor , Humanos , Dor/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico
9.
Nat Cell Biol ; 6(1): 52-8, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14688793

RESUMO

nt signalling pathways regulate cell proliferation, cell fate and morphogenetic movements. Here, we demonstrate that the Frizzled (Fz) family of Wnt receptors, similarly to G-protein-coupled receptors (GPCRs), form specific homo- and hetero-oligomers. Two lines of evidence suggest that oligomerization occurs in the endoplasmic reticulum: first, a mutant allele of Fz4, encoding a truncated protein that is retained in the endoplasmic reticulum, is linked to the autosomal-dominant retinal degenerative disease, familial exudative vitreoretinopathy (FEVR). We show that this mutant form of Fz4 oligomerizes with wild-type Fz4, retains it in the endoplasmic reticulum and inhibits its signalling. Second, a derivative of Fz1 targeted to the endoplasmic reticulum traps wild-type Fz1 in the endoplasmic reticulum and blocks its signalling. These data support the hypothesis that oligomerization of mutant and wild-type Fz proteins occurs in the endoplasmic reticulum and may explain the genetic dominance of this FEVR allele.


Assuntos
Retículo Endoplasmático/metabolismo , Proteínas/genética , Proteínas/metabolismo , Vitreorretinopatia Proliferativa/genética , Proteínas de Peixe-Zebra , Alelos , Animais , Células COS , Receptores Frizzled , Genes Dominantes/genética , Humanos , Mutação/genética , Fragmentos de Peptídeos/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G , Receptores de Neurotransmissores/metabolismo , Transdução de Sinais/genética , Proteínas Wnt
10.
Genome Biol ; 20(1): 142, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31315641

RESUMO

We develop CellSIUS (Cell Subtype Identification from Upregulated gene Sets) to fill a methodology gap for rare cell population identification for scRNA-seq data. CellSIUS outperforms existing algorithms for specificity and selectivity for rare cell types and their transcriptomic signature identification in synthetic and complex biological data. Characterization of a human pluripotent cell differentiation protocol recapitulating deep-layer corticogenesis using CellSIUS reveals unrecognized complexity in human stem cell-derived cellular populations. CellSIUS enables identification of novel rare cell populations and their signature genes providing the means to study those populations in vitro in light of their role in health and disease.


Assuntos
Análise de Célula Única/métodos , Transcriptoma , Algoritmos , Linhagem Celular , Humanos , Neurônios/citologia
11.
Nat Neurosci ; 22(3): 374-385, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30718903

RESUMO

Synapse density is reduced in postmortem cortical tissue from schizophrenia patients, which is suggestive of increased synapse elimination. Using a reprogrammed in vitro model of microglia-mediated synapse engulfment, we demonstrate increased synapse elimination in patient-derived neural cultures and isolated synaptosomes. This excessive synaptic pruning reflects abnormalities in both microglia-like cells and synaptic structures. Further, we find that schizophrenia risk-associated variants within the human complement component 4 locus are associated with increased neuronal complement deposition and synapse uptake; however, they do not fully explain the observed increase in synapse uptake. Finally, we demonstrate that the antibiotic minocycline reduces microglia-mediated synapse uptake in vitro and its use is associated with a modest decrease in incident schizophrenia risk compared to other antibiotics in a cohort of young adults drawn from electronic health records. These findings point to excessive pruning as a potential target for delaying or preventing the onset of schizophrenia in high-risk individuals.


Assuntos
Microglia/fisiologia , Plasticidade Neuronal , Esquizofrenia/fisiopatologia , Sinapses/fisiologia , Adolescente , Adulto , Idoso , Antibacterianos/administração & dosagem , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Masculino , Microglia/efeitos dos fármacos , Pessoa de Meia-Idade , Minociclina/administração & dosagem , Células-Tronco Neurais/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Fatores de Risco , Sinapses/efeitos dos fármacos , Adulto Jovem
12.
Cell Rep ; 27(2): 616-630.e6, 2019 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-30970262

RESUMO

Human pluripotent stem cells (hPSCs) generate a variety of disease-relevant cells that can be used to improve the translation of preclinical research. Despite the potential of hPSCs, their use for genetic screening has been limited by technical challenges. We developed a scalable and renewable Cas9 and sgRNA-hPSC library in which loss-of-function mutations can be induced at will. Our inducible mutant hPSC library can be used for multiple genome-wide CRISPR screens in a variety of hPSC-induced cell types. As proof of concept, we performed three screens for regulators of properties fundamental to hPSCs: their ability to self-renew and/or survive (fitness), their inability to survive as single-cell clones, and their capacity to differentiate. We identified the majority of known genes and pathways involved in these processes, as well as a plethora of genes with unidentified roles. This resource will increase the understanding of human development and genetics. This approach will be a powerful tool to identify disease-modifying genes and pathways.


Assuntos
Sistemas CRISPR-Cas/genética , Testes Genéticos/métodos , Genoma/genética , Células-Tronco Pluripotentes/metabolismo , Humanos
13.
Nat Commun ; 9(1): 4307, 2018 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-30333485

RESUMO

Here we report Digital RNA with pertUrbation of Genes (DRUG-seq), a high-throughput platform for drug discovery. Pharmaceutical discovery relies on high-throughput screening, yet current platforms have limited readouts. RNA-seq is a powerful tool to investigate drug effects using transcriptome changes as a proxy, yet standard library construction is costly. DRUG-seq captures transcriptional changes detected in standard RNA-seq at 1/100th the cost. In proof-of-concept experiments profiling 433 compounds across 8 doses, transcription profiles generated from DRUG-seq successfully grouped compounds into functional clusters by mechanism of actions (MoAs) based on their intended targets. Perturbation differences reflected in transcriptome changes were detected for compounds engaging the same target, demonstrating the value of using DRUG-seq for understanding on and off-target activities. We demonstrate DRUG-seq captures common mechanisms, as well as differences between compound treatment and CRISPR on the same target. DRUG-seq provides a powerful tool for comprehensive transcriptome readout in a high-throughput screening environment.


Assuntos
Descoberta de Drogas/métodos , Perfilação da Expressão Gênica/métodos , Ensaios de Triagem em Larga Escala/métodos , Análise de Sequência de RNA , Linhagem Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Humanos
14.
Nat Med ; 24(7): 939-946, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29892062

RESUMO

CRISPR/Cas9 has revolutionized our ability to engineer genomes and conduct genome-wide screens in human cells1-3. Whereas some cell types are amenable to genome engineering, genomes of human pluripotent stem cells (hPSCs) have been difficult to engineer, with reduced efficiencies relative to tumour cell lines or mouse embryonic stem cells3-13. Here, using hPSC lines with stable integration of Cas9 or transient delivery of Cas9-ribonucleoproteins (RNPs), we achieved an average insertion or deletion (indel) efficiency greater than 80%. This high efficiency of indel generation revealed that double-strand breaks (DSBs) induced by Cas9 are toxic and kill most hPSCs. In previous studies, the toxicity of Cas9 in hPSCs was less apparent because of low transfection efficiency and subsequently low DSB induction3. The toxic response to DSBs was P53/TP53-dependent, such that the efficiency of precise genome engineering in hPSCs with a wild-type P53 gene was severely reduced. Our results indicate that Cas9 toxicity creates an obstacle to the high-throughput use of CRISPR/Cas9 for genome engineering and screening in hPSCs. Moreover, as hPSCs can acquire P53 mutations14, cell replacement therapies using CRISPR/Cas9-enginereed hPSCs should proceed with caution, and such engineered hPSCs should be monitored for P53 function.


Assuntos
Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Engenharia Genética , Células-Tronco Pluripotentes/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Quebras de DNA de Cadeia Dupla , Deleção de Genes , Humanos , RNA Guia de Cinetoplastídeos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica , Receptor fas/genética , Receptor fas/metabolismo
15.
Curr Biol ; 13(8): 680-5, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12699626

RESUMO

In addition to the canonical Wnt/beta-catenin signaling pathway, at least two noncanonical Wnt/Fz pathways have been described: the planar cell polarity (PCP) pathway in Drosophila [1] and the Wnt/calcium pathway in vertebrate embryos [2]. Recent work suggests that a vertebrate pathway homologous to the PCP pathway acts to regulate the convergent extension movements of gastrulation [3-7]. To further test this hypothesis, we have identified two zebrafish homologs of the Drosophila PCP gene prickle (pk) [8], both of which show discrete and dynamic expression patterns during gastrulation. Both gain and loss of pk1 function cause defects in convergent extension. Pk1 localizes to both the cytoplasm and the cell membrane, and its normal localization is partially dependent on its C-terminal prenylation motif. At the cell membrane, Pk1 is frequently localized asymmetrically around the cell and can colocalize with the signaling molecule Dishevelled (Dsh). In overexpression assays, Pk1 is able to activate AP-1-mediated transcription and inhibit activation of Wnt/beta-catenin signaling. Like noncanonical Wnts [9-10], overexpression of Pk1 increases the frequency of calcium transients in zebrafish blastulae. Our results support the idea that a vertebrate PCP pathway regulates gastrulation movements and suggest that there is overlap between the PCP and Wnt/calcium pathways.


Assuntos
Movimento Celular/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Gástrula/fisiologia , Transdução de Sinais/fisiologia , Peixe-Zebra/embriologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Movimento Celular/genética , Proteínas Desgrenhadas , Receptores Frizzled , Gástrula/metabolismo , Expressão Gênica , Glicoproteínas/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Proteínas com Domínio LIM , Fosfoproteínas/metabolismo , Proteínas/metabolismo , Proteínas Wnt , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra
16.
J Vis Exp ; (127)2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28994790

RESUMO

The recent emergence of Zika virus (ZIKV) in susceptible populations has led to an abrupt increase in microcephaly and other neurodevelopmental conditions in newborn infants. While mosquitos are the main route of viral transmission, it has also been shown to spread via sexual contact and vertical mother-to-fetus transmission. In this latter case of transmission, due to the unique viral tropism of ZIKV, the virus is believed to predominantly target the neural progenitor cells (NPCs) of the developing brain. Here a method for modeling ZIKV infection, and the resulting microcephaly, that occur when human cerebral organoids are exposed to live ZIKV is described. The organoids display high levels of virus within their neural progenitor population, and exhibit severe cell death and microcephaly over time. This three-dimensional cerebral organoid model allows researchers to conduct species-matched experiments to observe and potentially intervene with ZIKV infection of the developing human brain. The model provides improved relevance over standard two-dimensional methods, and contains human-specific cellular architecture and protein expression that are not possible in animal models.


Assuntos
Encéfalo/virologia , Células-Tronco Neurais/virologia , Organoides/virologia , Infecção por Zika virus/virologia , Encéfalo/patologia , Humanos , Células-Tronco Neurais/patologia
17.
Neurosci Lett ; 660: 109-114, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28923481

RESUMO

Triggering receptor expressed in myeloid cells (TREM2) is a member of the immunoglobulin superfamily and is expressed in macrophages, dendritic cells, microglia, and osteoclasts. TREM2 plays a role in phagocytosis, regulates release of cytokine, contributes to microglia maintenance, and its ectodomain is shed from the cell surface. Here, the question was addressed at which position sheddases cleave TREM2 and what are the proteases involved in this process. Using both pharmacological and genetic approaches we report that the main protease contributing to the release of TREM2 ectodomain is ADAM17, (a disintegrin and metalloproteinase domain containing protein, also called TACE, TNFα converting enzyme) while ADAM10 plays a minor role. Complementary biochemical experiments reveal that cleavage occurs between histidine 157 and serine 158. Shedding is not altered for the R47H-mutated TREM2 protein that confers an increased risk for the development of Alzheimers disease. These findings reveal a link between shedding of TREM2 and its regulation during inflammatory conditions or chronic neurodegenerative disease like AD in which activity or expression of sheddases might be altered.


Assuntos
Proteína ADAM17/metabolismo , Histidina/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo , Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Células CHO , Linhagem Celular , Cricetulus , Humanos , Proteínas de Membrana/metabolismo
18.
Cell Stem Cell ; 20(1): 120-134, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-28094016

RESUMO

During human brain development, multiple signaling pathways generate diverse cell types with varied regional identities. Here, we integrate single-cell RNA sequencing and clonal analyses to reveal lineage trees and molecular signals underlying early forebrain and mid/hindbrain cell differentiation from human embryonic stem cells (hESCs). Clustering single-cell transcriptomic data identified 41 distinct populations of progenitor, neuronal, and non-neural cells across our differentiation time course. Comparisons with primary mouse and human gene expression data demonstrated rostral and caudal progenitor and neuronal identities from early brain development. Bayesian analyses inferred a unified cell-type lineage tree that bifurcates between cortical and mid/hindbrain cell types. Two methods of clonal analyses confirmed these findings and further revealed the importance of Wnt/ß-catenin signaling in controlling this lineage decision. Together, these findings provide a rich transcriptome-based lineage map for studying human brain development and modeling developmental disorders.


Assuntos
Encéfalo/embriologia , Linhagem da Célula , Desenvolvimento Embrionário , Células-Tronco Embrionárias Humanas/citologia , Análise de Célula Única/métodos , Animais , Encéfalo/metabolismo , Linhagem Celular , Linhagem da Célula/genética , Células Clonais , Desenvolvimento Embrionário/genética , Humanos , Camundongos , Modelos Biológicos , Neurônios/citologia , Neurônios/metabolismo , Reprodutibilidade dos Testes , Análise de Sequência de RNA , Fatores de Transcrição/metabolismo , Transcriptoma/genética , Via de Sinalização Wnt/genética
19.
Cell Stem Cell ; 19(6): 703-708, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27912091

RESUMO

Zika virus (ZIKV) can cross the placental barrier, resulting in infection of the fetal brain and neurological defects including microcephaly. The cellular tropism of ZIKV and the identity of attachment factors used by the virus to gain access to key cell types involved in pathogenesis are under intense investigation. Initial studies suggested that ZIKV preferentially targets neural progenitor cells (NPCs), providing an explanation for the developmental phenotypes observed in some pregnancies. The AXL protein has been nominated as a key attachment factor for ZIKV in several cell types including NPCs. However, here we show that genetic ablation of AXL has no effect on ZIKV entry or ZIKV-mediated cell death in human induced pluripotent stem cell (iPSC)-derived NPCs or cerebral organoids. These findings call into question the utility of AXL inhibitors for preventing birth defects after infection and suggest that further studies of viral attachment factors in NPCs are needed.


Assuntos
Cérebro/patologia , Deleção de Genes , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/virologia , Neuroproteção , Organoides/virologia , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Infecção por Zika virus/prevenção & controle , Morte Celular , Técnicas de Inativação de Genes , Humanos , Células-Tronco Neurais/patologia , Organoides/metabolismo , Organoides/patologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Infecção por Zika virus/patologia , Receptor Tirosina Quinase Axl
20.
Nat Neurosci ; 19(12): 1743-1749, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27798629

RESUMO

A fundamental impediment to understanding the brain is the availability of inexpensive and robust methods for targeting and manipulating specific neuronal populations. The need to overcome this barrier is pressing because there are considerable anatomical, physiological, cognitive and behavioral differences between mice and higher mammalian species in which it is difficult to specifically target and manipulate genetically defined functional cell types. In particular, it is unclear the degree to which insights from mouse models can shed light on the neural mechanisms that mediate cognitive functions in higher species, including humans. Here we describe a novel recombinant adeno-associated virus that restricts gene expression to GABAergic interneurons within the telencephalon. We demonstrate that the viral expression is specific and robust, allowing for morphological visualization, activity monitoring and functional manipulation of interneurons in both mice and non-genetically tractable species, thus opening the possibility to study GABAergic function in virtually any vertebrate species.


Assuntos
Encéfalo/virologia , Dependovirus/isolamento & purificação , Neurônios GABAérgicos/virologia , Interneurônios/fisiologia , Vertebrados/virologia , Animais , Comportamento Animal , Encéfalo/metabolismo , Células Cultivadas , Dependovirus/genética , Feminino , Neurônios GABAérgicos/patologia , Vetores Genéticos/genética , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA