Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Methods ; 19(7): 812-822, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35710610

RESUMO

Transcription factor over-expression is a proven method for reprogramming cells to a desired cell type for regenerative medicine and therapeutic discovery. However, a general method for the identification of reprogramming factors to create an arbitrary cell type is an open problem. Here we examine the success rate of methods and data for differentiation by testing the ability of nine computational methods (CellNet, GarNet, EBseq, AME, DREME, HOMER, KMAC, diffTF and DeepAccess) to discover and rank candidate factors for eight target cell types with known reprogramming solutions. We compare methods that use gene expression, biological networks and chromatin accessibility data, and comprehensively test parameter and preprocessing of input data to optimize performance. We find the best factor identification methods can identify an average of 50-60% of reprogramming factors within the top ten candidates, and methods that use chromatin accessibility perform the best. Among the chromatin accessibility methods, complex methods DeepAccess and diffTF have higher correlation with the ranked significance of transcription factor candidates within reprogramming protocols for differentiation. We provide evidence that AME and diffTF are optimal methods for transcription factor recovery that will allow for systematic prioritization of transcription factor candidates to aid in the design of new reprogramming protocols.


Assuntos
Reprogramação Celular , Cromatina , Diferenciação Celular/genética , Reprogramação Celular/genética , Cromatina/genética , Regulação da Expressão Gênica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
Nucleic Acids Res ; 47(6): e35, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30953075

RESUMO

Chromatin interaction analysis by paired-end tag sequencing (ChIA-PET) is a method for the genome-wide de novo discovery of chromatin interactions. Existing computational methods typically fail to detect weak or dynamic interactions because they use a peak-calling step that ignores paired-end linkage information. We have developed a novel computational method called Chromatin Interaction Discovery (CID) to overcome this limitation with an unbiased clustering approach for interaction discovery. CID outperforms existing chromatin interaction detection methods with improved sensitivity, replicate consistency, and concordance with other chromatin interaction datasets. In addition, CID also outperforms other methods in discovering chromatin interactions from HiChIP data. We expect that the CID method will be valuable in characterizing 3D chromatin interactions and in understanding the functional consequences of disease-associated distal genetic variations.


Assuntos
Imunoprecipitação da Cromatina/métodos , Cromatina/química , Cromatina/metabolismo , Biologia Computacional/métodos , Análise de Sequência de DNA/métodos , Algoritmos , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/metabolismo , Conjuntos de Dados como Assunto , Etiquetas de Sequências Expressas , Humanos , Ligação Proteica
3.
Nat Methods ; 8(12): 1056-8, 2011 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-22081127

RESUMO

The study of developmentally regulated transcription factors by chromatin immunoprecipitation and deep sequencing (ChIP-seq) faces two major obstacles: availability of ChIP-grade antibodies and access to sufficient number of cells. We describe versatile genome-wide analysis of transcription-factor binding sites by combining directed differentiation of embryonic stem cells and inducible expression of tagged proteins. We demonstrate its utility by mapping DNA-binding sites of transcription factors involved in motor neuron specification.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/genética , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/metabolismo , Estudo de Associação Genômica Ampla , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Análise de Sequência de DNA
4.
Sci Immunol ; 8(88): eadg2979, 2023 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-37862432

RESUMO

Loss of RNA homeostasis underlies numerous neurodegenerative and neuroinflammatory diseases. However, the molecular mechanisms that trigger neuroinflammation are poorly understood. Viral double-stranded RNA (dsRNA) triggers innate immune responses when sensed by host pattern recognition receptors (PRRs) present in all cell types. Here, we report that human neurons intrinsically carry exceptionally high levels of immunostimulatory dsRNAs and identify long 3'UTRs as giving rise to neuronal dsRNA structures. We found that the neuron-enriched ELAVL family of genes (ELAVL2, ELAVL3, and ELAVL4) can increase (i) 3'UTR length, (ii) dsRNA load, and (iii) activation of dsRNA-sensing PRRs such as MDA5, PKR, and TLR3. In wild-type neurons, neuronal dsRNAs signaled through PRRs to induce tonic production of the antiviral type I interferon. Depleting ELAVL2 in WT neurons led to global shortening of 3'UTR length, reduced immunostimulatory dsRNA levels, and rendered WT neurons susceptible to herpes simplex virus and Zika virus infection. Neurons deficient in ADAR1, a dsRNA-editing enzyme mutated in the neuroinflammatory disorder Aicardi-Goutières syndrome, exhibited intolerably high levels of dsRNA that triggered PRR-mediated toxic inflammation and neuronal death. Depleting ELAVL2 in ADAR1 knockout neurons led to prolonged neuron survival by reducing immunostimulatory dsRNA levels. In summary, neurons are specialized cells where PRRs constantly sense "self" dsRNAs to preemptively induce protective antiviral immunity, but maintaining RNA homeostasis is paramount to prevent pathological neuroinflammation.


Assuntos
Infecção por Zika virus , Zika virus , Humanos , Regiões 3' não Traduzidas/genética , RNA de Cadeia Dupla , Doenças Neuroinflamatórias , Inflamação , Receptores de Reconhecimento de Padrão/genética , Neurônios
5.
Nat Commun ; 13(1): 5427, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36109497

RESUMO

Neurons born in the embryo can undergo a protracted period of maturation lasting well into postnatal life. How gene expression changes are regulated during maturation and whether they can be recapitulated in cultured neurons remains poorly understood. Here, we show that mouse motor neurons exhibit pervasive changes in gene expression and accessibility of associated regulatory regions from embryonic till juvenile age. While motifs of selector transcription factors, ISL1 and LHX3, are enriched in nascent regulatory regions, motifs of NFI factors, activity-dependent factors, and hormone receptors become more prominent in maturation-dependent enhancers. Notably, stem cell-derived motor neurons recapitulate ~40% of the maturation expression program in vitro, with neural activity playing only a modest role as a late-stage modulator. Thus, the genetic maturation program consists of a core hardwired subprogram that is correctly executed in vitro and an extrinsically-controlled subprogram that is dependent on the in vivo context of the maturing organism.


Assuntos
Neurônios Motores , Neurogênese , Animais , Hormônios/metabolismo , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Neurônios Motores/metabolismo , Neurogênese/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
6.
Neuron ; 110(1): 70-85.e6, 2022 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-34727520

RESUMO

Proper assembly and function of the nervous system requires the generation of a uniquely diverse population of neurons expressing a cell-type-specific combination of effector genes that collectively define neuronal morphology, connectivity, and function. How countless partially overlapping but cell-type-specific patterns of gene expression are controlled at the genomic level remains poorly understood. Here we show that neuronal genes are associated with highly complex gene regulatory systems composed of independent cell-type- and cell-stage-specific regulatory elements that reside in expanded non-coding genomic domains. Mapping enhancer-promoter interactions revealed that motor neuron enhancers are broadly distributed across the large chromatin domains. This distributed regulatory architecture is not a unique property of motor neurons but is employed throughout the nervous system. The number of regulatory elements increased dramatically during the transition from invertebrates to vertebrates, suggesting that acquisition of new enhancers might be a fundamental process underlying the evolutionary increase in cellular complexity.


Assuntos
Elementos Facilitadores Genéticos , Vertebrados , Animais , Cromatina/genética , Cromatina/metabolismo , Elementos Facilitadores Genéticos/genética , Genômica , Neurônios Motores/metabolismo , Vertebrados/genética
7.
Neuron ; 92(6): 1252-1265, 2016 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-27939581

RESUMO

Generic spinal motor neuron identity is established by cooperative binding of programming transcription factors (TFs), Isl1 and Lhx3, to motor-neuron-specific enhancers. How expression of effector genes is maintained following downregulation of programming TFs in maturing neurons remains unknown. High-resolution exonuclease (ChIP-exo) mapping revealed that the majority of enhancers established by programming TFs are rapidly deactivated following Lhx3 downregulation in stem-cell-derived hypaxial motor neurons. Isl1 is released from nascent motor neuron enhancers and recruited to new enhancers bound by clusters of Onecut1 in maturing neurons. Synthetic enhancer reporter assays revealed that Isl1 operates as an integrator factor, translating the density of Lhx3 or Onecut1 binding sites into transient enhancer activity. Importantly, independent Isl1/Lhx3- and Isl1/Onecut1-bound enhancers contribute to sustained expression of motor neuron effector genes, demonstrating that outwardly stable expression of terminal effector genes in postmitotic neurons is controlled by a dynamic relay of stage-specific enhancers.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Fator 6 Nuclear de Hepatócito/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Neurônios Motores/metabolismo , Neurogênese/genética , Fatores de Transcrição/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Imunoprecipitação da Cromatina , Regulação para Baixo , Elementos Facilitadores Genéticos , Camundongos , Células-Tronco Embrionárias Murinas , Proteínas do Tecido Nervoso/metabolismo
8.
PLoS One ; 10(5): e0122420, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25970635

RESUMO

RNA Polymerase II ChIA-PET data has revealed enhancers that are active in a profiled cell type and the genes that the enhancers regulate through chromatin interactions. The most commonly used computational method for analyzing ChIA-PET data, the ChIA-PET Tool, discovers interaction anchors at a spatial resolution that is insufficient to accurately identify individual enhancers. We introduce Germ, a computational method that estimates the likelihood that any two narrowly defined genomic locations are jointly occupied by RNA Polymerase II. Germ takes a blind deconvolution approach to simultaneously estimate the likelihood of RNA Polymerase II occupation as well as a model of the arrangement of read alignments relative to locations occupied by RNA Polymerase II. Both types of information are utilized to estimate the likelihood that RNA Polymerase II jointly occupies any two genomic locations. We apply Germ to RNA Polymerase II ChIA-PET data from embryonic stem cells to identify the genomic locations that are jointly occupied along with transcription start sites. We show that these genomic locations align more closely with features of active enhancers measured by ChIP-Seq than the locations identified using the ChIA-PET Tool. We also apply Germ to RNA Polymerase II ChIA-PET data from motor neuron progenitors. Based on the Germ results, we observe that a combination of cell type specific and cell type independent regulatory interactions are utilized by cells to regulate gene expression.


Assuntos
Células-Tronco Embrionárias/metabolismo , Elementos Facilitadores Genéticos , Genoma , Regiões Promotoras Genéticas , RNA Polimerase II/genética , Software , Animais , Cromatina/química , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Mapeamento Cromossômico/métodos , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica , Loci Gênicos , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Cultura Primária de Células , RNA Polimerase II/metabolismo , Sítio de Iniciação de Transcrição , Transcrição Gênica
9.
Nat Neurosci ; 16(9): 1219-27, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23872598

RESUMO

Efficient transcriptional programming promises to open new frontiers in regenerative medicine. However, mechanisms by which programming factors transform cell fate are unknown, preventing more rational selection of factors to generate desirable cell types. Three transcription factors, Ngn2, Isl1 and Lhx3, were sufficient to program rapidly and efficiently spinal motor neuron identity when expressed in differentiating mouse embryonic stem cells. Replacement of Lhx3 by Phox2a led to specification of cranial, rather than spinal, motor neurons. Chromatin immunoprecipitation-sequencing analysis of Isl1, Lhx3 and Phox2a binding sites revealed that the two cell fates were programmed by the recruitment of Isl1-Lhx3 and Isl1-Phox2a complexes to distinct genomic locations characterized by a unique grammar of homeodomain binding motifs. Our findings suggest that synergistic interactions among transcription factors determine the specificity of their recruitment to cell type-specific binding sites and illustrate how a single transcription factor can be repurposed to program different cell types.


Assuntos
Diferenciação Celular/fisiologia , Neurônios Motores/fisiologia , Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos , Células-Tronco Embrionárias , Expressão Gênica , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Antígeno Ki-67/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Neurônios Motores/citologia , Proteínas do Tecido Nervoso/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Estrutura Terciária de Proteína , Medula Espinal/citologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Fatores de Tempo , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa