Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nature ; 582(7810): 109-114, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32494068

RESUMO

Advances in genetics and sequencing have identified a plethora of disease-associated and disease-causing genetic alterations. To determine causality between genetics and disease, accurate models for molecular dissection are required; however, the rapid expansion of transcriptional populations identified through single-cell analyses presents a major challenge for accurate comparisons between mutant and wild-type cells. Here we generate mouse models of human severe congenital neutropenia (SCN) using patient-derived mutations in the GFI1 transcription factor. To determine the effects of SCN mutations, we generated single-cell references for granulopoietic genomic states with linked epitopes1, aligned mutant cells to their wild-type equivalents and identified differentially expressed genes and epigenetic loci. We find that GFI1-target genes are altered sequentially, as cells go through successive states of differentiation. These insights facilitated the genetic rescue of granulocytic specification but not post-commitment defects in innate immune effector function, and underscore the importance of evaluating the effects of mutations and therapy within each relevant cell state.


Assuntos
Modelos Animais de Doenças , Células Precursoras de Granulócitos/patologia , Mutação , Neutropenia/genética , Neutropenia/patologia , Neutrófilos/patologia , Animais , Candida albicans/imunologia , Candida albicans/patogenicidade , Linhagem da Célula , Proteínas de Ligação a DNA/genética , Feminino , Humanos , Imunidade Inata , Masculino , Camundongos , Camundongos Transgênicos , Neutropenia/congênito , Neutropenia/imunologia , Neutrófilos/imunologia , Fatores de Transcrição/genética
2.
Nat Methods ; 18(3): 272-282, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33589839

RESUMO

The paired measurement of RNA and surface proteins in single cells with cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) is a promising approach to connect transcriptional variation with cell phenotypes and functions. However, combining these paired views into a unified representation of cell state is made challenging by the unique technical characteristics of each measurement. Here we present Total Variational Inference (totalVI; https://scvi-tools.org ), a framework for end-to-end joint analysis of CITE-seq data that probabilistically represents the data as a composite of biological and technical factors, including protein background and batch effects. To evaluate totalVI's performance, we profiled immune cells from murine spleen and lymph nodes with CITE-seq, measuring over 100 surface proteins. We demonstrate that totalVI provides a cohesive solution for common analysis tasks such as dimensionality reduction, the integration of datasets with different measured proteins, estimation of correlations between molecules and differential expression testing.


Assuntos
Linfonodos/metabolismo , Proteínas/análise , Análise de Célula Única/métodos , Baço/metabolismo , Transcriptoma/genética , Animais , Células Cultivadas , Análise de Dados , Feminino , Ensaios de Triagem em Larga Escala/métodos , Linfonodos/citologia , Camundongos , Camundongos Endogâmicos C57BL , RNA/análise , RNA/genética , Baço/citologia
3.
Nat Methods ; 18(8): 903-911, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34354295

RESUMO

The development of DNA-barcoded antibodies to tag cell surface molecules has enabled the use of droplet-based single-cell sequencing (dsc-seq) to profile protein abundances from thousands of cells simultaneously. As compared to flow and mass cytometry, the high per cell cost of current dsc-seq-based workflows precludes their use in clinical applications and large-scale pooled screens. Here, we introduce SCITO-seq, a workflow that uses splint oligonucleotides (oligos) to enable combinatorially indexed dsc-seq of DNA-barcoded antibodies from over 105 cells per reaction using commercial microfluidics. By encoding sample barcodes into splint oligos, we demonstrate that multiplexed SCITO-seq produces reproducible estimates of cellular composition and surface protein expression comparable to those from mass cytometry. We further demonstrate two modified splint oligo designs that extend SCITO-seq to achieve compatibility with commercial DNA-barcoded antibodies and simultaneous expression profiling of the transcriptome and surface proteins from the same cell. These results demonstrate SCITO-seq as a flexible and ultra-high-throughput platform for sequencing-based single-cell protein and multimodal profiling.


Assuntos
Citometria de Fluxo/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Microfluídica/métodos , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Transcriptoma , Estudos de Casos e Controles , Perfilação da Expressão Gênica , Humanos
4.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33833060

RESUMO

Parkinson's disease is characterized by accumulation of α-synuclein (αSyn). Release of oligomeric/fibrillar αSyn from damaged neurons may potentiate neuronal death in part via microglial activation. Heretofore, it remained unknown if oligomeric/fibrillar αSyn could activate the nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome in human microglia and whether anti-αSyn antibodies could prevent this effect. Here, we show that αSyn activates the NLRP3 inflammasome in human induced pluripotent stem cell (hiPSC)-derived microglia (hiMG) via dual stimulation involving Toll-like receptor 2 (TLR2) engagement and mitochondrial damage. In vitro, hiMG can be activated by mutant (A53T) αSyn secreted from hiPSC-derived A9-dopaminergic neurons. Surprisingly, αSyn-antibody complexes enhanced rather than suppressed inflammasome-mediated interleukin-1ß (IL-1ß) secretion, indicating these complexes are neuroinflammatory in a human context. A further increase in inflammation was observed with addition of oligomerized amyloid-ß peptide (Aß) and its cognate antibody. In vivo, engraftment of hiMG with αSyn in humanized mouse brain resulted in caspase-1 activation and neurotoxicity, which was exacerbated by αSyn antibody. These findings may have important implications for antibody therapies aimed at depleting misfolded/aggregated proteins from the human brain, as they may paradoxically trigger inflammation in human microglia.


Assuntos
Inflamassomos/metabolismo , Microglia/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Doença de Parkinson/imunologia , alfa-Sinucleína/imunologia , Peptídeos beta-Amiloides/imunologia , Anticorpos/imunologia , Diferenciação Celular , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Microglia/citologia , Receptor 2 Toll-Like/metabolismo , alfa-Sinucleína/genética
5.
Hum Mol Genet ; 25(R2): R182-R189, 2016 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-27439388

RESUMO

Next generation sequencing (NGS) has ignited an unprecedented pace of discovery in the biomedical sciences that is fundamentally transforming the way that we understand, diagnose and treat disease, and has motivated the belief that true precision medicine - medicine that is tailored to an individual's genetic, biochemical and exposure profile - will be a reality in the near term. With minimal sample requirement, NGS can enable the concurrent genome-wide study of genetic variations, transcriptomes, and certain epigenetic modifications. However, interrogating proteins as efficiently as DNA and RNA can be interrogated with NGS is lacking and this hampers more comprehensive views of molecular physiology and limits advances in biomedical science and precision medicine. The fact is that innovations in proteomic technologies pale in comparison to the advances in NGS, with current methodologies suffering from issues related to reproducibility, sensitivity, sample requirements, and limited multiplexing capacity. The development of proteomic technologies to overcome these limitations would fill the void in systems biology research, catalyze clinical innovations, and expedite the realization of precision medicine.

6.
Genome Res ; 25(8): 1091-103, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26055160

RESUMO

Duplication of the genome in mammalian cells occurs in a defined temporal order referred to as its replication-timing (RT) program. RT changes dynamically during development, regulated in units of 400-800 kb referred to as replication domains (RDs). Changes in RT are generally coordinated with transcriptional competence and changes in subnuclear position. We generated genome-wide RT profiles for 26 distinct human cell types, including embryonic stem cell (hESC)-derived, primary cells and established cell lines representing intermediate stages of endoderm, mesoderm, ectoderm, and neural crest (NC) development. We identified clusters of RDs that replicate at unique times in each stage (RT signatures) and confirmed global consolidation of the genome into larger synchronously replicating segments during differentiation. Surprisingly, transcriptome data revealed that the well-accepted correlation between early replication and transcriptional activity was restricted to RT-constitutive genes, whereas two-thirds of the genes that switched RT during differentiation were strongly expressed when late replicating in one or more cell types. Closer inspection revealed that transcription of this class of genes was frequently restricted to the lineage in which the RT switch occurred, but was induced prior to a late-to-early RT switch and/or down-regulated after an early-to-late RT switch. Analysis of transcriptional regulatory networks showed that this class of genes contains strong regulators of genes that were only expressed when early replicating. These results provide intriguing new insight into the complex relationship between transcription and RT regulation during human development.


Assuntos
Linhagem da Célula , Período de Replicação do DNA , Perfilação da Expressão Gênica/métodos , Células-Tronco Pluripotentes/fisiologia , Diferenciação Celular , Células Cultivadas , Análise por Conglomerados , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Genoma Humano , Humanos , Células-Tronco Pluripotentes/citologia
7.
Development ; 142(23): 4010-25, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26483210

RESUMO

Mechanisms of initial cell fate decisions differ among species. To gain insights into lineage allocation in humans, we derived ten human embryonic stem cell lines (designated UCSFB1-10) from single blastomeres of four 8-cell embryos and one 12-cell embryo from a single couple. Compared with numerous conventional lines from blastocysts, they had unique gene expression and DNA methylation patterns that were, in part, indicative of trophoblast competence. At a transcriptional level, UCSFB lines from different embryos were often more closely related than those from the same embryo. As predicted by the transcriptomic data, immunolocalization of EOMES, T brachyury, GDF15 and active ß-catenin revealed differential expression among blastomeres of 8- to 10-cell human embryos. The UCSFB lines formed derivatives of the three germ layers and CDX2-positive progeny, from which we derived the first human trophoblast stem cell line. Our data suggest heterogeneity among early-stage blastomeres and that the UCSFB lines have unique properties, indicative of a more immature state than conventional lines.


Assuntos
Blastômeros/citologia , Técnicas de Cultura Embrionária , Células-Tronco Embrionárias/citologia , Trofoblastos/citologia , Blastocisto/citologia , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Metilação de DNA , Endoderma/metabolismo , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Fator 15 de Diferenciação de Crescimento/metabolismo , Humanos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Células-Tronco Neurais/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Transcrição Gênica , Transcriptoma , beta Catenina/metabolismo
8.
Brain ; 140(3): 582-598, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28137726

RESUMO

New research suggests that common pathways are altered in many neurodevelopmental disorders including autism spectrum disorder; however, little is known about early molecular events that contribute to the pathology of these diseases. The study of monogenic, neurodevelopmental disorders with a high incidence of autistic behaviours, such as fragile X syndrome, has the potential to identify genes and pathways that are dysregulated in autism spectrum disorder as well as fragile X syndrome. In vitro generation of human disease-relevant cell types provides the ability to investigate aspects of disease that are impossible to study in patients or animal models. Differentiation of human pluripotent stem cells recapitulates development of the neocortex, an area affected in both fragile X syndrome and autism spectrum disorder. We have generated induced human pluripotent stem cells from several individuals clinically diagnosed with fragile X syndrome and autism spectrum disorder. When differentiated to dorsal forebrain cell fates, our fragile X syndrome human pluripotent stem cell lines exhibited reproducible aberrant neurogenic phenotypes. Using global gene expression and DNA methylation profiling, we have analysed the early stages of neurogenesis in fragile X syndrome human pluripotent stem cells. We discovered aberrant DNA methylation patterns at specific genomic regions in fragile X syndrome cells, and identified dysregulated gene- and network-level correlates of fragile X syndrome that are associated with developmental signalling, cell migration, and neuronal maturation. Integration of our gene expression and epigenetic analysis identified altered epigenetic-mediated transcriptional regulation of a distinct set of genes in fragile X syndrome. These fragile X syndrome-aberrant networks are significantly enriched for genes associated with autism spectrum disorder, giving support to the idea that underlying similarities exist among these neurodevelopmental diseases.


Assuntos
Diferenciação Celular/genética , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/patologia , Regulação da Expressão Gênica/genética , Modelos Biológicos , Células-Tronco Pluripotentes/fisiologia , Animais , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/patologia , Movimento Celular/genética , Células Cultivadas , Metilação de DNA/genética , Feto , Síndrome do Cromossomo X Frágil/genética , Redes Reguladoras de Genes , Humanos , Masculino , Camundongos , Neurogênese , Transfecção , Repetições de Trinucleotídeos/genética
9.
Nature ; 482(7384): 216-20, 2012 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-22278060

RESUMO

Our understanding of Alzheimer's disease pathogenesis is currently limited by difficulties in obtaining live neurons from patients and the inability to model the sporadic form of the disease. It may be possible to overcome these challenges by reprogramming primary cells from patients into induced pluripotent stem cells (iPSCs). Here we reprogrammed primary fibroblasts from two patients with familial Alzheimer's disease, both caused by a duplication of the amyloid-ß precursor protein gene (APP; termed APP(Dp)), two with sporadic Alzheimer's disease (termed sAD1, sAD2) and two non-demented control individuals into iPSC lines. Neurons from differentiated cultures were purified with fluorescence-activated cell sorting and characterized. Purified cultures contained more than 90% neurons, clustered with fetal brain messenger RNA samples by microarray criteria, and could form functional synaptic contacts. Virtually all cells exhibited normal electrophysiological activity. Relative to controls, iPSC-derived, purified neurons from the two APP(Dp) patients and patient sAD2 exhibited significantly higher levels of the pathological markers amyloid-ß(1-40), phospho-tau(Thr 231) and active glycogen synthase kinase-3ß (aGSK-3ß). Neurons from APP(Dp) and sAD2 patients also accumulated large RAB5-positive early endosomes compared to controls. Treatment of purified neurons with ß-secretase inhibitors, but not γ-secretase inhibitors, caused significant reductions in phospho-Tau(Thr 231) and aGSK-3ß levels. These results suggest a direct relationship between APP proteolytic processing, but not amyloid-ß, in GSK-3ß activation and tau phosphorylation in human neurons. Additionally, we observed that neurons with the genome of one sAD patient exhibited the phenotypes seen in familial Alzheimer's disease samples. More generally, we demonstrate that iPSC technology can be used to observe phenotypes relevant to Alzheimer's disease, even though it can take decades for overt disease to manifest in patients.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Neurônios/metabolismo , Idoso de 80 Anos ou mais , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Astrócitos/citologia , Biomarcadores/metabolismo , Células Cultivadas , Reprogramação Celular , Técnicas de Cocultura , Endossomos/metabolismo , Ativação Enzimática , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fragmentos de Peptídeos/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Proteólise , Sinapsinas/metabolismo , Proteínas tau/metabolismo
11.
Development ; 140(19): 3965-76, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24004950

RESUMO

The placenta is a transient organ that is necessary for proper fetal development. Its main functional component is the trophoblast, which is derived from extra-embryonic ectoderm. Little is known about early trophoblast differentiation in the human embryo, owing to lack of a proper in vitro model system. Human embryonic stem cells (hESCs) differentiate into functional trophoblast following BMP4 treatment in the presence of feeder-conditioned media; however, this model has not been widely accepted, in part owing to a lack of proof for a trophoblast progenitor population. We have previously shown that p63, a member of the p53 family of nuclear proteins, is expressed in proliferative cytotrophoblast (CTB), precursors to terminally differentiated syncytiotrophoblast (STB) in chorionic villi and extravillous trophoblast (EVT) at the implantation site. Here, we show that BMP4-treated hESCs differentiate into bona fide CTB by direct comparison with primary human placental tissues and isolated CTB through gene expression profiling. We show that, in primary CTB, p63 levels are reduced as cells differentiate into STB, and that forced expression of p63 maintains cyclin B1 and inhibits STB differentiation. We also establish that, similar to in vivo events, hESC differentiation into trophoblast is characterized by a p63(+)/KRT7(+) CTB stem cell state, followed by formation of functional KLF4(+) STB and HLA-G(+) EVT. Finally, we illustrate that downregulation of p63 by shRNA inhibits differentiation of hESCs into functional trophoblast. Taken together, our results establish that BMP4-treated hESCs are an excellent model of human trophoblast differentiation, closely mimicking the in vivo progression from p63(+) CTB stem cells to terminally differentiated trophoblast subtypes.


Assuntos
Proteína Morfogenética Óssea 4/farmacologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Trofoblastos/citologia , Trofoblastos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Citometria de Fluxo , Perfilação da Expressão Gênica , Idade Gestacional , Humanos , Técnicas In Vitro , Fator 4 Semelhante a Kruppel , Placenta/citologia , Gravidez , Reação em Cadeia da Polimerase em Tempo Real
12.
Circ Res ; 115(2): 311-24, 2014 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-24989490

RESUMO

The precise, temporal order of gene expression during development is critical to ensure proper lineage commitment, cell fate determination, and ultimately, organogenesis. Epigenetic regulation of chromatin structure is fundamental to the activation or repression of genes during embryonic development. In recent years, there has been an explosion of research relating to various modes of epigenetic regulation, such as DNA methylation, post-translational histone tail modifications, noncoding RNA control of chromatin structure, and nucleosome remodeling. Technological advances in genome-wide epigenetic profiling and pluripotent stem cell differentiation have been primary drivers for elucidating the epigenetic control of cellular identity during development and nuclear reprogramming. Not only do epigenetic mechanisms regulate transcriptional states in a cell-type-specific manner but also they establish higher order genomic topology and nuclear architecture. Here, we review the epigenetic control of pluripotency and changes associated with pluripotent stem cell differentiation. We focus on DNA methylation, DNA demethylation, and common histone tail modifications. Finally, we briefly discuss epigenetic heterogeneity among pluripotent stem cell lines and the influence of epigenetic patterns on genome topology.


Assuntos
Epigênese Genética/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células-Tronco Pluripotentes/citologia , Acetilação , Animais , Diferenciação Celular , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Dioxigenases/metabolismo , Elementos Facilitadores Genéticos/genética , Elementos Facilitadores Genéticos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Histonas/metabolismo , Humanos , Camundongos , Modelos Biológicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fosforilação , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/fisiologia , Processamento de Proteína Pós-Traducional , Inativação do Cromossomo X/genética , Inativação do Cromossomo X/fisiologia
13.
J Cell Sci ; 126(Pt 17): 3848-61, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23813959

RESUMO

The differentiation of human pluripotent stem cells (hPSCs) to insulin-expressing beta islet-like cells is a promising in vitro model system for studying the molecular signaling pathways underlying beta cell differentiation, as well as a potential source of cells for the treatment of type 1 diabetes. MicroRNAs (miRNAs) are a class of small non-coding RNAs that regulate many biological processes, including cellular differentiation. We studied the miRNA and mRNA expression profiles of hPSCs at five stages of in vitro differentiation along the pancreatic beta cell lineage (definitive endoderm, primitive gut tube, posterior foregut, pancreatic progenitor and hormone-expressing endocrine cells) in the context of samples of primary human fetal pancreas and purified adult islet cells using microarray analysis. Bioinformatic analysis of the resulting data identified a unique miRNA signature in differentiated beta islet cells, and predicted the effects of key miRNAs on mRNA expression. Many of the predicted miRNA-mRNA interactions involved mRNAs known to play key roles in the epithelial-mesenchymal transition process and pancreatic differentiation. We validated a subset of the predictions using qRT-PCR, luciferase reporter assays and western blotting, including the known interaction between miR-200 and ZEB2 (involved in epithelial-mesenchymal transition) and the novel interaction between miR-200 and SOX17 (a key transcription factor in specification of definitive endoderm). In addition, we found that miR-30d and let-7e, two miRNAs induced during differentiation, regulated the expression of RFX6, a transcription factor that directs pancreatic islet formation. These findings suggest that precise control of target mRNA expression by miRNAs ensures proper lineage specification during pancreatic development.


Assuntos
Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/metabolismo , MicroRNAs/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOXF/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem da Célula , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Diabetes Mellitus Tipo 1/terapia , Transição Epitelial-Mesenquimal/fisiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , MicroRNAs/genética , Pâncreas/embriologia , RNA Mensageiro/genética , Fatores de Transcrição de Fator Regulador X , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética , Fatores de Transcrição SOXF/biossíntese , Fatores de Transcrição SOXF/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Homeobox 2 de Ligação a E-box com Dedos de Zinco
14.
Ann Neurol ; 76(4): 489-508, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25159818

RESUMO

OBJECTIVE: To investigate whether a histone deacetylase inhibitor (HDACi) would be effective in an in vitro model for the neurodegenerative disease Friedreich ataxia (FRDA) and to evaluate safety and surrogate markers of efficacy in a phase I clinical trial in patients. METHODS: We used a human FRDA neuronal cell model, derived from patient induced pluripotent stem cells, to determine the efficacy of a 2-aminobenzamide HDACi (109) as a modulator of FXN gene expression and chromatin histone modifications. FRDA patients were dosed in 4 cohorts, ranging from 30mg/day to 240mg/day of the formulated drug product of HDACi 109, RG2833. Patients were monitored for adverse effects as well as for increases in FXN mRNA, frataxin protein, and chromatin modification in blood cells. RESULTS: In the neuronal cell model, HDACi 109/RG2833 increases FXN mRNA levels and frataxin protein, with concomitant changes in the epigenetic state of the gene. Chromatin signatures indicate that histone H3 lysine 9 is a key residue for gene silencing through methylation and reactivation through acetylation, mediated by the HDACi. Drug treatment in FRDA patients demonstrated increased FXN mRNA and H3 lysine 9 acetylation in peripheral blood mononuclear cells. No safety issues were encountered. INTERPRETATION: Drug exposure inducing epigenetic changes in neurons in vitro is comparable to the exposure required in patients to see epigenetic changes in circulating lymphoid cells and increases in gene expression. These findings provide a proof of concept for the development of an epigenetic therapy for this fatal neurological disease.


Assuntos
Ataxia de Friedreich/tratamento farmacológico , Ataxia de Friedreich/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/uso terapêutico , Proteínas de Ligação ao Ferro/genética , Administração Oral , Adolescente , Adulto , Aminocaproatos/farmacologia , Aminocaproatos/uso terapêutico , Área Sob a Curva , Benzamidas/farmacologia , Benzamidas/uso terapêutico , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Transformada , Imunoprecipitação da Cromatina , Estudos de Coortes , Estudos Transversais , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Ataxia de Friedreich/patologia , Regulação da Expressão Gênica/genética , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Células-Tronco Pluripotentes , Expansão das Repetições de Trinucleotídeos/genética , Adulto Jovem , Frataxina
15.
Nature ; 461(7260): 91-4, 2009 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-19672243

RESUMO

Recent landmark experiments have shown that transient overexpression of a small number of transcription factors can reprogram differentiated cells into induced pluripotent stem (iPS) cells that resemble embryonic stem (ES) cells. These iPS cells hold great promise for medicine because they have the potential to generate patient-specific cell types for cell replacement therapy and produce in vitro models of disease, without requiring embryonic tissues or oocytes. Although current iPS cell lines resemble ES cells, they have not passed the most stringent test of pluripotency by generating full-term or adult mice in tetraploid complementation assays, raising questions as to whether they are sufficiently potent to generate all of the cell types in an organism. Whether this difference between iPS and ES cells reflects intrinsic limitations of direct reprogramming is not known. Here we report fertile adult mice derived entirely from iPS cells that we generated by inducible genetic reprogramming of mouse embryonic fibroblasts. Producing adult mice derived entirely from a reprogrammed fibroblast shows that all features of a differentiated cell can be restored to an embryonic level of pluripotency without exposure to unknown ooplasmic factors. Comparing these fully pluripotent iPS cell lines to less developmentally potent lines may reveal molecular markers of different pluripotent states. Furthermore, mice derived entirely from iPS cells will provide a new resource to assess the functional and genomic stability of cells and tissues derived from iPS cells, which is important to validate their utility in cell replacement therapy and research applications.


Assuntos
Envelhecimento , Células-Tronco Pluripotentes/fisiologia , Técnicas Reprodutivas , Animais , Desdiferenciação Celular , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Feminino , Fibroblastos/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células-Tronco Pluripotentes/citologia , Gravidez , Taxa de Sobrevida
16.
Genomics ; 104(5): 358-67, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25179373

RESUMO

5-hydroxymethylcytosine (5hmC), an oxidized derivative of 5-methylcytosine (5mC), has been implicated as an important epigenetic regulator of mammalian development. Current procedures use DNA sequencing methods to discriminate 5hmC from 5mC, limiting their accessibility to the scientific community. Here we report a method that combines TET-assisted bisulfite conversion with Illumina 450K DNA methylation arrays for a low-cost high-throughput approach that distinguishes 5hmC and 5mC signals at base resolution. Implementing this approach, termed "TAB-array", we assessed DNA methylation dynamics in the differentiation of human pluripotent stem cells into cardiovascular progenitors and neural precursor cells. With the ability to discriminate 5mC and 5hmC, we identified a large number of novel dynamically methylated genomic regions that are implicated in the development of these lineages. The increased resolution and accuracy afforded by this approach provides a powerful means to investigate the distinct contributions of 5mC and 5hmC in human development and disease.


Assuntos
5-Metilcitosina/metabolismo , Citosina/análogos & derivados , Células-Tronco Pluripotentes/metabolismo , Análise de Sequência de DNA/métodos , Diferenciação Celular , Células Cultivadas , Citosina/metabolismo , Metilação de DNA , Epigênese Genética , Humanos , Dados de Sequência Molecular , Mioblastos Cardíacos/metabolismo , Células-Tronco Neurais
17.
Nat Biotechnol ; 39(10): 1246-1258, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34083792

RESUMO

Recent technological advances have enabled massively parallel chromatin profiling with scATAC-seq (single-cell assay for transposase accessible chromatin by sequencing). Here we present ATAC with select antigen profiling by sequencing (ASAP-seq), a tool to simultaneously profile accessible chromatin and protein levels. Our approach pairs sparse scATAC-seq data with robust detection of hundreds of cell surface and intracellular protein markers and optional capture of mitochondrial DNA for clonal tracking, capturing three distinct modalities in single cells. ASAP-seq uses a bridging approach that repurposes antibody:oligonucleotide conjugates designed for existing technologies that pair protein measurements with single-cell RNA sequencing. Together with DOGMA-seq, an adaptation of CITE-seq (cellular indexing of transcriptomes and epitopes by sequencing) for measuring gene activity across the central dogma of gene regulation, we demonstrate the utility of systematic multi-omic profiling by revealing coordinated and distinct changes in chromatin, RNA and surface proteins during native hematopoietic differentiation and peripheral blood mononuclear cell stimulation and as a combinatorial decoder and reporter of multiplexed perturbations in primary T cells.


Assuntos
RNA-Seq/métodos , Análise de Célula Única/métodos , Diferenciação Celular , Linhagem da Célula , Cromatina/genética , Cromatina/metabolismo , DNA Mitocondrial/genética , Epigenômica , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hematopoese , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Proteínas/genética , Proteínas/metabolismo , Linfócitos T/citologia , Linfócitos T/metabolismo
18.
NAR Genom Bioinform ; 2(2): lqaa025, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33575582

RESUMO

The development of single-cell transcriptomic technologies yields large datasets comprising multimodal informations, such as transcriptomes and immunophenotypes. Despite the current explosion of methods for pre-processing and integrating multimodal single-cell data, there is currently no user-friendly software to display easily and simultaneously both immunophenotype and transcriptome-based UMAP/t-SNE plots from the pre-processed data. Here, we introduce Single-Cell Virtual Cytometer, an open-source software for flow cytometry-like visualization and exploration of pre-processed multi-omics single cell datasets. Using an original CITE-seq dataset of PBMC from an healthy donor, we illustrate its use for the integrated analysis of transcriptomes and epitopes of functional maturation in human peripheral T lymphocytes. So this free and open-source algorithm constitutes a unique resource for biologists seeking for a user-friendly analytic tool for multimodal single cell datasets.

20.
Cell Rep ; 26(10): 2566-2579.e10, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30840882

RESUMO

In this report we describe a human pluripotent stem cell-derived vascular progenitor (MesoT) cell of the mesothelium lineage. MesoT cells are multipotent and generate smooth muscle cells, endothelial cells, and pericytes and self-assemble into vessel-like networks in vitro. MesoT cells transplanted into mechanically damaged neonatal mouse heart migrate into the injured tissue and contribute to nascent coronary vessels in the repair zone. When seeded onto decellularized vascular scaffolds, MesoT cells differentiate into the major vascular lineages and self-assemble into vasculature capable of supporting peripheral blood flow following transplantation. These findings demonstrate in vivo functionality and the potential utility of MesoT cells in vascular engineering applications.


Assuntos
Epitélio/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Medicina Regenerativa/métodos , Engenharia Tecidual/métodos , Linhagem da Célula , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA