Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell ; 158(4): 874-888, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25126791

RESUMEN

Stem cells are highly abundant during early development but become a rare population in most adult organs. The molecular mechanisms causing stem cells to exit proliferation at a specific time are not well understood. Here, we show that changes in energy metabolism induced by the steroid hormone ecdysone and the Mediator initiate an irreversible cascade of events leading to cell-cycle exit in Drosophila neural stem cells. We show that the timely induction of oxidative phosphorylation and the mitochondrial respiratory chain are required in neuroblasts to uncouple the cell cycle from cell growth. This results in a progressive reduction in neuroblast cell size and ultimately in terminal differentiation. Brain tumor mutant neuroblasts fail to undergo this shrinkage process and continue to proliferate until adulthood. Our findings show that cell size control can be modified by systemic hormonal signaling and reveal a unique connection between metabolism and proliferation in stem cells.


Asunto(s)
Proliferación Celular , Drosophila melanogaster/citología , Ecdisona/metabolismo , Células-Madre Neurales/citología , Animales , Tamaño de la Célula , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Metabolismo Energético , Genoma de los Insectos , Complejo Mediador/metabolismo , Células-Madre Neurales/metabolismo
2.
Cell ; 156(6): 1259-1273, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24630726

RESUMEN

Members of the SWI/SNF chromatin-remodeling complex are among the most frequently mutated genes in human cancer, but how they suppress tumorigenesis is currently unclear. Here, we use Drosophila neuroblasts to demonstrate that the SWI/SNF component Osa (ARID1) prevents tumorigenesis by ensuring correct lineage progression in stem cell lineages. We show that Osa induces a transcriptional program in the transit-amplifying population that initiates temporal patterning, limits self-renewal, and prevents dedifferentiation. We identify the Prdm protein Hamlet as a key component of this program. Hamlet is directly induced by Osa and regulates the progression of progenitors through distinct transcriptional states to limit the number of transit-amplifying divisions. Our data provide a mechanistic explanation for the widespread tumor suppressor activity of SWI/SNF. Because the Hamlet homologs Evi1 and Prdm16 are frequently mutated in cancer, this mechanism could well be conserved in human stem cell lineages. PAPERCLIP:


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Factores de Transcripción/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Regulación de la Expresión Génica , Genes Supresores de Tumor , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/genética , Transcripción Genética
3.
Nature ; 621(7978): 373-380, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37704762

RESUMEN

The development of the human brain involves unique processes (not observed in many other species) that can contribute to neurodevelopmental disorders1-4. Cerebral organoids enable the study of neurodevelopmental disorders in a human context. We have developed the CRISPR-human organoids-single-cell RNA sequencing (CHOOSE) system, which uses verified pairs of guide RNAs, inducible CRISPR-Cas9-based genetic disruption and single-cell transcriptomics for pooled loss-of-function screening in mosaic organoids. Here we show that perturbation of 36 high-risk autism spectrum disorder genes related to transcriptional regulation uncovers their effects on cell fate determination. We find that dorsal intermediate progenitors, ventral progenitors and upper-layer excitatory neurons are among the most vulnerable cell types. We construct a developmental gene regulatory network of cerebral organoids from single-cell transcriptomes and chromatin modalities and identify autism spectrum disorder-associated and perturbation-enriched regulatory modules. Perturbing members of the BRG1/BRM-associated factor (BAF) chromatin remodelling complex leads to enrichment of ventral telencephalon progenitors. Specifically, mutating the BAF subunit ARID1B affects the fate transition of progenitors to oligodendrocyte and interneuron precursor cells, a phenotype that we confirmed in patient-specific induced pluripotent stem cell-derived organoids. Our study paves the way for high-throughput phenotypic characterization of disease susceptibility genes in organoid models with cell state, molecular pathway and gene regulatory network readouts.


Asunto(s)
Trastorno del Espectro Autista , Encéfalo , Discapacidades del Desarrollo , Organoides , Análisis de Expresión Génica de una Sola Célula , Humanos , Trastorno del Espectro Autista/complicaciones , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Trastorno Autístico/complicaciones , Trastorno Autístico/genética , Trastorno Autístico/patología , Encéfalo/citología , Encéfalo/metabolismo , Linaje de la Célula/genética , Cromatina/genética , Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas , Discapacidades del Desarrollo/complicaciones , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Edición Génica , Mutación con Pérdida de Función , Mosaicismo , Neuronas/metabolismo , Neuronas/patología , Organoides/citología , Organoides/metabolismo , ARN Guía de Sistemas CRISPR-Cas , Transcripción Genética
4.
Hepatology ; 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38728662

RESUMEN

BACKGROUND AND AIMS: HEV is estimated to be responsible for 70,000 deaths annually, yet therapy options remain limited. In the pursuit of effective antiviral therapies, targeting viral entry holds promise and has proven effective for other viruses. However, the precise mechanisms and host factors required during HEV entry remain unclear. Cellular proteases have emerged as host factors required for viral surface protein activation and productive cell entry by many viruses. Hence, we investigated the functional requirement and therapeutic potential of cellular protease during HEV infection. APPROACH AND RESULTS: Using our established HEV cell culture model and subgenomic HEV replicons, we found that blocking lysosomal cathepsins (CTS) with small molecule inhibitors impedes HEV infection without affecting replication. Most importantly, the pan-cathepsin inhibitor K11777 suppressed HEV infections with an EC 50 of ~0.02 nM. Inhibition by K11777, devoid of notable toxicity in hepatoma cells, was also observed in HepaRG and primary human hepatocytes. Furthermore, through time-of-addition and RNAscope experiments, we confirmed that HEV entry is blocked by inhibition of cathepsins. Cathepsin L (CTSL) knockout cells were less permissive to HEV, suggesting that CTSL is critical for HEV infection. Finally, we observed cleavage of the glycosylated ORF2 protein and virus particles by recombinant CTSL. CONCLUSIONS: In summary, our study highlights the pivotal role of lysosomal cathepsins, especially CTSL, in the HEV entry process. The profound anti-HEV efficacy of the pan-cathepsin inhibitor K11777, especially with its notable safety profile in primary cells, further underscores its potential as a therapeutic candidate.

5.
Hepatology ; 77(6): 2104-2117, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36745934

RESUMEN

BACKGROUND AND AIMS: Being the most common cause of acute viral hepatitis with >20 million cases per year and 70,000 deaths annually, HEV presents a long-neglected and underinvestigated health burden. Although the entry process of viral particles is an attractive target for pharmacological intervention, druggable host factors to restrict HEV entry have not been identified so far. APPROACH AND RESULTS: Here we identify the EGF receptor (EGFR) as a novel host factor for HEV and reveal the significance of EGFR for the HEV entry process. By utilizing RNAi, chemical modulation with Food and Drug Administration-approved drugs, and ectopic expression of EGFR, we revealed that EGFR is critical for HEV infection without affecting HEV RNA replication or assembly of progeny virus. We further unveiled that EGFR itself and its ligand-binding domain, rather than its signaling function, is responsible for the proviral effect. Modulation of EGF expression in HepaRG cells and primary human hepatocytes affected HEV infection. CONCLUSIONS: Taken together, our study provides novel insights into the life cycle of HEV and identified EGFR as a possible target for future antiviral strategies against HEV.


Asunto(s)
Virus de la Hepatitis E , Hepatocitos , Humanos , Hepatocitos/metabolismo , Antivirales/farmacología , Receptores ErbB/metabolismo , Interferencia de ARN , Transducción de Señal , Virus de la Hepatitis E/genética , Replicación Viral
7.
Nat Methods ; 17(7): 708-716, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32514112

RESUMEN

CRISPR-Cas9 screens have emerged as a transformative approach to systematically probe gene functions. The quality and success of these screens depends on the frequencies of loss-of-function alleles, particularly in negative-selection screens widely applied for probing essential genes. Using optimized screening workflows, we performed essentialome screens in cancer cell lines and embryonic stem cells and achieved dropout efficiencies that could not be explained by common frameshift frequencies. We find that these superior effect sizes are mainly determined by the impact of in-frame mutations on protein function, which can be predicted based on amino acid composition and conservation. We integrate protein features into a 'Bioscore' and fuse it with improved predictors of single-guide RNA activity and indel formation to establish a score that captures all relevant processes in CRISPR-Cas9 mutagenesis. This Vienna Bioactivity CRISPR score (www.vbc-score.org) outperforms previous prediction tools and enables the selection of sgRNAs that effectively produce loss-of-function alleles.


Asunto(s)
Alelos , Sistemas CRISPR-Cas/genética , ARN Guía de Kinetoplastida/genética , Animales , Benchmarking , Proteína 9 Asociada a CRISPR/genética , Conjuntos de Datos como Asunto , Humanos , Ratones , Mutación
8.
Nat Immunol ; 12(7): 624-30, 2011 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-21642987

RESUMEN

Antiviral innate immunity relies on the recognition of microbial structures. One such structure is viral RNA that carries a triphosphate group on its 5' terminus (PPP-RNA). By an affinity proteomics approach with PPP-RNA as the 'bait', we found that the antiviral protein IFIT1 (interferon-induced protein with tetratricopeptide repeats 1) mediated binding of a larger protein complex containing other IFIT family members. IFIT1 bound PPP-RNA with nanomolar affinity and required the arginine at position 187 in a highly charged carboxy-terminal groove of the protein. In the absence of IFIT1, the growth and pathogenicity of viruses containing PPP-RNA was much greater. In contrast, IFIT proteins were dispensable for the clearance of pathogens that did not generate PPP-RNA. On the basis of this specificity and the great abundance of IFIT proteins after infection, we propose that the IFIT complex antagonizes viruses by sequestering specific viral nucleic acids.


Asunto(s)
Arginina/inmunología , Proteínas Portadoras/inmunología , ARN Viral/inmunología , Virus/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Arginina/química , Arginina/genética , Proteínas Portadoras/química , Proteínas Portadoras/genética , Femenino , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión al ARN
9.
Nature ; 550(7674): 114-118, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28953874

RESUMEN

The ability to directly uncover the contributions of genes to a given phenotype is fundamental for biology research. However, ostensibly homogeneous cell populations exhibit large clonal variance that can confound analyses and undermine reproducibility. Here we used genome-saturated mutagenesis to create a biobank of over 100,000 individual haploid mouse embryonic stem (mES) cell lines targeting 16,970 genes with genetically barcoded, conditional and reversible mutations. This Haplobank is, to our knowledge, the largest resource of hemi/homozygous mutant mES cells to date and is available to all researchers. Reversible mutagenesis overcomes clonal variance by permitting functional annotation of the genome directly in sister cells. We use the Haplobank in reverse genetic screens to investigate the temporal resolution of essential genes in mES cells, and to identify novel genes that control sprouting angiogenesis and lineage specification of blood vessels. Furthermore, a genome-wide forward screen with Haplobank identified PLA2G16 as a host factor that is required for cytotoxicity by rhinoviruses, which cause the common cold. Therefore, clones from the Haplobank combined with the use of reversible technologies enable high-throughput, reproducible, functional annotation of the genome.


Asunto(s)
Bancos de Muestras Biológicas , Genómica/métodos , Haploidia , Células Madre Embrionarias de Ratones/metabolismo , Mutación , Animales , Vasos Sanguíneos/citología , Linaje de la Célula/genética , Resfriado Común/genética , Resfriado Común/virología , Genes Esenciales/genética , Pruebas Genéticas , Células HEK293 , Homocigoto , Humanos , Ratones , Células Madre Embrionarias de Ratones/citología , Neovascularización Fisiológica/genética , Fosfolipasas A2 Calcio-Independiente/genética , Fosfolipasas A2 Calcio-Independiente/metabolismo , Rhinovirus/patogenicidad
10.
Mol Cell ; 59(2): 203-16, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26145176

RESUMEN

Uridylation of RNA species represents an emerging theme in post-transcriptional gene regulation. In the microRNA pathway, such modifications regulate small RNA biogenesis and stability in plants, worms, and mammals. Here, we report Tailor, an uridylyltransferase that is required for the majority of 3' end modifications of microRNAs in Drosophila and predominantly targets precursor hairpins. Uridylation modulates the characteristic two-nucleotide 3' overhang of microRNA hairpins, which regulates processing by Dicer-1 and destabilizes RNA hairpins. Tailor preferentially uridylates mirtron hairpins, thereby impeding the production of non-canonical microRNAs. Mirtron selectivity is explained by primary sequence specificity of Tailor, selecting substrates ending with a 3' guanosine. In contrast to mirtrons, conserved Drosophila precursor microRNAs are significantly depleted in 3' guanosine, thereby escaping regulatory uridylation. Our data support the hypothesis that evolutionary adaptation to Tailor-directed uridylation shapes the nucleotide composition of precursor microRNA 3' ends. Hence, hairpin uridylation may serve as a barrier for the de novo creation of microRNAs in Drosophila.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , MicroARNs/química , MicroARNs/metabolismo , ARN Nucleotidiltransferasas/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Femenino , Fertilidad/genética , Fertilidad/fisiología , Técnicas de Silenciamiento del Gen , Genes de Insecto , Masculino , MicroARNs/genética , Datos de Secuencia Molecular , Mutación , Conformación de Ácido Nucleico , ARN Nucleotidiltransferasas/antagonistas & inhibidores , ARN Nucleotidiltransferasas/genética , Procesamiento Postranscripcional del ARN , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Especificidad por Sustrato
11.
Proc Natl Acad Sci U S A ; 117(48): 30370-30379, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33199607

RESUMEN

Nibbler (Nbr) is a 3'-to-5' exoribonuclease whose catalytic 3'-end trimming activity impacts microRNA (miRNA) and PIWI-interacting RNA (piRNA) biogenesis. Here, we report on structural and functional studies to decipher the contributions of Nbr's N-terminal domain (NTD) and exonucleolytic domain (EXO) in miRNA 3'-end trimming. We have solved the crystal structures of the NTD core and EXO domains of Nbr, both in the apo-state. The NTD-core domain of Aedes aegypti Nbr adopts a HEAT-like repeat scaffold with basic patches constituting an RNA-binding surface exhibiting a preference for binding double-strand RNA (dsRNA) over single-strand RNA (ssRNA). Structure-guided functional assays in Drosophila S2 cells confirmed a principal role of the NTD in exonucleolytic miRNA trimming, which depends on basic surface patches. Gain-of-function experiments revealed a potential role of the NTD in recruiting Nbr to Argonaute-bound small RNA substrates. The EXO domain of A. aegypti and Drosophila melanogaster Nbr adopt a mixed α/ß-scaffold with a deep pocket lined by a DEDDy catalytic cleavage motif. We demonstrate that Nbr's EXO domain exhibits Mn2+-dependent ssRNA-specific 3'-to-5' exoribonuclease activity. Modeling of a 3' terminal Uridine into the catalytic pocket of Nbr EXO indicates that 2'-O-methylation of the 3'-U would result in a steric clash with a tryptophan side chain, suggesting that 2'-O-methylation protects small RNAs from Nbr-mediated trimming. Overall, our data establish that Nbr requires its NTD as a substrate recruitment platform to execute exonucleolytic miRNA maturation, catalyzed by the ribonuclease EXO domain.


Asunto(s)
Región de Flanqueo 3' , Proteínas de Drosophila/química , Exorribonucleasas/química , MicroARNs/química , MicroARNs/genética , Procesamiento Postranscripcional del ARN , Relación Estructura-Actividad , Animales , Proteínas Argonautas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Exorribonucleasas/metabolismo , MicroARNs/metabolismo , Modelos Biológicos , Modelos Moleculares , Conformación Molecular , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo
12.
Proc Natl Acad Sci U S A ; 117(3): 1731-1741, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31896581

RESUMEN

Hepatitis E virus (HEV) is the causative agent of hepatitis E in humans and the leading cause for acute viral hepatitis worldwide. The virus is classified as a member of the genus Orthohepevirus A within the Hepeviridae family. Due to the absence of a robust cell culture model for HEV infection, the analysis of the viral life cycle, the development of effective antivirals and a vaccine is severely limited. In this study, we established a protocol based on the HEV genotype 3 p6 (Kernow C-1) and the human hepatoma cell lines HepG2 and HepG2/C3A with different media conditions to produce intracellular HEV cell culture-derived particles (HEVcc) with viral titers between 105 and 106 FFU/mL. Viral titers could be further enhanced by an HEV variant harboring a mutation in the RNA-dependent RNA polymerase. These HEVcc particles were characterized in density gradients and allowed the trans-complementation of subgenomic reporter HEV replicons. In addition, in vitro produced intracellular-derived particles were infectious in liver-humanized mice with high RNA copy numbers detectable in serum and feces. Efficient infection of primary human and swine hepatocytes using the developed protocol could be observed and was inhibited by ribavirin. Finally, RNA sequencing studies of HEV-infected primary human hepatocytes demonstrated a temporally structured transcriptional defense response. In conclusion, this robust cell culture model of HEV infection provides a powerful tool for studying viral-host interactions that should facilitate the discovery of antiviral drugs for this important zoonotic pathogen.


Asunto(s)
Virus de la Hepatitis E/genética , Virus de la Hepatitis E/fisiología , Hepatitis E/metabolismo , Hepatocitos/virología , Animales , Antivirales/farmacología , Carcinoma Hepatocelular , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Genotipo , Células Hep G2 , Hepatitis E/virología , Virus de la Hepatitis E/efectos de los fármacos , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Ratones , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Replicón , Ribavirina/metabolismo , Porcinos , Carga Viral , Replicación Viral
13.
Development ; 146(23)2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31748204

RESUMEN

During central nervous system development, spatiotemporal gene expression programs mediate specific lineage decisions to generate neuronal and glial cell types from neural stem cells (NSCs). However, little is known about the epigenetic landscape underlying these highly complex developmental events. Here, we perform ChIP-seq on distinct subtypes of Drosophila FACS-purified NSCs and their differentiated progeny to dissect the epigenetic changes accompanying the major lineage decisions in vivo By analyzing active and repressive histone modifications, we show that stem cell identity genes are silenced during differentiation by loss of their activating marks and not via repressive histone modifications. Our analysis also uncovers a new set of genes specifically required for altering lineage patterns in type II neuroblasts (NBs), one of the two main Drosophila NSC identities. Finally, we demonstrate that this subtype specification in NBs, unlike NSC differentiation, requires Polycomb-group-mediated repression.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proteínas de Drosophila/metabolismo , Histonas/metabolismo , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Células-Madre Neurales/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Neoplasias Encefálicas/patología , Drosophila melanogaster , Células Madre Neoplásicas/patología , Células-Madre Neurales/patología
14.
Transfusion ; 62(11): 2200-2204, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36125237

RESUMEN

BACKGROUND AND OBJECTIVES: Photodynamic treatment with methylene blue (MB) and visible light is a well-established pathogen inactivation system for human plasma. This technique is routinely used in different countries. MB/light treatment was shown to inactivate several transfusion-transmittable viruses, but its efficiency for the inactivation of the quasi-enveloped hepatitis E virus (HEV) has not yet been investigated. MATERIALS AND METHODS: Plasma units were spiked with cell culture-derived HEV and treated with the THERAFLEX MB-Plasma system using various light doses (30, 60, 90, and 120 J/cm2 ). HEV titers in pre- and post-treatment samples were determined by virus titration and a large-volume plating assay to improve the detection limit of the virus assay. RESULTS: THERAFLEX MB-Plasma efficiently inactivated HEV in human plasma. Even the lowest light dose of 30 J/cm2 inactivated HEV down to the limit of detection, with a mean log reduction factor of greater than 2.4 for the total process. CONCLUSION: Our study demonstrates that the THERAFLEX MB-Plasma system effectively inactivates HEV in human plasma.


Asunto(s)
Virus de la Hepatitis E , Azul de Metileno , Humanos , Azul de Metileno/farmacología , Inactivación de Virus , Rayos Ultravioleta , Luz
15.
Nat Methods ; 15(8): 631-639, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30038414

RESUMEN

Brain tumors are among the most lethal and devastating cancers. Their study is limited by genetic heterogeneity and the incompleteness of available laboratory models. Three-dimensional organoid culture models offer innovative possibilities for the modeling of human disease. Here we establish a 3D in vitro model called a neoplastic cerebral organoid (neoCOR), in which we recapitulate brain tumorigenesis by introducing oncogenic mutations in cerebral organoids via transposon- and CRISPR-Cas9-mediated mutagenesis. By screening clinically relevant mutations identified in cancer genome projects, we defined mutation combinations that result in glioblastoma-like and central nervous system primitive neuroectodermal tumor (CNS-PNET)-like neoplasms. We demonstrate that neoCORs are suitable for use in investigations of aspects of tumor biology such as invasiveness, and for evaluation of drug effects in the context of specific DNA aberrations. NeoCORs will provide a valuable complement to the current basic and preclinical models used to study brain tumor biology.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Organoides/patología , Animales , Modelos Animales de Enfermedad , Genes myc , Ingeniería Genética , Glioblastoma/genética , Glioblastoma/patología , Xenoinjertos , Células Madre Embrionarias Humanas , Humanos , Masculino , Ratones , Ratones Desnudos , Mutación , Tumores Neuroectodérmicos Primitivos/genética , Tumores Neuroectodérmicos Primitivos/patología , Oncogenes , Transcriptoma , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Nat Methods ; 15(9): 748, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30135560

RESUMEN

In the originally published paper, the "before" image for the afatinib condition in Fig. 6c was incorrect. Instead of an image displaying a GBM-3 neoplastic organoid before afatinib treatment, this panel showed an image from the GBM-2 control (DMSO) group before treatment. This error has now been corrected in the HTML and PDF versions of the article; the "before, afatinib" panel in Fig. 6c now shows a representative image from the indicated experiment. The color of all error bars in Fig. 6 has also been changed to black, for consistency. All statistical analysis and all conclusions presented in the article are unaffected by this error. Nevertheless, we apologize for the mistake.

17.
Nat Methods ; 15(4): 283-289, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29481550

RESUMEN

MicroRNAs (miRNAs) play an essential role in the post-transcriptional regulation of animal development and physiology. However, in vivo studies aimed at linking miRNA function to the biology of distinct cell types within complex tissues remain challenging, partly because in vivo miRNA-profiling methods lack cellular resolution. We report microRNome by methylation-dependent sequencing (mime-seq), an in vivo enzymatic small-RNA-tagging approach that enables high-throughput sequencing of tissue- and cell-type-specific miRNAs in animals. The method combines cell-type-specific 3'-terminal 2'-O-methylation of animal miRNAs by a genetically encoded, plant-specific methyltransferase (HEN1), with chemoselective small-RNA cloning and high-throughput sequencing. We show that mime-seq uncovers the miRNomes of specific cells within Caenorhabditis elegans and Drosophila at unprecedented specificity and sensitivity, enabling miRNA profiling with single-cell resolution in whole animals. Mime-seq overcomes current challenges in cell-type-specific small-RNA profiling and provides novel entry points for understanding the function of miRNAs in spatially restricted physiological settings.


Asunto(s)
Caenorhabditis elegans/genética , Drosophila/genética , MicroARNs/genética , Análisis de Secuencia de ARN/métodos , Animales , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Clonación Molecular , Neuronas/metabolismo
18.
EMBO J ; 35(22): 2417-2434, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27729457

RESUMEN

The posttranscriptional addition of nucleotides to the 3' end of RNA regulates the maturation, function, and stability of RNA species in all domains of life. Here, we show that in flies, 3' terminal RNA uridylation triggers the processive, 3'-to-5' exoribonucleolytic decay via the RNase II/R enzyme CG16940, a homolog of the human Perlman syndrome exoribonuclease Dis3l2. Together with the TUTase Tailor, dmDis3l2 forms the cytoplasmic, terminal RNA uridylation-mediated processing (TRUMP) complex that functionally cooperates in the degradation of structured RNA RNA immunoprecipitation and high-throughput sequencing reveals a variety of TRUMP complex substrates, including abundant non-coding RNA, such as 5S rRNA, tRNA, snRNA, snoRNA, and the essential RNase MRP Based on genetic and biochemical evidence, we propose a key function of the TRUMP complex in the cytoplasmic quality control of RNA polymerase III transcripts. Together with high-throughput biochemical characterization of dmDis3l2 and bacterial RNase R, our results imply a conserved molecular function of RNase II/R enzymes as "readers" of destabilizing posttranscriptional marks-uridylation in eukaryotes and adenylation in prokaryotes-that play important roles in RNA surveillance.


Asunto(s)
Citoplasma/química , Citoplasma/metabolismo , Drosophila/metabolismo , Exorribonucleasas/metabolismo , Procesamiento Postranscripcional del ARN , Estabilidad del ARN , Animales , Línea Celular
19.
Development ; 144(21): 3932-3945, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28935704

RESUMEN

Stem cells need to balance self-renewal and differentiation for correct tissue development and homeostasis. Defects in this balance can lead to developmental defects or tumor formation. In recent years, mRNA splicing has emerged as an important mechanism regulating cell fate decisions. Here we address the role of the evolutionarily conserved splicing co-factor Barricade (Barc)/Tat-SF1/CUS2 in Drosophila neural stem cell (neuroblast) lineage formation. We show that Barc is required for the generation of neurons during Drosophila brain development by ensuring correct neural progenitor proliferation and differentiation. Barc associates with components of the U2 small nuclear ribonucleoprotein (snRNP) complex, and its depletion causes alternative splicing in the form of intron retention in a subset of genes. Using bioinformatics analysis and a cell culture-based splicing assay, we found that Barc-dependent introns share three major traits: they are short, GC rich and have weak 3' splice sites. Our results show that Barc, together with the U2 snRNP complex, plays an important role in regulating neural stem cell lineage progression during brain development and facilitates correct splicing of a subset of introns.


Asunto(s)
Ciclo Celular , Linaje de la Célula , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Factores de Transcripción/metabolismo , Empalme Alternativo/genética , Animales , Composición de Base/genética , Secuencia de Bases , Tipificación del Cuerpo/genética , Encéfalo/anatomía & histología , Recuento de Células , Proliferación Celular , Células Clonales , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Técnicas de Silenciamiento del Gen , Intrones/genética , Ratones , Modelos Biológicos , Mutación/genética , Neuronas/citología , Neuronas/metabolismo , Fenotipo , Unión Proteica , Interferencia de ARN , Sitios de Empalme de ARN/genética , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Factores de Tiempo
20.
Nat Methods ; 14(12): 1198-1204, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28945705

RESUMEN

Gene expression profiling by high-throughput sequencing reveals qualitative and quantitative changes in RNA species at steady state but obscures the intracellular dynamics of RNA transcription, processing and decay. We developed thiol(SH)-linked alkylation for the metabolic sequencing of RNA (SLAM seq), an orthogonal-chemistry-based RNA sequencing technology that detects 4-thiouridine (s4U) incorporation in RNA species at single-nucleotide resolution. In combination with well-established metabolic RNA labeling protocols and coupled to standard, low-input, high-throughput RNA sequencing methods, SLAM seq enabled rapid access to RNA-polymerase-II-dependent gene expression dynamics in the context of total RNA. We validated the method in mouse embryonic stem cells by showing that the RNA-polymerase-II-dependent transcriptional output scaled with Oct4/Sox2/Nanog-defined enhancer activity, and we provide quantitative and mechanistic evidence for transcript-specific RNA turnover mediated by post-transcriptional gene regulatory pathways initiated by microRNAs and N6-methyladenosine. SLAM seq facilitates the dissection of fundamental mechanisms that control gene expression in an accessible, cost-effective and scalable manner.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , ARN/genética , Compuestos de Sulfhidrilo/química , Alquilación , Células Madre Embrionarias/metabolismo , Redes Reguladoras de Genes , ARN/química , ARN Polimerasa II/genética , Procesamiento Postranscripcional del ARN , Tiouridina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA