Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 183(5): 1325-1339.e21, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33080218

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a recently identified coronavirus that causes the respiratory disease known as coronavirus disease 2019 (COVID-19). Despite the urgent need, we still do not fully understand the molecular basis of SARS-CoV-2 pathogenesis. Here, we comprehensively define the interactions between SARS-CoV-2 proteins and human RNAs. NSP16 binds to the mRNA recognition domains of the U1 and U2 splicing RNAs and acts to suppress global mRNA splicing upon SARS-CoV-2 infection. NSP1 binds to 18S ribosomal RNA in the mRNA entry channel of the ribosome and leads to global inhibition of mRNA translation upon infection. Finally, NSP8 and NSP9 bind to the 7SL RNA in the signal recognition particle and interfere with protein trafficking to the cell membrane upon infection. Disruption of each of these essential cellular functions acts to suppress the interferon response to viral infection. Our results uncover a multipronged strategy utilized by SARS-CoV-2 to antagonize essential cellular processes to suppress host defenses.


Asunto(s)
COVID-19/metabolismo , Interacciones Huésped-Patógeno , Biosíntesis de Proteínas , Empalme del ARN , SARS-CoV-2/metabolismo , Proteínas no Estructurales Virales/metabolismo , Células A549 , Animales , COVID-19/virología , Chlorocebus aethiops , Células HEK293 , Humanos , Interferones/metabolismo , Transporte de Proteínas , ARN Mensajero/metabolismo , ARN Ribosómico 18S/metabolismo , ARN Citoplasmático Pequeño/química , ARN Citoplasmático Pequeño/metabolismo , Partícula de Reconocimiento de Señal/química , Partícula de Reconocimiento de Señal/metabolismo , Células Vero , Proteínas no Estructurales Virales/química
2.
Cell ; 182(4): 872-885.e19, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32783915

RESUMEN

Cell function and activity are regulated through integration of signaling, epigenetic, transcriptional, and metabolic pathways. Here, we introduce INs-seq, an integrated technology for massively parallel recording of single-cell RNA sequencing (scRNA-seq) and intracellular protein activity. We demonstrate the broad utility of INs-seq for discovering new immune subsets by profiling different intracellular signatures of immune signaling, transcription factor combinations, and metabolic activity. Comprehensive mapping of Arginase 1-expressing cells within tumor models, a metabolic immune signature of suppressive activity, discovers novel Arg1+ Trem2+ regulatory myeloid (Mreg) cells and identifies markers, metabolic activity, and pathways associated with these cells. Genetic ablation of Trem2 in mice inhibits accumulation of intra-tumoral Mreg cells, leading to a marked decrease in dysfunctional CD8+ T cells and reduced tumor growth. This study establishes INs-seq as a broadly applicable technology for elucidating integrated transcriptional and intra-cellular maps and identifies the molecular signature of myeloid suppressive cells in tumors.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Neoplasias/patología , ARN Citoplasmático Pequeño/química , Receptores Inmunológicos/metabolismo , Animales , Arginasa/genética , Arginasa/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Neoplasias/metabolismo , ARN Citoplasmático Pequeño/metabolismo , Receptores Inmunológicos/genética , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Microambiente Tumoral , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
3.
Genome Res ; 34(3): 484-497, 2024 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-38580401

RESUMEN

Transcriptional regulation controls cellular functions through interactions between transcription factors (TFs) and their chromosomal targets. However, understanding the fate conversion potential of multiple TFs in an inducible manner remains limited. Here, we introduce iTF-seq as a method for identifying individual TFs that can alter cell fate toward specific lineages at a single-cell level. iTF-seq enables time course monitoring of transcriptome changes, and with biotinylated individual TFs, it provides a multi-omics approach to understanding the mechanisms behind TF-mediated cell fate changes. Our iTF-seq study in mouse embryonic stem cells identified multiple TFs that trigger rapid transcriptome changes indicative of differentiation within a day of induction. Moreover, cells expressing these potent TFs often show a slower cell cycle and increased cell death. Further analysis using bioChIP-seq revealed that GCM1 and OTX2 act as pioneer factors and activators by increasing gene accessibility and activating the expression of lineage specification genes during cell fate conversion. iTF-seq has utility in both mapping cell fate conversion and understanding cell fate conversion mechanisms.


Asunto(s)
Diferenciación Celular , Factores de Transcripción , Animales , Ratones , Diferenciación Celular/genética , Linaje de la Célula/genética , Perfilación de la Expresión Génica/métodos , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/citología , Multiómica , ARN Citoplasmático Pequeño/genética , ARN Citoplasmático Pequeño/metabolismo , RNA-Seq/métodos , Análisis de Secuencia de ARN/métodos , Análisis de Expresión Génica de una Sola Célula , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Transcriptoma
4.
Nat Rev Mol Cell Biol ; 16(12): 727-41, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26530390

RESUMEN

Plant genomes encode various small RNAs that function in distinct, yet overlapping, genetic and epigenetic silencing pathways. However, the abundance and diversity of small-RNA classes varies among plant species, suggesting coevolution between environmental adaptations and gene-silencing mechanisms. Biogenesis of small RNAs in plants is well understood, but we are just beginning to uncover their intricate regulation and activity. Here, we discuss the biogenesis of plant small RNAs, such as microRNAs, secondary siRNAs and heterochromatic siRNAs, and their diverse cellular and developmental functions, including in reproductive transitions, genomic imprinting and paramutation. We also discuss the diversification of small-RNA-directed silencing pathways through the expansion of RNA-dependent RNA polymerases, DICER proteins and ARGONAUTE proteins.


Asunto(s)
Arabidopsis/metabolismo , MicroARNs/metabolismo , ARN de Planta/metabolismo , ARN Citoplasmático Pequeño/metabolismo , ARN Interferente Pequeño/metabolismo , Arabidopsis/genética , Proteínas Argonautas/metabolismo , Regulación de la Expresión Génica de las Plantas , MicroARNs/genética , Interferencia de ARN , Empalme del ARN/genética , ARN de Planta/genética , ARN Citoplasmático Pequeño/genética , ARN Interferente Pequeño/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Ribonucleasa III/metabolismo
5.
Development ; 148(14)2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34184026

RESUMEN

Transcription factor 4 (TCF4) is a crucial regulator of neurodevelopment and has been linked to the pathogenesis of autism, intellectual disability and schizophrenia. As a class I bHLH transcription factor (TF), it is assumed that TCF4 exerts its neurodevelopmental functions through dimerization with proneural class II bHLH TFs. Here, we aim to identify TF partners of TCF4 in the control of interhemispheric connectivity formation. Using a new bioinformatic strategy integrating TF expression levels and regulon activities from single cell RNA-sequencing data, we find evidence that TCF4 interacts with non-bHLH TFs and modulates their transcriptional activity in Satb2+ intercortical projection neurons. Notably, this network comprises regulators linked to the pathogenesis of neurodevelopmental disorders, e.g. FOXG1, SOX11 and BRG1. In support of the functional interaction of TCF4 with non-bHLH TFs, we find that TCF4 and SOX11 biochemically interact and cooperatively control commissure formation in vivo, and regulate the transcription of genes implicated in this process. In addition to identifying new candidate interactors of TCF4 in neurodevelopment, this study illustrates how scRNA-Seq data can be leveraged to predict TF networks in neurodevelopmental processes.


Asunto(s)
ARN Citoplasmático Pequeño/metabolismo , Análisis de la Célula Individual , Factor de Transcripción 4/genética , Factor de Transcripción 4/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , ADN Helicasas , Embrión de Mamíferos , Factores de Transcripción Forkhead , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Discapacidad Intelectual , Proteínas de Unión a la Región de Fijación a la Matriz , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso , Neuronas/fisiología , Proteínas Nucleares , Dominios y Motivos de Interacción de Proteínas , ARN Citoplasmático Pequeño/genética , Factores de Transcripción SOXC , Esquizofrenia/genética , Esquizofrenia/metabolismo
6.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34880136

RESUMEN

Identification of type 1 innate lymphoid cells (ILC1s) has been problematic. The transcription factor Hobit encoded by Zfp683 has been proposed as a major driver of ILC1 programs. Using Zfp683 reporter mice, we showed that correlation of Hobit expression with ILC1s is tissue- and context-dependent. In liver and intestinal mucosa, Zfp683 expression correlated well with ILC1s; in salivary glands, Zfp683 was coexpressed with the natural killer (NK) master transcription factors Eomes and TCF1 in a unique cell population, which we call ILC1-like NK cells; during viral infection, Zfp683 was induced in conventional NK cells of spleen and liver. The impact of Zfp683 deletion on ILC1s and NK cells was also multifaceted, including a marked decrease in granzyme- and interferon-gamma (IFNγ)-producing ILC1s in the liver, slightly fewer ILC1s and more Eomes+ TCF1+ ILC1-like NK cells in salivary glands, and only reduced production of granzyme B by ILC1 in the intestinal mucosa. NK cell-mediated control of viral infection was unaffected. We conclude that Hobit has two major impacts on ILC1s: It sustains liver ILC1 numbers, while promoting ILC1 functional maturation in other tissues by controlling TCF1, Eomes, and granzyme expression.


Asunto(s)
Inmunidad Celular/fisiología , Inmunidad Innata/fisiología , Subgrupos Linfocitarios/clasificación , Subgrupos Linfocitarios/fisiología , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Animales , Antígenos CD , Biomarcadores , Eliminación de Gen , Regulación de la Expresión Génica/fisiología , Granzimas/genética , Granzimas/metabolismo , Interferón gamma/genética , Interferón gamma/metabolismo , Hígado/metabolismo , Proteínas de la Membrana/genética , Ratones , ARN Citoplasmático Pequeño/genética , ARN Citoplasmático Pequeño/metabolismo , RNA-Seq , Proteínas de Dominio T Box/genética , Factores de Transcripción/genética
7.
Biol Chem ; 404(11-12): 1123-1136, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37632732

RESUMEN

Small non-coding RNAs (sncRNA) are involved in many steps of the gene expression cascade and regulate processing and expression of mRNAs by the formation of ribonucleoprotein complexes (RNP) such as the RNA-induced silencing complex (RISC). By analyzing small RNA Seq data sets, we identified a sncRNA annotated as piR-hsa-1254, which is likely derived from the 3'-end of 7SL RNA2 (RN7SL2), herein referred to as snc7SL RNA. The 7SL RNA is an abundant long non-coding RNA polymerase III transcript and serves as structural component of the cytoplasmic signal recognition particle (SRP). To evaluate a potential functional role of snc7SL RNA, we aimed to define its cellular localization by live cell imaging. Therefore, a Molecular Beacon (MB)-based method was established to compare the subcellular localization of snc7SL RNA with its precursor 7SL RNA. We designed and characterized several MBs in vitro and tested those by live cell fluorescence microscopy. Using a multiplex approach, we show that 7SL RNA localizes mainly to the endoplasmic reticulum (ER), as expected for the SRP, whereas snc7SL RNA predominately localizes to the nucleus. This finding suggests a fundamentally different function of 7SL RNA and its derivate snc7SL RNA.


Asunto(s)
ARN Citoplasmático Pequeño , Partícula de Reconocimiento de Señal , Partícula de Reconocimiento de Señal/genética , ARN , ARN Citoplasmático Pequeño/genética , ARN Citoplasmático Pequeño/metabolismo , ARN Mensajero
8.
Brief Bioinform ; 22(4)2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-33201180

RESUMEN

The prevalence of dropout events is a serious problem for single-cell Hi-C (scHiC) data due to insufficient sequencing depth and data coverage, which brings difficulties in downstream studies such as clustering and structural analysis. Complicating things further is the fact that dropouts are confounded with structural zeros due to underlying properties, leading to observed zeros being a mixture of both types of events. Although a great deal of progress has been made in imputing dropout events for single cell RNA-sequencing (RNA-seq) data, little has been done in identifying structural zeros and imputing dropouts for scHiC data. In this paper, we adapted several methods from the single-cell RNA-seq literature for inference on observed zeros in scHiC data and evaluated their effectiveness. Through an extensive simulation study and real data analysis, we have shown that a couple of the adapted single-cell RNA-seq algorithms can be powerful for correctly identifying structural zeros and accurately imputing dropout values. Downstream analysis using the imputed values showed considerable improvement for clustering cells of the same types together over clustering results before imputation.


Asunto(s)
Algoritmos , Simulación por Computador , ARN Citoplasmático Pequeño , RNA-Seq , Análisis de la Célula Individual , Programas Informáticos , Humanos , ARN Citoplasmático Pequeño/genética , ARN Citoplasmático Pequeño/metabolismo
9.
Development ; 146(12)2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31249009

RESUMEN

Complex 3D tissues arise during development following tightly organized events in space and time. In particular, gene regulatory networks and local interactions between single cells lead to emergent properties at the tissue and organism levels. To understand the design principles of tissue organization, we need to characterize individual cells at given times, but we also need to consider the collective behavior of multiple cells across different spatial and temporal scales. In recent years, powerful single cell methods have been developed to characterize cells in tissues and to address the challenging questions of how different tissues are formed throughout development, maintained in homeostasis, and repaired after injury and disease. These approaches have led to a massive increase in data pertaining to both mRNA and protein abundances in single cells. As we review here, these new technologies, in combination with in toto live imaging, now allow us to bridge spatial and temporal information quantitatively at the single cell level and generate a mechanistic understanding of tissue development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Homeostasis , Regeneración , Análisis de la Célula Individual/métodos , Animales , Linaje de la Célula , Biología Evolutiva , Humanos , Hibridación Fluorescente in Situ , Ratones , Proteoma , ARN Mensajero/metabolismo , ARN Citoplasmático Pequeño/metabolismo
10.
Development ; 146(21)2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31582414

RESUMEN

The ability of men to remain fertile throughout their lives depends upon establishment of a spermatogonial stem cell (SSC) pool from gonocyte progenitors, and thereafter balancing SSC renewal versus terminal differentiation. Here, we report that precise regulation of the cell cycle is crucial for this balance. Whereas cyclin-dependent kinase 2 (Cdk2) is not necessary for mouse viability or gametogenesis stages prior to meiotic prophase I, mice bearing a deregulated allele (Cdk2Y15S ) are severely deficient in spermatogonial differentiation. This allele disrupts an inhibitory phosphorylation site (Tyr15) for the kinase WEE1. Remarkably, Cdk2Y15S/Y15S mice possess abnormal clusters of mitotically active SSC-like cells, but these are eventually removed by apoptosis after failing to differentiate properly. Analyses of lineage markers, germ cell proliferation over time, and single cell RNA-seq data revealed delayed and defective differentiation of gonocytes into SSCs. Biochemical and genetic data demonstrated that Cdk2Y15S is a gain-of-function allele causing elevated kinase activity, which underlies these differentiation defects. Our results demonstrate that precise regulation of CDK2 kinase activity in male germ cell development is crucial for the gonocyte-to-spermatogonia transition and long-term spermatogenic homeostasis.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Quinasa 2 Dependiente de la Ciclina/metabolismo , Células Germinativas/enzimología , Espermatogonias/citología , Alelos , Animales , Apoptosis , Sistemas CRISPR-Cas , Proliferación Celular , Análisis por Conglomerados , Cruzamientos Genéticos , Células Germinativas/citología , Heterocigoto , Homeostasis , Masculino , Espectrometría de Masas , Meiosis , Ratones , Mutagénesis Sitio-Dirigida , Fenotipo , Fosforilación , ARN Citoplasmático Pequeño/metabolismo , Túbulos Seminíferos/metabolismo , Espermatogénesis , Espermatogonias/metabolismo , Testículo/metabolismo , Transcriptoma
11.
PLoS Comput Biol ; 17(1): e1008585, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33428615

RESUMEN

Experimental single-cell approaches are becoming widely used for many purposes, including investigation of the dynamic behaviour of developing biological systems. Consequently, a large number of computational methods for extracting dynamic information from such data have been developed. One example is RNA velocity analysis, in which spliced and unspliced RNA abundances are jointly modeled in order to infer a 'direction of change' and thereby a future state for each cell in the gene expression space. Naturally, the accuracy and interpretability of the inferred RNA velocities depend crucially on the correctness of the estimated abundances. Here, we systematically compare five widely used quantification tools, in total yielding thirteen different quantification approaches, in terms of their estimates of spliced and unspliced RNA abundances in five experimental droplet scRNA-seq data sets. We show that there are substantial differences between the quantifications obtained from different tools, and identify typical genes for which such discrepancies are observed. We further show that these abundance differences propagate to the downstream analysis, and can have a large effect on estimated velocities as well as the biological interpretation. Our results highlight that abundance quantification is a crucial aspect of the RNA velocity analysis workflow, and that both the definition of the genomic features of interest and the quantification algorithm itself require careful consideration.


Asunto(s)
Biología Computacional/métodos , Perfilación de la Expresión Génica/métodos , ARN Mensajero , ARN Citoplasmático Pequeño , Análisis de Secuencia de ARN/métodos , Algoritmos , Animales , Bases de Datos Genéticas , Ratones , ARN Mensajero/análisis , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Citoplasmático Pequeño/análisis , ARN Citoplasmático Pequeño/genética , ARN Citoplasmático Pequeño/metabolismo , Análisis de la Célula Individual/métodos
12.
Nucleic Acids Res ; 48(1): 373-389, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31732748

RESUMEN

7SK RNA, as part of the 7SK ribonucleoprotein complex, is crucial to the regulation of transcription by RNA-polymerase II, via its interaction with the positive transcription elongation factor P-TEFb. The interaction is induced by binding of the protein HEXIM to the 5' hairpin (HP1) of 7SK RNA. Four distinct structural models have been obtained experimentally for HP1. Here, we employ computational methods to investigate the relative stability of these structures, transitions between them, and the effects of mutations on the observed structural ensembles. We further analyse the results with respect to mutational binding assays, and hypothesize a mechanism for HEXIM binding. Our results indicate that the dominant structure in the wild type exhibits a triplet involving the unpaired nucleotide U40 and the base pair A43-U66 in the GAUC/GAUC repeat. This conformation leads to an open major groove with enough potential binding sites for peptide recognition. Sequence mutations of the RNA change the relative stability of the different structural ensembles. Binding affinity is consequently lost if these changes alter the dominant structure.


Asunto(s)
Factor B de Elongación Transcripcional Positiva/química , ARN Polimerasa II/química , ARN Citoplasmático Pequeño/química , Proteínas de Unión al ARN/química , Factores de Transcripción/química , Transcripción Genética , Sitios de Unión , Humanos , Simulación de Dinámica Molecular , Conformación de Ácido Nucleico , Motivos de Nucleótidos , Péptidos/genética , Péptidos/metabolismo , Factor B de Elongación Transcripcional Positiva/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , Unión Proteica , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , ARN Citoplasmático Pequeño/genética , ARN Citoplasmático Pequeño/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Termodinámica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Mediators Inflamm ; 2022: 4955761, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35909659

RESUMEN

Interleukin- (IL-) 33 contributes to various inflammatory processes. IL-33/ST2 activation participates in systemic lupus erythematous via binding to the receptor of Suppression of Tumorigenicity 2 protein (ST2). However, whether IL-33/ST2 interferes with the nosogenesis of cutaneous lupus erythematosus (CLE) has not been reported so far. Herein, we proposed to disclose the impacts on IL-33/ST2 activation and Ro60 on CLE and their potential implications in the photosensitization of CLE cells. IL-33, ST2, and Ro60 in CLE patients' skin lesions were detected. Murine keratinocytes stimulated with or without IL-33 were irradiated by ultraviolet B (UVB), and the levels of Ro60 and inflammation markers were determined. Keratinocytes were cocultured with J774.2 macrophages and stimulated with IL-33 for analysis of chemostasis. The results identified that IL-33, ST2, and downstream inflammation markers were significantly upregulated in CLE lesions with Ro60 overexpression. Additionally, IL-33 treatment promoted the upregulation of Ro60 induced by UVB treatment in murine keratinocytes. Moreover, IL-33 stimulates keratinocytes to induce macrophage migration via enhancing the generation of the chemokine (C-C motif) ligands 17 and 22. Meanwhile, the silencing of ST2 or nuclear factor-kappa B (NF-κB) suppression abolished IL-33-induced upregulation of Ro60 in keratinocytes. Similarly, the inhibition of SOX17 expression was followed by downregulation of Ro60 in keratinocytes following IL-33 stimulation. In addition, UVB irradiation upregulated SOX17 in keratinocytes. Conclusively, the IL-33/ST2 axis interferes with Ro60-regulated photosensitization via activating the NF-κB- and PI3K/Akt- and SOX17-related pathways.


Asunto(s)
Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33 , Lupus Eritematoso Cutáneo , Animales , Autoantígenos/genética , Autoantígenos/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/genética , Interleucina-33/metabolismo , Queratinocitos/metabolismo , Lupus Eritematoso Cutáneo/complicaciones , Lupus Eritematoso Cutáneo/genética , Lupus Eritematoso Cutáneo/metabolismo , Ratones , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Trastornos por Fotosensibilidad/etiología , Trastornos por Fotosensibilidad/genética , Trastornos por Fotosensibilidad/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Citoplasmático Pequeño/genética , ARN Citoplasmático Pequeño/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Factores de Transcripción SOXF/metabolismo , Rayos Ultravioleta/efectos adversos
14.
Plant J ; 103(3): 1155-1173, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32369637

RESUMEN

In recent years, research has increasingly focused on the key role of post-transcriptional regulation of messenger ribonucleoprotein (mRNP) function and turnover. As a result of the complexity and dynamic nature of mRNPs, the full composition of a single mRNP complex remains unrevealed and mRNPs are poorly described in plants. Here we identify canonical Sm proteins as part of the cytoplasmic mRNP complex, indicating their function in the post-transcriptional regulation of gene expression in plants. Sm proteins comprise an evolutionarily ancient family of small RNA-binding proteins involved in pre-mRNA splicing. The latest research indicates that Sm could also impact on mRNA at subsequent stages of its life cycle. In this work we show that in the microsporocyte cytoplasm of Larix decidua, the European larch, Sm proteins accumulate within distinct cytoplasmic bodies, also containing polyadenylated RNA. To date, several types of cytoplasmic bodies involved in the post-transcriptional regulation of gene expression have been described, mainly in animal cells. Their role and molecular composition in plants remain less well established, however. A total of 222 mRNA transcripts have been identified as cytoplasmic partners for Sm proteins. The specific colocalization of these mRNAs with Sm proteins within cytoplasmic bodies has been confirmed via microscopic analysis. The results from this work support the hypothesis, that evolutionarily conserved Sm proteins have been adapted to perform a whole repertoire of functions related to the post-transcriptional regulation of gene expression in Eukaryota. This adaptation presumably enabled them to coordinate the interdependent processes of splicing element assembly, mRNA maturation and processing, and mRNA translation regulation, and its degradation.


Asunto(s)
Proteínas de Plantas/metabolismo , ARN de Planta/metabolismo , ARN Citoplasmático Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/metabolismo , Empalmosomas/metabolismo , Citoplasma/metabolismo , Regulación de la Expresión Génica de las Plantas , Larix/metabolismo , ARN Mensajero/metabolismo
15.
Biochem Biophys Res Commun ; 566: 53-58, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34116357

RESUMEN

The signal recognition particle (SRP) plays an essential role in protein translocation across biological membranes. Stable complexation of two GTPases in the signal recognition particle (SRP) and its receptor (SR) control the delivery of nascent polypeptide to the membrane translocon. In archaea, protein targeting is mediated by the SRP54/SRP19/7S RNA ribonucleoprotein complex (SRP) and the FtsY protein (SR). In the present study, using fluorescence resonance energy transfer (FRET), we demonstrate that archaeal 7S RNA stabilizes the SRP54·FtsY targeting complex (TC). Moreover, we show that archaeal SRP19 further assists 7S RNA in stabilizing the targeting complex (TC). These results suggest that archaeal 7S RNA and SRP19 modulate the conformation of the targeting complex and thereby reinforce TC to execute protein translocation via concomitant GTP hydrolysis.


Asunto(s)
Proteínas Arqueales/metabolismo , ARN Citoplasmático Pequeño/metabolismo , Partícula de Reconocimiento de Señal/metabolismo , Sulfolobus acidocaldarius/metabolismo , Guanosina Trifosfato/metabolismo , Hidrólisis , Modelos Moleculares
16.
PLoS Comput Biol ; 16(2): e1007644, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32069291

RESUMEN

Methods for the analysis of time series single cell expression data (scRNA-Seq) either do not utilize information about transcription factors (TFs) and their targets or only study these as a post-processing step. Using such information can both, improve the accuracy of the reconstructed model and cell assignments, while at the same time provide information on how and when the process is regulated. We developed the Continuous-State Hidden Markov Models TF (CSHMM-TF) method which integrates probabilistic modeling of scRNA-Seq data with the ability to assign TFs to specific activation points in the model. TFs are assumed to influence the emission probabilities for cells assigned to later time points allowing us to identify not just the TFs controlling each path but also their order of activation. We tested CSHMM-TF on several mouse and human datasets. As we show, the method was able to identify known and novel TFs for all processes, assigned time of activation agrees with both expression information and prior knowledge and combinatorial predictions are supported by known interactions. We also show that CSHMM-TF improves upon prior methods that do not utilize TF-gene interaction.


Asunto(s)
ARN Citoplasmático Pequeño/metabolismo , RNA-Seq , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Algoritmos , Animales , Inmunoprecipitación de Cromatina , Biología Computacional , Bases de Datos Factuales , Perfilación de la Expresión Génica , Humanos , Hígado/metabolismo , Pulmón/metabolismo , Cadenas de Markov , Ratones , Modelos Estadísticos , Probabilidad , Transcripción Genética
17.
RNA Biol ; 18(8): 1152-1159, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33103602

RESUMEN

Bacterial small-RNA (sRNA) sequences are functional RNAs, which play an important role in regulating the expression of a diverse class of genes. It is thus critical to identify such sRNA sequences and their probable mRNA targets. Here, we discuss new procedures to identify and characterize sRNA and their targets via the introduction of an integrated online platform 'PresRAT'. PresRAT uses the primary and secondary structural attributes of sRNA sequences to predict sRNA from a given sequence or bacterial genome. PresRAT also finds probable target mRNAs of sRNA sequences from a given bacterial chromosome and further concentrates on the identification of the probable sRNA-mRNA binding regions. Using PresRAT, we have identified a total of 66,209 potential sRNA sequences from 292 bacterial genomes and 2247 potential targets from 13 bacterial genomes. We have also implemented a protocol to build and refine 3D models of sRNA and sRNA-mRNA duplex regions and generated 3D models of 50 known sRNAs and 81 sRNA-mRNA duplexes using this platform. Along with the server part, PresRAT also contains a database section, which enlists the predicted sRNA sequences, sRNA targets, and their corresponding 3D models with structural dynamics information.


Asunto(s)
Bacterias/genética , ARN Bacteriano/química , ARN Mensajero/química , ARN Citoplasmático Pequeño/química , ARN Nuclear Pequeño/química , Programas Informáticos , Bacterias/metabolismo , Emparejamiento Base , Benchmarking , Cromosomas Bacterianos/química , Bases de Datos de Ácidos Nucleicos , Conformación de Ácido Nucleico , Hibridación de Ácido Nucleico , ARN Bacteriano/clasificación , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , ARN Mensajero/clasificación , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Citoplasmático Pequeño/genética , ARN Citoplasmático Pequeño/metabolismo , ARN Nuclear Pequeño/clasificación , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo
18.
J Immunol ; 202(1): 48-55, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30518570

RESUMEN

Given that diseases associated with anti-SSA/Ro autoantibodies, such as systemic lupus erythematosus and Sjögren syndrome, are linked with an upregulation of IFN and type I IFN-stimulated genes, including sialic acid-binding Ig-like lectin 1 (Siglec-1), a receptor on monocytes/macrophages, recent attention has focused on a potential role for IFN and IFN-stimulated genes in the pathogenesis of congenital heart block (CHB). Accordingly, three approaches were leveraged to address the association of IFN, IFN-stimulated genes, and the phenotype of macrophages in affected fetal cardiac tissue: 1) cultured healthy human macrophages transfected with hY3, an anti-SSA/Ro-associated ssRNA, 2) RNA isolated from freshly sorted human leukocytes/macrophages after Langendorff perfusion of three fetal hearts dying with CHB and three healthy gestational age-matched hearts, and 3) autopsy tissue from three additional human CHB hearts and one healthy heart. TLR ligation of macrophages with hY3 led to the upregulation of a panel of IFN transcripts, including SIGLEC1, a result corroborated using quantitative PCR. Using independent and agnostic bioinformatics approaches, CD45+CD11c+ and CD45+CD11c- human leukocytes flow sorted from the CHB hearts highly expressed type I IFN response genes inclusive of SIGLEC1. Furthermore, Siglec-1 expression was identified in the septal region of several affected fetal hearts. These data now provide a link between IFN, IFN-stimulated genes, and the inflammatory and possibly fibrosing components of CHB, positioning Siglec-1-positive macrophages as integral to the process.


Asunto(s)
Bloqueo Cardíaco/congénito , Tabiques Cardíacos/metabolismo , Lupus Eritematoso Sistémico/inmunología , Macrófagos/fisiología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/metabolismo , Síndrome de Sjögren/inmunología , Adulto , Anticuerpos Antinucleares/metabolismo , Autoantígenos/genética , Autoantígenos/metabolismo , Autoinmunidad , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Bloqueo Cardíaco/inmunología , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , ARN Citoplasmático Pequeño/genética , ARN Citoplasmático Pequeño/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Lectina 1 Similar a Ig de Unión al Ácido Siálico/genética
19.
Mol Cell ; 52(4): 517-28, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24184211

RESUMEN

Metazoan gene expression is often regulated after the recruitment of RNA polymerase II (Pol II) to promoters, through the controlled release of promoter-proximally paused Pol II into productive RNA synthesis. Despite the prevalence of paused Pol II, very little is known about the dynamics of these early elongation complexes or the fate of the short transcription start site-associated (tss) RNAs they produce. Here, we demonstrate that paused elongation complexes can be remarkably stable, with half-lives exceeding 15 min at genes with inefficient pause release. Promoter-proximal termination by Pol II is infrequent, and released tssRNAs are targeted for rapid degradation. Further, we provide evidence that the predominant tssRNA species observed are nascent RNAs held within early elongation complexes. We propose that stable pausing of polymerase provides a temporal window of opportunity for recruitment of factors to modulate gene expression and that the nascent tssRNA represents an appealing target for these interactions.


Asunto(s)
Proteínas de Drosophila/genética , ARN Polimerasa II/fisiología , ARN Citoplasmático Pequeño/metabolismo , Animales , Secuencia de Bases , Línea Celular , Cromatina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Regulación de la Expresión Génica , Regiones Promotoras Genéticas , Estabilidad del ARN , Transducción de Señal , Elongación de la Transcripción Genética
20.
Reprod Domest Anim ; 56(5): 801-811, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33624340

RESUMEN

The new technology of high-throughput single-cell RNA sequencing (10 × scRNA-seq) was developed recently with many advantages. However, it was not commonly used in farm animal research. There are few reports for the gene expression of goat ovarian follicle granulosa cells (GCs) during different developmental stages. In the current investigation, the gene expression of follicle GCs at different stages from two populations of Ji'ning grey goats: high litter size (HL; ≥3/L; 2 L) and low litter size (LL; ≤2 /L; 2 L) were analysed by scRNA-seq. Many GC marker genes were identified, and the pseudo-time showed that GCs developed during the time course which reflected the follicular development and differentiation trajectory. Moreover, the gene expression difference between the two populations HL versus LL was very clear at different developmental stages. Many marker genes differentially expressed at different developmental stages. ASIP and ASPN were found to be highly expressed in the early stage of GCs, INHA, INHBA, MFGE8 and HSD17B1 were identified to be highly expressed in the growing stage of GCs, while IGFBP2, IGFBP5 and CYP11A1 were found to be highly expressed in late stage. These marker genes could be used as reference genes of goat follicle GC development. This investigation for the first time discovered the gene expression patterns in goat follicle GCs in high- or low-fertility populations (based on litter size) by scRNA-seq which may be useful for uncovering the oocyte development potential.


Asunto(s)
Cabras/genética , Tamaño de la Camada/genética , Animales , Femenino , Fertilidad/genética , Perfilación de la Expresión Génica , Células de la Granulosa , Folículo Ovárico , ARN Citoplasmático Pequeño/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA