Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Syst ; 15(2): 109-133.e10, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38335955

RESUMEN

Pluripotency can be induced in somatic cells by the expression of OCT4, KLF4, SOX2, and MYC. Usually only a rare subset of cells reprogram, and the molecular characteristics of this subset remain unknown. We apply retrospective clone tracing to identify and characterize the rare human fibroblasts primed for reprogramming. These fibroblasts showed markers of increased cell cycle speed and decreased fibroblast activation. Knockdown of a fibroblast activation factor identified by our analysis increased the reprogramming efficiency. We provide evidence for a unified model in which cells can move into and out of the primed state over time, explaining how reprogramming appears deterministic at short timescales and stochastic at long timescales. Furthermore, inhibiting the activity of LSD1 enlarged the pool of cells that were primed for reprogramming. Thus, even homogeneous cell populations can exhibit heritable molecular variability that can dictate whether individual rare cells will reprogram or not.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Estudios Retrospectivos , Fibroblastos
2.
Nature ; 620(7974): 651-659, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37468627

RESUMEN

Even among genetically identical cancer cells, resistance to therapy frequently emerges from a small subset of those cells1-7. Molecular differences in rare individual cells in the initial population enable certain cells to become resistant to therapy7-9; however, comparatively little is known about the variability in the resistance outcomes. Here we develop and apply FateMap, a framework that combines DNA barcoding with single-cell RNA sequencing, to reveal the fates of hundreds of thousands of clones exposed to anti-cancer therapies. We show that resistant clones emerging from single-cell-derived cancer cells adopt molecularly, morphologically and functionally distinct resistant types. These resistant types are largely predetermined by molecular differences between cells before drug addition and not by extrinsic factors. Changes in the dose and type of drug can switch the resistant type of an initial cell, resulting in the generation and elimination of certain resistant types. Samples from patients show evidence for the existence of these resistant types in a clinical context. We observed diversity in resistant types across several single-cell-derived cancer cell lines and cell types treated with a variety of drugs. The diversity of resistant types as a result of the variability in intrinsic cell states may be a generic feature of responses to external cues.


Asunto(s)
Antineoplásicos , Células Clonales , Resistencia a Antineoplásicos , Neoplasias , Humanos , Células Clonales/efectos de los fármacos , Células Clonales/metabolismo , Células Clonales/patología , Código de Barras del ADN Taxonómico , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , RNA-Seq , Análisis de Expresión Génica de una Sola Célula , Células Tumorales Cultivadas , Antineoplásicos/farmacología
3.
bioRxiv ; 2023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-37461472

RESUMEN

The ability of a virus to infect a cell type is at least in part determined by the presence of host factors required for the viral life cycle. However, even within cell types that express known factors needed for infection, not every cell is equally susceptible, suggesting that our knowledge of the full spectrum of factors that promote infection is incomplete. Profiling the most susceptible subsets of cells within a population may reveal additional factors that promote infection. However, because viral infection dramatically alters the state of the cell, new approaches are needed to reveal the state of these cells prior to infection with virus. Here, we used single-cell clone tracing to retrospectively identify and characterize lung epithelial cells that are highly susceptible to infection with SARS-CoV-2. The transcriptional state of these highly susceptible cells includes markers of retinoic acid signaling and epithelial differentiation. Loss of candidate factors identified by our approach revealed that many of these factors play roles in viral entry. Moreover, a subset of these factors exert control over the infectable cell state itself, regulating the expression of key factors associated with viral infection and entry. Analysis of patient samples revealed the heterogeneous expression of these factors across both cells and patients in vivo. Further, the expression of these factors is upregulated in particular inflammatory pathologies. Altogether, our results show that the variable expression of intrinsic cell states is a major determinant of whether a cell can be infected by SARS-CoV-2.

4.
bioRxiv ; 2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36798299

RESUMEN

Pluripotency can be induced in somatic cells by the expression of the four "Yamanaka" factors OCT4, KLF4, SOX2, and MYC. However, even in homogeneous conditions, usually only a rare subset of cells admit reprogramming, and the molecular characteristics of this subset remain unknown. Here, we apply retrospective clone tracing to identify and characterize the individual human fibroblast cells that are primed for reprogramming. These fibroblasts showed markers of increased cell cycle speed and decreased fibroblast activation. Knockdown of a fibroblast activation factor identified by our analysis led to increased reprogramming efficiency, identifying it as a barrier to reprogramming. Changing the frequency of reprogramming by inhibiting the activity of LSD1 led to an enlarging of the pool of cells that were primed for reprogramming. Our results show that even homogeneous cell populations can exhibit heritable molecular variability that can dictate whether individual rare cells will reprogram or not.

5.
Nat Struct Mol Biol ; 29(11): 1136-1144, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36369346

RESUMEN

Transcription at most promoters is divergent, initiating at closely spaced oppositely oriented core promoters to produce sense transcripts along with often unstable upstream antisense transcripts (uasTrx). How antisense transcription is regulated and to what extent it is coordinated with sense transcription is not well understood. Here, by combining acute degradation of the multi-functional transcription factor CTCF and nascent transcription measurements, we find that CTCF specifically suppresses antisense but not sense transcription at hundreds of divergent promoters. Primary transcript RNA-FISH shows that CTCF lowers burst fraction but not burst intensity of uasTrx and that co-bursting of sense and antisense transcripts is disfavored. Genome editing, chromatin conformation studies and high-resolution transcript mapping revealed that precisely positioned CTCF directly suppresses the initiation of uasTrx, in a manner independent of its architectural function. In sum, CTCF shapes the transcriptional landscape in part by suppressing upstream antisense transcription.


Asunto(s)
Cromatina , Transcripción Genética , Regiones Promotoras Genéticas , Cromatina/genética , ARN sin Sentido/genética , Regulación de la Expresión Génica
6.
Cell Syst ; 13(7): 547-560.e3, 2022 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-35705097

RESUMEN

Organoids recapitulate complex 3D organ structures and represent a unique opportunity to probe the principles of self-organization. While we can alter an organoid's morphology by manipulating the culture conditions, the morphology of an organoid often resembles that of its original organ, suggesting that organoid morphologies are governed by a set of tissue-specific constraints. Here, we establish a framework to identify constraints on an organoid's morphological features by quantifying them from microscopy images of organoids exposed to a range of perturbations. We apply this framework to Madin-Darby canine kidney cysts and show that they obey a number of constraints taking the form of scaling relationships or caps on certain parameters. For example, we found that the number, but not size, of cells increases with increasing cyst size. We also find that these constraints vary with cyst age and can be altered by varying the culture conditions. We observed similar sets of constraints in intestinal organoids. This quantitative framework for identifying constraints on organoid morphologies may inform future efforts to engineer organoids.


Asunto(s)
Quistes , Organoides , Animales , Perros , Fenotipo
7.
Cell Syst ; 12(9): 885-899.e8, 2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34352221

RESUMEN

Identifying the particular transcription factors that maintain cell type in vitro is important for manipulating cell type. Identifying such transcription factors by their cell-type-specific expression or their involvement in developmental regulation has had limited success. We hypothesized that because cell type is often resilient to perturbations, the transcriptional response to perturbations would reveal identity-maintaining transcription factors. We developed perturbation panel profiling (P3) as a framework for perturbing cells across many conditions and measuring gene expression responsiveness transcriptome-wide. In human iPSC-derived cardiac myocytes, P3 showed that transcription factors important for cardiac myocyte differentiation and maintenance were among the most frequently upregulated (most responsive). We reasoned that one function of responsive genes may be to maintain cellular identity. We identified responsive transcription factors in fibroblasts using P3 and found that suppressing their expression led to enhanced reprogramming. We propose that responsiveness to perturbations is a property of transcription factors that help maintain cellular identity in vitro. A record of this paper's transparent peer review process is included in the supplemental information.


Asunto(s)
Células Madre Pluripotentes Inducidas , Factores de Transcripción , Diferenciación Celular/genética , Fibroblastos/metabolismo , Humanos , Miocitos Cardíacos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Mol Cell ; 81(8): 1666-1681.e6, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33823140

RESUMEN

Nuclear speckles are prominent nuclear bodies that contain proteins and RNA involved in gene expression. Although links between nuclear speckles and gene activation are emerging, the mechanisms regulating association of genes with speckles are unclear. We find that speckle association of p53 target genes is driven by the p53 transcription factor. Focusing on p21, a key p53 target, we demonstrate that speckle association boosts expression by elevating nascent RNA amounts. p53-regulated speckle association did not depend on p53 transactivation functions but required an intact proline-rich domain and direct DNA binding, providing mechanisms within p53 for regulating gene-speckle association. Beyond p21, a substantial subset of p53 targets have p53-regulated speckle association. Strikingly, speckle-associating p53 targets are more robustly activated and occupy a distinct niche of p53 biology compared with non-speckle-associating p53 targets. Together, our findings illuminate regulated speckle association as a mechanism used by a transcription factor to boost gene expression.


Asunto(s)
Núcleo Celular/genética , Regulación de la Expresión Génica/genética , Proteínas Nucleares/genética , ARN/genética , Activación Transcripcional/genética , Proteína p53 Supresora de Tumor/genética , ADN/genética , Células HEK293 , Humanos , Cuerpos de Inclusión Intranucleares/genética , Unión Proteica/genética , Factores de Transcripción/genética , Transcripción Genética/genética
9.
Nat Methods ; 16(7): 633-639, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31235883

RESUMEN

Mammalian genomes are folded into tens of thousands of long-range looping interactions. The cause-and-effect relationship between looping and genome function is poorly understood, and the extent to which loops are dynamic on short time scales remains an unanswered question. Here, we engineer a new class of synthetic architectural proteins for directed rearrangement of the three-dimensional genome using blue light. We target our light-activated-dynamic-looping (LADL) system to two genomic anchors with CRISPR guide RNAs and induce their spatial colocalization via light-induced heterodimerization of cryptochrome 2 and a dCas9-CIBN fusion protein. We apply LADL to redirect a stretch enhancer (SE) away from its endogenous Klf4 target gene and to the Zfp462 promoter. Using single-molecule RNA-FISH, we demonstrate that de novo formation of the Zfp462-SE loop correlates with a modest increase in Zfp462 expression. LADL facilitates colocalization of genomic loci without exogenous chemical cofactors and will enable future efforts to engineer reversible and oscillatory loops on short time scales.


Asunto(s)
Regulación de la Expresión Génica , Ingeniería de Proteínas , Animales , Proteínas Portadoras/genética , Células Cultivadas , Proteínas de Unión al ADN , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Luz , Masculino , Ratones , Proteínas del Tejido Nervioso/genética , Regiones Promotoras Genéticas , ARN Guía de Kinetoplastida/genética
10.
Cancer Res ; 78(17): 4957-4970, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29976575

RESUMEN

The tumor microenvironment (TME) plays a major role in the pathogenesis of multiple cancer types, including upper-gastrointestinal (GI) cancers that currently lack effective therapeutic options. Cancer-associated fibroblasts (CAF) are an essential component of the TME, contributing to tumorigenesis by secreting growth factors, modifying the extracellular matrix, supporting angiogenesis, and suppressing antitumor immune responses. Through an unbiased approach, we have established that IL-6 mediates cross-talk between tumor cells and CAF not only by supporting tumor cell growth, but also by promoting fibroblast activation. As a result, IL-6 receptor (IL6Rα) and downstream effectors offer opportunities for targeted therapy in upper-GI cancers. IL-6 loss suppressed tumorigenesis in physiologically relevant three-dimensional (3D) organotypic and 3D tumoroid models and murine models of esophageal cancer. Tocilizumab, an anti-IL6Rα antibody, suppressed tumor growth in vivo in part via inhibition of STAT3 and MEK/ERK signaling. Analysis of a pan-cancer TCGA dataset revealed an inverse correlation between IL-6 and IL6Rα overexpression and patient survival. Therefore, we expanded evaluation of tocilizumab to head and neck squamous cell carcinoma patient-derived xenografts and gastric adenocarcinoma xenografts, demonstrating suppression of tumor growth and altered STAT3 and ERK1/2 gene signatures. We used small-molecule inhibitors of STAT3 and MEK1/2 signaling to suppress tumorigenesis in the 3D organotypic model of esophageal cancer. We demonstrate that IL6 is a major contributor to the dynamic cross-talk between tumor cells and CAF in the TME. Our findings provide a translational rationale for inhibition of IL6Rα and downstream signaling pathways as a novel targeted therapy in oral-upper-GI cancers.Significance: These findings demonstrate the interaction of esophageal cancer and cancer-associated fibroblasts through IL-6 signaling, providing rationale for a novel therapeutic approach to target these cancers. Cancer Res; 78(17); 4957-70. ©2018 AACR.


Asunto(s)
Neoplasias Esofágicas/genética , Neoplasias Gastrointestinales/genética , Interleucina-6/genética , Receptores de Interleucina-6/genética , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Carcinogénesis/genética , Línea Celular Tumoral , Neoplasias Esofágicas/patología , Neoplasias Gastrointestinales/patología , Humanos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Factor de Transcripción STAT3/genética , Transducción de Señal , Microambiente Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Proc Natl Acad Sci U S A ; 114(46): E9873-E9882, 2017 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-29078295

RESUMEN

RNAs have been shown to undergo transfer between mammalian cells, although the mechanism behind this phenomenon and its overall importance to cell physiology is not well understood. Numerous publications have suggested that RNAs (microRNAs and incomplete mRNAs) undergo transfer via extracellular vesicles (e.g., exosomes). However, in contrast to a diffusion-based transfer mechanism, we find that full-length mRNAs undergo direct cell-cell transfer via cytoplasmic extensions characteristic of membrane nanotubes (mNTs), which connect donor and acceptor cells. By employing a simple coculture experimental model and using single-molecule imaging, we provide quantitative data showing that mRNAs are transferred between cells in contact. Examples of mRNAs that undergo transfer include those encoding GFP, mouse ß-actin, and human Cyclin D1, BRCA1, MT2A, and HER2. We show that intercellular mRNA transfer occurs in all coculture models tested (e.g., between primary cells, immortalized cells, and in cocultures of immortalized human and murine cells). Rapid mRNA transfer is dependent upon actin but is independent of de novo protein synthesis and is modulated by stress conditions and gene-expression levels. Hence, this work supports the hypothesis that full-length mRNAs undergo transfer between cells through a refined structural connection. Importantly, unlike the transfer of miRNA or RNA fragments, this process of communication transfers genetic information that could potentially alter the acceptor cell proteome. This phenomenon may prove important for the proper development and functioning of tissues as well as for host-parasite or symbiotic interactions.


Asunto(s)
Comunicación Celular , Nanotubos , Transporte de Proteínas/fisiología , ARN Mensajero/fisiología , Actinina/genética , Actinina/metabolismo , Actinas/metabolismo , Animales , Comunicación Celular/genética , Línea Celular , Técnicas de Cocultivo , Ciclina D1/metabolismo , Exosomas/metabolismo , Fibroblastos , Regulación de la Expresión Génica/genética , Interacciones Huésped-Parásitos/fisiología , Humanos , Metalotioneína/metabolismo , Ratones , MicroARNs/genética , MicroARNs/fisiología , Biosíntesis de Proteínas/genética , Transporte de Proteínas/genética , Proteoma , ARN Mensajero/genética , Receptor ErbB-2/metabolismo , Simbiosis/fisiología , Ubiquitina-Proteína Ligasas/metabolismo
13.
Immunity ; 47(3): 435-449.e8, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28930659

RESUMEN

Commitment to the innate lymphoid cell (ILC) lineage is determined by Id2, a transcriptional regulator that antagonizes T and B cell-specific gene expression programs. Yet how Id2 expression is regulated in each ILC subset remains poorly understood. We identified a cis-regulatory element demarcated by a long non-coding RNA (lncRNA) that controls the function and lineage identity of group 1 ILCs, while being dispensable for early ILC development and homeostasis of ILC2s and ILC3s. The locus encoding this lncRNA, which we termed Rroid, directly interacted with the promoter of its neighboring gene, Id2, in group 1 ILCs. Moreover, the Rroid locus, but not the lncRNA itself, controlled the identity and function of ILC1s by promoting chromatin accessibility and deposition of STAT5 at the promoter of Id2 in response to interleukin (IL)-15. Thus, non-coding elements responsive to extracellular cues unique to each ILC subset represent a key regulatory layer for controlling the identity and function of ILCs.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad Innata/genética , Linfocitos/metabolismo , ARN Largo no Codificante/genética , Secuencias Reguladoras de Ácidos Nucleicos , Animales , Diferenciación Celular , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Ensamble y Desensamble de Cromatina , Femenino , Perfilación de la Expresión Génica , Sitios Genéticos , Homeostasis , Proteína 2 Inhibidora de la Diferenciación/genética , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Linfocitos/inmunología , Masculino , Ratones , Regiones Promotoras Genéticas , Factor de Transcripción STAT5/metabolismo , Transcripción Genética
14.
Nature ; 546(7658): 431-435, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28607484

RESUMEN

Therapies that target signalling molecules that are mutated in cancers can often have substantial short-term effects, but the emergence of resistant cancer cells is a major barrier to full cures. Resistance can result from secondary mutations, but in other cases there is no clear genetic cause, raising the possibility of non-genetic rare cell variability. Here we show that human melanoma cells can display profound transcriptional variability at the single-cell level that predicts which cells will ultimately resist drug treatment. This variability involves infrequent, semi-coordinated transcription of a number of resistance markers at high levels in a very small percentage of cells. The addition of drug then induces epigenetic reprogramming in these cells, converting the transient transcriptional state to a stably resistant state. This reprogramming begins with a loss of SOX10-mediated differentiation followed by activation of new signalling pathways, partially mediated by the activity of the transcription factors JUN and/or AP-1 and TEAD. Our work reveals the multistage nature of the acquisition of drug resistance and provides a framework for understanding resistance dynamics in single cells. We find that other cell types also exhibit sporadic expression of many of these same marker genes, suggesting the existence of a general program in which expression is displayed in rare subpopulations of cells.


Asunto(s)
Reprogramación Celular/efectos de los fármacos , Reprogramación Celular/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Melanoma/genética , Melanoma/patología , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética/efectos de los fármacos , Receptores ErbB/metabolismo , Femenino , Marcadores Genéticos/efectos de los fármacos , Marcadores Genéticos/genética , Humanos , Hibridación Fluorescente in Situ , Indoles/farmacología , Masculino , Proteínas Nucleares/metabolismo , Proteína Oncogénica p65(gag-jun)/metabolismo , Factores de Transcripción SOXE/deficiencia , Factores de Transcripción SOXE/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Análisis de la Célula Individual , Sulfonamidas/farmacología , Factores de Transcripción de Dominio TEA , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Vemurafenib , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Nature ; 537(7619): 239-243, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27525555

RESUMEN

Neutrophils, eosinophils and 'classical' monocytes collectively account for about 70% of human blood leukocytes and are among the shortest-lived cells in the body. Precise regulation of the lifespan of these myeloid cells is critical to maintain protective immune responses and minimize the deleterious consequences of prolonged inflammation. However, how the lifespan of these cells is strictly controlled remains largely unknown. Here we identify a long non-coding RNA that we termed Morrbid, which tightly controls the survival of neutrophils, eosinophils and classical monocytes in response to pro-survival cytokines in mice. To control the lifespan of these cells, Morrbid regulates the transcription of the neighbouring pro-apoptotic gene, Bcl2l11 (also known as Bim), by promoting the enrichment of the PRC2 complex at the Bcl2l11 promoter to maintain this gene in a poised state. Notably, Morrbid regulates this process in cis, enabling allele-specific control of Bcl2l11 transcription. Thus, in these highly inflammatory cells, changes in Morrbid levels provide a locus-specific regulatory mechanism that allows rapid control of apoptosis in response to extracellular pro-survival signals. As MORRBID is present in humans and dysregulated in individuals with hypereosinophilic syndrome, this long non-coding RNA may represent a potential therapeutic target for inflammatory disorders characterized by aberrant short-lived myeloid cell lifespan.


Asunto(s)
Proteína 11 Similar a Bcl2/genética , Células Mieloides/citología , Células Mieloides/metabolismo , ARN Largo no Codificante/genética , Alelos , Animales , Antígenos Ly/metabolismo , Apoptosis , Proteína 11 Similar a Bcl2/biosíntesis , Supervivencia Celular , Regulación hacia Abajo , Eosinófilos/citología , Eosinófilos/metabolismo , Femenino , Humanos , Masculino , Ratones , Monocitos/citología , Monocitos/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo , Regiones Promotoras Genéticas
16.
Nat Commun ; 7: 10865, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26936319

RESUMEN

Mesenchymal stem cells (MSCs) display substantial cell-to-cell heterogeneity, complicating their use in regenerative medicine. However, conventional bulk assays mask this variability. Here we show that both chondrocytes and chondrogenically induced MSCs exhibit substantial mRNA expression heterogeneity. Single-molecule RNA FISH to measure mRNA expression of differentiation markers in single cells reveals that sister cell pairs have high levels of mRNA variability, suggesting that marker expression is not heritable. Surprisingly, this variability does not correlate with cell-to-cell differences in cartilage-like matrix production. Transcriptome-wide analysis suggests that no combination of markers can predict functional potential. De-differentiating chondrocytes also show a disconnect between mRNA expression of the cartilage marker aggrecan and cartilage-like matrix accumulation. Altogether, these quantitative analyses suggest that sorting subpopulations based on these markers would only marginally enrich the progenitor population for 'superior' MSCs. Our results suggest that instantaneous mRNA abundance of canonical markers is tenuously linked to the chondrogenic phenotype at the single-cell level.


Asunto(s)
Condrocitos/fisiología , Regulación de la Expresión Génica/fisiología , Células Madre Mesenquimatosas/fisiología , Animales , Biomarcadores/metabolismo , Bovinos , Diferenciación Celular/fisiología , Matriz Extracelular , Hibridación Fluorescente in Situ , ARN Mensajero
17.
Genome Biol ; 16: 20, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25630241

RESUMEN

BACKGROUND: Long non-coding RNAs (lncRNAs) have been implicated in diverse biological processes. In contrast to extensive genomic annotation of lncRNA transcripts, far fewer have been characterized for subcellular localization and cell-to-cell variability. Addressing this requires systematic, direct visualization of lncRNAs in single cells at single-molecule resolution. RESULTS: We use single-molecule RNA-FISH to systematically quantify and categorize the subcellular localization patterns of a representative set of 61 lncRNAs in three different cell types. Our survey yields high-resolution quantification and stringent validation of the number and spatial positions of these lncRNA, with an mRNA set for comparison. Using this highly quantitative image-based dataset, we observe a variety of subcellular localization patterns, ranging from bright sub-nuclear foci to almost exclusively cytoplasmic localization. We also find that the low abundance of lncRNAs observed from cell population measurements cannot be explained by high expression in a small subset of 'jackpot' cells. Additionally, nuclear lncRNA foci dissolve during mitosis and become widely dispersed, suggesting these lncRNAs are not mitotic bookmarking factors. Moreover, we see that divergently transcribed lncRNAs do not always correlate with their cognate mRNA, nor do they have a characteristic localization pattern. CONCLUSIONS: Our systematic, high-resolution survey of lncRNA localization reveals aspects of lncRNAs that are similar to mRNAs, such as cell-to-cell variability, but also several distinct properties. These characteristics may correspond to particular functional roles. Our study also provides a quantitative description of lncRNAs at the single-cell level and a universally applicable framework for future study and validation of lncRNAs.


Asunto(s)
Hibridación Fluorescente in Situ , ARN Largo no Codificante/metabolismo , Análisis de la Célula Individual , Animales , Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Células HeLa , Humanos , Masculino , Ratones , Mitosis , Transporte de ARN/genética , ARN Largo no Codificante/genética , ARN Mensajero/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA