Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 13(1): 6874, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37106015

RESUMEN

DNA methylation is important for establishing and maintaining cell identity and for genomic stability. This is achieved by regulating the accessibility of regulatory and transcriptional elements and the compaction of subtelomeric, centromeric, and other inactive genomic regions. Carcinogenesis is accompanied by a global loss in DNA methylation, which facilitates the transformation of cells. Cancer hypomethylation may also cause genomic instability, for example through interference with the protective function of telomeres and centromeres. However, understanding the role(s) of hypomethylation in tumor evolution is incomplete because the precise mutational consequences of global hypomethylation have thus far not been systematically assessed. Here we made genome-wide inventories of all possible genetic variation that accumulates in single cells upon the long-term global hypomethylation by CRISPR interference-mediated conditional knockdown of DNMT1. Depletion of DNMT1 resulted in a genomewide reduction in DNA methylation. The degree of DNA methylation loss was similar to that observed in many cancer types. Hypomethylated cells showed reduced proliferation rates, increased transcription of genes, reactivation of the inactive X-chromosome and abnormal nuclear morphologies. Prolonged hypomethylation was accompanied by increased chromosomal instability. However, there was no increase in mutational burden, enrichment for certain mutational signatures or accumulation of structural variation to the genome. In conclusion, the primary consequence of hypomethylation is genomic instability, which in cancer leads to increased tumor heterogeneity and thereby fuels cancer evolution.


Asunto(s)
Metilación de ADN , Inestabilidad Genómica , Humanos , Mutación , Carcinogénesis , ADN
2.
Commun Biol ; 4(1): 1301, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34795391

RESUMEN

Inflammatory liver disease increases the risk of developing primary liver cancer. The mechanism through which liver disease induces tumorigenesis remains unclear, but is thought to occur via increased mutagenesis. Here, we performed whole-genome sequencing on clonally expanded single liver stem cells cultured as intrahepatic cholangiocyte organoids (ICOs) from patients with alcoholic cirrhosis, non-alcoholic steatohepatitis (NASH), and primary sclerosing cholangitis (PSC). Surprisingly, we find that these precancerous liver disease conditions do not result in a detectable increased accumulation of mutations, nor altered mutation types in individual liver stem cells. This finding contrasts with the mutational load and typical mutational signatures reported for liver tumors, and argues against the hypothesis that liver disease drives tumorigenesis via a direct mechanism of induced mutagenesis. Disease conditions in the liver may thus act through indirect mechanisms to drive the transition from healthy to cancerous cells, such as changes to the microenvironment that favor the outgrowth of precancerous cells.


Asunto(s)
Colangitis Esclerosante/genética , Cirrosis Hepática Alcohólica/genética , Hepatopatías/genética , Mutagénesis , Enfermedad del Hígado Graso no Alcohólico/genética , Lesiones Precancerosas/genética , Células Madre/metabolismo , Humanos , Hígado/fisiología , Organoides/metabolismo
3.
Nat Genet ; 53(8): 1187-1195, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34211178

RESUMEN

Central to tumor evolution is the generation of genetic diversity. However, the extent and patterns by which de novo karyotype alterations emerge and propagate within human tumors are not well understood, especially at single-cell resolution. Here, we present 3D Live-Seq-a protocol that integrates live-cell imaging of tumor organoid outgrowth and whole-genome sequencing of each imaged cell to reconstruct evolving tumor cell karyotypes across consecutive cell generations. Using patient-derived colorectal cancer organoids and fresh tumor biopsies, we demonstrate that karyotype alterations of varying complexity are prevalent and can arise within a few cell generations. Sub-chromosomal acentric fragments were prone to replication and collective missegregation across consecutive cell divisions. In contrast, gross genome-wide karyotype alterations were generated in a single erroneous cell division, providing support that aneuploid tumor genomes can evolve via punctuated evolution. Mapping the temporal dynamics and patterns of karyotype diversification in cancer enables reconstructions of evolutionary paths to malignant fitness.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Análisis de la Célula Individual/métodos , Proliferación Celular/genética , Cromatina/genética , Cromosomas Humanos , Dosificación de Gen , Humanos , Cariotipo , Cariotipificación , Microscopía Confocal , Mitosis , Organoides/crecimiento & desarrollo , Organoides/patología , Huso Acromático/genética
4.
Nat Commun ; 11(1): 3932, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32753580

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Sci Adv ; 6(16): eaaz7602, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32494621

RESUMEN

Genomic instability is common in human embryos, but the underlying causes are largely unknown. Here, we examined the consequences of sperm DNA damage on the embryonic genome by single-cell whole-genome sequencing of individual blastomeres from bovine embryos produced with sperm damaged by γ-radiation. Sperm DNA damage primarily leads to fragmentation of the paternal chromosomes followed by random distribution of the chromosomal fragments over the two sister cells in the first cell division. An unexpected secondary effect of sperm DNA damage is the induction of direct unequal cleavages, which include the poorly understood heterogoneic cell divisions. As a result, chaotic mosaicism is common in embryos derived from fertilizations with damaged sperm. The mosaic aneuploidies, uniparental disomies, and de novo structural variation induced by sperm DNA damage may compromise fertility and lead to rare congenital disorders when embryos escape developmental arrest.


Asunto(s)
Desarrollo Embrionario , Espermatozoides , Animales , Bovinos , Daño del ADN , Desarrollo Embrionario/genética , Femenino , Inestabilidad Genómica , Humanos , Masculino , Mosaicismo , Embarazo
6.
Nat Commun ; 11(1): 2493, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32427826

RESUMEN

Genetic changes acquired during in vitro culture pose a risk for the successful application of stem cells in regenerative medicine. To assess the genetic risks induced by culturing, we determined all mutations in individual human stem cells by whole genome sequencing. Individual pluripotent, intestinal, and liver stem cells accumulate 3.5 ± 0.5, 7.2 ± 1.1 and 8.3 ± 3.6 base substitutions per population doubling, respectively. The annual in vitro mutation accumulation rate of adult stem cells is nearly 40-fold higher than the in vivo mutation accumulation rate. Mutational signature analysis reveals that in vitro induced mutations are caused by oxidative stress. Reducing oxygen tension in culture lowers the mutational load. We use the mutation rates, spectra, and genomic distribution to model the accumulation of oncogenic mutations during typical in vitro expansion, manipulation or screening experiments using human stem cells. Our study provides empirically defined parameters to assess the mutational risk of stem cell based therapies.


Asunto(s)
Células Madre Adultas/metabolismo , Análisis Mutacional de ADN/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Mutación , Adulto , Células Madre Adultas/citología , Algoritmos , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/citología , Intestinos/citología , Hígado/citología , Hígado/metabolismo , Modelos Genéticos , Acumulación de Mutaciones , Tasa de Mutación , Medicina Regenerativa/métodos , Secuenciación Completa del Genoma/métodos
7.
Genome Med ; 11(1): 79, 2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31801603

RESUMEN

BACKGROUND: Genomic structural variants (SVs) can affect many genes and regulatory elements. Therefore, the molecular mechanisms driving the phenotypes of patients carrying de novo SVs are frequently unknown. METHODS: We applied a combination of systematic experimental and bioinformatic methods to improve the molecular diagnosis of 39 patients with multiple congenital abnormalities and/or intellectual disability harboring apparent de novo SVs, most with an inconclusive diagnosis after regular genetic testing. RESULTS: In 7 of these cases (18%), whole-genome sequencing analysis revealed disease-relevant complexities of the SVs missed in routine microarray-based analyses. We developed a computational tool to predict the effects on genes directly affected by SVs and on genes indirectly affected likely due to the changes in chromatin organization and impact on regulatory mechanisms. By combining these functional predictions with extensive phenotype information, candidate driver genes were identified in 16/39 (41%) patients. In 8 cases, evidence was found for the involvement of multiple candidate drivers contributing to different parts of the phenotypes. Subsequently, we applied this computational method to two cohorts containing a total of 379 patients with previously detected and classified de novo SVs and identified candidate driver genes in 189 cases (50%), including 40 cases whose SVs were previously not classified as pathogenic. Pathogenic position effects were predicted in 28% of all studied cases with balanced SVs and in 11% of the cases with copy number variants. CONCLUSIONS: These results demonstrate an integrated computational and experimental approach to predict driver genes based on analyses of WGS data with phenotype association and chromatin organization datasets. These analyses nominate new pathogenic loci and have strong potential to improve the molecular diagnosis of patients with de novo SVs.


Asunto(s)
Estudios de Asociación Genética , Enfermedades Genéticas Congénitas/diagnóstico , Enfermedades Genéticas Congénitas/genética , Predisposición Genética a la Enfermedad , Variación Genética , Fenotipo , Biología Computacional/métodos , Variaciones en el Número de Copia de ADN , Genoma Humano , Variación Estructural del Genoma , Humanos , Anotación de Secuencia Molecular , Secuenciación Completa del Genoma
8.
Nat Commun ; 10(1): 4571, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31594944

RESUMEN

5-Fluorouracil (5-FU) is a chemotherapeutic drug commonly used for the treatment of solid cancers. It is proposed that 5-FU interferes with nucleotide synthesis and incorporates into DNA, which may have a mutational impact on both surviving tumor and healthy cells. Here, we treat intestinal organoids with 5-FU and find a highly characteristic mutational pattern that is dominated by T>G substitutions in a CTT context. Tumor whole genome sequencing data confirms that this signature is also identified in vivo in colorectal and breast cancer patients who have received 5-FU treatment. Taken together, our results demonstrate that 5-FU is mutagenic and may drive tumor evolution and increase the risk of secondary malignancies. Furthermore, the identified signature shows a strong resemblance to COSMIC signature 17, the hallmark signature of treatment-naive esophageal and gastric tumors, which indicates that distinct endogenous and exogenous triggers can converge onto highly similar mutational signatures.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Fluorouracilo/efectos adversos , Neoplasias/genética , Mutación Puntual/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Adulto , Edad de Inicio , Anciano , Biopsia , Carcinogénesis/genética , Técnicas de Cultivo de Célula , Línea Celular , Ensayos Clínicos como Asunto , Estudios de Cohortes , Análisis Mutacional de ADN , Femenino , Humanos , Intestinos/citología , Masculino , Persona de Mediana Edad , Modelos Genéticos , Tasa de Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Organoides , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Células Madre , Transcriptoma/genética , Secuenciación Completa del Genoma , Adulto Joven
9.
Sci Adv ; 5(5): eaaw1271, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31149636

RESUMEN

A developing human fetus needs to balance rapid cellular expansion with maintaining genomic stability. Here, we accurately quantified and characterized somatic mutation accumulation in fetal tissues by analyzing individual stem cells from human fetal liver and intestine. Fetal mutation rates were about fivefold higher than in tissue-matched adult stem cells. The mutational landscape of fetal intestinal stem cells resembled that of adult intestinal stem cells, while the mutation spectrum of fetal liver stem cells is distinct from stem cells of the fetal intestine and the adult liver. Our analyses indicate that variation in mutational mechanisms, including oxidative stress and spontaneous deamination of methylated cytosines, contributes to the observed divergence in mutation accumulation patterns and drives genetic mosaicism in humans.


Asunto(s)
Feto/fisiología , Mutación , Células Madre Adultas/fisiología , Feto/citología , Humanos , Intestinos/citología , Intestinos/embriología , Hígado/citología , Hígado/embriología , Tasa de Mutación , Especificidad de Órganos , Piel/citología , Piel/embriología
10.
Genome Res ; 29(7): 1067-1077, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31221724

RESUMEN

Nucleotide excision repair (NER) is one of the main DNA repair pathways that protect cells against genomic damage. Disruption of this pathway can contribute to the development of cancer and accelerate aging. Mutational characteristics of NER-deficiency may reveal important diagnostic opportunities, as tumors deficient in NER are more sensitive to certain treatments. Here, we analyzed the genome-wide somatic mutational profiles of adult stem cells (ASCs) from NER-deficient Ercc1 -/Δ mice. Our results indicate that NER-deficiency increases the base substitution load twofold in liver but not in small intestinal ASCs, which coincides with the tissue-specific aging pathology observed in these mice. Moreover, NER-deficient ASCs of both tissues show an increased contribution of Signature 8 mutations, which is a mutational pattern with unknown etiology that is recurrently observed in various cancer types. The scattered genomic distribution of the base substitutions indicates that deficiency of global-genome NER (GG-NER) underlies the observed mutational consequences. In line with this, we observe increased Signature 8 mutations in a GG-NER-deficient human organoid culture, in which XPC was deleted using CRISPR-Cas9 gene-editing. Furthermore, genomes of NER-deficient breast tumors show an increased contribution of Signature 8 mutations compared with NER-proficient tumors. Elevated levels of Signature 8 mutations could therefore contribute to a predictor of NER-deficiency based on a patient's mutational profile.


Asunto(s)
Reparación del ADN/genética , Mutación , Neoplasias/genética , Células Madre Adultas , Animales , Neoplasias de la Mama/genética , Estudios de Cohortes , Análisis Mutacional de ADN , ADN de Neoplasias , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Femenino , Humanos , Ratones , Organoides , Técnicas de Cultivo de Tejidos , Secuenciación Completa del Genoma
11.
Circ Genom Precis Med ; 12(2): e002328, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30681347

RESUMEN

BACKGROUND: Regulatory elements may be involved in the mechanisms by which 52 loci influence myocardial mass, reflected by abnormal amplitude and duration of the QRS complex on the ECG. Functional annotation thus far did not take into account how these elements are affected in disease context. METHODS: We generated maps of regulatory elements on hypertrophic cardiomyopathy patients (ChIP-seq N=14 and RNA-seq N=11) and nondiseased hearts (ChIP-seq N=4 and RNA-seq N=11). We tested enrichment of QRS-associated loci on elements differentially acetylated and directly regulating differentially expressed genes between hypertrophic cardiomyopathy patients and controls. We further performed functional annotation on QRS-associated loci using these maps of differentially active regulatory elements. RESULTS: Regions differentially affected in disease showed a stronger enrichment ( P=8.6×10-5) for QRS-associated variants than those not showing differential activity ( P=0.01). Promoters of genes differentially regulated between hypertrophic cardiomyopathy patients and controls showed more enrichment ( P=0.001) than differentially acetylated enhancers ( P=0.8) and super-enhancers ( P=0.025). We also identified 74 potential causal variants overlapping these differential regulatory elements. Eighteen of the genes mapped confirmed previous findings, now also pinpointing the potentially affected regulatory elements and candidate causal variants. Fourteen new genes were also mapped. CONCLUSIONS: Our results suggest differentially active regulatory elements between hypertrophic cardiomyopathy patients and controls can offer more insights into the mechanisms of QRS-associated loci than elements not affected by disease.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Miocardio/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Acetilación , Adolescente , Adulto , Cardiomiopatía Hipertrófica/patología , Proteínas Portadoras/genética , Estudios de Casos y Controles , Electrocardiografía , Femenino , Sitios Genéticos , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Adulto Joven
12.
Nat Protoc ; 13(1): 59-78, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29215633

RESUMEN

Characterization of mutational processes in adult stem cells (ASCs) will improve our understanding of aging-related diseases, such as cancer and organ failure, and may ultimately help prevent the development of these diseases. Here, we present a method for cataloging mutations in individual human ASCs without the necessity of using error-prone whole-genome amplification. Single ASCs are expanded in vitro into clonal organoid cultures to generate sufficient DNA for accurate whole-genome sequencing (WGS) analysis. We developed a data-analysis pipeline that identifies with high confidence somatic variants that accumulated in vivo in the original ASC. These genome-wide mutation catalogs are valuable resources for the characterization of the underlying mutational mechanisms. In addition, this protocol can be used to determine the effects of culture conditions or mutagen exposure on mutation accumulation in ASCs in vitro. Here, we describe a protocol for human liver ASCs that can be completed over a period of 3-4 months with hands-on time of ∼5 d.


Asunto(s)
Células Madre Adultas/citología , Acumulación de Mutaciones , Mutación/genética , Organoides/citología , Secuenciación Completa del Genoma/métodos , Células Cultivadas , ADN/análisis , ADN/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hígado/citología
13.
Nature ; 538(7624): 260-264, 2016 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-27698416

RESUMEN

The gradual accumulation of genetic mutations in human adult stem cells (ASCs) during life is associated with various age-related diseases, including cancer. Extreme variation in cancer risk across tissues was recently proposed to depend on the lifetime number of ASC divisions, owing to unavoidable random mutations that arise during DNA replication. However, the rates and patterns of mutations in normal ASCs remain unknown. Here we determine genome-wide mutation patterns in ASCs of the small intestine, colon and liver of human donors with ages ranging from 3 to 87 years by sequencing clonal organoid cultures derived from primary multipotent cells. Our results show that mutations accumulate steadily over time in all of the assessed tissue types, at a rate of approximately 40 novel mutations per year, despite the large variation in cancer incidence among these tissues. Liver ASCs, however, have different mutation spectra compared to those of the colon and small intestine. Mutational signature analysis reveals that this difference can be attributed to spontaneous deamination of methylated cytosine residues in the colon and small intestine, probably reflecting their high ASC division rate. In liver, a signature with an as-yet-unknown underlying mechanism is predominant. Mutation spectra of driver genes in cancer show high similarity to the tissue-specific ASC mutation spectra, suggesting that intrinsic mutational processes in ASCs can initiate tumorigenesis. Notably, the inter-individual variation in mutation rate and spectra are low, suggesting tissue-specific activity of common mutational processes throughout life.


Asunto(s)
Células Madre Adultas/metabolismo , Envejecimiento/genética , Acumulación de Mutaciones , Tasa de Mutación , Especificidad de Órganos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Niño , Preescolar , Colon/metabolismo , Análisis Mutacional de ADN , Femenino , Genes Relacionados con las Neoplasias/genética , Humanos , Incidencia , Intestino Delgado/metabolismo , Hígado/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Células Madre Multipotentes/metabolismo , Neoplasias/epidemiología , Neoplasias/genética , Organoides/metabolismo , Mutación Puntual/genética , Adulto Joven
14.
BMC Genomics ; 16: 357, 2015 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-25943489

RESUMEN

BACKGROUND: Since the completion of the rat reference genome in 2003, whole-genome sequencing data from more than 40 rat strains have become available. These data represent the broad range of strains that are used in rat research including commonly used substrains. Currently, this wealth of information cannot be used to its full extent, because the variety of different variant calling algorithms employed by different groups impairs comparison between strains. In addition, all rat whole genome sequencing studies to date used an outdated reference genome for analysis (RGSC3.4 released in 2004). RESULTS: Here we present a comprehensive, multi-sample and uniformly called set of genetic variants in 40 rat strains, including 19 substrains. We reanalyzed all primary data using a recent version of the rat reference assembly (RGSC5.0 released in 2012) and identified over 12 million genomic variants (SNVs, indels and structural variants) among the 40 strains. 28,318 SNVs are specific to individual substrains, which may be explained by introgression from other unsequenced strains and ongoing evolution by genetic drift. Substrain SNVs may have a larger predicted functional impact compared to older shared SNVs. CONCLUSIONS: In summary we present a comprehensive catalog of uniformly analyzed genetic variants among 40 widely used rat inbred strains based on the RGSC5.0 assembly. This represents a valuable resource, which will facilitate rat functional genomic research. In line with previous observations, our genome-wide analyses do not show evidence for contribution of multiple ancestral founder rat subspecies to the currently used rat inbred strains, as is the case for mouse. In addition, we find that the degree of substrain variation is highly variable between strains, which is of importance for the correct interpretation of experimental data from different labs.


Asunto(s)
Genómica , Ratas/genética , Animales , Perros , Evolución Molecular , Flujo Genético , Mutación INDEL , Ratones , Polimorfismo de Nucleótido Simple , Especificidad de la Especie
15.
Cell Rep ; 9(6): 2001-10, 2014 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-25497101

RESUMEN

Genomic rearrangements are a common cause of human congenital abnormalities. However, their origin and consequences are poorly understood. We performed molecular analysis of two patients with congenital disease who carried de novo genomic rearrangements. We found that the rearrangements in both patients hit genes that are recurrently rearranged in cancer (ETV1, FOXP1, and microRNA cluster C19MC) and drive formation of fusion genes similar to those described in cancer. Subsequent analysis of a large set of 552 de novo germline genomic rearrangements underlying congenital disorders revealed enrichment for genes rearranged in cancer and overlap with somatic cancer breakpoints. Breakpoints of common (inherited) germline structural variations also overlap with cancer breakpoints but are depleted for cancer genes. We propose that the same genomic positions are prone to genomic rearrangements in germline and soma but that timing and context of breakage determines whether developmental defects or cancer are promoted.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas Humanos/genética , Anomalías Congénitas/genética , Reordenamiento Génico , Genoma Humano , Mutación de Línea Germinal , Animales , Puntos de Rotura del Cromosoma , Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead/genética , Células HEK293 , Humanos , MicroARNs/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Pez Cebra
16.
BMC Res Notes ; 7: 34, 2014 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-24418292

RESUMEN

BACKGROUND: Small RNAs are important regulators of genome function, yet their prediction in genomes is still a major computational challenge. Statistical analyses of pre-miRNA sequences indicated that their 2D structure tends to have a minimal free energy (MFE) significantly lower than MFE values of equivalently randomized sequences with the same nucleotide composition, in contrast to other classes of non-coding RNA. The computation of many MFEs is, however, too intensive to allow for genome-wide screenings. RESULTS: Using a local grid infrastructure, MFE distributions of random sequences were pre-calculated on a large scale. These distributions follow a normal distribution and can be used to determine the MFE distribution for any given sequence composition by interpolation. It allows on-the-fly calculation of the normal distribution for any candidate sequence composition. CONCLUSION: The speedup achieved makes genome-wide screening with this characteristic of a pre-miRNA sequence practical. Although this particular property alone will not be able to distinguish miRNAs from other sequences sufficiently discriminative, the MFE-based P-value should be added to the parameters of choice to be included in the selection of potential miRNA candidates for experimental verification.


Asunto(s)
Secuencia de Bases , Biología Computacional/métodos , Entropía , MicroARNs/genética , Herpesvirus Humano 4/genética , Secuencias Invertidas Repetidas , MicroARNs/química , Datos de Secuencia Molecular , Distribución Normal , Conformación de Ácido Nucleico
17.
Genome Biol ; 15(1): R6, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24393600

RESUMEN

BACKGROUND: Long noncoding RNAs (lncRNAs) form an abundant class of transcripts, but the function of the majority of them remains elusive. While it has been shown that some lncRNAs are bound by ribosomes, it has also been convincingly demonstrated that these transcripts do not code for proteins. To obtain a comprehensive understanding of the extent to which lncRNAs bind ribosomes, we performed systematic RNA sequencing on ribosome-associated RNA pools obtained through ribosomal fractionation and compared the RNA content with nuclear and (non-ribosome bound) cytosolic RNA pools. RESULTS: The RNA composition of the subcellular fractions differs significantly from each other, but lncRNAs are found in all locations. A subset of specific lncRNAs is enriched in the nucleus but surprisingly the majority is enriched in the cytosol and in ribosomal fractions. The ribosomal enriched lncRNAs include H19 and TUG1. CONCLUSIONS: Most studies on lncRNAs have focused on the regulatory function of these transcripts in the nucleus. We demonstrate that only a minority of all lncRNAs are nuclear enriched. Our findings suggest that many lncRNAs may have a function in cytoplasmic processes, and in particular in ribosome complexes.


Asunto(s)
Citosol/química , ARN Largo no Codificante/genética , Ribosomas/genética , Línea Celular Tumoral , Núcleo Celular/química , Núcleo Celular/genética , Biblioteca de Genes , Humanos , ARN Largo no Codificante/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ribosomas/metabolismo , Análisis de Secuencia de ARN
18.
Cell Stem Cell ; 13(6): 653-8, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24315439

RESUMEN

Single murine and human intestinal stem cells can be expanded in culture over long time periods as genetically and phenotypically stable epithelial organoids. Increased cAMP levels induce rapid swelling of such organoids by opening the cystic fibrosis transmembrane conductor receptor (CFTR). This response is lost in organoids derived from cystic fibrosis (CF) patients. Here we use the CRISPR/Cas9 genome editing system to correct the CFTR locus by homologous recombination in cultured intestinal stem cells of CF patients. The corrected allele is expressed and fully functional as measured in clonally expanded organoids. This study provides proof of concept for gene correction by homologous recombination in primary adult stem cells derived from patients with a single-gene hereditary defect.


Asunto(s)
Proteínas Asociadas a CRISPR/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Intestinos/patología , Organoides/metabolismo , Células Madre/metabolismo , Adulto , Células Madre Adultas/metabolismo , Animales , Secuencia de Bases , Terapia Genética , Humanos , Ratones , Datos de Secuencia Molecular , Edición de ARN/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA