Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 83(3): 320-323, 2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-36736305

RESUMEN

The Central Dogma has been a useful conceptualization of the transfer of genetic information, and our understanding of the detailed mechanisms involved in that transfer continues to evolve. Here, we speak to several scientists about their research, how it influences our understanding of information transfer, and questions for the future.

2.
Cell ; 161(2): 228-39, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25860606

RESUMEN

Somatic LINE-1 (L1) retrotransposition during neurogenesis is a potential source of genotypic variation among neurons. As a neurogenic niche, the hippocampus supports pronounced L1 activity. However, the basal parameters and biological impact of L1-driven mosaicism remain unclear. Here, we performed single-cell retrotransposon capture sequencing (RC-seq) on individual human hippocampal neurons and glia, as well as cortical neurons. An estimated 13.7 somatic L1 insertions occurred per hippocampal neuron and carried the sequence hallmarks of target-primed reverse transcription. Notably, hippocampal neuron L1 insertions were specifically enriched in transcribed neuronal stem cell enhancers and hippocampus genes, increasing their probability of functional relevance. In addition, bias against intronic L1 insertions sense oriented relative to their host gene was observed, perhaps indicating moderate selection against this configuration in vivo. These experiments demonstrate pervasive L1 mosaicism at genomic loci expressed in hippocampal neurons.


Asunto(s)
Hipocampo/citología , Elementos de Nucleótido Esparcido Largo , Mosaicismo , Neuronas/citología , Variación Genética , Humanos , Neurogénesis , Reacción en Cadena de la Polimerasa , Bancos de Tejidos
3.
Mol Cell ; 82(9): 1610-1612, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35523126

RESUMEN

Garland et al. (2022) discover a physical and functional connection between the HUSH silencing complex and the NEXT nuclear RNA decay complex in mouse embryonic stem cells and conclude HUSH recruits NEXT to degrade prematurely terminated retrotransposon RNAs.


Asunto(s)
ARN , Retroelementos , Animales , Ratones , ARN/genética , Retroelementos/genética
4.
Nature ; 620(7975): 863-872, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37587336

RESUMEN

Cells undergo a major epigenome reconfiguration when reprogrammed to human induced pluripotent stem cells (hiPS cells). However, the epigenomes of hiPS cells and human embryonic stem (hES) cells differ significantly, which affects hiPS cell function1-8. These differences include epigenetic memory and aberrations that emerge during reprogramming, for which the mechanisms remain unknown. Here we characterized the persistence and emergence of these epigenetic differences by performing genome-wide DNA methylation profiling throughout primed and naive reprogramming of human somatic cells to hiPS cells. We found that reprogramming-induced epigenetic aberrations emerge midway through primed reprogramming, whereas DNA demethylation begins early in naive reprogramming. Using this knowledge, we developed a transient-naive-treatment (TNT) reprogramming strategy that emulates the embryonic epigenetic reset. We show that the epigenetic memory in hiPS cells is concentrated in cell of origin-dependent repressive chromatin marked by H3K9me3, lamin-B1 and aberrant CpH methylation. TNT reprogramming reconfigures these domains to a hES cell-like state and does not disrupt genomic imprinting. Using an isogenic system, we demonstrate that TNT reprogramming can correct the transposable element overexpression and differential gene expression seen in conventional hiPS cells, and that TNT-reprogrammed hiPS and hES cells show similar differentiation efficiencies. Moreover, TNT reprogramming enhances the differentiation of hiPS cells derived from multiple cell types. Thus, TNT reprogramming corrects epigenetic memory and aberrations, producing hiPS cells that are molecularly and functionally more similar to hES cells than conventional hiPS cells. We foresee TNT reprogramming becoming a new standard for biomedical and therapeutic applications and providing a novel system for studying epigenetic memory.


Asunto(s)
Reprogramación Celular , Epigénesis Genética , Células Madre Pluripotentes Inducidas , Humanos , Cromatina/genética , Cromatina/metabolismo , Desmetilación del ADN , Metilación de ADN , Elementos Transponibles de ADN , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Lamina Tipo B
5.
Cell ; 153(1): 101-11, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23540693

RESUMEN

LINE-1 (L1) retrotransposons are mobile genetic elements comprising ~17% of the human genome. New L1 insertions can profoundly alter gene function and cause disease, though their significance in cancer remains unclear. Here, we applied enhanced retrotransposon capture sequencing (RC-seq) to 19 hepatocellular carcinoma (HCC) genomes and elucidated two archetypal L1-mediated mechanisms enabling tumorigenesis. In the first example, 4/19 (21.1%) donors presented germline retrotransposition events in the tumor suppressor mutated in colorectal cancers (MCC). MCC expression was ablated in each case, enabling oncogenic ß-catenin/Wnt signaling. In the second example, suppression of tumorigenicity 18 (ST18) was activated by a tumor-specific L1 insertion. Experimental assays confirmed that the L1 interrupted a negative feedback loop by blocking ST18 repression of its enhancer. ST18 was also frequently amplified in HCC nodules from Mdr2(-/-) mice, supporting its assignment as a candidate liver oncogene. These proof-of-principle results substantiate L1-mediated retrotransposition as an important etiological factor in HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Análisis Mutacional de ADN , Genes Supresores de Tumor , Neoplasias Hepáticas/genética , Elementos de Nucleótido Esparcido Largo , Mutagénesis Insercional , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Adulto , Anciano , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Proteínas Represoras/genética , Proteínas Supresoras de Tumor/genética , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
6.
Mol Cell ; 80(5): 915-928.e5, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33186547

RESUMEN

Transposable elements (TEs) drive genome evolution and are a notable source of pathogenesis, including cancer. While CpG methylation regulates TE activity, the locus-specific methylation landscape of mobile human TEs has to date proven largely inaccessible. Here, we apply new computational tools and long-read nanopore sequencing to directly infer CpG methylation of novel and extant TE insertions in hippocampus, heart, and liver, as well as paired tumor and non-tumor liver. As opposed to an indiscriminate stochastic process, we find pronounced demethylation of young long interspersed element 1 (LINE-1) retrotransposons in cancer, often distinct to the adjacent genome and other TEs. SINE-VNTR-Alu (SVA) retrotransposons, including their internal tandem repeat-associated CpG island, are near-universally methylated. We encounter allele-specific TE methylation and demethylation of aberrantly expressed young LINE-1s in normal tissues. Finally, we recover the complete sequences of tumor-specific LINE-1 insertions and their retrotransposition hallmarks, demonstrating how long-read sequencing can simultaneously survey the epigenome and detect somatic TE mobilization.


Asunto(s)
Metilación de ADN , Elementos Transponibles de ADN , ADN de Neoplasias , Epigénesis Genética , Epigenoma , Regulación Neoplásica de la Expresión Génica , Elementos de Nucleótido Esparcido Largo , Secuenciación de Nanoporos , Neoplasias , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Persona de Mediana Edad , Neoplasias/genética , Neoplasias/metabolismo , Especificidad de Órganos
7.
Development ; 151(1)2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38063486

RESUMEN

Cholinergic signaling plays a crucial role in the regulation of adult hippocampal neurogenesis; however, the mechanisms by which acetylcholine mediates neurogenic effects are not completely understood. Here, we report the expression of muscarinic acetylcholine receptor subtype M4 (M4 mAChR) on a subpopulation of neural precursor cells (NPCs) in the adult mouse hippocampus, and demonstrate that its pharmacological stimulation promotes their proliferation, thereby enhancing the production of new neurons in vivo. Using a targeted ablation approach, we also show that medial septum (MS) and the diagonal band of Broca (DBB) cholinergic neurons support both the survival and morphological maturation of adult-born neurons in the mouse hippocampus. Although the systemic administration of an M4-selective allosteric potentiator fails to fully rescue the MS/DBB cholinergic lesion-induced decrease in hippocampal neurogenesis, it further exacerbates the impairment in the morphological maturation of adult-born neurons. Collectively, these findings reveal stage-specific roles of M4 mAChRs in regulating adult hippocampal neurogenesis, uncoupling their positive role in enhancing the production of new neurons from the M4-induced inhibition of their morphological maturation, at least in the context of cholinergic signaling dysfunction.


Asunto(s)
Células-Madre Neurales , Receptor Muscarínico M4 , Ratones , Animales , Receptor Muscarínico M4/metabolismo , Células-Madre Neurales/metabolismo , Hipocampo/metabolismo , Neurogénesis/genética , Colinérgicos/metabolismo , Colinérgicos/farmacología , Proliferación Celular
8.
Mol Cell ; 75(3): 590-604.e12, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31230816

RESUMEN

Epigenetic silencing defends against LINE-1 (L1) retrotransposition in mammalian cells. However, the mechanisms that repress young L1 families and how L1 escapes to cause somatic genome mosaicism in the brain remain unclear. Here we report that a conserved Yin Yang 1 (YY1) transcription factor binding site mediates L1 promoter DNA methylation in pluripotent and differentiated cells. By analyzing 24 hippocampal neurons with three distinct single-cell genomic approaches, we characterized and validated a somatic L1 insertion bearing a 3' transduction. The source (donor) L1 for this insertion was slightly 5' truncated, lacked the YY1 binding site, and was highly mobile when tested in vitro. Locus-specific bisulfite sequencing revealed that the donor L1 and other young L1s with mutated YY1 binding sites were hypomethylated in embryonic stem cells, during neurodifferentiation, and in liver and brain tissue. These results explain how L1 can evade repression and retrotranspose in the human body.


Asunto(s)
Represión Epigenética/genética , Elementos de Nucleótido Esparcido Largo/genética , Retroelementos/genética , Factor de Transcripción YY1/genética , Sitios de Unión/genética , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Genoma Humano/genética , Hipocampo/metabolismo , Humanos , Hígado/metabolismo , Neuronas/metabolismo , Análisis de la Célula Individual
9.
Genome Res ; 33(9): 1465-1481, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37798118

RESUMEN

Mice harbor ∼2800 intact copies of the retrotransposon Long Interspersed Element 1 (L1). The in vivo retrotransposition capacity of an L1 copy is defined by both its sequence integrity and epigenetic status, including DNA methylation of the monomeric units constituting young mouse L1 promoters. Locus-specific L1 methylation dynamics during development may therefore elucidate and explain spatiotemporal niches of endogenous retrotransposition but remain unresolved. Here, we interrogate the retrotransposition efficiency and epigenetic fate of source (donor) L1s, identified as mobile in vivo. We show that promoter monomer loss consistently attenuates the relative retrotransposition potential of their offspring (daughter) L1 insertions. We also observe that most donor/daughter L1 pairs are efficiently methylated upon differentiation in vivo and in vitro. We use Oxford Nanopore Technologies (ONT) long-read sequencing to resolve L1 methylation genome-wide and at individual L1 loci, revealing a distinctive "smile" pattern in methylation levels across the L1 promoter region. Using Pacific Biosciences (PacBio) SMRT sequencing of L1 5' RACE products, we then examine DNA methylation dynamics at the mouse L1 promoter in parallel with transcription start site (TSS) distribution at locus-specific resolution. Together, our results offer a novel perspective on the interplay between epigenetic repression, L1 evolution, and genome stability.


Asunto(s)
Desarrollo Embrionario , Elementos de Nucleótido Esparcido Largo , Ratones , Animales , Retroelementos/genética , Metilación de ADN , Regiones Promotoras Genéticas
10.
Nat Rev Genet ; 21(3): 191-201, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31848477

RESUMEN

Pseudogenes are defined as regions of the genome that contain defective copies of genes. They exist across almost all forms of life, and in mammalian genomes are annotated in similar numbers to recognized protein-coding genes. Although often presumed to lack function, growing numbers of pseudogenes are being found to play important biological roles. In consideration of their evolutionary origins and inherent limitations in genome annotation practices, we posit that pseudogenes have been classified on a scientifically unsubstantiated basis. We reflect that a broad misunderstanding of pseudogenes, perpetuated in part by the pejorative inference of the 'pseudogene' label, has led to their frequent dismissal from functional assessment and exclusion from genomic analyses. With the advent of technologies that simplify the study of pseudogenes, we propose that an objective reassessment of these genomic elements will reveal valuable insights into genome function and evolution.


Asunto(s)
Seudogenes , Animales , Evolución Molecular , Genómica , Humanos
11.
Genome Res ; 32(7): 1298-1314, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35728967

RESUMEN

The retrotransposon LINE-1 (L1) is central to the recent evolutionary history of the human genome and continues to drive genetic diversity and germline pathogenesis. However, the spatiotemporal extent and biological significance of somatic L1 activity are poorly defined and are virtually unexplored in other primates. From a single L1 lineage active at the divergence of apes and Old World monkeys, successive L1 subfamilies have emerged in each descendant primate germline. As revealed by case studies, the presently active human L1 subfamily can also mobilize during embryonic and brain development in vivo. It is unknown whether nonhuman primate L1s can similarly generate somatic insertions in the brain. Here we applied approximately 40× single-cell whole-genome sequencing (scWGS), as well as retrotransposon capture sequencing (RC-seq), to 20 hippocampal neurons from two rhesus macaques (Macaca mulatta). In one animal, we detected and PCR-validated a somatic L1 insertion that generated target site duplications, carried a short 5' transduction, and was present in ∼7% of hippocampal neurons but absent from cerebellum and nonbrain tissues. The corresponding donor L1 allele was exceptionally mobile in vitro and was embedded in PRDM4, a gene expressed throughout development and in neural stem cells. Nanopore long-read methylome and RNA-seq transcriptome analyses indicated young retrotransposon subfamily activation in the early embryo, followed by repression in adult tissues. These data highlight endogenous macaque L1 retrotransposition potential, provide prototypical evidence of L1-mediated somatic mosaicism in a nonhuman primate, and allude to L1 mobility in the brain over the past 30 million years of human evolution.


Asunto(s)
Encéfalo , Elementos de Nucleótido Esparcido Largo , Retroelementos , Animales , Proteínas de Unión al ADN/genética , Macaca mulatta/genética , Neuronas , Retroelementos/genética , Factores de Transcripción/genética
12.
Biochem Soc Trans ; 52(3): 1431-1447, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38747720

RESUMEN

Whole genome sequencing of viruses provides high-resolution molecular insights, enhancing our understanding of viral genome function and phylogeny. Beyond fundamental research, viral sequencing is increasingly vital for pathogen surveillance, epidemiology, and clinical applications. As sequencing methods rapidly evolve, the diversity of viral genomics applications and catalogued genomes continues to expand. Advances in long-read, single molecule, real-time sequencing methodologies present opportunities to sequence contiguous, haplotype resolved viral genomes in a range of research and applied settings. Here we present an overview of nucleic acid sequencing methods and their applications in studying viral genomes. We emphasise the advantages of different viral sequencing approaches, with a particular focus on the benefits of third-generation sequencing technologies in elucidating viral evolution, transmission networks, and pathogenesis.


Asunto(s)
Genoma Viral , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Virus/genética , Genómica/métodos , Filogenia , Secuenciación Completa del Genoma/métodos , Análisis de Secuencia de ADN/métodos
13.
Br J Cancer ; 128(7): 1236-1248, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36707636

RESUMEN

BACKGROUND: Molecular characterisation of hepatocellular carcinoma (HCC) is central to the development of novel therapeutic strategies for the disease. We have previously demonstrated mutagenic consequences of Long-Interspersed Nuclear Element-1 (LINE1s/L1) retrotransposition. However, the role of L1 in HCC, besides somatic mutagenesis, is not well understood. METHODS: We analysed L1 expression in the TCGA-HCC RNAseq dataset (n = 372) and explored potential relationships between L1 expression and clinical features. The findings were confirmed by immunohistochemical (IHC) analysis of an independent human HCC cohort (n = 48) and functional mechanisms explored using in vitro and in vivo model systems. RESULTS: We observed positive associations between L1 and activated TGFß-signalling, TP53 mutation, alpha-fetoprotein and tumour invasion. IHC confirmed a positive association between pSMAD3, a surrogate for TGFß-signalling status, and L1 ORF1p (P < 0.0001, n = 32). Experimental modulation of L1 ORF1p levels revealed an influence of L1 ORF1p on key hepatocarcinogenesis-related pathways. Reduction in cell migration and invasive capacity was observed upon L1 ORF1 knockdown, both in vitro and in vivo. In particular, L1 ORF1p increased PIN1 cytoplasmic localisation. Blocking PIN1 activity abrogated L1 ORF1p-induced NF-κB-mediated inflammatory response genes while further activated TGFß-signalling confirming differential alteration of PIN1 activity in cellular compartments by L1 ORF1p. DISCUSSION: Our data demonstrate a causal link between L1 ORF1p and key oncogenic pathways mediated by PIN1, presenting a novel therapeutic avenue.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Retroelementos , Carcinoma Hepatocelular/genética , Regulación hacia Arriba , Neoplasias Hepáticas/genética , Elementos de Nucleótido Esparcido Largo/genética , Factor de Crecimiento Transformador beta/genética , Peptidilprolil Isomerasa de Interacción con NIMA/genética
14.
Nat Rev Genet ; 23(8): 458-459, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35361925
15.
Bioessays ; 43(11): e2100186, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34569081

RESUMEN

Processed pseudogenes may serve as a genetic reservoir for evolutionary innovation. Here, we argue that through the activity of long interspersed element-1 retrotransposons, processed pseudogenes disperse coding and noncoding sequences rich with regulatory potential throughout the human genome. While these sequences may appear to be non-functional, a lack of contemporary function does not prohibit future development of biological activity. Here, we discuss the dynamic evolution of certain processed pseudogenes into coding and noncoding genes and regulatory elements, and their implication in wide-ranging biological and pathological processes. Also see the video abstract here: https://youtu.be/iUY_mteVoPI.


Asunto(s)
Seudogenes , Retroelementos , Evolución Molecular , Genoma Humano/genética , Humanos , Elementos de Nucleótido Esparcido Largo , Seudogenes/genética , Retroelementos/genética
16.
Annu Rev Genet ; 48: 1-27, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25036377

RESUMEN

Long interspersed element 1 (LINE-1 or L1) retrotransposons have generated one-third of the human genome, and their ongoing mobility is a source of inter- and intraindividual genetic diversity. Although retrotransposition in metazoans has long been considered a germline phenomenon, recent experiments using cultured cells, animal models, and human tissues have revealed extensive L1 mobilization in rodent and human neurons, as well as mobile element activity in the Drosophila brain. In this review, we evaluate the available evidence for L1 retrotransposition in the brain and discuss mechanisms that may regulate neuronal retrotransposition in vivo. We compare experimental strategies used to map de novo somatic retrotransposition events and present the optimal criteria to identify a somatic L1 insertion. Finally, we discuss the unresolved impact of L1-mediated somatic mosaicism upon normal neurobiology, as well as its potential to drive neurological disease.


Asunto(s)
Elementos de Nucleótido Esparcido Largo/genética , Mosaicismo , Neuronas/fisiología , Retroelementos/genética , Animales , Encéfalo/fisiología , Drosophila , Humanos , Roedores
17.
Genome Res ; 28(5): 639-653, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29643204

RESUMEN

The retrotransposon Long Interspersed Element 1 (LINE-1 or L1) is a continuing source of germline and somatic mutagenesis in mammals. Deregulated L1 activity is a hallmark of cancer, and L1 mutagenesis has been described in numerous human malignancies. We previously employed retrotransposon capture sequencing (RC-seq) to analyze hepatocellular carcinoma (HCC) samples from patients infected with hepatitis B or hepatitis C virus and identified L1 variants responsible for activating oncogenic pathways. Here, we have applied RC-seq and whole-genome sequencing (WGS) to an Abcb4 (Mdr2)-/- mouse model of hepatic carcinogenesis and demonstrated for the first time that L1 mobilization occurs in murine tumors. In 12 HCC nodules obtained from 10 animals, we validated four somatic L1 insertions by PCR and capillary sequencing, including TF subfamily elements, and one GF subfamily example. One of the TF insertions carried a 3' transduction, allowing us to identify its donor L1 and to demonstrate that this full-length TF element retained retrotransposition capacity in cultured cancer cells. Using RC-seq, we also identified eight tumor-specific L1 insertions from 25 HCC patients with a history of alcohol abuse. Finally, we used RC-seq and WGS to identify three tumor-specific L1 insertions among 10 intra-hepatic cholangiocarcinoma (ICC) patients, including one insertion traced to a donor L1 on Chromosome 22 known to be highly active in other cancers. This study reveals L1 mobilization as a common feature of hepatocarcinogenesis in mammals, demonstrating that the phenomenon is not restricted to human viral HCC etiologies and is encountered in murine liver tumors.


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Elementos de Nucleótido Esparcido Largo/genética , Retroelementos/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Transformación Celular Neoplásica/genética , Femenino , Humanos , Hígado/metabolismo , Hígado/patología , Masculino , Mamíferos/genética , Ratones Noqueados , Persona de Mediana Edad , Mutagénesis Insercional , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
18.
Nucleic Acids Res ; 47(14): 7262-7275, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31305886

RESUMEN

RNA-Seq is a powerful transcriptome profiling technology enabling transcript discovery and quantification. Whilst most commonly used for gene-level quantification, the data can be used for the analysis of transcript isoforms. However, when the underlying transcript assemblies are complex, current visualization approaches can be limiting, with splicing events a challenge to interpret. Here, we report on the development of a graph-based visualization method as a complementary approach to understanding transcript diversity from short-read RNA-Seq data. Following the mapping of reads to a reference genome, a read-to-read comparison is performed on all reads mapping to a given gene, producing a weighted similarity matrix between reads. This is used to produce an RNA assembly graph, where nodes represent reads and edges similarity scores between them. The resulting graphs are visualized in 3D space to better appreciate their sometimes large and complex topology, with other information being overlaid on to nodes, e.g. transcript models. Here we demonstrate the utility of this approach, including the unusual structure of these graphs and how they can be used to identify issues in assembly, repetitive sequences within transcripts and splice variants. We believe this approach has the potential to significantly improve our understanding of transcript complexity.


Asunto(s)
Empalme Alternativo , Gráficos por Computador , Perfilación de la Expresión Génica/métodos , ARN Mensajero/genética , Análisis de Secuencia de ARN/métodos , Genoma Humano/genética , Humanos , Modelos Genéticos , Modelos Moleculares , Conformación de Ácido Nucleico , Isoformas de ARN/química , Isoformas de ARN/genética , Isoformas de ARN/metabolismo , ARN Mensajero/química , ARN Mensajero/metabolismo
19.
Trends Genet ; 33(11): 802-816, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28797643

RESUMEN

The retrotransposon LINE-1 (long interspersed element 1, L1) is a transposable element that has extensively colonized the mammalian germline. L1 retrotransposition can also occur in somatic cells, causing genomic mosaicism, as well as in cancer. However, the extent of L1-driven mosaicism arising during ontogenesis is unclear. We discuss here recent experimental data which, at a minimum, fully substantiate L1 mosaicism in early embryonic development and neural cells, including post-mitotic neurons. We also consider the possible biological impact of somatic L1 insertions in neurons, the existence of donor L1s that are highly active ('hot') in specific spatiotemporal niches, and the evolutionary selection of donor L1s driving neuronal mosaicism.


Asunto(s)
Mamíferos/genética , Mosaicismo , Animales , Desarrollo Embrionario/genética , Humanos , Elementos de Nucleótido Esparcido Largo , Neuronas/metabolismo , Retroelementos
20.
Genome Res ; 27(8): 1395-1405, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28483779

RESUMEN

LINE-1 (L1) retrotransposons are a noted source of genetic diversity and disease in mammals. To expand its genomic footprint, L1 must mobilize in cells that will contribute their genetic material to subsequent generations. Heritable L1 insertions may therefore arise in germ cells and in pluripotent embryonic cells, prior to germline specification, yet the frequency and predominant developmental timing of such events remain unclear. Here, we applied mouse retrotransposon capture sequencing (mRC-seq) and whole-genome sequencing (WGS) to pedigrees of C57BL/6J animals, and uncovered an L1 insertion rate of ≥1 event per eight births. We traced heritable L1 insertions to pluripotent embryonic cells and, strikingly, to early primordial germ cells (PGCs). New L1 insertions bore structural hallmarks of target-site primed reverse transcription (TPRT) and mobilized efficiently in a cultured cell retrotransposition assay. Together, our results highlight the rate and evolutionary impact of heritable L1 retrotransposition and reveal retrotransposition-mediated genomic diversification as a fundamental property of pluripotent embryonic cells in vivo.


Asunto(s)
Embrión de Mamíferos/metabolismo , Elementos de Nucleótido Esparcido Largo , Animales , Embrión de Mamíferos/citología , Femenino , Genómica/métodos , Células Germinativas , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mosaicismo , Secuenciación Completa del Genoma/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA