Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Cell ; 186(2): 305-326.e27, 2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36638792

RESUMEN

All living things experience an increase in entropy, manifested as a loss of genetic and epigenetic information. In yeast, epigenetic information is lost over time due to the relocalization of chromatin-modifying proteins to DNA breaks, causing cells to lose their identity, a hallmark of yeast aging. Using a system called "ICE" (inducible changes to the epigenome), we find that the act of faithful DNA repair advances aging at physiological, cognitive, and molecular levels, including erosion of the epigenetic landscape, cellular exdifferentiation, senescence, and advancement of the DNA methylation clock, which can be reversed by OSK-mediated rejuvenation. These data are consistent with the information theory of aging, which states that a loss of epigenetic information is a reversible cause of aging.


Asunto(s)
Envejecimiento , Epigénesis Genética , Animales , Envejecimiento/genética , Metilación de ADN , Epigenoma , Mamíferos/genética , Nucleoproteínas , Saccharomyces cerevisiae/genética
3.
Cell ; 169(5): 836-848.e15, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28525754

RESUMEN

Myriad experiences produce transient memory, yet, contingent on the internal state of the organism and the saliency of the experience, only some memories persist over time. How experience and internal state influence the duration of memory at the molecular level remains unknown. A self-assembled aggregated state of Drosophila Orb2A protein is required specifically for long-lasting memory. We report that in the adult fly brain the mRNA encoding Orb2A protein exists in an unspliced non-protein-coding form. The convergence of experience and internal drive transiently increases the spliced protein-coding Orb2A mRNA. A screen identified pasilla, the fly ortholog of mammalian Nova-1/2, as a mediator of Orb2A mRNA processing. A single-nucleotide substitution in the intronic region that reduces Pasilla binding and intron removal selectively impairs long-term memory. We posit that pasilla-mediated processing of unspliced Orb2A mRNA integrates experience and internal state to control Orb2A protein abundance and long-term memory formation.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Intrones , Memoria a Largo Plazo , Ribonucleoproteínas/metabolismo , Factores de Transcripción/genética , Factores de Escisión y Poliadenilación de ARNm/genética , Animales , Secuencia de Bases , Conducta Animal , Encéfalo/metabolismo , Condicionamiento Psicológico , Proteínas de Drosophila/química , Drosophila melanogaster/genética , Aprendizaje , Modelos Animales , Motivación , Mutación , Isoformas de Proteínas/metabolismo , Empalme del ARN , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Factores de Escisión y Poliadenilación de ARNm/química , Factores de Escisión y Poliadenilación de ARNm/metabolismo
4.
Nature ; 601(7893): 434-439, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34937944

RESUMEN

The switch/sucrose non-fermentable (SWI/SNF) complex has a crucial role in chromatin remodelling1 and is altered in over 20% of cancers2,3. Here we developed a proteolysis-targeting chimera (PROTAC) degrader of the SWI/SNF ATPase subunits, SMARCA2 and SMARCA4, called AU-15330. Androgen receptor (AR)+ forkhead box A1 (FOXA1)+ prostate cancer cells are exquisitely sensitive to dual SMARCA2 and SMARCA4 degradation relative to normal and other cancer cell lines. SWI/SNF ATPase degradation rapidly compacts cis-regulatory elements bound by transcription factors that drive prostate cancer cell proliferation, namely AR, FOXA1, ERG and MYC, which dislodges them from chromatin, disables their core enhancer circuitry, and abolishes the downstream oncogenic gene programs. SWI/SNF ATPase degradation also disrupts super-enhancer and promoter looping interactions that wire supra-physiologic expression of the AR, FOXA1 and MYC oncogenes themselves. AU-15330 induces potent inhibition of tumour growth in xenograft models of prostate cancer and synergizes with the AR antagonist enzalutamide, even inducing disease remission in castration-resistant prostate cancer (CRPC) models without toxicity. Thus, impeding SWI/SNF-mediated enhancer accessibility represents a promising therapeutic approach for enhancer-addicted cancers.


Asunto(s)
Adenosina Trifosfatasas , ADN Helicasas , Proteínas Nucleares , Neoplasias de la Próstata , Factores de Transcripción , Adenosina Trifosfatasas/metabolismo , Animales , Benzamidas , ADN Helicasas/genética , Elementos de Facilitación Genéticos , Genes myc , Factor Nuclear 3-alfa del Hepatocito , Humanos , Masculino , Nitrilos , Proteínas Nucleares/genética , Oncogenes , Feniltiohidantoína , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Receptores Androgénicos , Factores de Transcripción/genética , Regulador Transcripcional ERG , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Genes Dev ; 27(4): 378-89, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23392611

RESUMEN

Transcription and pre-mRNA alternative splicing are highly regulated processes that play major roles in modulating eukaryotic gene expression. It is increasingly apparent that other pathways of RNA metabolism, including small RNA biogenesis, can regulate these processes. However, a direct link between alternative pre-mRNA splicing and small RNA pathways has remained elusive. Here we show that the small RNA pathway protein Argonaute-2 (Ago-2) regulates alternative pre-mRNA splicing patterns of specific transcripts in the Drosophila nucleus using genome-wide methods in conjunction with RNAi in cell culture and Ago-2 deletion or catalytic site mutations in Drosophila adults. Moreover, we show that nuclear Argonaute-2 binds to specific chromatin sites near gene promoters and negatively regulates the transcription of the Ago-2-associated target genes. These transcriptional target genes are also bound by Polycomb group (PcG) transcriptional repressor proteins and change during development, implying that Ago-2 may regulate Drosophila development. Importantly, both of these activities were independent of the catalytic activity of Ago-2, suggesting new roles for Ago-2 in the nucleus. Finally, we determined the nuclear RNA-binding profile of Ago-2, found it bound to several splicing target transcripts, and identified a G-rich RNA-binding site for Ago-2 that was enriched in these transcripts. These results suggest two new nuclear roles for Ago-2: one in pre-mRNA splicing and one in transcriptional repression.


Asunto(s)
Empalme Alternativo , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Núcleo Celular/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Drosophila/metabolismo , Regulación de la Expresión Génica , Animales , Línea Celular , Cromatina/metabolismo , Mutación , Unión Proteica , ARN/metabolismo , Transcriptoma
7.
Genome Res ; 26(3): 342-50, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26848124

RESUMEN

Long-range and highly accurate de novo assembly from short-read data is one of the most pressing challenges in genomics. Recently, it has been shown that read pairs generated by proximity ligation of DNA in chromatin of living tissue can address this problem, dramatically increasing the scaffold contiguity of assemblies. Here, we describe a simpler approach ("Chicago") based on in vitro reconstituted chromatin. We generated two Chicago data sets with human DNA and developed a statistical model and a new software pipeline ("HiRise") that can identify poor quality joins and produce accurate, long-range sequence scaffolds. We used these to construct a highly accurate de novo assembly and scaffolding of a human genome with scaffold N50 of 20 Mbp. We also demonstrated the utility of Chicago for improving existing assemblies by reassembling and scaffolding the genome of the American alligator. With a single library and one lane of Illumina HiSeq sequencing, we increased the scaffold N50 of the American alligator from 508 kbp to 10 Mbp.


Asunto(s)
Cromosomas , Biología Computacional/métodos , Ligamiento Genético , Biblioteca Genómica , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Animales , Línea Celular , Humanos , Reproducibilidad de los Resultados
8.
Nucleic Acids Res ; 45(7): 4051-4067, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-27928057

RESUMEN

We recently identified the 4-pyridinone-benzisothiazole carboxamide compound 1C8 as displaying strong anti-HIV-1 potency against a variety of clinical strains in vitro. Here we show that 1C8 decreases the expression of HIV-1 and alters splicing events involved in the production of HIV-1 mRNAs. Although 1C8 was designed to be a structural mimic of the fused tetracyclic indole compound IDC16 that targets SRSF1, it did not affect the splice site shifting activity of SRSF1. Instead, 1C8 altered splicing regulation mediated by SRSF10. Depleting SRSF10 by RNA interference affected viral splicing and, like 1C8, decreased expression of Tat, Gag and Env. Incubating cells with 1C8 promoted the dephosphorylation of SRSF10 and increased its interaction with hTra2ß, a protein previously implicated in the control of HIV-1 RNA splicing. While 1C8 affects the alternative splicing of cellular transcripts controlled by SRSF10 and hTra2ß, concentrations greater than those needed to inhibit HIV-1 replication were required to elicit significant alterations. Thus, the ability of 1C8 to alter the SRSF10-dependent splicing of HIV-1 transcripts, with minor effects on cellular splicing, supports the view that SRSF10 may be used as a target for the development of new anti-viral agents.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Fármacos Anti-VIH/farmacología , Benzotiazoles/farmacología , Proteínas de Ciclo Celular/metabolismo , VIH-1/efectos de los fármacos , Niacinamida/análogos & derivados , Proteínas Represoras/metabolismo , Factores de Empalme Serina-Arginina/metabolismo , Replicación Viral/efectos de los fármacos , Fármacos Anti-VIH/química , Benzotiazoles/química , Células Cultivadas , VIH-1/genética , VIH-1/metabolismo , VIH-1/fisiología , Células HeLa , Humanos , Niacinamida/química , Niacinamida/farmacología , Precursores del ARN/metabolismo , Factores de Empalme de ARN/metabolismo , ARN Mensajero/metabolismo , Transcripción Genética/efectos de los fármacos
9.
Genes Dev ; 25(6): 608-20, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21406555

RESUMEN

Alternative splicing of precursor mRNA (pre-mRNA) is a strategy employed by most eukaryotes to increase transcript and proteomic diversity. Many metazoan splicing factors are members of multigene families, with each member having different functions. How these highly related proteins evolve unique properties has been unclear. Here we characterize the evolution and function of a new Drosophila splicing factor, termed LS2 (Large Subunit 2), that arose from a gene duplication event of dU2AF(50), the large subunit of the highly conserved heterodimeric general splicing factor U2AF (U2-associated factor). The quickly evolving LS2 gene has diverged from the splicing-promoting, ubiquitously expressed dU2AF(50) such that it binds a markedly different RNA sequence, acts as a splicing repressor, and is preferentially expressed in testes. Target transcripts of LS2 are also enriched for performing testes-related functions. We therefore propose a path for the evolution of a new splicing factor in Drosophila that regulates specific pre-mRNAs and contributes to transcript diversity in a tissue-specific manner.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Evolución Molecular , Empalme de Proteína/genética , Animales , Línea Celular , Drosophila melanogaster/clasificación , Duplicación de Gen/genética , Regulación de la Expresión Génica , Masculino , Proteínas Nucleares/genética , Especificidad de Órganos , Filogenia , Ribonucleoproteínas/genética , Factor de Empalme U2AF , Testículo/metabolismo
10.
Nature ; 484(7393): 260-4, 2012 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-22446625

RESUMEN

In most eukaryotes, the progressive loss of chromosome-terminal DNA sequences is counteracted by the enzyme telomerase, a reverse transcriptase that uses part of an RNA subunit as template to synthesize telomeric repeats. Many cancer cells express high telomerase activity, and mutations in telomerase subunits are associated with degenerative syndromes including dyskeratosis congenita and aplastic anaemia. The therapeutic value of altering telomerase activity thus provides ample impetus to study the biogenesis and regulation of this enzyme in human cells and model systems. We have previously identified a precursor of the fission yeast telomerase RNA subunit (TER1) and demonstrated that the mature 3'-end is generated by the spliceosome in a single cleavage reaction akin to the first step of splicing. Directly upstream and partly overlapping with the spliceosomal cleavage site is a putative binding site for Sm proteins. Sm and like-Sm (LSm) proteins belong to an ancient family of RNA-binding proteins represented in all three domains of life. Members of this family form ring complexes on specific sets of target RNAs and have critical roles in their biogenesis, function and turnover. Here we demonstrate that the canonical Sm ring and the Lsm2-8 complex sequentially associate with fission yeast TER1. The Sm ring binds to the TER1 precursor, stimulates spliceosomal cleavage and promotes the hypermethylation of the 5'-cap by Tgs1. Sm proteins are then replaced by the Lsm2-8 complex, which promotes the association with the catalytic subunit and protects the mature 3'-end of TER1 from exonucleolytic degradation. Our findings define the sequence of events that occur during telomerase biogenesis and characterize roles for Sm and Lsm complexes as well as for the methylase Tgs1.


Asunto(s)
Complejos Multiproteicos/metabolismo , Proteínas de Unión al ARN/metabolismo , ARN/biosíntesis , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Empalmosomas/metabolismo , Telomerasa/biosíntesis , Secuencia de Bases , Cromosomas Fúngicos/genética , Cromosomas Fúngicos/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Metiltransferasas/metabolismo , Complejos Multiproteicos/química , Unión Proteica , ARN/genética , Empalme del ARN , ARN de Hongos/genética , ARN de Hongos/metabolismo , Schizosaccharomyces/enzimología , Proteínas de Schizosaccharomyces pombe/genética , Telomerasa/genética , Telómero/genética , Telómero/metabolismo , ARNt Metiltransferasas/metabolismo
11.
PLoS Genet ; 11(7): e1005308, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26176819

RESUMEN

The Scc2-Scc4 complex is essential for loading the cohesin complex onto DNA. Cohesin has important roles in chromosome segregation, DSB repair, and chromosome condensation. Here we report that Scc2 is important for gene expression in budding yeast. Scc2 and the transcriptional regulator Paf1 collaborate to promote the production of Box H/ACA snoRNAs which guide pseudouridylation of RNAs including ribosomal RNA. Mutation of SCC2 was associated with defects in the production of ribosomal RNA, ribosome assembly, and splicing. While the scc2 mutant does not have a general defect in protein synthesis, it shows increased frameshifting and reduced cap-independent translation. These findings suggest Scc2 normally promotes a gene expression program that supports translational fidelity. We hypothesize that translational dysfunction may contribute to the human disorder Cornelia de Lange syndrome, which is caused by mutations in NIPBL, the human ortholog of SCC2.


Asunto(s)
Proteínas Cromosómicas no Histona/biosíntesis , Síndrome de Cornelia de Lange/genética , Biosíntesis de Proteínas , Proteínas/genética , ARN Largo no Codificante/biosíntesis , Proteínas de Saccharomyces cerevisiae/biosíntesis , Proteínas de Ciclo Celular/genética , Cromatina/genética , Proteínas Cromosómicas no Histona/genética , Síndrome de Cornelia de Lange/patología , Regulación Fúngica de la Expresión Génica , Humanos , Empalme del ARN/genética , ARN Largo no Codificante/genética , ARN Ribosómico/biosíntesis , ARN Ribosómico/genética , ARN Nucleolar Pequeño/biosíntesis , ARN Nucleolar Pequeño/genética , Ribosomas/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Cohesinas
12.
RNA ; 21(1): 75-92, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25414008

RESUMEN

SR proteins are a well-conserved class of RNA-binding proteins that are essential for regulation of splice-site selection, and have also been implicated as key regulators during other stages of RNA metabolism. For many SR proteins, the complexity of the RNA targets and specificity of RNA-binding location are poorly understood. It is also unclear if general rules governing SR protein alternative pre-mRNA splicing (AS) regulation uncovered for individual SR proteins on few model genes, apply to the activity of all SR proteins on endogenous targets. Using RNA-seq, we characterize the global AS regulation of the eight Drosophila SR protein family members. We find that a majority of AS events are regulated by multiple SR proteins, and that all SR proteins can promote exon inclusion, but also exon skipping. Most coregulated targets exhibit cooperative regulation, but some AS events are antagonistically regulated. Additionally, we found that SR protein levels can affect alternative promoter choices and polyadenylation site selection, as well as overall transcript levels. Cross-linking and immunoprecipitation coupled with high-throughput sequencing (iCLIP-seq), reveals that SR proteins bind a distinct and functionally diverse class of RNAs, which includes several classes of noncoding RNAs, uncovering possible novel functions of the SR protein family. Finally, we find that SR proteins exhibit positional RNA binding around regulated AS events. Therefore, regulation of AS by the SR proteins is the result of combinatorial regulation by multiple SR protein family members on most endogenous targets, and SR proteins have a broader role in integrating multiple layers of gene expression regulation.


Asunto(s)
Poliadenilación , Empalme del ARN , ARN Mensajero/genética , Proteínas de Unión al ARN/fisiología , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Secuencia de Consenso , Drosophila melanogaster , Regulación de la Expresión Génica , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , Estabilidad del ARN , ARN Mensajero/metabolismo , Transcripción Genética
13.
Nature ; 471(7339): 473-9, 2011 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21179090

RESUMEN

Drosophila melanogaster is one of the most well studied genetic model organisms; nonetheless, its genome still contains unannotated coding and non-coding genes, transcripts, exons and RNA editing sites. Full discovery and annotation are pre-requisites for understanding how the regulation of transcription, splicing and RNA editing directs the development of this complex organism. Here we used RNA-Seq, tiling microarrays and cDNA sequencing to explore the transcriptome in 30 distinct developmental stages. We identified 111,195 new elements, including thousands of genes, coding and non-coding transcripts, exons, splicing and editing events, and inferred protein isoforms that previously eluded discovery using established experimental, prediction and conservation-based approaches. These data substantially expand the number of known transcribed elements in the Drosophila genome and provide a high-resolution view of transcriptome dynamics throughout development.


Asunto(s)
Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Transcripción Genética/genética , Empalme Alternativo/genética , Animales , Secuencia de Bases , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Exones/genética , Femenino , Genes de Insecto/genética , Genoma de los Insectos/genética , Masculino , MicroARNs/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Isoformas de Proteínas/genética , Edición de ARN/genética , ARN Mensajero/análisis , ARN Mensajero/genética , ARN Pequeño no Traducido/análisis , ARN Pequeño no Traducido/genética , Análisis de Secuencia , Caracteres Sexuales
14.
Mol Cell ; 33(4): 438-49, 2009 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-19250905

RESUMEN

Heterogeneous nuclear ribonucleoproteins (hnRNPs) have been traditionally seen as proteins packaging RNA nonspecifically into ribonucleoprotein particles (RNPs), but evidence suggests specific cellular functions on discrete target pre-mRNAs. Here we report genome-wide analysis of alternative splicing patterns regulated by four Drosophila homologs of the mammalian hnRNP A/B family (hrp36, hrp38, hrp40, and hrp48). Analysis of the global RNA-binding distributions of each protein revealed both small and extensively bound regions on target transcripts. A significant subset of RNAs were bound and regulated by more than one hnRNP protein, revealing a combinatorial network of interactions. In vitro RNA-binding site selection experiments (SELEX) identified distinct binding motif specificities for each protein, which were overrepresented in their respective regulated and bound transcripts. These results indicate that individual heterogeneous ribonucleoproteins have specific affinities for overlapping, but distinct, populations of target pre-mRNAs controlling their patterns of RNA processing.


Asunto(s)
Empalme Alternativo/genética , Drosophila/genética , Genoma de los Insectos , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Precursores del ARN/metabolismo , ARN Mensajero/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Drosophila/metabolismo , Datos de Secuencia Molecular , Precursores del ARN/genética
15.
RNA ; 17(3): 453-68, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21233220

RESUMEN

In Drosophila melanogaster, female-specific expression of Sex-lethal (SXL) and Transformer (TRA) proteins controls sex-specific alternative splicing and/or translation of a handful of regulatory genes responsible for sexual differentiation and behavior. Recent findings in 2009 by Telonis-Scott et al. document widespread sex-biased alternative splicing in fruitflies, including instances of tissue-restricted sex-specific splicing. Here we report results arguing that some of these novel sex-specific splicing events are regulated by mechanisms distinct from those established by female-specific expression of SXL and TRA. Bioinformatic analysis of SXL/TRA binding sites, experimental analysis of sex-specific splicing in S2 and Kc cells lines and of the effects of SXL knockdown in Kc cells indicate that SXL-dependent and SXL-independent regulatory mechanisms coexist within the same cell. Additional determinants of sex-specific splicing can be provided by sex-specific differences in the expression of RNA binding proteins, including Hrp40/Squid. We report that sex-specific alternative splicing of the gene hrp40/squid leads to sex-specific differences in the levels of this hnRNP protein. The significant overlap between sex-regulated alternative splicing changes and those induced by knockdown of hrp40/squid and the presence of related sequence motifs enriched near subsets of Hrp40/Squid-regulated and sex-regulated splice sites indicate that this protein contributes to sex-specific splicing regulation. A significant fraction of sex-specific splicing differences are absent in germline-less tudor mutant flies. Intriguingly, these include alternative splicing events that are differentially spliced in tissues distant from the germline. Collectively, our results reveal that distinct genetic programs control widespread sex-specific splicing in Drosophila melanogaster.


Asunto(s)
Empalme Alternativo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas Nucleares/genética , Proteínas de Unión al ARN/genética , Animales , Biomarcadores/metabolismo , Western Blotting , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Femenino , Perfilación de la Expresión Génica , Genes Reguladores , Masculino , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores Sexuales
16.
Nucleic Acids Res ; 39(6): 2344-56, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21071404

RESUMEN

Splicing factor 1 (SF1) binds to the branch point sequence (BPS) of mammalian introns and is believed to be important for the splicing of some, but not all, introns. To help identify BPSs, particularly those that depend on SF1, we generated a BPS profile model in which SF1 binding affinity data, validated by branch point mapping, were iteratively incorporated into computational models. We searched a data set of 117,499 human introns for best matches to the SF1 Affinity Model above a threshold, and counted the number of matches at each intronic position. After subtracting a background value, we found that 87.9% of remaining high-scoring matches identified were located in a region upstream of 3'-splice sites where BPSs are typically found. Since U2AF65 recognizes the polypyrimidine tract (PPT) and forms a cooperative RNA complex with SF1, we combined the SF1 model with a PPT model computed from high affinity binding sequences for U2AF65. The combined model, together with binding site location constraints, accurately identified introns bound by SF1 that are candidates for SF1-dependent splicing.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Intrones , Modelos Genéticos , Factores de Transcripción/metabolismo , Secuencia de Bases , Sitios de Unión , Humanos , Factores de Empalme de ARN , ARN Mensajero/química , Análisis de Secuencia de ARN
17.
Nat Commun ; 14(1): 939, 2023 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-36805596

RESUMEN

Alternative Lengthening of Telomeres (ALT) is an aberrant DNA recombination pathway which grants replicative immortality to approximately 10% of all cancers. Despite this high prevalence of ALT in cancer, the mechanism and genetics by which cells activate this pathway remain incompletely understood. A major challenge in dissecting the events that initiate ALT is the extremely low frequency of ALT induction in human cell systems. Guided by the genetic lesions that have been associated with ALT from cancer sequencing studies, we genetically engineered primary human pluripotent stem cells to deterministically induce ALT upon differentiation. Using this genetically defined system, we demonstrate that disruption of the p53 and Rb pathways in combination with ATRX loss-of-function is sufficient to induce all hallmarks of ALT and results in functional immortalization in a cell type-specific manner. We further demonstrate that ALT can be induced in the presence of telomerase, is neither dependent on telomere shortening nor crisis, but is rather driven by continuous telomere instability triggered by the induction of differentiation in ATRX-deficient stem cells.


Asunto(s)
Células Madre Pluripotentes , Telomerasa , Humanos , Homeostasis del Telómero/genética , Telómero/genética , Diferenciación Celular/genética , Telomerasa/genética , Proteína Nuclear Ligada al Cromosoma X/genética
18.
bioRxiv ; 2023 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-37961446

RESUMEN

DNA looping is vital for establishing many enhancer-promoter interactions. While CTCF is known to anchor many cohesin-mediated loops, the looped chromatin fiber appears to predominantly exist in a poorly characterized actively extruding state. To better characterize extruding chromatin loop structures, we used CTCF MNase HiChIP data to determine both CTCF binding at high resolution and 3D contact information. Here we present FactorFinder, a tool that identifies CTCF binding sites at near base-pair resolution. We leverage this substantial advance in resolution to determine that the fully extruded (CTCF-CTCF) state is rare genome-wide with locus-specific variation from ~1-10%. We further investigate the impact of chromatin state on loop extrusion dynamics, and find that active enhancers and RNA Pol II impede cohesin extrusion, facilitating an enrichment of enhancer-promoter contacts in the partially extruded loop state. We propose a model of topological regulation whereby the transient, partially extruded states play active roles in transcription.

19.
Nat Commun ; 14(1): 7111, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37932252

RESUMEN

Chromosomal rearrangements can initiate and drive cancer progression, yet it has been challenging to evaluate their impact, especially in genetically heterogeneous solid cancers. To address this problem we developed HiDENSEC, a new computational framework for analyzing chromatin conformation capture in heterogeneous samples that can infer somatic copy number alterations, characterize large-scale chromosomal rearrangements, and estimate cancer cell fractions. After validating HiDENSEC with in silico and in vitro controls, we used it to characterize chromosome-scale evolution during melanoma progression in formalin-fixed tumor samples from three patients. The resulting comprehensive annotation of the genomic events includes copy number neutral translocations that disrupt tumor suppressor genes such as NF1, whole chromosome arm exchanges that result in loss of CDKN2A, and whole-arm copy-number neutral loss of homozygosity involving PTEN. These findings show that large-scale chromosomal rearrangements occur throughout cancer evolution and that characterizing these events yields insights into drivers of melanoma progression.


Asunto(s)
Aberraciones Cromosómicas , Melanoma , Humanos , Variaciones en el Número de Copia de ADN , Cromosomas , Translocación Genética , Melanoma/genética
20.
Nat Struct Mol Biol ; 14(12): 1134-40, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21188797

RESUMEN

The Down syndrome cell adhesion molecule (Dscam) gene has essential roles in neural wiring and pathogen recognition in Drosophila melanogaster. Dscam encodes 38,016 distinct isoforms via extensive alternative splicing. The 95 alternative exons in Dscam are organized into clusters that are spliced in a mutually exclusive manner. The exon 6 cluster contains 48 variable exons and uses a complex system of competing RNA structures to ensure that only one variable exon is included. Here we show that the heterogeneous nuclear ribonucleoprotein hrp36 acts specifically within, and throughout, the exon 6 cluster to prevent the inclusion of multiple exons. Moreover, hrp36 prevents serine/arginine-rich proteins from promoting the ectopic inclusion of multiple exon 6 variants. Thus, the fidelity of mutually exclusive splicing in the exon 6 cluster is governed by an intricate combination of alternative RNA structures and a globally acting splicing repressor.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Exones/genética , Ribonucleoproteínas Nucleares Heterogéneas/fisiología , Sitios de Empalme de ARN/genética , Animales , Drosophila melanogaster/genética , Regulación de la Expresión Génica , Humanos , Proteínas Nucleares , Análisis por Matrices de Proteínas , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA