Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Genes Dev ; 26(22): 2512-23, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23109676

RESUMEN

Telomerase replenishes telomere tracts by reiteratively copying its RNA template, TER. Unlike other model organisms, Arabidopsis thaliana harbors two divergent TER genes. However, only TER1 is required for telomere maintenance. Here we examine the function of TER2. We show that TER2 is spliced and its 3' end is truncated in vivo to generate a third TER isoform, TER2(S). TERT preferentially associates with TER2 > TER1 > TER2(S). Moreover, TER2 and TER2(S) assemble with Ku and POT1b (protection of telomeres), forming RNP (ribonucleoprotein) complexes distinct from TER1 RNP. Plants null for TER2 display increased telomerase enzyme activity, while TER2 overexpression inhibits telomere synthesis from TER1 and leads to telomere shortening. These findings argue that TER2 negatively regulates telomerase by sequestering TERT in a nonproductive RNP complex. Introduction of DNA double-strand breaks by zeocin leads to an immediate and specific spike in TER2 and a concomitant decrease in telomerase enzyme activity. This response is not triggered by replication stress or telomere dysfunction and is abrogated in ter2 mutants. We conclude that Arabidopsis telomerase is modulated by TER2, a novel DNA damage-induced noncoding RNA that works in concert with the canonical TER to promote genome integrity.


Asunto(s)
Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimología , Arabidopsis/genética , Daño del ADN/genética , ARN/genética , ARN/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , Activación Enzimática , Regulación de la Expresión Génica de las Plantas/genética , Procesamiento de Término de ARN 3' , Empalme del ARN , Ribonucleoproteínas/metabolismo , Telómero/genética
2.
Proc Natl Acad Sci U S A ; 112(11): E1272-7, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25733895

RESUMEN

High-grade serous ovarian cancer (HGSC) is among the most lethal forms of cancer in women. Excessive genomic rearrangements, which are expected to create fusion oncogenes, are the hallmark of this cancer. Here we report a cancer-specific gene fusion between BCAM, a membrane adhesion molecule, and AKT2, a key kinase in the PI3K signaling pathway. This fusion is present in 7% of the 60 patient cancers tested, a significant frequency considering the highly heterogeneous nature of this malignancy. Further, we provide direct evidence that BCAM-AKT2 is translated into an in-frame fusion protein in the patient's tumor. The resulting AKT2 fusion kinase is membrane-associated, constitutively phosphorylated, and activated as a functional kinase in cells. Unlike endogenous AKT2, whose activity is tightly regulated by external stimuli, BCAM-AKT2 escapes the regulation from external stimuli. Moreover, a BCAM-AKT2 fusion gene generated via chromosomal translocation using the CRISPR/Cas9 system leads to focus formation in both OVCAR8 and HEK-293T cell lines, suggesting that BCAM-AKT2 is oncogenic. Together, the results indicate that BCAM-AKT2 expression is a new mechanism of AKT2 kinase activation in HGSC. BCAM-AKT2 is the only fusion gene in HGSC that is proven to translate an aberrant yet functional kinase fusion protein with oncogenic properties. This recurrent genomic alteration is a potential therapeutic target and marker of a clinically relevant subtype for tailored therapy of HGSC.


Asunto(s)
Cistadenocarcinoma Seroso/enzimología , Cistadenocarcinoma Seroso/genética , Neoplasias Glandulares y Epiteliales/enzimología , Neoplasias Glandulares y Epiteliales/genética , Proteínas de Fusión Oncogénica/genética , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Secuencia de Aminoácidos , Secuencia de Bases , Sistemas CRISPR-Cas/genética , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Membrana Celular/enzimología , Cromosomas Humanos/genética , Cistadenocarcinoma Seroso/patología , Activación Enzimática , Femenino , Regulación Neoplásica de la Expresión Génica , Reordenamiento Génico/genética , Humanos , Datos de Secuencia Molecular , Clasificación del Tumor , Neoplasias Glandulares y Epiteliales/patología , Proteínas de Fusión Oncogénica/química , Neoplasias Ováricas/patología , Fosforilación , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas c-akt/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transfección , Translocación Genética
3.
PLoS Genet ; 10(3): e1004216, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24675677

RESUMEN

Ovarian cancer is the fifth leading cause of cancer death in women. Almost 70% of ovarian cancer deaths are due to the high-grade serous subtype, which is typically detected only after it has metastasized. Characterization of high-grade serous cancer is further complicated by the significant heterogeneity and genome instability displayed by this cancer. Other than mutations in TP53, which is common to many cancers, highly recurrent recombinant events specific to this cancer have yet to be identified. Using high-throughput transcriptome sequencing of seven patient samples combined with experimental validation at DNA, RNA and protein levels, we identified a cancer-specific and inter-chromosomal fusion gene CDKN2D-WDFY2 that occurs at a frequency of 20% among sixty high-grade serous cancer samples but is absent in non-cancerous ovary and fallopian tube samples. This is the most frequent recombinant event identified so far in high-grade serous cancer implying a major cellular lineage in this highly heterogeneous cancer. In addition, the same fusion transcript was also detected in OV-90, an established high-grade serous type cell line. The genomic breakpoint was identified in intron 1 of CDKN2D and intron 2 of WDFY2 in patient tumor, providing direct evidence that this is a fusion gene. The parental gene, CDKN2D, is a cell-cycle modulator that is also involved in DNA repair, while WDFY2 is known to modulate AKT interactions with its substrates. Transfection of cloned fusion construct led to loss of wildtype CDKN2D and wildtype WDFY2 protein expression, and a gain of a short WDFY2 protein isoform that is presumably under the control of the CDKN2D promoter. The expression of short WDFY2 protein in transfected cells appears to alter the PI3K/AKT pathway that is known to play a role in oncogenesis. CDKN2D-WDFY2 fusion could be an important molecular signature for understanding and classifying sub-lineages among heterogeneous high-grade serous ovarian carcinomas.


Asunto(s)
Inhibidor p19 de las Quinasas Dependientes de la Ciclina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de Fusión Oncogénica/genética , Neoplasias Ováricas/genética , Biomarcadores de Tumor/genética , Trompas Uterinas/metabolismo , Trompas Uterinas/patología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Mutación , Clasificación del Tumor , Neoplasias Ováricas/patología , Translocación Genética/genética , Proteína p53 Supresora de Tumor/genética
4.
Bioinformatics ; 29(9): 1174-81, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23505294

RESUMEN

SUMMARY: Gene fusions are being discovered at an increasing rate using massively parallel sequencing technologies. Prioritization of cancer fusion drivers for validation cannot be performed using traditional single-gene based methods because fusions involve portions of two partner genes. To address this problem, we propose a novel network analysis method called fusion centrality that is specifically tailored for prioritizing gene fusions. We first propose a domain-based fusion model built on the theory of exon/domain shuffling. The model leads to a hypothesis that a fusion is more likely to be an oncogenic driver if its partner genes act like hubs in a network because the fusion mutation can deregulate normal functions of many other genes and their pathways. The hypothesis is supported by the observation that for most known cancer fusion genes, at least one of the fusion partners appears to be a hub in a network, and even for many fusions both partners appear to be hubs. Based on this model, we construct fusion centrality, a multi-gene-based network metric, and use it to score fusion drivers. We show that the fusion centrality outperforms other single gene-based methods. Specifically, the method successfully predicts most of 38 newly discovered fusions that had validated oncogenic importance. To our best knowledge, this is the first network-based approach for identifying fusion drivers. AVAILABILITY: Matlab code implementing the fusion centrality method is available upon request from the corresponding authors.


Asunto(s)
Redes Reguladoras de Genes , Genes Relacionados con las Neoplasias , Fusión de Oncogenes , Fusión Génica , Humanos , Modelos Genéticos , Mutación , Neoplasias/genética , Proteínas de Fusión Oncogénica/química , Estructura Terciaria de Proteína
5.
Proc Natl Acad Sci U S A ; 108(1): 73-78, 2011 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-21164032

RESUMEN

Telomerase is a ribonucleoprotein (RNP) reverse transcriptase whose essential RNA subunit (TER) functions as a template for telomere repeat synthesis. Here we report the identification of two divergent TER moieties in the flowering plant Arabidopsis thaliana. Although both TER1 and TER2 copurify with telomerase activity and serve as templates for telomerase in vitro, depletion of TER1, but not TER2, leads to decreased telomerase activity and progressive telomere shortening in vivo. Moreover, mutation of the templating domain in TER1 results in the incorporation of mutant telomere repeats on chromosome ends. Thus, TER1 provides the major template for telomerase in vivo. We also show that POT1a binds TER1 with a Kd of 2 × 10(-7) M and the two components assemble into an enzymatically active RNP in vivo. In contrast, TER1-POT1b and TER2-POT1a associations were not observed. In other organisms POT1 proteins bind telomeric DNA and provide chromosome end protection. We propose that duplication of TER and POT1 in Arabidopsis fueled the evolution of novel protein-nucleic acid interactions and the migration of POT1 from the telomere to the telomerase RNP.


Asunto(s)
Proteínas de Arabidopsis/genética , Arabidopsis/enzimología , Proteínas Nucleares/genética , Subunidades de Proteína/genética , ARN/genética , Telomerasa/genética , Secuencia de Bases , Northern Blotting , Western Blotting , Cartilla de ADN/genética , Duplicación de Gen/genética , Inmunoprecipitación , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación/genética , Técnicas de Amplificación de Ácido Nucleico , Oligonucleótidos/genética , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Telomerasa/metabolismo
6.
Proc Natl Acad Sci U S A ; 108(22): 9172-7, 2011 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-21571633

RESUMEN

Transcription-induced chimeric RNAs, possessing sequences from different genes, are expected to increase the proteomic diversity through chimeric proteins or altered regulation. Despite their importance, few studies have focused on chimeric RNAs especially regarding their presence/roles in human cancers. By deep sequencing the transcriptome of 20 human prostate cancer and 10 matched benign prostate tissues, we obtained 1.3 billion sequence reads, which led to the identification of 2,369 chimeric RNA candidates. Chimeric RNAs occurred in significantly higher frequency in cancer than in matched benign samples. Experimental investigation of a selected 46 set led to the confirmation of 32 chimeric RNAs, of which 27 were highly recurrent and previously undescribed in prostate cancer. Importantly, a subset of these chimeras was present in prostate cancer cell lines, but not detectable in primary human prostate epithelium cells, implying their associations with cancer. These chimeras contain discernable 5' and 3' splice sites at the RNA junction, indicating that their formation is mediated by splicing. Their presence is also largely independent of the expression of parental genes, suggesting that other factors are involved in their production and regulation. One chimera, TMEM79-SMG5, is highly differentially expressed in human cancer samples and therefore a potential biomarker. The prevalence of chimeric RNAs may allow the limited number of human genes to encode a substantially larger number of RNAs and proteins, forming an additional layer of cellular complexity. Together, our results suggest that chimeric RNAs are widespread, and increased chimeric RNA events could represent a unique class of molecular alteration in cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Neoplasias de la Próstata/metabolismo , Análisis de Secuencia de ADN , Empalme Alternativo , Estudios de Cohortes , Biología Computacional/métodos , Bases de Datos Genéticas , Perfilación de la Expresión Génica , Humanos , Masculino , Prostatectomía , Neoplasias de la Próstata/genética , ARN/metabolismo
7.
Cancers (Basel) ; 7(4): 2083-93, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26492273

RESUMEN

High-grade serous ovarian cancer (HGSC) is among the most lethal forms of cancer in women. By analyzing the mRNA-seq reads from The Cancer Genome Atlas (TCGA), we uncovered a novel cancer-enriched chimeric RNA as the result of splicing between MUC1, a highly glycosylated transmembrane mucin, TRIM46, a tripartite motif containing protein, and KRTCAP2, a keratinocyte associated protein. Experimental analyses by RT-PCR (reverse transcription PCR) and Sanger sequencing using an in-house cohort of 59 HGSC patient tumors revealed a total of six MUC1-TRIM46-KRTCAP2 isoforms joined by different annotated splice sites between these genes. These chimeric isoforms are not detected in non-cancerous ovaries, yet are present in three out of every four HGSC patient tumors, a significant frequency given the exceedingly heterogeneous nature of this disease. Transfection of the cDNA of MUC1-TRIM46-KRTCAP2 isoforms in mammalian cells led to the translation of mutant MUC1 fusion proteins that are unglycosylated and cytoplasmically localized as opposed to the cell membrane, a feature resembling the tumor-associated MUC1. Because the parental MUC1 is overexpressed in 90% of HGSC tumors and has been proposed as a clinical biomarker and therapeutic target, the chimeric MUC1-TRIM46-KRTCAP2 isoforms identified in this report could represent significantly better MUC1 variants for the same clinical utilities.

8.
Oncotarget ; 4(11): 2135-43, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24243830

RESUMEN

It is increasingly recognized that chimeric RNAs may exert a novel layer of cellular complexity that contributes to oncogenesis and cancer progression, and could be utilized as molecular biomarkers and therapeutic targets. To date yet no fusion chimeric RNAs have been identified in esophageal cancer, the 6th most frequent cause of cancer death in the world. While analyzing the expression of 32 recurrent cancer chimeric RNAs in esophageal squamous cell carcinoma (ESCC) from patients and cancer cell lines, we identified GOLM1-MAK10, as a highly cancer-enriched chimeric RNA in ESCC. In situ hybridization revealed that the expression of the chimera is largely restricted to cancer cells in patient tumors, and nearly undetectable in non-neoplastic esophageal tissue from normal subjects. The aberrant chimera closely correlated with histologic differentiation and lymph node metastasis. Furthermore, we demonstrate that chimera GOLM1-MAK10 encodes a secreted fusion protein. Mechanistic studies reveal that GOLM1-MAK10 is likely derived from transcription read-through/splicing rather than being generated from a fusion gene. Collectively, these findings provide novel insights into the molecular mechanism involved in ESCC and provide a novel potential target for future therapies. The secreted fusion protein translated from GOLM1-MAK10 could also serve as a unique protein signature detectable by standard non-invasive assays. These observations are critical as there is no clinically useful molecular signature available for detecting this deadly disease or monitoring the treatment response.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Proteínas de la Membrana/genética , Acetiltransferasa C N-Terminal/genética , Proteínas de Fusión Oncogénica/genética , ARN/genética , Anciano , Secuencia de Bases , Quimerismo , Estudios de Cohortes , Carcinoma de Células Escamosas de Esófago , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Hibridación in Situ , Masculino , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Acetiltransferasa C N-Terminal/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Pronóstico , Transcriptoma , Transfección
9.
Mol Cell Biol ; 28(7): 2332-41, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18212040

RESUMEN

Dyskerin binds the H/ACA box of human telomerase RNA and is a core telomerase subunit required for RNP biogenesis and enzyme function in vivo. Missense mutations in dyskerin result in dyskeratosis congenita, a complex syndrome characterized by bone marrow failure, telomerase enzyme deficiency, and progressive telomere shortening. Here we demonstrate that dyskerin also contributes to telomere maintenance in Arabidopsis thaliana. We report that both AtNAP57, the Arabidopsis dyskerin homolog, and AtTERT, the telomerase catalytic subunit, accumulate in the plant nucleolus, and AtNAP57 associates with active telomerase RNP particles in an RNA-dependent manner. Furthermore, AtNAP57 interacts in vitro with AtPOT1a, a novel component of Arabidopsis telomerase. Although a null mutation in AtNAP57 is lethal, AtNAP57, like AtTERT, is not haploinsufficient for telomere maintenance in Arabidopsis. However, introduction of an AtNAP57 allele containing a T66A mutation decreased telomerase activity in vitro, disrupted telomere length regulation on individual chromosome ends in vivo, and established a new, shorter telomere length set point. These results imply that T66A NAP57 behaves as a dominant-negative inhibitor of telomerase. We conclude that dyskerin is a conserved component of the telomerase RNP complex in higher eukaryotes that is required for maximal enzyme activity in vivo.


Asunto(s)
Proteínas de Arabidopsis/fisiología , Cromosomas de las Plantas/ultraestructura , Hidroliasas/fisiología , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/fisiología , Telomerasa/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Telómero/ultraestructura , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Células Cultivadas/metabolismo , Células Cultivadas/ultraestructura , Cromosomas de las Plantas/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Humanos , Hidroliasas/genética , Datos de Secuencia Molecular , Mutación Missense , Plantas Modificadas Genéticamente , Mutación Puntual , Mapeo de Interacción de Proteínas , Procesamiento Postranscripcional del ARN , ARN de Planta/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Complejo Shelterina , Telómero/metabolismo , Uridina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA