Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Neoplasia ; 42: 100910, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37267699

RESUMEN

BACKGROUND: Biliary tract cancers (BTCs) including intrahepatic, perihilar, and distal cholangiocarcinoma as well as gallbladder cancer, are rare but aggressive malignancies with few effective standard of care therapies. METHODS: We implemented integrative clinical sequencing of advanced BTC tumors from 124 consecutive patients who progressed on standard therapies (N=92 with MI-ONCOSEQ and N=32 with commercial gene panels) enrolled between 2011-2020. RESULTS: Genomic profiling of paired tumor and normal DNA and tumor transcriptome (RNA) sequencing identified actionable somatic and germline genomic alterations in 54 patients (43.5%), and potentially actionable alterations in 79 (63.7%) of the cohort. Of these, patients who received matched targeted therapy (22; 40.7%) had a median overall survival of 28.1 months compared to 13.3 months in those who did not receive matched targeted therapy (32; P < 0.01), or 13.9 months in those without actionable mutations (70; P < 0.01). Additionally, we discovered recurrent activating mutations in FGFR2, and a novel association between KRAS and BRAF mutant tumors with high expression of immune modulatory protein NT5E (CD73) that may represent novel therapeutic avenues. CONCLUSIONS: Overall, the identification of actionable/ potentially actionable aberrations in a large proportion of cases, and improvement in survival with precision oncology supports molecular analysis and clinical sequencing for all patients with advanced BTC.


Asunto(s)
Neoplasias de los Conductos Biliares , Neoplasias del Sistema Biliar , Humanos , Medicina de Precisión , Neoplasias de los Conductos Biliares/genética , Neoplasias del Sistema Biliar/genética , Neoplasias del Sistema Biliar/terapia , Neoplasias del Sistema Biliar/patología , Mutación , Genómica , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología
2.
Neoplasia ; 36: 100872, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36621024

RESUMEN

PURPOSE: Glioblastoma(GBM) is a lethal disease characterized by inevitable recurrence. Here we investigate the molecular pathways mediating resistance, with the goal of identifying novel therapeutic opportunities. EXPERIMENTAL DESIGN: We developed a longitudinal in vivo recurrence model utilizing patient-derived explants to produce paired specimens(pre- and post-recurrence) following temozolomide(TMZ) and radiation(IR). These specimens were evaluated for treatment response and to identify gene expression pathways driving treatment resistance. Findings were clinically validated using spatial transcriptomics of human GBMs. RESULTS: These studies reveal in replicate cohorts, a gene expression profile characterized by upregulation of mesenchymal and stem-like genes at recurrence. Analyses of clinical databases revealed significant association of this transcriptional profile with worse overall survival and upregulation at recurrence. Notably, gene expression analyses identified upregulation of TGFß signaling, and more than one-hundred-fold increase in THY1 levels at recurrence. Furthermore, THY1-positive cells represented <10% of cells in treatment-naïve tumors, compared to 75-96% in recurrent tumors. We then isolated THY1-positive cells from treatment-naïve patient samples and determined that they were inherently resistant to chemoradiation in orthotopic models. Additionally, using image-guided biopsies from treatment-naïve human GBM, we conducted spatial transcriptomic analyses. This revealed rare THY1+ regions characterized by mesenchymal/stem-like gene expression, analogous to our recurrent mouse model, which co-localized with macrophages within the perivascular niche. We then inhibited TGFBRI activity in vivo which decreased mesenchymal/stem-like protein levels, including THY1, and restored sensitivity to TMZ/IR in recurrent tumors. CONCLUSIONS: These findings reveal that GBM recurrence may result from tumor repopulation by pre-existing, therapy-resistant, THY1-positive, mesenchymal cells within the perivascular niche.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Animales , Ratones , Humanos , Glioblastoma/metabolismo , Línea Celular Tumoral , Neoplasias Encefálicas/patología , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/tratamiento farmacológico , Temozolomida/farmacología , Resistencia a Antineoplásicos/genética , Antineoplásicos Alquilantes/farmacología
3.
PNAS Nexus ; 1(3): pgac084, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35923912

RESUMEN

Activating mutations in RAS GTPases drive nearly 30% of all human cancers. Our prior work described an essential role for Argonaute 2 (AGO2), of the RNA-induced silencing complex, in mutant KRAS-driven cancers. Here, we identified a novel endogenous interaction between AGO2 and RAS in both wild-type (WT) and mutant HRAS/NRAS cells. This interaction was regulated through EGFR-mediated phosphorylation of Y393-AGO2, and utilizing molecular dynamic simulation, we identified a conformational change in pY393-AGO2 protein structure leading to disruption of the RAS binding site. Knockdown of AGO2 led to a profound decrease in proliferation of mutant HRAS/NRAS-driven cell lines but not WT RAS cells. These cells demonstrated oncogene-induced senescence (OIS) as evidenced by ß-galactosidase staining and induction of multiple downstream senescence effectors. Mechanistically, we discovered that the senescent phenotype was mediated via induction of reactive oxygen species. Intriguingly, we further identified that loss of AGO2 promoted a novel feed forward pathway leading to inhibition of the PTP1B phosphatase and activation of EGFR-MAPK signaling, consequently resulting in OIS. Taken together, our study demonstrates that the EGFR-AGO2-RAS signaling axis is essential for maintaining mutant HRAS and NRAS-driven malignancies.

4.
Biochem Biophys Rep ; 29: 101191, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34988297

RESUMEN

Oncogenic mutations in KRAS result in a constitutively active, GTP-bound form that in turn activates many proliferative pathways. However, because of its compact and simple architecture, directly targeting KRAS with small molecule drugs has been challenging. Another approach is to identify targetable proteins that interact with KRAS. Argonaute 2 (AGO2) was recently identified as a protein that facilitates RAS-driven oncogenesis. Whereas previous studies described the in vivo effect of AGO2 on cancer progression in cells harboring mutated KRAS, here we sought to examine their direct interaction using purified proteins. We show that full length AGO2 co-immunoprecipitates with KRAS using purified components, however, a complex between FL AGO2 and KRAS could not be isolated. We also generated a smaller N-terminal fragment of AGO2 (NtAGO2) which is believed to represent the primary binding site of KRAS. A complex with NtAGO2 could be detected via ion-mobility mass spectrometry and size exclusion chromatography. However, the data suggest that the interaction of KRAS with purified AGO2 (NtAGO2 or FL AGO2) is weak and likely requires additional cellular components or proteo-forms of AGO2 that are not readily available in our purified assay systems. Future studies are needed to determine what conformation or modifications of AGO2 are necessary to enrich KRAS association and regulate its activities.

5.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33972443

RESUMEN

Lung cancer is the deadliest malignancy in the United States. Non-small cell lung cancer (NSCLC) accounts for 85% of cases and is frequently driven by activating mutations in the gene encoding the KRAS GTPase (e.g., KRASG12D). Our previous work demonstrated that Argonaute 2 (AGO2)-a component of the RNA-induced silencing complex (RISC)-physically interacts with RAS and promotes its downstream signaling. We therefore hypothesized that AGO2 could promote KRASG12D-dependent NSCLC in vivo. To test the hypothesis, we evaluated the impact of Ago2 knockout in the KPC (LSL-KrasG12D/+;p53f/f;Cre) mouse model of NSCLC. In KPC mice, intratracheal delivery of adenoviral Cre drives lung-specific expression of a stop-floxed KRASG12D allele and biallelic ablation of p53 Simultaneous biallelic ablation of floxed Ago2 inhibited KPC lung nodule growth while reducing proliferative index and improving pathological grade. We next applied the KPHetC model, in which the Clara cell-specific CCSP-driven Cre activates KRASG12D and ablates a single p53 allele. In these mice, Ago2 ablation also reduced tumor size and grade. In both models, Ago2 knockout inhibited ERK phosphorylation (pERK) in tumor cells, indicating impaired KRAS signaling. RNA sequencing (RNA-seq) of KPC nodules and nodule-derived organoids demonstrated impaired canonical KRAS signaling with Ago2 ablation. Strikingly, accumulation of pERK in KPC organoids depended on physical interaction of AGO2 and KRAS. Taken together, our data demonstrate a pathogenic role for AGO2 in KRAS-dependent NSCLC. Given the prevalence of this malignancy and current difficulties in therapeutically targeting KRAS signaling, our work may have future translational relevance.


Asunto(s)
Proteínas Argonautas/fisiología , Carcinoma de Pulmón de Células no Pequeñas/etiología , Neoplasias Pulmonares/etiología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Neoplasias Pulmonares/genética , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología
6.
Nat Commun ; 11(1): 2817, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32499547

RESUMEN

Both KRAS and EGFR are essential mediators of pancreatic cancer development and interact with Argonaute 2 (AGO2) to perturb its function. Here, in a mouse model of mutant KRAS-driven pancreatic cancer, loss of AGO2 allows precursor lesion (PanIN) formation yet prevents progression to pancreatic ductal adenocarcinoma (PDAC). Precursor lesions with AGO2 ablation undergo oncogene-induced senescence with altered microRNA expression and EGFR/RAS signaling, bypassed by loss of p53. In mouse and human pancreatic tissues, PDAC progression is associated with increased plasma membrane localization of RAS/AGO2. Furthermore, phosphorylation of AGO2Y393 disrupts both the wild-type and oncogenic KRAS-AGO2 interaction, albeit under different conditions. ARS-1620 (G12C-specific inhibitor) disrupts the KRASG12C-AGO2 interaction, suggesting that the interaction is targetable. Altogether, our study supports a biphasic model of pancreatic cancer development: an AGO2-independent early phase of PanIN formation reliant on EGFR-RAS signaling, and an AGO2-dependent phase wherein the mutant KRAS-AGO2 interaction is critical for PDAC progression.


Asunto(s)
Proteínas Argonautas/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Alelos , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Senescencia Celular , Progresión de la Enfermedad , Receptores ErbB/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Genotipo , Humanos , Masculino , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Neoplasias Pancreáticas/patología , Fosforilación , Unión Proteica , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo
7.
Cell Rep ; 14(6): 1448-1461, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26854235

RESUMEN

Oncogenic mutations in RAS provide a compelling yet intractable therapeutic target. Using co-immunoprecipitation mass spectrometry, we uncovered an interaction between RAS and Argonaute 2 (AGO2). Endogenously, RAS and AGO2 co-sediment and co-localize in the endoplasmic reticulum. The AGO2 N-terminal domain directly binds the Switch II region of KRAS, agnostic of nucleotide (GDP/GTP) binding. Functionally, AGO2 knockdown attenuates cell proliferation in mutant KRAS-dependent cells and AGO2 overexpression enhances KRAS(G12V)-mediated transformation. Using AGO2-/- cells, we demonstrate that the RAS-AGO2 interaction is required for maximal mutant KRAS expression and cellular transformation. Mechanistically, oncogenic KRAS attenuates AGO2-mediated gene silencing. Overall, the functional interaction with AGO2 extends KRAS function beyond its canonical role in signaling.


Asunto(s)
Proteínas Argonautas/genética , Transformación Celular Neoplásica/genética , Retículo Endoplásmico/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Proteínas Argonautas/antagonistas & inhibidores , Proteínas Argonautas/metabolismo , Secuencia de Bases , Carboxipeptidasas/genética , Carboxipeptidasas/metabolismo , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Silenciador del Gen , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Datos de Secuencia Molecular , Mutación , Células 3T3 NIH , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transgenes
8.
Neoplasia ; 16(2): 115-28, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24709419

RESUMEN

Attempts to target mutant KRAS have been unsuccessful. Here, we report the identification of Smad ubiquitination regulatory factor 2 (SMURF2) and UBCH5 as a critical E3:E2 complex maintaining KRAS protein stability. Loss of SMURF2 either by small interfering RNA/short hairpin RNA (siRNA/shRNA) or by overexpression of a catalytically inactive mutant causes KRAS degradation, whereas overexpression of wild-type SMURF2 enhances KRAS stability. Importantly, mutant KRAS is more susceptible to SMURF2 loss where protein half-life decreases from >12 hours in control siRNA-treated cells to <3 hours on Smurf2 silencing, whereas only marginal differences were noted for wild-type protein. This loss of mutant KRAS could be rescued by overexpressing a siRNA-resistant wild-type SMURF2. Our data further show that SMURF2 monoubiquitinates UBCH5 at lysine 144 to form an active complex required for efficient degradation of a RAS-family E3, ß-transducing repeat containing protein 1 (ß-TrCP1). Conversely, ß-TrCP1 is accumulated on SMURF2 loss, leading to increased KRAS degradation. Therefore, as expected, ß-TrCP1 knockdown following Smurf2 siRNA treatment rescues mutant KRAS loss. Further, we identify two conserved proline (P) residues in UBCH5 critical for SMURF2 interaction; mutation of either of these P to alanine also destabilizes KRAS. As a proof of principle, we demonstrate that Smurf2 silencing reduces the clonogenic survival in vitro and prolongs tumor latency in vivo in cancer cells including mutant KRAS-driven tumors. Taken together, we show that SMURF2:UBCH5 complex is critical in maintaining KRAS protein stability and propose that targeting such complex may be a unique strategy to degrade mutant KRAS to kill cancer cells.


Asunto(s)
Proteínas Proto-Oncogénicas/metabolismo , Enzimas Ubiquitina-Conjugadoras/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Proteínas con Repetición de beta-Transducina/fisiología , Proteínas ras/metabolismo , Adenocarcinoma , Adenocarcinoma del Pulmón , Secuencias de Aminoácidos , Animales , Femenino , Expresión Génica , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Neoplasias Pulmonares , Ratones Desnudos , Trasplante de Neoplasias , Unión Proteica , Estabilidad Proteica , Proteolisis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Ubiquitinación , Proteínas ras/genética
9.
Nat Commun ; 4: 2617, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24135919

RESUMEN

Global 'multi-omics' profiling of cancer cells harbours the potential for characterizing the signalling networks associated with specific oncogenes. Here we profile the transcriptome, proteome and phosphoproteome in a panel of non-small cell lung cancer (NSCLC) cell lines in order to reconstruct targetable networks associated with KRAS dependency. We develop a two-step bioinformatics strategy addressing the challenge of integrating these disparate data sets. We first define an 'abundance-score' combining transcript, protein and phospho-protein abundances to nominate differentially abundant proteins and then use the Prize Collecting Steiner Tree algorithm to identify functional sub-networks. We identify three modules centred on KRAS and MET, LCK and PAK1 and ß-Catenin. We validate activation of these proteins in KRAS-dependent (KRAS-Dep) cells and perform functional studies defining LCK as a critical gene for cell proliferation in KRAS-Dep but not KRAS-independent NSCLCs. These results suggest that LCK is a potential druggable target protein in KRAS-Dep lung cancers.


Asunto(s)
Antineoplásicos/farmacología , Biología Computacional , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Fosfoproteínas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Algoritmos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Redes Reguladoras de Genes , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Terapia Molecular Dirigida , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , Transducción de Señal , Transcriptoma , beta Catenina/genética , beta Catenina/metabolismo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Proteínas ras/metabolismo
10.
Cancer Discov ; 3(3): 280-93, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23384775

RESUMEN

Protein kinases represent the most effective class of therapeutic targets in cancer; therefore, determination of kinase aberrations is a major focus of cancer genomic studies. Here, we analyzed transcriptome sequencing data from a compendium of 482 cancer and benign samples from 25 different tissue types, and defined distinct "outlier kinases" in individual breast and pancreatic cancer samples, based on highest levels of absolute and differential expression. Frequent outlier kinases in breast cancer included therapeutic targets like ERBB2 and FGFR4, distinct from MET, AKT2, and PLK2 in pancreatic cancer. Outlier kinases imparted sample-specific dependencies in various cell lines, as tested by siRNA knockdown and/or pharmacologic inhibition. Outlier expression of polo-like kinases was observed in a subset of KRAS-dependent pancreatic cancer cell lines, and conferred increased sensitivity to the pan-PLK inhibitor BI-6727. Our results suggest that outlier kinases represent effective precision therapeutic targets that are readily identifiable through RNA sequencing of tumors.


Asunto(s)
Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/biosíntesis , Análisis de Secuencia de ARN/métodos , Animales , Línea Celular Tumoral , Perfilación de la Expresión Génica/métodos , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Quinasas/genética , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Interferencia de ARN , Distribución Aleatoria , Receptor ErbB-2/biosíntesis , Receptor ErbB-2/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/biosíntesis , Proteínas ras/genética
11.
Neoplasia ; 14(8): 702-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22952423

RESUMEN

Application of high-throughput transcriptome sequencing has spurred highly sensitive detection and discovery of gene fusions in cancer, but distinguishing potentially oncogenic fusions from random, "passenger" aberrations has proven challenging. Here we examine a distinctive group of gene fusions that involve genes present in the loci of chromosomal amplifications--a class of oncogenic aberrations that are widely prevalent in breast cancers. Integrative analysis of a panel of 14 breast cancer cell lines comparing gene fusions discovered by high-throughput transcriptome sequencing and genome-wide copy number aberrations assessed by array comparative genomic hybridization, led to the identification of 77 gene fusions, of which more than 60% were localized to amplicons including 17q12, 17q23, 20q13, chr8q, and others. Many of these fusions appeared to be recurrent or involved highly expressed oncogenic drivers, frequently fused with multiple different partners, but sometimes displaying loss of functional domains. As illustrative examples of the "amplicon-associated" gene fusions, we examined here a recurrent gene fusion involving the mediator of mammalian target of rapamycin signaling, RPS6KB1 kinase in BT-474, and the therapeutically important receptor tyrosine kinase EGFR in MDA-MB-468 breast cancer cell line. These gene fusions comprise a minor allelic fraction relative to the highly expressed full-length transcripts and encode chimera lacking the kinase domains, which do not impart dependence on the respective cells. Our study suggests that amplicon-associated gene fusions in breast cancer primarily represent a by-product of chromosomal amplifications, which constitutes a subset of passenger aberrations and should be factored accordingly during prioritization of gene fusion candidates.


Asunto(s)
Neoplasias de la Mama/genética , Receptores ErbB/genética , Amplificación de Genes , Fusión Génica , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Secuencia de Bases , Línea Celular Tumoral , Mapeo Cromosómico , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN , Femenino , Dosificación de Gen , Perfilación de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Interferencia de ARN , ARN Interferente Pequeño , Análisis de Secuencia de ADN
12.
Cell ; 149(7): 1622-34, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22726445

RESUMEN

Pseudogene transcripts can provide a novel tier of gene regulation through generation of endogenous siRNAs or miRNA-binding sites. Characterization of pseudogene expression, however, has remained confined to anecdotal observations due to analytical challenges posed by the extremely close sequence similarity with their counterpart coding genes. Here, we describe a systematic analysis of pseudogene "transcription" from an RNA-Seq resource of 293 samples, representing 13 cancer and normal tissue types, and observe a surprisingly prevalent, genome-wide expression of pseudogenes that could be categorized as ubiquitously expressed or lineage and/or cancer specific. Further, we explore disease subtype specificity and functions of selected expressed pseudogenes. Taken together, we provide evidence that transcribed pseudogenes are a significant contributor to the transcriptional landscape of cells and are positioned to play significant roles in cellular differentiation and cancer progression, especially in light of the recently described ceRNA networks. Our work provides a transcriptome resource that enables high-throughput analyses of pseudogene expression.


Asunto(s)
Estudio de Asociación del Genoma Completo , Neoplasias/genética , Seudogenes/genética , Transcriptoma , Secuencia de Aminoácidos , Secuencia de Bases , Neoplasias de la Mama/genética , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Neoplasias de la Próstata/genética , Análisis de Secuencia de ARN
13.
Cancer Discov ; 1(1): 35-43, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22140652

RESUMEN

UNLABELLED: Using an integrative genomics approach called amplification breakpoint ranking and assembly analysis, we nominated KRAS as a gene fusion with the ubiquitin-conjugating enzyme UBE2L3 in the DU145 cell line, originally derived from prostate cancer metastasis to the brain. Interestingly, analysis of tissues revealed that 2 of 62 metastatic prostate cancers harbored aberrations at the KRAS locus. In DU145 cells, UBE2L3-KRAS produces a fusion protein, a specific knockdown of which attenuates cell invasion and xenograft growth. Ectopic expression of the UBE2L3-KRAS fusion protein exhibits transforming activity in NIH 3T3 fibroblasts and RWPE prostate epithelial cells in vitro and in vivo. In NIH 3T3 cells, UBE2L3-KRAS attenuates MEK/ERK signaling, commonly engaged by oncogenic mutant KRAS, and instead signals via AKT and p38 mitogen-activated protein kinase (MAPK) pathways. This is the first report of a gene fusion involving the Ras family, suggesting that this aberration may drive metastatic progression in a rare subset of prostate cancers. SIGNIFICANCE: This is the first description of an oncogenic gene fusion of KRAS, one of the most studied proto-oncogenes. KRAS rearrangement may represent the driving mutation in a rare subset of metastatic prostate cancers, emphasizing the importance of RAS-RAF-MAPK signaling in this disease.


Asunto(s)
Reordenamiento Génico , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Animales , Línea Celular Tumoral , Quinasas MAP Reguladas por Señal Extracelular/genética , Fusión Génica , Células HEK293 , Humanos , Quinasas Quinasa Quinasa PAM/genética , Masculino , Ratones , Mutación , Células 3T3 NIH , Proteínas Proto-Oncogénicas c-akt/genética , Enzimas Ubiquitina-Conjugadoras/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
14.
Nat Med ; 17(12): 1646-51, 2011 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-22101766

RESUMEN

Breast cancer is a heterogeneous disease that has a wide range of molecular aberrations and clinical outcomes. Here we used paired-end transcriptome sequencing to explore the landscape of gene fusions in a panel of breast cancer cell lines and tissues. We observed that individual breast cancers have a variety of expressed gene fusions. We identified two classes of recurrent gene rearrangements involving genes encoding microtubule-associated serine-threonine kinase (MAST) and members of the Notch family. Both MAST and Notch-family gene fusions have substantial phenotypic effects in breast epithelial cells. Breast cancer cell lines harboring Notch gene rearrangements are uniquely sensitive to inhibition of Notch signaling, and overexpression of MAST1 or MAST2 gene fusions has a proliferative effect both in vitro and in vivo. These findings show that recurrent gene rearrangements have key roles in subsets of carcinomas and suggest that transcriptome sequencing could identify individuals with rare, targetable gene fusions.


Asunto(s)
Neoplasias de la Mama/genética , Reordenamiento Génico , Proteínas Asociadas a Microtúbulos/genética , Proteínas Serina-Treonina Quinasas/genética , Receptores Notch/genética , Alelos , Animales , Carcinoma/genética , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Fusión Génica , Humanos , Ratones , Ratones SCID , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos , Familia de Multigenes , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptor Notch2/genética , Receptor Notch2/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Transcriptoma
15.
Genome Res ; 21(7): 1028-41, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21724842

RESUMEN

Beginning with precursor lesions, aberrant DNA methylation marks the entire spectrum of prostate cancer progression. We mapped the global DNA methylation patterns in select prostate tissues and cell lines using MethylPlex-next-generation sequencing (M-NGS). Hidden Markov model-based next-generation sequence analysis identified ∼68,000 methylated regions per sample. While global CpG island (CGI) methylation was not differential between benign adjacent and cancer samples, overall promoter CGI methylation significantly increased from ~12.6% in benign samples to 19.3% and 21.8% in localized and metastatic cancer tissues, respectively (P-value < 2 × 10(-16)). We found distinct patterns of promoter methylation around transcription start sites, where methylation occurred not only on the CGIs, but also on flanking regions and CGI sparse promoters. Among the 6691 methylated promoters in prostate tissues, 2481 differentially methylated regions (DMRs) are cancer-specific, including numerous novel DMRs. A novel cancer-specific DMR in the WFDC2 promoter showed frequent methylation in cancer (17/22 tissues, 6/6 cell lines), but not in the benign tissues (0/10) and normal PrEC cells. Integration of LNCaP DNA methylation and H3K4me3 data suggested an epigenetic mechanism for alternate transcription start site utilization, and these modifications segregated into distinct regions when present on the same promoter. Finally, we observed differences in repeat element methylation, particularly LINE-1, between ERG gene fusion-positive and -negative cancers, and we confirmed this observation using pyrosequencing on a tissue panel. This comprehensive methylome map will further our understanding of epigenetic regulation in prostate cancer progression.


Asunto(s)
Metilación de ADN , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Neoplasias de la Próstata/genética , Línea Celular Tumoral , Islas de CpG , ADN de Neoplasias/genética , Epigenómica , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Biblioteca de Genes , Humanos , Masculino , Cadenas de Markov , Metástasis de la Neoplasia , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Análisis de Secuencia de ARN , Sitio de Iniciación de la Transcripción
16.
Clin Cancer Res ; 17(12): 3924-32, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21555375

RESUMEN

PURPOSE: Bone is the most common metastatic site for prostate cancer, and osseous metastases are the leading cause of morbidity from this disease. Recent autopsy studies prove that 100% of men who die of prostate cancer have bone involvement. Understanding the biology of prostate cancer and its evolution to an incurable androgen-independent phenotype requires an understanding of the genetic and cellular alterations that lead to the seeding and proliferation of tumor foci in bone, as well as the microenvironment in which these metastases arise. No intensive studies, however, have been conducted on osseous metastatic tissues from patients with metastatic prostate cancer due to lack of access to such tissues for profiling and other research. EXPERIMENTAL DESIGN: We show, for the first time, a reproducible methodology to obtain high quality clinical tumor tissues metastatic to the bone. This technique allowed the procurement of viable metastatic tumor tissue from involved bones in 13 recent autopsies conducted at the University of Michigan and analyzed the gene expression of these tissues using real-time PCR and microarrays. RESULTS: We present here the discovery of nonossified bone metastases from multiple patients with advanced prostate cancer and their subsequent characterization and comparison to nonosseous metastases from the same patients. CONCLUSION: This represents a versatile and practical approach that may be employed to characterize the steps in metastasis and the phenotypic characteristics of osseous metastasis of prostate cancer and to profile RNA, DNA, and cDNA from tumor samples metastatic to the bone.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Próstata/patología , Anciano , Autopsia , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Huesos/patología , Hibridación Genómica Comparativa , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/genética
17.
Optometry ; 81(12): 688-94, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21111378

RESUMEN

BACKGROUND: Tilted disc syndrome (TDS) is associated with characteristic ocular findings. The purpose of this study was to evaluate the ocular, refractive, and biometric characteristics in patients with TDS. METHODS: This case-control study included 41 eyes of 25 patients who had established TDS and 40 eyes of 20 healthy control subjects. All participants underwent a complete ocular examination, including refraction and analysis using Fourier transformation, slit lamp biomicroscopy, pachymetry, keratometry, and ocular biometry. Corneal topography examinations were performed in the syndrome group only. RESULTS: There were no significant differences in spherical equivalent (P = 0.13) and total astigmatism (P = 0.37) between groups. However, mean best spectacle-corrected visual acuity (Log Mar) was significantly worse in TDS patients (P = 0.003). The lenticular astigmatism was greater in the syndrome group, whereas the corneal component was greater in controls (P = 0.059 and P = 0.028, respectively). The measured biometric features were the same in both groups, except for the lens thickness and lens-axial length factor, which were greater in the TDS group (P = 0.007 and P = 0.055, respectively). CONCLUSIONS: Clinically significant lenticular astigmatism, more oblique corneal astigmatism, and thicker lenses were characteristic findings in patients with TDS.


Asunto(s)
Astigmatismo/etiología , Córnea/patología , Cristalino/patología , Disco Óptico/anomalías , Adolescente , Adulto , Astigmatismo/diagnóstico por imagen , Astigmatismo/patología , Estudios de Casos y Controles , Niño , Córnea/diagnóstico por imagen , Técnicas de Diagnóstico Oftalmológico , Femenino , Humanos , Cristalino/diagnóstico por imagen , Masculino , Persona de Mediana Edad , Disco Óptico/diagnóstico por imagen , Análisis de Regresión , Síndrome , Ultrasonografía
18.
Nat Med ; 16(7): 793-8, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20526349

RESUMEN

Although recurrent gene fusions involving erythroblastosis virus E26 transformation-specific (ETS) family transcription factors are common in prostate cancer, their products are considered 'undruggable' by conventional approaches. Recently, rare targetable gene fusions involving the anaplastic lymphoma receptor tyrosine kinase (ALK) gene, have been identified in 1-5% of lung cancers, suggesting that similar rare gene fusions may occur in other common epithelial cancers, including prostate cancer. Here we used paired-end transcriptome sequencing to screen ETS rearrangement-negative prostate cancers for targetable gene fusions and identified the SLC45A3-BRAF (solute carrier family 45, member 3-v-raf murine sarcoma viral oncogene homolog B1) and ESRP1-RAF1 (epithelial splicing regulatory protein-1-v-raf-1 murine leukemia viral oncogene homolog-1) gene fusions. Expression of SLC45A3-BRAF or ESRP1-RAF1 in prostate cells induced a neoplastic phenotype that was sensitive to RAF and mitogen-activated protein kinase kinase (MAP2K1) inhibitors. Screening a large cohort of patients, we found that, although rare, recurrent rearrangements in the RAF pathway tend to occur in advanced prostate cancers, gastric cancers and melanoma. Taken together, our results emphasize the key role of RAF family gene rearrangements in cancer, suggest that RAF and MEK inhibitors may be useful in a subset of gene fusion-harboring solid tumors and demonstrate that sequencing of tumor transcriptomes and genomes may lead to the identification of rare targetable fusions across cancer types.


Asunto(s)
Melanoma/genética , Proteínas de Fusión Oncogénica/genética , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas de Unión al ARN/genética , Neoplasias Gástricas/genética , Translocación Genética , Humanos , Masculino , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Monosacáridos , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Transducción de Señal/genética
19.
Cancer Cell ; 17(5): 443-54, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20478527

RESUMEN

Chromosomal rearrangements fusing the androgen-regulated gene TMPRSS2 to the oncogenic ETS transcription factor ERG occur in approximately 50% of prostate cancers, but how the fusion products regulate prostate cancer remains unclear. Using chromatin immunoprecipitation coupled with massively parallel sequencing, we found that ERG disrupts androgen receptor (AR) signaling by inhibiting AR expression, binding to and inhibiting AR activity at gene-specific loci, and inducing repressive epigenetic programs via direct activation of the H3K27 methyltransferase EZH2, a Polycomb group protein. These findings provide a working model in which TMPRSS2-ERG plays a critical role in cancer progression by disrupting lineage-specific differentiation of the prostate and potentiating the EZH2-mediated dedifferentiation program.


Asunto(s)
Proteínas de Unión al ADN/genética , Fusión Génica , Proteínas de Fusión Oncogénica/genética , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Factores de Transcripción/genética , Inmunoprecipitación de Cromatina , Progresión de la Enfermedad , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Masculino , Complejo Represivo Polycomb 2 , Neoplasias de la Próstata/genética , Transducción de Señal
20.
Science ; 322(5908): 1695-9, 2008 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-19008416

RESUMEN

Enhancer of zeste homolog 2 (EZH2) is a mammalian histone methyltransferase that contributes to the epigenetic silencing of target genes and regulates the survival and metastasis of cancer cells. EZH2 is overexpressed in aggressive solid tumors by mechanisms that remain unclear. Here we show that the expression and function of EZH2 in cancer cell lines are inhibited by microRNA-101 (miR-101). Analysis of human prostate tumors revealed that miR-101 expression decreases during cancer progression, paralleling an increase in EZH2 expression. One or both of the two genomic loci encoding miR-101 were somatically lost in 37.5% of clinically localized prostate cancer cells (6 of 16) and 66.7% of metastatic disease cells (22 of 33). We propose that the genomic loss of miR-101 in cancer leads to overexpression of EZH2 and concomitant dysregulation of epigenetic pathways, resulting in cancer progression.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , MicroARNs/genética , Neoplasias/genética , Neoplasias de la Próstata/genética , Factores de Transcripción/genética , Regiones no Traducidas 3' , Algoritmos , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Femenino , Genoma Humano , Humanos , Lisina/metabolismo , Masculino , Metilación , MicroARNs/metabolismo , Metástasis de la Neoplasia , Neoplasias/metabolismo , Complejo Represivo Polycomb 2 , Regiones Promotoras Genéticas , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA