Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Cell Chem Biol ; 31(6): 1039-1043, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38906108

RESUMEN

Velcrins are molecular glues that induce complex formation between PDE3A and SLFN12. The PDE3A-SLFN12 complex activates the SLFN12 RNase, resulting in cleavage of the specific substrate, tRNA-Leu-TAA, global inhibition of translation, and death of cells expressing sufficient levels of both proteins. Here, unanswered questions about the mechanism of action and therapeutic promise of velcrin compounds are discussed.


Asunto(s)
Endorribonucleasas , Humanos , Endorribonucleasas/metabolismo , Proteínas Bacterianas , Endodesoxirribonucleasas , Proteínas Asociadas a CRISPR
2.
Nat Chem Biol ; 19(3): 301-310, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36302897

RESUMEN

Velcrin compounds kill cancer cells expressing high levels of phosphodiesterase 3A (PDE3A) and Schlafen family member 12 (SLFN12) by inducing complex formation between these two proteins, but the mechanism of cancer cell killing by the PDE3A-SLFN12 complex is not fully understood. Here, we report that the physiological substrate of SLFN12 RNase is tRNALeu(TAA). SLFN12 selectively digests tRNALeu(TAA), and velcrin treatment promotes the cleavage of tRNALeu(TAA) by inducing PDE3A-SLFN12 complex formation in vitro. We found that distinct sequences in the variable loop and acceptor stem of tRNALeu(TAA) are required for substrate digestion. Velcrin treatment of sensitive cells results in downregulation of tRNALeu(TAA), ribosome pausing at Leu-TTA codons and global inhibition of protein synthesis. Velcrin-induced cleavage of tRNALeu(TAA) by SLFN12 and the concomitant global inhibition of protein synthesis thus define a new mechanism of apoptosis initiation.


Asunto(s)
Neoplasias , ARN de Transferencia de Leucina , Línea Celular Tumoral , Muerte Celular , Apoptosis , Biosíntesis de Proteínas
3.
Cell Chem Biol ; 29(6): 925-927, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35714590

RESUMEN

PDE3A-SLFN12 complex formation activates the SLFN12 RNase, but the biochemical details of RNase activation remain mysterious. In this issue of Cell Chemical Biology, Yan and colleagues report that two phosphoserines on SLFN12 are dephosphorylated in response to PDE3A binding, and this dephosphorylation is required for activation of the SLFN12 RNase.


Asunto(s)
Ribonucleasas , Línea Celular Tumoral , Fosforilación
4.
Nat Commun ; 12(1): 4375, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34272366

RESUMEN

DNMDP and related compounds, or velcrins, induce complex formation between the phosphodiesterase PDE3A and the SLFN12 protein, leading to a cytotoxic response in cancer cells that express elevated levels of both proteins. The mechanisms by which velcrins induce complex formation, and how the PDE3A-SLFN12 complex causes cancer cell death, are not fully understood. Here, we show that PDE3A and SLFN12 form a heterotetramer stabilized by binding of DNMDP. Interactions between the C-terminal alpha helix of SLFN12 and residues near the active site of PDE3A are required for complex formation, and are further stabilized by interactions between SLFN12 and DNMDP. Moreover, we demonstrate that SLFN12 is an RNase, that PDE3A binding increases SLFN12 RNase activity, and that SLFN12 RNase activity is required for DNMDP response. This new mechanistic understanding will facilitate development of velcrin compounds into new cancer therapies.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/química , Péptidos y Proteínas de Señalización Intracelular/química , Piridazinas/química , Adenosina Monofosfato/química , Rastreo Diferencial de Calorimetría , Dominio Catalítico , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Microscopía por Crioelectrón , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Endorribonucleasas/química , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Cinética , Espectrometría de Masas , Complejos Multienzimáticos/ultraestructura , Mutación , Unión Proteica , Conformación Proteica en Hélice alfa , Multimerización de Proteína , Piridazinas/farmacología , Proteínas Recombinantes , Tetrahidroisoquinolinas/química
6.
Nat Cancer ; 1(2): 235-248, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32613204

RESUMEN

Anti-cancer uses of non-oncology drugs have occasionally been found, but such discoveries have been serendipitous. We sought to create a public resource containing the growth inhibitory activity of 4,518 drugs tested across 578 human cancer cell lines. We used PRISM, a molecular barcoding method, to screen drugs against cell lines in pools. An unexpectedly large number of non-oncology drugs selectively inhibited subsets of cancer cell lines in a manner predictable from the cell lines' molecular features. Our findings include compounds that killed by inducing PDE3A-SLFN12 complex formation; vanadium-containing compounds whose killing depended on the sulfate transporter SLC26A2; the alcohol dependence drug disulfiram, which killed cells with low expression of metallothioneins; and the anti-inflammatory drug tepoxalin, which killed via the multi-drug resistance protein ABCB1. The PRISM drug repurposing resource (https://depmap.org/repurposing) is a starting point to develop new oncology therapeutics, and more rarely, for potential direct clinical translation.


Asunto(s)
Neoplasias , Línea Celular , Disulfiram , Reposicionamiento de Medicamentos , Humanos , Neoplasias/tratamiento farmacológico
7.
J Biol Chem ; 295(11): 3431-3446, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32005668

RESUMEN

Cytotoxic molecules can kill cancer cells by disrupting critical cellular processes or by inducing novel activities. 6-(4-(Diethylamino)-3-nitrophenyl)-5-methyl-4,5-dihydropyridazin-3(2H)-one (DNMDP) is a small molecule that kills cancer cells by generation of novel activity. DNMDP induces complex formation between phosphodiesterase 3A (PDE3A) and schlafen family member 12 (SLFN12) and specifically kills cancer cells expressing elevated levels of these two proteins. Here, we examined the characteristics and covariates of the cancer cell response to DNMDP. On average, the sensitivity of human cancer cell lines to DNMDP is correlated with PDE3A expression levels. However, DNMDP could also bind the related protein, PDE3B, and PDE3B supported DNMDP sensitivity in the absence of PDE3A expression. Although inhibition of PDE3A catalytic activity did not account for DNMDP sensitivity, we found that expression of the catalytic domain of PDE3A in cancer cells lacking PDE3A is sufficient to confer sensitivity to DNMDP, and substitutions in the PDE3A active site abolish compound binding. Moreover, a genome-wide CRISPR screen identified the aryl hydrocarbon receptor-interacting protein (AIP), a co-chaperone protein, as required for response to DNMDP. We determined that AIP is also required for PDE3A-SLFN12 complex formation. Our results provide mechanistic insights into how DNMDP induces PDE3A-SLFN12 complex formation, thereby killing cancer cells with high levels of PDE3A and SLFN12 expression.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias/patología , Secuencia de Bases , Biomarcadores de Tumor/metabolismo , Sistemas CRISPR-Cas/genética , Dominio Catalítico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/química , Mutación del Sistema de Lectura/genética , Genoma , Heterocigoto , Humanos , Unión Proteica/efectos de los fármacos , Piridazinas/farmacología
8.
Int J Cancer ; 146(8): 2194-2200, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-31290142

RESUMEN

Somatic mutations of epidermal growth factor receptor (EGFR) occur in ~3% of colorectal cancer (CRC) patients. Here, through systematic functional screening of 21 recurrent EGFR mutations selected from public data sets, we show that 11 colon cancer-derived EGFR mutants (G63R, E114K, R165Q, R222C, S492R, P596L, K708R, E709K, G719S, G724S and L858R) are oncogenic and able to transform cells in a ligand-independent manner. We demonstrate that cellular transformation by these mutants requires receptor dimerization. Importantly, the EGF-induced and constitutive oncogenic potential of these EGFR mutants are inhibited by cetuximab or panitumumab in vivo and in vitro. Taken together, we propose that a subset of EGFR mutations can serve as genomic predictors for response to anti-EGFR antibodies and that metastatic CRC patients with such mutations may benefit from these drugs as part of the first-line therapy.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Cetuximab/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Panitumumab/farmacología , Adenocarcinoma/enzimología , Adenocarcinoma/genética , Animales , Antineoplásicos Inmunológicos/farmacología , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/genética , Dimerización , Receptores ErbB/genética , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Células HT29 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Terapia Molecular Dirigida , Mutación , Células 3T3 NIH , Ensayos Antitumor por Modelo de Xenoinjerto
9.
ACS Med Chem Lett ; 10(11): 1537-1542, 2019 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-31749907

RESUMEN

6-(4-(Diethylamino)-3-nitrophenyl)-5-methyl-4,5-dihydropyridazin-3(2H)-one, or DNMDP, potently and selectively inhibits phosphodiesterases 3A and 3B (PDE3A and PDE3B) and kills cancer cells by inducing PDE3A/B interactions with SFLN12. The structure-activity relationship (SAR) of DNMDP analogs was evaluated using a phenotypic viability assay, resulting in several compounds with suitable pharmacokinetic properties for in vivo analysis. One of these compounds, BRD9500, was active in an SK-MEL-3 xenograft model of cancer.

11.
Nat Biotechnol ; 34(5): 539-46, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27088724

RESUMEN

Systematic efforts to sequence the cancer genome have identified large numbers of mutations and copy number alterations in human cancers. However, elucidating the functional consequences of these variants, and their interactions to drive or maintain oncogenic states, remains a challenge in cancer research. We developed REVEALER, a computational method that identifies combinations of mutually exclusive genomic alterations correlated with functional phenotypes, such as the activation or gene dependency of oncogenic pathways or sensitivity to a drug treatment. We used REVEALER to uncover complementary genomic alterations associated with the transcriptional activation of ß-catenin and NRF2, MEK-inhibitor sensitivity, and KRAS dependency. REVEALER successfully identified both known and new associations, demonstrating the power of combining functional profiles with extensive characterization of genomic alterations in cancer genomes.


Asunto(s)
Biomarcadores de Tumor/genética , Mapeo Cromosómico/métodos , Estudio de Asociación del Genoma Completo/métodos , Proteínas de Neoplasias/genética , Neoplasias/genética , Polimorfismo de Nucleótido Simple/genética , Resistencia a Antineoplásicos/genética , Genes Relacionados con las Neoplasias/genética , Predisposición Genética a la Enfermedad/genética , Genoma Humano/genética , Humanos , Mutación/genética , Neoplasias/diagnóstico , Transducción de Señal/genética
12.
Nat Chem Biol ; 12(2): 102-8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26656089

RESUMEN

High cancer death rates indicate the need for new anticancer therapeutic agents. Approaches to discovering new cancer drugs include target-based drug discovery and phenotypic screening. Here, we identified phosphodiesterase 3A modulators as cell-selective cancer cytotoxic compounds through phenotypic compound library screening and target deconvolution by predictive chemogenomics. We found that sensitivity to 6-(4-(diethylamino)-3-nitrophenyl)-5-methyl-4,5-dihydropyridazin-3(2H)-one, or DNMDP, across 766 cancer cell lines correlates with expression of the gene PDE3A, encoding phosphodiesterase 3A. Like DNMDP, a subset of known PDE3A inhibitors kill selected cancer cells, whereas others do not. Furthermore, PDE3A depletion leads to DNMDP resistance. We demonstrated that DNMDP binding to PDE3A promotes an interaction between PDE3A and Schlafen 12 (SLFN12), suggestive of a neomorphic activity. Coexpression of SLFN12 with PDE3A correlates with DNMDP sensitivity, whereas depletion of SLFN12 results in decreased DNMDP sensitivity. Our results implicate PDE3A modulators as candidate cancer therapeutic agents and demonstrate the power of predictive chemogenomics in small-molecule discovery.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Citotoxinas/farmacología , Neoplasias/terapia , Piridazinas/química , Piridazinas/farmacología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citotoxinas/química , Citotoxinas/aislamiento & purificación , Sistemas de Liberación de Medicamentos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Genómica , Humanos , Immunoblotting
13.
Mol Cancer ; 13: 141, 2014 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-24894453

RESUMEN

BACKGROUND: Inhibition of the activated epidermal growth factor receptor (EGFR) with either enzymatic kinase inhibitors or anti-EGFR antibodies such as cetuximab, is an effective modality of treatment for multiple human cancers. Enzymatic EGFR inhibitors are effective for lung adenocarcinomas with somatic kinase domain EGFR mutations while, paradoxically, anti-EGFR antibodies are more effective in colon and head and neck cancers where EGFR mutations occur less frequently. In colorectal cancer, anti-EGFR antibodies are routinely used as second-line therapy of KRAS wild-type tumors. However, detailed mechanisms and genomic predictors for pharmacological response to these antibodies in colon cancer remain unclear. FINDINGS: We describe a case of colorectal adenocarcinoma, which was found to harbor a kinase domain mutation, G724S, in EGFR through whole genome sequencing. We show that G724S mutant EGFR is oncogenic and that it differs from classic lung cancer derived EGFR mutants in that it is cetuximab responsive in vitro, yet relatively insensitive to small molecule kinase inhibitors. Through biochemical and cellular pharmacologic studies, we have determined that cells harboring the colon cancer-derived G719S and G724S mutants are responsive to cetuximab therapy in vitro and found that the requirement for asymmetric dimerization of these mutant EGFR to promote cellular transformation may explain their greater inhibition by cetuximab than small-molecule kinase inhibitors. CONCLUSION: The colon-cancer derived G719S and G724S mutants are oncogenic and sensitive in vitro to cetuximab. These data suggest that patients with these mutations may benefit from the use of anti-EGFR antibodies as part of the first-line therapy.


Asunto(s)
Adenocarcinoma/genética , Anticuerpos Monoclonales Humanizados/uso terapéutico , Neoplasias Colorrectales/genética , Receptores ErbB/genética , Mutación , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Antineoplásicos/uso terapéutico , Cetuximab , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/química , Receptores ErbB/metabolismo , Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inhibidores de Proteínas Quinasas/uso terapéutico , Multimerización de Proteína , Estructura Terciaria de Proteína
14.
J Clin Invest ; 124(4): 1582-6, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24569458

RESUMEN

Targeted cancer therapies often induce "outlier" responses in molecularly defined patient subsets. One patient with advanced-stage lung adenocarcinoma, who was treated with oral sorafenib, demonstrated a near-complete clinical and radiographic remission for 5 years. Whole-genome sequencing and RNA sequencing of primary tumor and normal samples from this patient identified a somatic mutation, ARAF S214C, present in the cancer genome and expressed at high levels. Additional mutations affecting this residue of ARAF and a nearby residue in the related kinase RAF1 were demonstrated across 1% of an independent cohort of lung adenocarcinoma cases. The ARAF mutations were shown to transform immortalized human airway epithelial cells in a sorafenib-sensitive manner. These results suggest that mutant ARAF is an oncogenic driver in lung adenocarcinoma and an indicator of sorafenib response.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Antineoplásicos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Mutación Missense , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Proteínas Proto-Oncogénicas A-raf/genética , Adenocarcinoma/enzimología , Adenocarcinoma del Pulmón , Anciano , Sustitución de Aminoácidos , Transformación Celular Neoplásica/genética , ADN de Neoplasias/genética , Femenino , Humanos , Neoplasias Pulmonares/enzimología , Terapia Molecular Dirigida , Niacinamida/uso terapéutico , Oncogenes , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-raf/genética , Sorafenib
15.
Nature ; 506(7488): 371-5, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24390348

RESUMEN

Cervical cancer is responsible for 10-15% of cancer-related deaths in women worldwide. The aetiological role of infection with high-risk human papilloma viruses (HPVs) in cervical carcinomas is well established. Previous studies have also implicated somatic mutations in PIK3CA, PTEN, TP53, STK11 and KRAS as well as several copy-number alterations in the pathogenesis of cervical carcinomas. Here we report whole-exome sequencing analysis of 115 cervical carcinoma-normal paired samples, transcriptome sequencing of 79 cases and whole-genome sequencing of 14 tumour-normal pairs. Previously unknown somatic mutations in 79 primary squamous cell carcinomas include recurrent E322K substitutions in the MAPK1 gene (8%), inactivating mutations in the HLA-B gene (9%), and mutations in EP300 (16%), FBXW7 (15%), NFE2L2 (4%), TP53 (5%) and ERBB2 (6%). We also observe somatic ELF3 (13%) and CBFB (8%) mutations in 24 adenocarcinomas. Squamous cell carcinomas have higher frequencies of somatic nucleotide substitutions occurring at cytosines preceded by thymines (Tp*C sites) than adenocarcinomas. Gene expression levels at HPV integration sites were statistically significantly higher in tumours with HPV integration compared with expression of the same genes in tumours without viral integration at the same site. These data demonstrate several recurrent genomic alterations in cervical carcinomas that suggest new strategies to combat this disease.


Asunto(s)
Genoma Humano/genética , Mutación/genética , Neoplasias del Cuello Uterino/genética , Adenocarcinoma/genética , Adenocarcinoma/virología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/virología , Estudios de Casos y Controles , Proteínas de Ciclo Celular/genética , Subunidad beta del Factor de Unión al Sitio Principal/genética , Variaciones en el Número de Copia de ADN/genética , Análisis Mutacional de ADN , Proteínas de Unión al ADN/genética , Proteína p300 Asociada a E1A/genética , Exoma/genética , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Genómica , Antígenos HLA-B/genética , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Factor 2 Relacionado con NF-E2/genética , Papillomaviridae/genética , Papillomaviridae/fisiología , Infecciones por Papillomavirus/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-ets , Receptor ErbB-2/genética , Factores de Transcripción/genética , Transcriptoma/genética , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Neoplasias del Cuello Uterino/virología , Integración Viral/genética
16.
Cancer Res ; 73(22): 6770-9, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24063894

RESUMEN

Kinase domain mutations of the EGF receptor (EGFR) are common oncogenic events in lung adenocarcinoma. Here, we explore the dependency upon asymmetric dimerization of the kinase domain for activation of lung cancer-derived EGFR mutants. We show that whereas wild-type EGFR and the L858R mutant require dimerization for activation and oncogenic transformation, the exon 19 deletion, exon 20 insertion, and L858R/T790M EGFR mutants do not require dimerization. In addition, treatment with the monoclonal antibody, cetuximab, shrinks mouse lung tumors induced by the dimerization-dependent L858R mutant, but exerts only a modest effect on tumors driven by dimerization-independent EGFR mutants. These data imply that different EGFR mutants show differential requirements for dimerization and that disruption of dimerization may be among the antitumor mechanisms of cetuximab.


Asunto(s)
Adenocarcinoma/genética , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Receptores ErbB/metabolismo , Neoplasias Pulmonares/genética , Multimerización de Proteína/efectos de los fármacos , Adenocarcinoma/metabolismo , Sustitución de Aminoácidos , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Cetuximab , Receptores ErbB/química , Receptores ErbB/genética , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Células 3T3 NIH , Conformación Proteica/efectos de los fármacos
17.
Front Oncol ; 3: 86, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23630663

RESUMEN

Despite the ongoing "war on cancer," cancer remains one of the major causes of human morbidity and mortality. A new paradigm of targeted therapies holds the most promise for the future, making identification of tumor-specific therapeutic targets of prime importance. ERBB2/HER2, best known for its role in breast cancer tumorigenesis, can be targeted by two types of pharmacological manipulation: antibody therapy against the extracellular receptor domain and small molecule compounds against the intracellular tyrosine kinase domain. Aberrant activation of ERBB2 by gene amplification has been shown to participate in the pathophysiology of breast, ovarian, gastric, colorectal, lung, brain, and head and neck tumors. However, the advent of next-generation sequencing technologies has enabled efficient identification of activating molecular alterations of ERBB2. In this review, we will focus on the functional role of these somatic mutations that cause ERBB2 receptor activation. We will additionally discuss the current preclinical and clinical therapeutic strategies for targeting mutationally activated ERBB2.

18.
Cell ; 150(6): 1107-20, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22980975

RESUMEN

Lung adenocarcinoma, the most common subtype of non-small cell lung cancer, is responsible for more than 500,000 deaths per year worldwide. Here, we report exome and genome sequences of 183 lung adenocarcinoma tumor/normal DNA pairs. These analyses revealed a mean exonic somatic mutation rate of 12.0 events/megabase and identified the majority of genes previously reported as significantly mutated in lung adenocarcinoma. In addition, we identified statistically recurrent somatic mutations in the splicing factor gene U2AF1 and truncating mutations affecting RBM10 and ARID1A. Analysis of nucleotide context-specific mutation signatures grouped the sample set into distinct clusters that correlated with smoking history and alterations of reported lung adenocarcinoma genes. Whole-genome sequence analysis revealed frequent structural rearrangements, including in-frame exonic alterations within EGFR and SIK2 kinases. The candidate genes identified in this study are attractive targets for biological characterization and therapeutic targeting of lung adenocarcinoma.


Asunto(s)
Adenocarcinoma/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Genes Relacionados con las Neoplasias , Secuenciación de Nucleótidos de Alto Rendimiento , Neoplasias Pulmonares/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/patología , Estudios de Cohortes , Exoma , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Mutación , Tasa de Mutación
19.
Proc Natl Acad Sci U S A ; 109(36): 14476-81, 2012 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-22908275

RESUMEN

We assessed somatic alleles of six receptor tyrosine kinase genes mutated in lung adenocarcinoma for oncogenic activity. Five of these genes failed to score in transformation assays; however, novel recurring extracellular domain mutations of the receptor tyrosine kinase gene ERBB2 were potently oncogenic. These ERBB2 extracellular domain mutants were activated by two distinct mechanisms, characterized by elevated C-terminal tail phosphorylation or by covalent dimerization mediated by intermolecular disulfide bond formation. These distinct mechanisms of receptor activation converged upon tyrosine phosphorylation of cellular proteins, impacting cell motility. Survival of Ba/F3 cells transformed to IL-3 independence by the ERBB2 extracellular domain mutants was abrogated by treatment with small-molecule inhibitors of ERBB2, raising the possibility that patients harboring such mutations could benefit from ERBB2-directed therapy.


Asunto(s)
Adenocarcinoma/enzimología , Neoplasias Pulmonares/enzimología , Mutación/genética , Receptor ErbB-2/genética , Adenocarcinoma/genética , Adenocarcinoma del Pulmón , Alelos , Animales , Movimiento Celular/fisiología , Clonación Molecular , Cartilla de ADN/genética , Dimerización , Immunoblotting , Neoplasias Pulmonares/genética , Ratones , Células 3T3 NIH , Fosforilación , Estructura Terciaria de Proteína/genética , Retroviridae , Espectrometría de Masas en Tándem
20.
Nature ; 488(7409): 106-10, 2012 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-22820256

RESUMEN

Medulloblastomas are the most common malignant brain tumours in children. Identifying and understanding the genetic events that drive these tumours is critical for the development of more effective diagnostic, prognostic and therapeutic strategies. Recently, our group and others described distinct molecular subtypes of medulloblastoma on the basis of transcriptional and copy number profiles. Here we use whole-exome hybrid capture and deep sequencing to identify somatic mutations across the coding regions of 92 primary medulloblastoma/normal pairs. Overall, medulloblastomas have low mutation rates consistent with other paediatric tumours, with a median of 0.35 non-silent mutations per megabase. We identified twelve genes mutated at statistically significant frequencies, including previously known mutated genes in medulloblastoma such as CTNNB1, PTCH1, MLL2, SMARCA4 and TP53. Recurrent somatic mutations were newly identified in an RNA helicase gene, DDX3X, often concurrent with CTNNB1 mutations, and in the nuclear co-repressor (N-CoR) complex genes GPS2, BCOR and LDB1. We show that mutant DDX3X potentiates transactivation of a TCF promoter and enhances cell viability in combination with mutant, but not wild-type, ß-catenin. Together, our study reveals the alteration of WNT, hedgehog, histone methyltransferase and now N-CoR pathways across medulloblastomas and within specific subtypes of this disease, and nominates the RNA helicase DDX3X as a component of pathogenic ß-catenin signalling in medulloblastoma.


Asunto(s)
Neoplasias Cerebelosas/genética , Exoma/genética , Genoma Humano/genética , Meduloblastoma/genética , Mutación/genética , Neoplasias Cerebelosas/clasificación , Niño , ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , ADN Helicasas/química , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Proteínas Hedgehog/metabolismo , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM/genética , Meduloblastoma/clasificación , Modelos Moleculares , Proteínas de Neoplasias/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Receptores Patched , Receptor Patched-1 , Regiones Promotoras Genéticas/genética , Estructura Terciaria de Proteína/genética , Proteínas Proto-Oncogénicas/genética , Receptores de Superficie Celular/genética , Proteínas Represoras/genética , Transducción de Señal , Factores de Transcripción TCF/metabolismo , Factores de Transcripción/química , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...