Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cancer Metastasis Rev ; 40(3): 661-674, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33555482

RESUMEN

One way to understand ductal adenocarcinoma of the pancreas (pancreatic cancer) is to view it as unimaginably large numbers of evolving living organisms interacting with their environment. This "evolutionary view" creates both expected and surprising perspectives in all stages of neoplastic progression. Advances in the field will require greater attention to this critical evolutionary prospective.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Carcinoma Ductal Pancreático/genética , Humanos , Páncreas , Neoplasias Pancreáticas/genética , Estudios Prospectivos
2.
Mod Pathol ; 33(12): 2544-2563, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32704031

RESUMEN

The publication of the "Pan-Cancer Atlas" by the Pan-Cancer Analysis of Whole Genomes Consortium, a partnership formed by The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC), provides a wonderful opportunity to reflect on where we stand in our understanding of the genetics of pancreatic cancer, as well as on the opportunities to translate this understanding to patient care. From germline variants that predispose to the development of pancreatic cancer, to somatic mutations that are therapeutically targetable, genetics is now providing hope, where there once was no hope, for those diagnosed with pancreatic cancer.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma Ductal Pancreático/genética , Variación Genética , Genómica/tendencias , Técnicas de Diagnóstico Molecular/tendencias , Neoplasias Pancreáticas/genética , Animales , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/terapia , Difusión de Innovaciones , Predicción , Predisposición Genética a la Enfermedad , Humanos , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Fenotipo , Pronóstico
3.
Nature ; 475(7354): 106-9, 2011 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-21734707

RESUMEN

Reactive oxygen species (ROS) are mutagenic and may thereby promote cancer. Normally, ROS levels are tightly controlled by an inducible antioxidant program that responds to cellular stressors and is predominantly regulated by the transcription factor Nrf2 (also known as Nfe2l2) and its repressor protein Keap1 (refs 2-5). In contrast to the acute physiological regulation of Nrf2, in neoplasia there is evidence for increased basal activation of Nrf2. Indeed, somatic mutations that disrupt the Nrf2-Keap1 interaction to stabilize Nrf2 and increase the constitutive transcription of Nrf2 target genes were recently identified, indicating that enhanced ROS detoxification and additional Nrf2 functions may in fact be pro-tumorigenic. Here, we investigated ROS metabolism in primary murine cells following the expression of endogenous oncogenic alleles of Kras, Braf and Myc, and found that ROS are actively suppressed by these oncogenes. K-Ras(G12D), B-Raf(V619E) and Myc(ERT2) each increased the transcription of Nrf2 to stably elevate the basal Nrf2 antioxidant program and thereby lower intracellular ROS and confer a more reduced intracellular environment. Oncogene-directed increased expression of Nrf2 is a new mechanism for the activation of the Nrf2 antioxidant program, and is evident in primary cells and tissues of mice expressing K-Ras(G12D) and B-Raf(V619E), and in human pancreatic cancer. Furthermore, genetic targeting of the Nrf2 pathway impairs K-Ras(G12D)-induced proliferation and tumorigenesis in vivo. Thus, the Nrf2 antioxidant and cellular detoxification program represents a previously unappreciated mediator of oncogenesis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Factor 2 Relacionado con NF-E2/metabolismo , Oncogenes/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Alelos , Animales , Antioxidantes/metabolismo , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Células Cultivadas , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibroblastos/metabolismo , Genes myc/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch , Sistema de Señalización de MAP Quinasas , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Factor 2 Relacionado con NF-E2/deficiencia , Factor 2 Relacionado con NF-E2/genética , Células 3T3 NIH , Oxidación-Reducción , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
4.
Hum Mutat ; 36(2): 260-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25451944

RESUMEN

The greatest interpretive challenge of modern medicine may be to functionally annotate the vast variation of human genomes. Demonstrating a proposed approach, we created a library of BRCA2 exon 27 shotgun-mutant plasmids including solitary and multiplex mutations to generate human knockin clones using homologous recombination. This 55-mutation, 13-clone syngeneic variance library (SyVaL) comprised severely affected clones having early-stop nonsense mutations, functionally hypomorphic clones having multiple missense mutations emphasizing the potential to identify and assess hypomorphic mutations in novel proteomic and epidemiologic studies, and neutral clones having multiple missense mutations. Efficient coverage of nonessential amino acids was provided by mutation multiplexing. Severe mutations were distinguished from hypomorphic or neutral changes by chemosensitivity assays (hypersensitivity to mitomycin C and acetaldehyde), by analysis of RAD51 focus formation, and by mitotic multipolarity. A multiplex unbiased approach of generating all-human SyVaLs in medically important genes, with random mutations in native genes, would provide databases of variants that could be functionally annotated without concerns arising from exogenous cDNA constructs or interspecies interactions, as a basis for subsequent proteomic domain mapping or clinical calibration if desired. Such gene-irrelevant approaches could be scaled up for multiple genes of clinical interest, providing distributable cellular libraries linked to public-shared functional databases.


Asunto(s)
Proteína BRCA2/genética , Anotación de Secuencia Molecular , Sustitución de Aminoácidos , Línea Celular , Bases de Datos Genéticas , Biblioteca de Genes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Hemicigoto , Recombinación Homóloga , Humanos , Mitosis , Recombinasa Rad51/genética
5.
Am J Pathol ; 184(1): 260-70, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24200853

RESUMEN

Large-magnitude numerical distinctions (>10-fold) among drug responses of genetically contrasting cancers were crucial for guiding the development of some targeted therapies. Similar strategies brought epidemiological clues and prevention goals for genetic diseases. Such numerical guides, however, were incomplete or low magnitude for Fanconi anemia pathway (FANC) gene mutations relevant to cancer in FANC-mutation carriers (heterozygotes). We generated a four-gene FANC-null cancer panel, including the engineering of new PALB2/FANCN-null cancer cells by homologous recombination. A characteristic matching of FANCC-null, FANCG-null, BRCA2/FANCD1-null, and PALB2/FANCN-null phenotypes was confirmed by uniform tumor regression on single-dose cross-linker therapy in mice and by shared chemical hypersensitivities to various inter-strand cross-linking agents and γ-radiation in vitro. Some compounds, however, had contrasting magnitudes of sensitivity; a strikingly high (19- to 22-fold) hypersensitivity was seen among PALB2-null and BRCA2-null cells for the ethanol metabolite, acetaldehyde, associated with widespread chromosomal breakage at a concentration not producing breaks in parental cells. Because FANC-defective cancer cells can share or differ in their chemical sensitivities, patterns of selective hypersensitivity hold implications for the evolutionary understanding of this pathway. Clinical decisions for cancer-relevant prevention and management of FANC-mutation carriers could be modified by expanded studies of high-magnitude sensitivities.


Asunto(s)
Acetaldehído/farmacología , Resistencia a Antineoplásicos/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Animales , Western Blotting , Línea Celular Tumoral , Anemia de Fanconi/genética , Humanos , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Am J Pathol ; 183(1): 296-303, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23665203

RESUMEN

Like the p16, SMAD4, and RB1 genes, FAM190A (alias CCSER1) lies at a consensus site of homogeneous genomic deletions in human cancer. FAM190A transcripts in 40% of cancers also contain in-frame deletions of evolutionarily conserved exons. Its gene function was unknown. We found an internal deletion of the FAM190A gene in a pancreatic cancer having prominent focal multinuclearity. The experimental knockdown of FAM190A expression by shRNA caused focal cytokinesis defects, multipolar mitosis, and multinuclearity as observed in time-lapse microscopy. FAM190A was localized to the γ-tubulin ring complex of early mitosis and to the midbody in late cytokinesis by immunofluorescence assay and was present in the nuclear fraction of unsynchronized cells by immunoblot. FAM190A interacted with EXOC1 and Ndel1, which function in cytoskeletal organization and the cell division cycle. Levels of FAM190A protein peaked 12 hours after release from thymidine block, corresponding to M-phase. Slower-migrating phosphorylated forms accumulated toward M-phase and disappeared after release from a mitotic block and before cytokinesis. Studies of FAM190A alterations may provide mechanistic insights into mitotic dysregulation and multinuclearity in cancer. We propose that FAM190A is a regulator or structural component required for normal mitosis and that both the rare truncating mutations and common in-frame deletion alteration of FAM190A may contribute to the chromosomal instability of cancer.


Asunto(s)
Biomarcadores de Tumor/deficiencia , Proteínas de Ciclo Celular/deficiencia , División Celular/fisiología , Neoplasias/metabolismo , Biomarcadores de Tumor/genética , Proteínas de Ciclo Celular/genética , División Celular/genética , Línea Celular Tumoral , Inestabilidad Cromosómica , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Mutación , Neoplasias/genética , Neoplasias/patología
7.
J Proteome Res ; 11(11): 5301-10, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-23025254

RESUMEN

Although significant effort is expended on identifying transcripts/proteins that are up-regulated in cancer, there are few reports on systematic elucidation of transcriptional mechanisms underlying such druggable cancer-specific targets. The mesothelin (MSLN) gene offers a promising subject, being expressed in a restricted pattern normally, yet highly overexpressed in almost one-third of human malignancies and a target of cancer immunotherapeutic trials. CanScript, a cis promoter element, appears to control MSLN cancer-specific expression; its related genomic sequences may up-regulate other cancer markers. CanScript is a 20-nt bipartite element consisting of an SP1-like motif and a consensus MCAT sequence. The latter recruits TEAD (TEA domain) family members, which are universally expressed. Exploration of the active CanScript element, especially the proteins binding to the SP1-like motif, thus could reveal cancer-specific features having diagnostic or therapeutic interest. The efficient identification of sequence-specific DNA-binding proteins at a given locus, however, has lagged in biomarker explorations. We used two orthogonal proteomics approaches--unbiased SILAC (stable isotope labeling by amino acids in cell culture)/DNA affinity-capture/mass spectrometry survey (SD-MS) and a large transcription factor protein microarray (TFM)--and functional validation to explore systematically the CanScript interactome. SD-MS produced nine candidates, and TFM, 18. The screens agreed in confirming binding by TEAD proteins and by newly identified NAB1 and NFATc. Among other identified candidates, we found functional roles for ZNF24, NAB1 and RFX1 in MSLN expression by cancer cells. Combined interactome screens yield an efficient, reproducible, sensitive, and unbiased approach to identify sequence-specific DNA-binding proteins and other participants in disease-specific DNA elements.


Asunto(s)
Proteínas Ligadas a GPI/metabolismo , Neoplasias/metabolismo , Secuencia de Bases , Western Blotting , Línea Celular , ADN , Humanos , Luciferasas/genética , Mesotelina , Datos de Secuencia Molecular , Neoplasias/diagnóstico , Neoplasias/terapia , Espectrometría de Masas en Tándem
8.
J Biol Chem ; 286(14): 11960-9, 2011 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21288909

RESUMEN

Mesothelin (MSLN) may be the most "dramatic" of the tumor markers, being strongly overexpressed in nearly one-third of human malignancies. The biochemical cause is unclear. We previously ascribed this cancer-specific overexpression to an element, Canscript, residing around 50 bp 5' of the transcription start site in cancer (Hucl, T., Brody, J. R., Gallmeier, E., Iacobuzio-Donahue, C. A., Farrance, I. K., and Kern, S. E. (2007) Cancer Res. 67, 9055-9065). Herein, we found a Canscript promoter activity elevated over 100-fold in cancer cells. In addition to a highly conserved TEAD1 (TEA domain family member 1)-binding MCAT motif, nucleotide substitution revealed the consensus core sequence (WCYCCACCC) of an SP1-like motif in Canscript. The unknown transcription factor binding to the SP1-like motif may hold the key for the cancer specificity of Canscript. SP1, GLI1, and RUNX1, -2, and -3 appeared unlikely to be the direct transcription factors acting at the SP1-like motif, but KLF6 had some features of such a candidate. YAP1, a TEAD1-binding protein, appeared necessary, but not sufficient, for Canscript activity; knockdown of YAP1 by small interfering RNAs greatly reduced MSLN levels in MSLN-overexpressing cells, but overexpressing YAP1 in MSLN-negative cells did not induce MSLN expression. Cansript-like sequences were found in other genes up-regulated in pancreatic cancer; reporters driven by the sequences from FXYD3, MUC1, and TIMP1 had activities more than 2 times that of the control. This suggested that the cause of MSLN overexpression might also contribute mechanistically to the overexpression of other tumor markers.


Asunto(s)
Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Western Blotting , Ciclo Celular , Línea Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Células HEK293 , Células HeLa , Humanos , Factor 6 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mesotelina , Mucina-1/genética , Mucina-1/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Técnicas de Amplificación de Ácido Nucleico , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Interferente Pequeño , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Factores de Transcripción de Dominio TEA , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP , Proteína con Dedos de Zinc GLI1
9.
J Pathol ; 223(2): 295-306, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21125682

RESUMEN

The directions of differentiation and the molecular features of ductal pancreatic cancer have by now been explored in reasonable detail. Already, diagnoses and therapeutic strategies benefit from observations distinguishing the major variant types of pancreatic cancer and the differing stages of disease at presentation. Additionally, individual patients differ within each variant type. In certain high-risk groups, this permits focused screening efforts. The tumorigenic influences that characterize individual patients are increasingly considered appropriate in defining clinical treatment plans. As a result, multiple variables affect success when individualizing screening or therapy. These competing variables often limit the potential for success: some variables dominate and should receive greater consideration than others. Simplistic expectations, often falsely optimistic, for individualized care may fail to 'pan out' in the real world. The development of individualized care will be efficient only when the full complexity of the disease is embraced.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Terapia Molecular Dirigida/métodos , Neoplasias Pancreáticas/genética , Antineoplásicos/uso terapéutico , Carcinoma Ductal Pancreático/clasificación , Carcinoma Ductal Pancreático/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica , Humanos , Mutación , Neoplasias Pancreáticas/clasificación , Neoplasias Pancreáticas/tratamiento farmacológico , Lesiones Precancerosas/genética
11.
Clin Cancer Res ; 14(2): 412-8, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18223216

RESUMEN

PURPOSE: The role of E-cadherin in carcinogenesis is of great interest, but few studies have examined its relevance to pancreatic carcinoma. EXPERIMENTAL DESIGN: We evaluated E-cadherin protein expression by immunohistochemistry in pancreatobiliary cancers having a noncohesive histologic phenotype (21 undifferentiated adenocarcinomas and 7 signet ring carcinomas), comparing the results with pancreatic cancers having a cohesive phenotype (25 moderately differentiated and 14 poorly differentiated adenocarcinomas). RESULTS: Twenty of 21 undifferentiated cancers had complete absence of E-cadherin expression, as did two signet ring carcinomas. In contrast, cohesive cancers (n = 39) had E-cadherin labeling at the plasma membrane (P < 0.001). Subsets of cancers were also evaluated for beta-catenin expression. All of the cohesive lesions (n = 28) showed a membranous beta-catenin expression pattern, whereas noncohesive foci (n = 7) were characterized by either cytoplasmic labeling or complete absence of beta-catenin protein expression, suggestive of a deficient zonula adherens in noncohesive cancers. E-cadherin promoter hypermethylation was observed in an undifferentiated pancreatic cancer cell line, MiaPaCa-2, whereas two pancreatic cancer cell lines derived from differentiated lesions lacked any evidence of E-cadherin promoter methylation. No pattern of E-cadherin promoter methylation could be determined in three primary cancers having mixed histologic patterns (contained both cohesive and noncohesive foci). No somatic mutations in E-cadherin were identified in noncohesive pancreatic cancers having inactivated E-cadherin. CONCLUSIONS: Noncohesive pancreatic cancers were characterized by the loss of E-cadherin protein expression. Promoter hypermethylation is a possible mechanism of E-cadherin gene silencing in a subset of these cancers.


Asunto(s)
Cadherinas/metabolismo , Carcinoma de Células en Anillo de Sello/metabolismo , Neoplasias Pancreáticas/metabolismo , beta Catenina/metabolismo , Cadherinas/genética , Carcinoma de Células en Anillo de Sello/genética , Carcinoma de Células en Anillo de Sello/patología , Metilación de ADN , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Regiones Promotoras Genéticas
12.
Genes Chromosomes Cancer ; 47(3): 253-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18064648

RESUMEN

Germ line gain-of-function mutations in several members of the RAS/ERK pathway, including PTPN11, KRAS, and RAF1, cause the autosomal dominant genetic disorder Noonan Syndrome (NS). NS patients are at increased risk of leukemia/myeloproliferative disease and possibly some solid tumors, such as neuroblastoma. Recently, SOS1 gain of function mutations have also been shown to cause NS. Somatic PTPN11, KRAS, and RAF1 mutations occur (although at different frequencies) in a variety of sporadic neoplasms, but whether SOS1 mutations are associated with human cancer has not been evaluated. We sequenced DNA from a total of 810 primary malignancies, including pancreatic, lung, breast, and colon carcinomas, and acute myelogenous leukemia, as well as several neuroblastoma cell lines. From this large, diverse series, missense SOS1 mutations were identified in a single pancreatic tumor, one lung adenocarcinoma, and a T-cell acute lymphoblastic leukemia cell line. Our findings suggest that SOS1 is not a significant human oncogene in most cancers. Furthermore, NS patients with SOS1 mutations may not be at increased risk of developing cancer.


Asunto(s)
Mutación , Neoplasias/genética , Síndrome de Noonan/complicaciones , Proteína SOS1/genética , ADN de Neoplasias , Predisposición Genética a la Enfermedad , Humanos , Neoplasias/etiología , Síndrome de Noonan/genética , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN
13.
Cancer Res ; 67(19): 8985-8, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17908998

RESUMEN

Apparently effective therapeutic agents very often fail to cure cancer patients. It is therefore attractive to wonder whether a specific resistant cell subset should be recognized and separately targeted. In solid tumors, such as carcinomas, a minor population of "cancer stem cells" has been proposed and sought experimentally in human tumors and isolated cell populations. It is often overlooked that the rationale and supportive data are essentially numerical and can be evaluated as such. A reevaluation of the published studies and related claims within awarded U.S. patents suggests that the mathematical support for the concept of therapeutically useful stem cells is weak and may even invalidate the foundations of these publications and patent claims. Mathematical arguments should be used more consistently, because they can serve as a guide for interpreting studies into cancer stem cells of solid tumors.


Asunto(s)
Lógica Difusa , Modelos Biológicos , Células Madre Neoplásicas/patología , Humanos , Patentes como Asunto
14.
Cancer Res ; 67(19): 9055-65, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17909009

RESUMEN

Identification of genes with cancer-specific overexpression offers the potential to efficiently discover cancer-specific activities in an unbiased manner. We apply this paradigm to study mesothelin (MSLN) overexpression, a nearly ubiquitous, diagnostically and therapeutically useful characteristic of pancreatic cancer. We identified an 18-bp upstream enhancer, termed CanScript, strongly activating transcription from an otherwise weak tissue-nonspecific promoter and operating selectively in cells having aberrantly elevated cancer-specific MSLN transcription. Introducing mutations into CanScript showed two functionally distinct sites: an Sp1-like site and an MCAT element. Gel retardation and chromatin immunoprecipitation assays showed the MCAT element to be bound by transcription enhancer factor (TEF)-1 (TEAD1) in vitro and in vivo. The presence of TEF-1 was required for MSLN protein overexpression as determined by TEF-1 knockdown experiments. The cancer specificity seemed to be provided by a putative limiting cofactor of TEF-1 that could be outcompeted by exogenous TEF-1 only in a MSLN-overexpressing cell line. A CanScript concatemer offered enhanced activity. These results identify a TEF family member as a major regulator of MSLN overexpression, a fundamental characteristic of pancreatic and other cancers, perhaps due to an upstream and highly frequent aberrant cellular activity. The CanScript sequence represents a modular element for cancer-specific targeting, potentially suitable for nearly a third of human malignancies.


Asunto(s)
Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos , Glicoproteínas de Membrana/biosíntesis , Proteínas Nucleares/genética , Factores de Transcripción/genética , Secuencia de Bases , Línea Celular Tumoral , ADN Complementario/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Proteínas Ligadas a GPI , Células HeLa , Humanos , Inmunohistoquímica , Glicoproteínas de Membrana/genética , Mesotelina , Datos de Secuencia Molecular , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción de Dominio TEA , Factores de Transcripción/deficiencia , Factores de Transcripción/metabolismo , Transcripción Genética , Transfección
15.
Cancer Res ; 67(5): 2169-77, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17332347

RESUMEN

Inactivation of the Fanconi anemia (FA) pathway occurs in diverse human tumors among the general population and renders those tumors hypersensitive to DNA interstrand-cross-linking (ICL) agents. The identification of novel agents to which FA pathway-deficient cells were hypersensitive could provide new therapeutic opportunities and improve our molecular understanding of the FA genes. Using high-throughput screening, we assessed the growth of isogenic human cancer cells that differed only in the presence or absence of single FA genes upon treatment with 880 active drugs and 40,000 diverse compounds. We identified several compounds to which FA pathway-deficient cells were more sensitive than FA pathway-proficient cells, including two groups of structurally related compounds. We further investigated the compound eliciting the strongest effect, termed 80136342. Its mechanism of action was distinct from that of ICL agents; 80136342 did not cause increased chromosomal aberrations, enhanced FANCD2 monoubiquitination, H2AX phosphorylation, p53 activation, or ICL induction. Similar to ICL agents, however, 80136342 caused a pronounced G(2) arrest in FA pathway-deficient cells. When applied in combination with ICL agents, 80136342 had at least additive toxic effects, excluding interferences on ICL-induced toxicity and facilitating a combinational application. Finally, we identified one particular methyl group necessary for the effects of 80136342 on FA-deficient cells. In conclusion, using high-throughput screening in an isogenic human FA cancer model, we explored a novel approach to identify agents eliciting hypersensitivity in FA pathway-deficient cells. We discovered several attractive candidates to serve as lead compounds for evaluating structure-activity relationships and developing therapeutics selectively targeting FA pathway-deficient tumors.


Asunto(s)
Antineoplásicos/análisis , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Neoplasias/genética , Transducción de Señal/efectos de los fármacos , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Rotura Cromosómica/efectos de los fármacos , Reactivos de Enlaces Cruzados/farmacología , Evaluación Preclínica de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Histonas/metabolismo , Humanos , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Piridinas/uso terapéutico , Quinolinas/uso terapéutico , Células Tumorales Cultivadas
16.
Clin Cancer Res ; 13(1): 4-10, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17200332

RESUMEN

Defects in the Fanconi anemia (FA) pathway occur in subsets of diverse human cancers. The hypersensitivity of FA pathway-deficient cells to DNA interstrand cross-linking and possibly other agents renders these genes attractive targets for a genotype-based, individualized anticancer therapy. A prerequisite before clinical trials is the validation and quantification of this hypersensitivity in suitable preclinical pharmacogenomic models. In addition, the effects of combinational therapy need to be evaluated and novel agents sought. We discuss here the pitfalls and limitations in the interpretation of common FA models when applied to the validation of FA gene defects as therapeutic targets. In general, all preclinical models are prone to certain artifacts and, thus, promising results in a single or few models rarely translate into clinical success. Nevertheless, the extraordinary robustness of FA pathway-deficient cells to interstrand cross-linking agents, which are observable in virtually any model independent of species, cell type, or technique used to engineer the gene defect, in various in vitro and in vivo settings, renders these gene defects particularly attractive for targeted therapy. Clinical trials are now under way.


Asunto(s)
Proteína BRCA2/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/fisiología , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Neoplasias/metabolismo , Farmacogenética/métodos , Animales , Línea Celular Tumoral , Reactivos de Enlaces Cruzados/farmacología , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Humanos , Ratones , Modelos Biológicos , Mutación
17.
Cancer Res ; 66(11): 5560-4, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16740690

RESUMEN

Tumor-suppressors have commanded attention due to the selection for their inactivating mutations in human tumors. However, relatively little is understood about the inverse, namely, that tumors do not select for a large proportion of seemingly favorable mutations in tumor-suppressor genes. This could be explained by a detrimental phenotype accruing in a cell type-specific manner to most cells experiencing a biallelic loss. For example, MKK4, a tumor suppressor gene distinguished by a remarkably consistent mutational rate across diverse tumor types and an unusually high rate of loss of heterozygosity, has the surprisingly low rate of genetic inactivation of only approximately 5%. To explore this incongruity, we engineered a somatic gene knockout of MKK4 in human cancer cells. Although the null cells resembled the wild-type cells regarding in vitro viability and proliferation in plastic dishes, there was a marked difference in a more relevant in vivo model of experimental metastasis and tumorigenesis. MKK4(-/-) clones injected i.v. produced fewer lung metastases than syngeneic MKK4-competent cells (P = 0.0034). These findings show how cell type-specific detrimental phenotypes can offer a paradoxical and yet key counterweight to the selective advantage attained by cells as they experiment with genetic null states during tumorigenesis, the resultant balance then determining the observed biallelic mutation rate for a given tumor-suppressor gene.


Asunto(s)
Genes Supresores de Tumor , MAP Quinasa Quinasa 4/deficiencia , MAP Quinasa Quinasa 4/genética , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Animales , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Activación Enzimática , Exones , Femenino , Eliminación de Gen , Humanos , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Neoplasias Pancreáticas/patología , Fenotipo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Oncogene ; 24(22): 3652-6, 2005 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-15806175

RESUMEN

Germline BRCA2 mutations predispose to the development of pancreatic cancer. A polymorphic stop codon in the coding region of BRCA2 (K3326X) has been described, and although an initial epidemiological study suggested it was not disease causing, subsequent studies have been inconclusive. To investigate the biological significance of the K3326X polymorphism, we determined its prevalence in patients with sporadic and familial pancreatic cancer. Using a case-control design, we studied 250 patients with resected sporadic pancreatic adenocarcinomas, 144 patients with familial pancreatic adenocarcinoma, 115 spouses of patients with pancreatic cancer, and a disease control group of 135 patients without a personal history of cancer who had undergone cholecystectomy for non-neoplastic disease. The K3326X polymorphism was detected using heteroduplex analysis and DNA sequencing. The BRCA2 K3326X polymorphism was significantly more prevalent in individuals with familial pancreatic cancer: 8/144 (5.6%) vs 3/250 controls (1.2%) (odds ratio, 4.84; 95% CI, 1.27-18.55, P<0.01). One K3326X carrier with familial pancreatic cancer carried an alteration (IVS 16-2A>G) suspected to be deleterious. Excluding this case did not alter the significance of the association (OR: 4.24, P<0.01). In contrast, there was no difference in prevalence among individuals with sporadic pancreatic cancer - 7/250 (OR: 2.37, 95% CI: 0.61-9.27). The increased prevalence of the BRCA2 K3326X polymorphism in patients with familial pancreatic cancer suggests that this polymorphism is deleterious and contributes to pancreatic cancer risk.


Asunto(s)
Adenocarcinoma/epidemiología , Adenocarcinoma/genética , Genes BRCA2 , Neoplasias Pancreáticas/epidemiología , Neoplasias Pancreáticas/genética , Adulto , Anciano , Estudios de Casos y Controles , Codón de Terminación , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Polimorfismo Genético , Prevalencia
20.
Hum Mutat ; 27(10): 1060-1, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16941478

RESUMEN

Tyrosine kinases are major regulators of signal transduction cascades involved in cellular proliferation and have important roles in tumorigenesis. We have recently analyzed the tyrosine kinase gene family for alterations in human colorectal cancers and identified somatic mutations in seven members of this gene family. In this study we have used high-throughput sequencing approaches to further evaluate this subset of genes for genetic alterations in other human tumors. We identified somatic mutations in GUCY2F, EPHA3, and NTRK3 in breast, lung, and pancreatic cancers. Our results implicate these tyrosine kinase genes in the pathogenesis of other tumor types and suggest that they may be useful targets for diagnostic and therapeutic intervention in selected patients.


Asunto(s)
Guanilato Ciclasa/genética , Mutación/genética , Neoplasias/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptor trkC/genética , Receptores de Superficie Celular/genética , Adenocarcinoma/genética , Secuencia de Bases , Neoplasias de la Mama/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Células Pequeñas/genética , Carcinoma de Células Escamosas/genética , Análisis Mutacional de ADN , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pancreáticas/genética , Reacción en Cadena de la Polimerasa , Receptor EphA3
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA