Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Mol Cell Proteomics ; 14(7): 1959-76, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25953087

RESUMEN

Mutations in PIK3CA, the gene encoding the p110α catalytic subunit of phosphoinositide 3-kinase (PI3K) have been shown to transform human mammary epithelial cells (MECs). These mutations are present in all breast cancer subtypes, including basal-like breast cancer (BLBC). Using liquid chromatography-tandem mass spectrometry (LC-MS/MS), we identified 72 protein expression changes in human basal-like MECs with knock-in E545K or H1047R PIK3CA mutations versus isogenic MECs with wild-type PIK3CA. Several of these were secreted proteins, cell surface receptors or ECM interacting molecules and were required for growth of PIK3CA mutant cells as well as adjacent cells with wild-type PIK3CA. The proteins identified by MS were enriched among human BLBC cell lines and pointed to a PI3K-dependent amphiregulin/EGFR/ERK signaling axis that is activated in BLBC. Proteins induced by PIK3CA mutations correlated with EGFR signaling and reduced relapse-free survival in BLBC. Treatment with EGFR inhibitors reduced growth of PIK3CA mutant BLBC cell lines and murine mammary tumors driven by a PIK3CA mutant transgene, all together suggesting that PIK3CA mutations promote tumor growth in part by inducing protein changes that activate EGFR.


Asunto(s)
Neoplasias de la Mama/genética , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Mutación/genética , Comunicación Paracrina , Fosfatidilinositol 3-Quinasas/genética , Transducción de Señal , Anfirregulina/metabolismo , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromatografía Liquida , Fosfatidilinositol 3-Quinasa Clase I , Supervivencia sin Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/antagonistas & inhibidores , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Femenino , Humanos , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Comunicación Paracrina/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteómica , Transducción de Señal/efectos de los fármacos , Espectrometría de Masas en Tándem , Regulación hacia Arriba/efectos de los fármacos
2.
Mol Cell Proteomics ; 14(9): 2357-74, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25693799

RESUMEN

There is an increasing need in biology and clinical medicine to robustly and reliably measure tens to hundreds of peptides and proteins in clinical and biological samples with high sensitivity, specificity, reproducibility, and repeatability. Previously, we demonstrated that LC-MRM-MS with isotope dilution has suitable performance for quantitative measurements of small numbers of relatively abundant proteins in human plasma and that the resulting assays can be transferred across laboratories while maintaining high reproducibility and quantitative precision. Here, we significantly extend that earlier work, demonstrating that 11 laboratories using 14 LC-MS systems can develop, determine analytical figures of merit, and apply highly multiplexed MRM-MS assays targeting 125 peptides derived from 27 cancer-relevant proteins and seven control proteins to precisely and reproducibly measure the analytes in human plasma. To ensure consistent generation of high quality data, we incorporated a system suitability protocol (SSP) into our experimental design. The SSP enabled real-time monitoring of LC-MRM-MS performance during assay development and implementation, facilitating early detection and correction of chromatographic and instrumental problems. Low to subnanogram/ml sensitivity for proteins in plasma was achieved by one-step immunoaffinity depletion of 14 abundant plasma proteins prior to analysis. Median intra- and interlaboratory reproducibility was <20%, sufficient for most biological studies and candidate protein biomarker verification. Digestion recovery of peptides was assessed and quantitative accuracy improved using heavy-isotope-labeled versions of the proteins as internal standards. Using the highly multiplexed assay, participating laboratories were able to precisely and reproducibly determine the levels of a series of analytes in blinded samples used to simulate an interlaboratory clinical study of patient samples. Our study further establishes that LC-MRM-MS using stable isotope dilution, with appropriate attention to analytical validation and appropriate quality control measures, enables sensitive, specific, reproducible, and quantitative measurements of proteins and peptides in complex biological matrices such as plasma.


Asunto(s)
Proteínas de Neoplasias/sangre , Neoplasias/metabolismo , Péptidos/análisis , Proteómica/métodos , Cromatografía Liquida/métodos , Humanos , Marcaje Isotópico , Espectrometría de Masas/métodos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/aislamiento & purificación , Neoplasias/sangre , Péptidos/química , Reproducibilidad de los Resultados
3.
Mol Cell Proteomics ; 12(2): 343-55, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23161513

RESUMEN

Activating mutations in KRAS occur in 30% to 40% of colorectal cancers. How mutant KRAS alters cancer cell behavior has been studied intensively, but non-cell autonomous effects of mutant KRAS are less understood. We recently reported that exosomes isolated from mutant KRAS-expressing colon cancer cells enhanced the invasiveness of recipient cells relative to exosomes purified from wild-type KRAS-expressing cells, leading us to hypothesize mutant KRAS might affect neighboring and distant cells by regulating exosome composition and behavior. Herein, we show the results of a comprehensive proteomic analysis of exosomes from parental DLD-1 cells that contain both wild-type and G13D mutant KRAS alleles and isogenically matched derivative cell lines, DKO-1 (mutant KRAS allele only) and DKs-8 (wild-type KRAS allele only). Mutant KRAS status dramatically affects the composition of the exosome proteome. Exosomes from mutant KRAS cells contain many tumor-promoting proteins, including KRAS, EGFR, SRC family kinases, and integrins. DKs-8 cells internalize DKO-1 exosomes, and, notably, DKO-1 exosomes transfer mutant KRAS to DKs-8 cells, leading to enhanced three-dimensional growth of these wild-type KRAS-expressing non-transformed cells. These results have important implications for non-cell autonomous effects of mutant KRAS, such as field effect and tumor progression.


Asunto(s)
Neoplasias del Colon/química , Neoplasias del Colon/patología , Exosomas/química , Invasividad Neoplásica , Proteínas de Neoplasias/química , Proteoma/química , Proteínas Proto-Oncogénicas/química , Proteínas ras/química , Alelos , Línea Celular Tumoral , Proliferación Celular , Cromatografía Liquida , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Exosomas/metabolismo , Humanos , Mutación , Proteínas de Neoplasias/metabolismo , Transporte de Proteínas , Proteoma/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , Transducción de Señal , Espectrometría de Masas en Tándem , Microambiente Tumoral , Proteínas ras/genética , Proteínas ras/metabolismo
4.
Mol Cell Proteomics ; 12(9): 2623-39, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23689285

RESUMEN

Multiple reaction monitoring (MRM) mass spectrometry coupled with stable isotope dilution (SID) and liquid chromatography (LC) is increasingly used in biological and clinical studies for precise and reproducible quantification of peptides and proteins in complex sample matrices. Robust LC-SID-MRM-MS-based assays that can be replicated across laboratories and ultimately in clinical laboratory settings require standardized protocols to demonstrate that the analysis platforms are performing adequately. We developed a system suitability protocol (SSP), which employs a predigested mixture of six proteins, to facilitate performance evaluation of LC-SID-MRM-MS instrument platforms, configured with nanoflow-LC systems interfaced to triple quadrupole mass spectrometers. The SSP was designed for use with low multiplex analyses as well as high multiplex approaches when software-driven scheduling of data acquisition is required. Performance was assessed by monitoring of a range of chromatographic and mass spectrometric metrics including peak width, chromatographic resolution, peak capacity, and the variability in peak area and analyte retention time (RT) stability. The SSP, which was evaluated in 11 laboratories on a total of 15 different instruments, enabled early diagnoses of LC and MS anomalies that indicated suboptimal LC-MRM-MS performance. The observed range in variation of each of the metrics scrutinized serves to define the criteria for optimized LC-SID-MRM-MS platforms for routine use, with pass/fail criteria for system suitability performance measures defined as peak area coefficient of variation <0.15, peak width coefficient of variation <0.15, standard deviation of RT <0.15 min (9 s), and the RT drift <0.5min (30 s). The deleterious effect of a marginally performing LC-SID-MRM-MS system on the limit of quantification (LOQ) in targeted quantitative assays illustrates the use and need for a SSP to establish robust and reliable system performance. Use of a SSP helps to ensure that analyte quantification measurements can be replicated with good precision within and across multiple laboratories and should facilitate more widespread use of MRM-MS technology by the basic biomedical and clinical laboratory research communities.


Asunto(s)
Cromatografía Liquida/instrumentación , Cromatografía Liquida/métodos , Espectrometría de Masas/instrumentación , Espectrometría de Masas/métodos , Secuencia de Aminoácidos , Animales , Bovinos , Límite de Detección , Datos de Secuencia Molecular , Péptidos/química , Péptidos/metabolismo , Estándares de Referencia , Programas Informáticos , Factores de Tiempo
5.
Am J Pathol ; 181(5): 1560-72, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22944598

RESUMEN

Early diagnosis and curative resection are the predominant factors associated with increased survival in patients with gastric cancer. However, most gastric cancer cases are still diagnosed at later stages. Since most pathologic specimens are archived as FFPE samples, the ability to use them to generate expression profiles can greatly improve cancer biomarker discovery. We sought to uncover new biomarkers for stomach preneoplastic metaplasias and neoplastic lesions by generating proteome profiles using FFPE samples. We combined peptide isoelectric focusing and liquid chromatography-tandem mass spectrometry analysis to generate proteomic profiles from FFPE samples of intestinal-type gastric cancer, metaplasia, and normal mucosa. The expression patterns of selected proteins were analyzed by immunostaining first in single tissue sections from normal stomach, metaplasia, and gastric cancer and later in larger tissue array cohorts. We detected 60 proteins up-regulated and 87 proteins down-regulated during the progression from normal mucosa to metaplasia to gastric cancer. Two of the up-regulated proteins, LTF and DMBT1, were validated as specific markers for spasmolytic polypeptide-expressing metaplasia and intestinal metaplasia, respectively. In cancers, significantly lower levels of DMBT1 or LTF correlated with more advanced disease and worse prognosis. Thus, proteomic profiling using FFPE samples has led to the identification of two novel markers for stomach metaplasias and gastric cancer prognosis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Mucosa Gástrica/metabolismo , Adhesión en Parafina , Proteómica/métodos , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Estómago/patología , Proteínas de Unión al Calcio , Linaje de la Célula , Clusterina/metabolismo , Proteínas de Unión al ADN , Progresión de la Enfermedad , Humanos , Péptidos y Proteínas de Señalización Intercelular , Lactoferrina/metabolismo , Metaplasia , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Estadificación de Neoplasias , Péptidos/metabolismo , Receptores de Superficie Celular/metabolismo , Factor Trefoil-2 , Proteínas Supresoras de Tumor , Regulación hacia Arriba
6.
J Proteome Res ; 10(7): 2896-904, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21520941

RESUMEN

In shotgun proteomics, protein identification by tandem mass spectrometry relies on bioinformatics tools. Despite recent improvements in identification algorithms, a significant number of high quality spectra remain unidentified for various reasons. Here we present ScanRanker, an open-source tool that evaluates the quality of tandem mass spectra via sequence tagging with reliable performance in data from different instruments. The superior performance of ScanRanker enables it not only to find unassigned high quality spectra that evade identification through database search but also to select spectra for de novo sequencing and cross-linking analysis. In addition, we demonstrate that the distribution of ScanRanker scores predicts the richness of identifiable spectra among multiple LC-MS/MS runs in an experiment, and ScanRanker scores assist the process of peptide assignment validation to increase confident spectrum identifications. The source code and executable versions of ScanRanker are available from http://fenchurch.mc.vanderbilt.edu.


Asunto(s)
Algoritmos , Biología Computacional , Fragmentos de Péptidos/análisis , Proteínas/análisis , Proteómica/métodos , Espectrometría de Masas en Tándem/métodos , Animales , Cromatografía Liquida , Bases de Datos de Proteínas , Humanos , Fragmentos de Péptidos/química , Proteínas/química , Proyectos de Investigación , Análisis de Secuencia de Proteína
7.
PLoS Pathog ; 2(8): e87, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16933993

RESUMEN

Staphylococcus aureus pathogenesis is significantly influenced by the iron status of the host. However, the regulatory impact of host iron sources on S. aureus gene expression remains unknown. In this study, we combine multivariable difference gel electrophoresis and mass spectrometry with multivariate statistical analyses to systematically cluster cellular protein response across distinct iron-exposure conditions. Quadruplicate samples were simultaneously analyzed for alterations in protein abundance and/or post-translational modification state in response to environmental (iron chelation, hemin treatment) or genetic (Deltafur) alterations in bacterial iron exposure. We identified 120 proteins representing several coordinated biochemical pathways that are affected by changes in iron-exposure status. Highlighted in these experiments is the identification of the heme-regulated transport system (HrtAB), a novel transport system which plays a critical role in staphylococcal heme metabolism. Further, we show that regulated overproduction of acidic end-products brought on by iron starvation decreases local pH resulting in the release of iron from the host iron-sequestering protein transferrin. These findings reveal novel strategies used by S. aureus to acquire scarce nutrients in the hostile host environment and begin to define the iron and heme-dependent regulons of S. aureus.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Hierro/metabolismo , Proteínas Represoras/metabolismo , Staphylococcus aureus/metabolismo , Proteínas Bacterianas/genética , Disponibilidad Biológica , Electroforesis en Gel Bidimensional , Hemo/metabolismo , Concentración de Iones de Hidrógeno , Deficiencias de Hierro , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo , Espectrometría de Masas , Proteómica , Proteínas Represoras/genética , Staphylococcus aureus/genética , Staphylococcus aureus/crecimiento & desarrollo , Transferrina/metabolismo
8.
Mol Cell Proteomics ; 6(1): 150-69, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17028091

RESUMEN

Multivariable DIGE/MS was used to investigate proteins altered in expression and/or post-translational modification in response to activation of transforming growth factor (TGF)-beta receptors in MCF10A mammary epithelial cells overexpressing the HER2/Neu (ErbB2) oncogene. Proteome changes were monitored in response to exogenous TGF-beta over time (0, 8, 24, and 40 h), and proteins were resolved using medium range (pH 4-7) and narrow range (pH 5.3-6.5) isoelectric focusing combined with up to 2 mg of protein to allow inspection of lower abundance proteins. Triplicate samples were prepared independently and analyzed together across multiple DIGE gels using a pooled sample internal standard to quantify expression changes with statistical confidence. Unsupervised principle component analysis and hierarchical clustering of the individual DIGE proteome expression maps provided independent confirmation of distinct expression patterns from the individual experiments and demonstrated high reproducibility between replicate samples. Fifty-nine proteins (including some isoforms) that exhibited significant kinetic expression changes were identified using mass spectrometry and database interrogation and were mapped to existing biological networks involved in TGF-beta signaling. Several proteins with a potential role in breast cancer, such as maspin and cathepsin D, were identified as novel molecules associated with TGF-beta signaling.


Asunto(s)
Electroforesis en Gel Bidimensional/métodos , Espectrometría de Masas/métodos , Receptor ErbB-2/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología , Secuencia de Aminoácidos , Catepsina D/química , Catepsina D/metabolismo , Análisis por Conglomerados , Genes Supresores de Tumor , Humanos , Concentración de Iones de Hidrógeno , Redes y Vías Metabólicas , Datos de Secuencia Molecular , Análisis Multivariante , Análisis de Componente Principal , Proteoma/química , Reproducibilidad de los Resultados , Serpinas/química , Serpinas/metabolismo , Factores de Tiempo
9.
J Cell Physiol ; 210(3): 740-9, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17133351

RESUMEN

Activating mutations of the RAS family of small GTPases are among the most common genetic events in human tumorigenesis. Constitutive activation of the three canonical family members, KRAS, NRAS, and HRAS segregate strongly by tissue type. Of these, KRAS mutations predominate in human tumors, including those arising from the colon and lung. We sought to compare the oncogenic contributions of different RAS isoforms in a comparable genetic setting and to explore downstream molecular changes that may explain the apparent differential oncogenic effects of the various RAS family members. We utilized colorectal cancer cell lines characterized by oncogenic KRAS in parallel with isogenically derived lines in which the mutant allele has been disrupted. We additionally attempted to reconstitute the isogenic derivatives with oncogenic forms of other RAS family members and analyze them in parallel. Pairwise analysis of HCT 116 and DLD-1 cell lines as well as their isogenic derivatives reveals distinct K-RAS(G13D) signatures despite the genetic similarities of these cell lines. In DLD-1, for example, oncogenic K-RAS enhances the motility of these cells by downregulation of Rap1 activity, yet is not associated with increased ERK1/2 phosphorylation. In HCT 116, however, ERK1/2 phosphorylation is elevated relative to the isogenic derivative, but Rap1 activity is unchanged. K-RAS is uniquely oncogenic in the colonic epithelium, though the molecular aspects of its oncogenic contribution are not necessarily conserved across cell lines. We therefore conclude that the oncogenic contribution of K-RAS is a function of its multifaceted functionality and is highly context-dependent.


Asunto(s)
Colon/metabolismo , Neoplasias Colorrectales/genética , Genes ras/genética , Mucosa Intestinal/metabolismo , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Colon/patología , Neoplasias Colorrectales/fisiopatología , Regulación Neoplásica de la Expresión Génica , Genes ras/fisiología , Humanos , Mucosa Intestinal/patología , Isoenzimas , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Transducción de Señal/fisiología , Proteínas de Unión al GTP rap1/genética , Proteínas de Unión al GTP rap1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA