Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Transl Lung Cancer Res ; 12(1): 109-126, 2023 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-36762061

RESUMEN

Background: Circulating tumor cells (CTCs) are a promising non-invasive tool for monitoring therapy response. The only Food and Drug Administration (FDA)-approved test is limited to enumeration of epithelial CTC without further characterization and is not approved for the management of non-small cell lung cancer (NSCLC). Here we use a MicroCavity Array (MCA) system to capture CTC agnostic of epithelial markers for further molecular testing in NSCLC. Methods: CTCs were enumerated by fluorescent microscopy as longitudinal sampling throughout disease management from 213 NSCLC patients. CTC-enriched samples from a subset of 127 patients were interrogated for gene expression by reverse transcription polymerase chain reaction (RT-PCR) using a customized pre-selected panel of 20 genes. Results: At least 1 CTC was detected by enumeration in 53.8% of samples. Most patients had fewer than 5 CTCs (91%) and the highest observed count was 35 CTCs. Enumeration of single CTCs was not prognostic, although detection of CTC clusters at any time point was associated with increased risk of progression [hazard ratio (HR) 3.00, 95% confidence interval (CI): 1.1-8.2, P=0.0318]. In contrast, 124 (97.6%) patients with samples interrogated for gene expression had at least 1 gene detectable in at least 1 sample, and 101 (79.5%) had at least one elevated epithelial gene in at least one timepoint. High expression of BCL2, CD274 [programmed death-ligand 1 (PD-L1)], CDH1, EPCAM, FGFR1, FN1, KRT18, MET and MUC1 were associated with poor prognosis. Patients with CTCs positive for at least 3 epithelial genes at baseline all progressed within 10 months (HR 8.2, P<0.001, 95% CI: 3.2-21.1). BCL2, CD274 (PD-L1), EPCAM and MUC1 remained significant independent prognostic factors in multivariate, time-dependent analyses of progression and death. Conclusions: The selective profile of CTC genes and identification of CTC clusters better correlated with prognosis than enumeration of enriched CTC in NSCLC patients in this study.

2.
Cancers (Basel) ; 15(5)2023 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-36900406

RESUMEN

Circulating tumor cells (CTCs) are indicators of metastatic spread and progression. In a longitudinal, single-center trial of patients with metastatic breast cancer starting a new line of treatment, a microcavity array was used to enrich CTCs from 184 patients at up to 9 timepoints at 3-month intervals. CTCs were analyzed in parallel samples from the same blood draw by imaging and by gene expression profiling to capture CTC phenotypic plasticity. Enumeration of CTCs by image analysis relying primarily on epithelial markers from samples obtained before therapy or at 3-month follow-up identified the patients at the highest risk of progression. CTC counts decreased with therapy, and progressors had higher CTC counts than non-progressors. CTC count was prognostic primarily at the start of therapy in univariate and multivariate analyses but had less prognostic utility at 6 months to 1 year later. In contrast, gene expression, including both epithelial and mesenchymal markers, identified high-risk patients after 6-9 months of treatment, and progressors had a shift towards mesenchymal CTC gene expression on therapy. Cross-sectional analysis showed higher CTC-related gene expression in progressors 6-15 months after baseline. Furthermore, patients with higher CTC counts and CTC gene expression experienced more progression events. Longitudinal time-dependent multivariate analysis indicated that CTC count, triple-negative status, and CTC expression of FGFR1 significantly correlated with inferior progression-free survival while CTC count and triple-negative status correlated with inferior overall survival. This highlights the utility of protein-agnostic CTC enrichment and multimodality analysis to capture the heterogeneity of CTCs.

3.
Nat Commun ; 13(1): 1791, 2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35379811

RESUMEN

Current gold standard for absolute quantitation of a specific DNA sequence is droplet digital PCR (ddPCR), which has been applied to copy number variation (CNV) detection. However, the number of quantitation modules in ddPCR is limited by fluorescence channels, which thus limits the CNV sensitivity due to sampling error following Poisson distribution. Here we develop a PCR-based molecular barcoding NGS approach, quantitative amplicon sequencing (QASeq), for accurate absolute quantitation scalable to over 200 quantitation modules. By attaching barcodes to individual target molecules with high efficiency, 2-plex QASeq exhibits higher and more consistent conversion yield than ddPCR in absolute molecule count quantitation. Multiplexed QASeq improves CNV sensitivity allowing confident distinguishment of 2.05 ploidy from normal 2.00 ploidy. We apply multiplexed QASeq to serial longitudinal plasma cfDNA samples from patients with metastatic ERBB2+ (HER2+ ) breast cancer seeking association with tumor progression. We further show an RNA QASeq panel for targeted expression profiling.


Asunto(s)
Neoplasias de la Mama , Ácidos Nucleicos Libres de Células , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Variaciones en el Número de Copia de ADN , Femenino , Humanos , Reacción en Cadena de la Polimerasa , ARN/análisis
4.
Cancers (Basel) ; 14(21)2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-36358657

RESUMEN

Circulating tumor cells (CTCs) captured from the blood of cancer patients may serve as a surrogate source of tumor material that can be obtained via a venipuncture (also known as a liquid biopsy) and used to better understand tumor characteristics. However, the only FDA-cleared CTC assay has been limited to the enumeration of surface marker-defined cells and not further characterization of the CTCs. In this study, we tested the ability of a semi-automated device capable of capturing and harvesting CTCs from peripheral blood based on cell size and deformability, agnostic of cell-surface markers (the Parsortix® PC1 System), to yield CTCs for evaluation by downstream techniques commonly available in clinical laboratories. The data generated from this study were used to support a De Novo request (DEN200062) for the classification of this device, which the FDA recently granted. As part of a multicenter clinical trial, peripheral blood samples from 216 patients with metastatic breast cancer (MBC) and 205 healthy volunteers were subjected to CTC enrichment. A board-certified pathologist enumerated the CTCs from each participant by cytologic evaluation of Wright-Giemsa-stained slides. As proof of principle, cells harvested from a concurrent parallel sample provided by each participant were evaluated using one of three additional evaluation techniques: molecular profiling by qRT-PCR, RNA sequencing, or cytogenetic analysis of HER2 amplification by FISH. The study demonstrated that the Parsortix® PC1 System can effectively capture and harvest CTCs from the peripheral blood of MBC patients and that the harvested cells can be evaluated using orthogonal methodologies such as gene expression and/or Fluorescence In Situ Hybridization (FISH).

5.
PLoS One ; 15(10): e0241123, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33095819

RESUMEN

Circulating tumor cells (CTC) isolated from the peripheral blood of cancer patients by a minimally invasive procedure provide surrogate markers of the tumor that can be repeatedly sampled. However, the selection and enumeration of CTCs by traditional methods based on surface proteins like EPCAM may not detect CTCs with a mesenchymal phenotype. Here, we employed an antibody-agnostic platform, the Parsortix® PR1 system, which enriches CTCs based on cell size and membrane deformability. We evaluated the linearity, sensitivity, and specificity of the Parsortix PR1 system in tandem with 3 downstream molecular characterization techniques using healthy donor blood spiked with cultured cell lines. Signal amplification of mRNA using a QuantiGene 25-gene assay was able to quantitate multiple epithelial genes, including CDH1, EGFR, ERBB2, KRT18, and MUC1, from high numbers of spiked cells and was able to detect KRT18 when only 50 MCF-7 or SUM190 cells were spiked into healthy donor blood. However, target amplification of mRNA by quantitative polymerase chain reaction (qPCR) showed better sensitivity; qPCR without pre-amplification was able to detect CTC-related genes in Parsortix PR1-enriched cells when as few as 5 SKBR3 cells were spiked into blood. Finally, the HTG EdgeSeq nuclease protection assay was able to profile mRNA expression of over 2,560 cancer-related genes from Parsortix PR1 enriched cells, showing enrichment in cancer signaling pathways and ERBB2, KRT19, and KRT7. Overall, the Parsortix PR1 platform may be amenable to transition into routine clinical workflows.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Separación Celular/métodos , Neoplasias/diagnóstico , Células Neoplásicas Circulantes/inmunología , Biomarcadores de Tumor/aislamiento & purificación , Recuento de Células , Línea Celular Tumoral , Separación Celular/instrumentación , Perfilación de la Expresión Génica/métodos , Voluntarios Sanos , Humanos , Microfluídica/instrumentación , Microfluídica/métodos , Neoplasias/sangre , Neoplasias/inmunología , Células Neoplásicas Circulantes/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Sensibilidad y Especificidad
6.
Transl Lung Cancer Res ; 9(5): 1974-1985, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33209617

RESUMEN

BACKGROUND: Various methods of liquid biopsy through the sampling of blood in cancer patients allow access to minuscule amounts of tumor that can easily be sampled repeatedly throughout therapy. Circulating tumor cells (CTCs) represent shed tumor cells that can be characterized by imaging or molecular techniques using an amenable enrichment platform. Here we validate the Hitachi Chemical Micro Cavity Array (MCA) for the enrichment of CTCs from the blood of patients diagnosed with stage III non-small cell lung cancer (NSCLC). MCA is a semi-automated filtration system that enriches CTCs on the basis of size and membrane deformability rather than a biased selection of surface antigens. METHODS: CTCs were enriched from the peripheral blood of 38 patients diagnosed with stage III NSCLC at the start of chemoradiation. Two tubes of EDTA blood were collected from each patient and processed through MCA in parallel. In the first tube, CTCs were identified as pan-cytokeratin (CK)+ CD45- nucleated cells and enumerated. The second tube was depleted of leukocytes using CD45 antibody-coated magnetic microbeads before enrichment by MCA, followed by quantitative reverse transcription polymerase chain reaction (qRT-PCR) to interrogate CTC-enriched lysates for expression of 16 target mRNAs from a panel of epithelial, mesenchymal, stem-like, and cancer signaling-related genes. CTC-enriched lysates from similarly prepared peripheral blood samples from 18 healthy donors were used to define positive gene expression. RESULTS: CTCs were identified by imaging in 30 of 38 patient samples (79%). At least 1 target gene was positively expressed in 23 of 25 (92%) patient samples that was subjected to molecular characterization. A CTC count of ≥7 was associated with poor progression-free survival (PFS) [hazard ratio (HR) 4.24, 95% confidence interval (CI), 1.73-10.40, P=0.020] and poor overall survival (HR 8.17, 95% CI, 2.87-23.26, P<0.001). Expression of BCL2 by MCA-enriched CTCs was associated with poor PFS (HR 3.11, 95% CI, 1.18-8.22, P=0.022). Individually, CTC count and expression of BCL2 each remained statistically significant predictors of disease progression and overall survival in multivariate analysis. CONCLUSIONS: This is the first demonstration that lysates of MCA-enriched CTCs are amenable to molecular characterization. CTCs enriched by MCA are an independent prognostic marker in NSCLC.

7.
Cancer Res ; 67(2): 709-17, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17234782

RESUMEN

FUS1 is a novel tumor suppressor gene identified in human chromosome 3p21.3 region. Loss of expression and deficiency of posttranslational modification of FUS1 protein have been found in a majority of human lung cancers. Restoration of wild-type FUS1 in 3p21.3-deficient human lung cancer cells exhibited a potent tumor suppression function in vitro and in vivo. In this study, we evaluated the combined effects of FUS1 and tumor suppressor p53 on antitumor activity and explored the molecular mechanisms of their mutual actions in human non-small cell lung cancer (NSCLC) cells. We found that coexpression of FUS1 and p53 by N-[1-(2,3-dioleoyloxyl)propyl]-NNN-trimethylammoniummethyl sulfate:cholesterol nanoparticle-mediated gene transfer significantly and synergistically inhibited NSCLC cell growth and induced apoptosis in vitro. We also found that a systemic treatment with a combination of FUS1 and p53 nanoparticles synergistically suppressed the development and growth of tumors in a human H322 lung cancer orthotopic mouse model. Furthermore, we showed that the observed synergistic tumor suppression by FUS1 and p53 concurred with the FUS1-mediated down-regulation of murine double minute-2 (MDM2) expression, the accumulation and stabilization of p53 protein, as well as the activation of the apoptotic protease-activating factor 1 (Apaf-1)-dependent apoptotic pathway in human NSCLC cells. Our results therefore provide new insights into the molecular mechanism of FUS1-mediated tumor suppression activity and imply that a molecular therapy combining two or more functionally synergistic tumor suppressors may constitute a novel and effective strategy for cancer treatment.


Asunto(s)
Factor Apoptótico 1 Activador de Proteasas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogénicas c-mdm2/biosíntesis , Proteína p53 Supresora de Tumor/biosíntesis , Proteínas Supresoras de Tumor/biosíntesis , Animales , Apoptosis/genética , Factor Apoptótico 1 Activador de Proteasas/biosíntesis , Factor Apoptótico 1 Activador de Proteasas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Terapia Genética/métodos , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Ratones , Ratones Desnudos , Nanopartículas/administración & dosificación , Plásmidos/administración & dosificación , Plásmidos/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Transfección , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética
8.
Cancer Res ; 67(13): 6293-303, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17616688

RESUMEN

101F6 is a candidate tumor suppressor gene harbored on chromosome 3p21.3, a region with frequent and early allele loss and genetic alterations in many human cancers. We previously showed that enforced expression of wild-type 101F6 by adenoviral vector-mediated gene transfer significantly inhibited tumor cell growth in 3p21.3-deficient non-small cell lung cancer (NSCLC) cells in vitro and in vivo. The molecular mechanism of 101F6-mediated tumor suppression is largely unknown. A computer-aided structural and functional model predicts the 101F6 protein to be a member of the cytochrome b561 protein family that is involved in the regeneration of the antioxidant ascorbate. 101F6 protein is expressed in normal lung bronchial epithelial cells and fibroblasts but is lost in most lung cancers. Treatment with 101F6 nanoparticle-mediated gene transfer in combination with a subpharmacologic dose (200-500 micromol/L) of ascorbate synergistically and selectively inhibited lung cancer cell growth in vitro. Systemic injection of 101F6 nanoparticles plus the i.p. injection of ascorbate synergistically inhibited both tumor formation and growth in human NSCLC H322 orthotopic lung cancer mouse models (P<0.001). Furthermore, exogenous expression of 101F6 enhanced intracellular uptake of ascorbate, leading to an accumulation of cytotoxic H(2)O(2) and a synergistic killing of tumor cells through caspase-independent apoptotic and autophagic pathways. The antitumor synergism showed by the combination treatment with systemic administration of 101F6 nanoparticles and ascorbate on lung cancer offers an attractive therapeutic strategy for future clinical trials in cancer prevention and treatment.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Autofagia , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Grupo Citocromo b/metabolismo , Genes Supresores de Tumor , Neoplasias Pulmonares/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Antioxidantes/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Inhibidores de Caspasas , Línea Celular Tumoral , Proliferación Celular , Mapeo Cromosómico , Grupo Citocromo b/genética , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Humanos , Neoplasias Pulmonares/genética , Nanopartículas , Proteínas Supresoras de Tumor/genética
9.
PLoS One ; 10(6): e0123967, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26053020

RESUMEN

Expression of the tumor suppressor gene TUSC2 is reduced or absent in most lung cancers and is associated with worse overall survival. In this study, we restored TUSC2 gene expression in several wild type EGFR non-small cell lung cancer (NSCLC) cell lines resistant to the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor erlotinib and analyzed their sensitivity to erlotinib in vitro and in vivo. A significant inhibition of cell growth and colony formation was observed with TUSC2 transient and stable expression. TUSC2-erlotinib cooperativity in vitro could be reproduced in vivo in subcutaneous tumor growth and lung metastasis formation lung cancer xenograft mouse models. Combination treatment with intravenous TUSC2 nanovesicles and erlotinib synergistically inhibited tumor growth and metastasis, and increased apoptotic activity. High-throughput qRT-PCR array analysis enabling multi-parallel expression profile analysis of eighty six receptor and non-receptor tyrosine kinase genes revealed a significant decrease of FGFR2 expression level, suggesting a potential role of FGFR2 in TUSC2-enhanced sensitivity to erlotinib. Western blots showed inhibition of FGFR2 by TUSC2 transient transfection, and marked increase of PARP, an apoptotic marker, cleavage level after TUSC2-erlotinb combined treatment. Suppression of FGFR2 by AZD4547 or gene knockdown enhanced sensitivity to erlotinib in some but not all tested cell lines. TUSC2 inhibits mTOR activation and the latter cell lines were responsive to the mTOR inhibitor rapamycin combined with erlotinib. These results suggest that TUSC2 restoration in wild type EGFR NSCLC may overcome erlotinib resistance, and identify FGFR2 and mTOR as critical regulators of this activity in varying cellular contexts. The therapeutic activity of TUSC2 could extend the use of erlotinib to lung cancer patients with wildtype EGFR.


Asunto(s)
Receptores ErbB/metabolismo , Clorhidrato de Erlotinib/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Transducción de Señal/efectos de los fármacos , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Benzamidas/farmacología , Benzamidas/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Clorhidrato de Erlotinib/farmacología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/patología , Ratones , Metástasis de la Neoplasia , Piperazinas/farmacología , Piperazinas/uso terapéutico , Pirazoles/farmacología , Pirazoles/uso terapéutico , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Resultado del Tratamiento , Ensayo de Tumor de Célula Madre
10.
J Insect Sci ; 2: 21, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-15455055

RESUMEN

Homologous transfection systems provide a useful tool for characterizing promoters and other regulatory elements from cloned genes. We have used cultured Aedes albopictus C7-10 mosquito cells to evaluate expression of 20-hydroxyecdysone-inducible genes. Although this cell line has previously been shown to synthesize components of the ecdysteroid receptor and ecdysone-inducible proteins, the well-characterized ecdysteroid response element (EcRE) from the Drosophilahsp27 promoter failed to confer a substantial 20-hydroxyecdysone mediated induction in transfected mosquito cells. Recovery of stably transformed clones was also reduced in a DNA dependent manner when the EcREs were in the sense orientation, relative to control plasmids lacking the EcREs or containing an antisense construct. Finally, when tandem EcREs were placed within the hsp70 promoter, CAT activity was detected only after prolonged enzyme incubation, suggesting that the DNA interfered with cellular metabolism. In these constructs, we noted that the promoter DNA contained several potential binding sites for the activator protein-1 (AP-1) transcription factor, one of which lay between the tandem EcREs. On southwestern blots, a 40 kDa nuclear protein from C7-10 cells bound to DNA containing AP-1 sites. A DNA affinity column was used to partially purify the 40 kDa protein, and western analysis showed that the mosquito protein cross-reacted with a heterologous antibody to JUN. Likewise, mRNA from C7-10 cells cross-hybridized with the jun cDNA from Drosophila. These results suggest that like estrogen, 20-hydroxyecdysone interfaces with AP-1 as a co-activator protein that modulates the overall hormone response.


Asunto(s)
Aedes/fisiología , Proteínas de Unión al ADN/fisiología , Proteínas de Insectos/fisiología , Elementos de Respuesta/fisiología , Aedes/citología , Aedes/genética , Animales , Complejo del Señalosoma COP9 , Supervivencia Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN/aislamiento & purificación , Ecdisteroides/genética , Ecdisteroides/fisiología , Ecdisterona/genética , Ecdisterona/fisiología , Expresión Génica/fisiología , Genes de Insecto/genética , Hormonas de Insectos/fisiología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/aislamiento & purificación , Péptido Hidrolasas/inmunología , Péptido Hidrolasas/aislamiento & purificación , Plásmidos/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Secuencias Reguladoras de Ácidos Nucleicos/genética , Elementos de Respuesta/genética , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/fisiología , Transfección/métodos
11.
Methods Mol Biol ; 818: 199-216, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22083825

RESUMEN

Protein phosphorylation is a dynamic post-translational modification that plays a critical role in the regulation of a wide spectrum of biological events and cellular functions including signal transduction, gene expression, cell proliferation, and apoptosis. Determination of the sites and magnitudes of protein phosphorylation has been an essential step in the analysis of the control of many biological systems. A high throughput analysis of phosphorylation of proteins would provide a simple, logical, and useful tool for a functional dissection and prediction of biological functions and signaling pathways in association with these important molecular events. We have developed a functional proteomics technique using the ProteinChip array-based SELDI-TOF-MS analysis for high throughput profiling of phosphoproteins/phosphopeptides in human serum for the early detection and diagnosis as well as for the molecular staging of human cancer. The methodology and experimental approach consists of five steps: (1) generation of a total peptide pool of serum proteins by a global trypsin digestion; (2) rapid isolation of phosphopeptides from the total serum peptide pool by an affinity selection, purification, and enrichment using a novel automated micro-bioprocessing system with phospho-antibody-conjugated paramagnetic beads and a hybrid magnet plate; (3) high throughput phosphopeptide analysis on ProteinChip arrays by automated SELDI-TOF-MS; and (4) bioinformatics and statistical methods for data analysis. This method with appropriate modifications may be equally applicable to serine-, threonine- and tyrosine-phosphorylated proteins and for selectively isolating, profiling, and identifying phosphopeptides present in a highly complex phosphor-peptide mixture prepared from various human specimens such as cells, tissue samples, and serum and other body fluids.


Asunto(s)
Fosfopéptidos/sangre , Fosfoproteínas/sangre , Proteómica/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Métodos Analíticos de la Preparación de la Muestra , Anticuerpos/química , Anticuerpos/inmunología , Humanos , Proteínas Inmovilizadas/sangre , Proteínas Inmovilizadas/inmunología , Proteínas Inmovilizadas/aislamiento & purificación , Proteínas Inmovilizadas/metabolismo , Imanes , Microesferas , Péptido Hidrolasas/metabolismo , Fosfopéptidos/inmunología , Fosfopéptidos/aislamiento & purificación , Fosfopéptidos/metabolismo , Fosfoproteínas/inmunología , Fosfoproteínas/aislamiento & purificación , Fosfoproteínas/metabolismo , Análisis por Matrices de Proteínas , Proteolisis , Estadística como Asunto
12.
Methods Mol Biol ; 818: 217-26, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22083826

RESUMEN

Protein-protein interactions are key elements in the assembly of cellular regulatory and signaling protein complexes that integrate and transmit signals and information in controlling and regulating various cellular processes and functions. Many conventional methods of studying protein-protein interaction, such as the immuno-precipitation and immuno-blotting assay and the affinity-column pull-down and chromatographic analysis, are very time-consuming and labor intensive and lack accuracy and sensitivity. We have developed a simple, rapid, and sensitive assay using a ProteinChip array and SELDI-TOF mass spectrometry to analyze protein-protein interactions and map the crucial elements that are directly involved in these interactions. First, a purified "bait" protein or a synthetic peptide of interest is immobilized onto the preactivated surface of a PS10 or PS20 ProteinChip and the unoccupied surfaces on the chip are protected by application of a layer ethanolamine to prevent them from binding to other non-interactive proteins. Then, the target-containing cellular protein lysate or synthetic peptide containing the predicted amino acid sequence of protein-interaction motif is applied to the protected array with immobilized bait protein/peptide. The nonspecific proteins/peptides are washed off under various stringent conditions and only the proteins specifically interacting with the bait protein/peptide remain on the chip. Last, the captured interacting protein/peptide complexes are then analyzed by SELDI-TOF mass spectrometry and their identities are confirmed by their predicted distinctive masses. This method can be used to unambiguously detect the specific protein-protein interaction of known proteins/peptides, to easily identify potential cellular targets of proteins of interest, and to accurately analyze and map the structural elements of a given protein and its target proteins using synthetic peptides with the predicted potential protein interaction motifs.


Asunto(s)
Análisis por Matrices de Proteínas/métodos , Mapeo de Interacción de Proteínas/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Secuencia de Aminoácidos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/metabolismo , Datos de Secuencia Molecular , Factores de Tiempo
13.
PLoS One ; 7(4): e34833, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22558101

RESUMEN

BACKGROUND: Tumor suppressor gene TUSC2/FUS1 (TUSC2) is frequently inactivated early in lung cancer development. TUSC2 mediates apoptosis in cancer cells but not normal cells by upregulation of the intrinsic apoptotic pathway. No drug strategies currently exist targeting loss-of-function genetic abnormalities. We report the first in-human systemic gene therapy clinical trial of tumor suppressor gene TUSC2. METHODS: Patients with recurrent and/or metastatic lung cancer previously treated with platinum-based chemotherapy were treated with escalating doses of intravenous N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTAP):cholesterol nanoparticles encapsulating a TUSC2 expression plasmid (DOTAP:chol-TUSC2) every 3 weeks. RESULTS: Thirty-one patients were treated at 6 dose levels (range 0.01 to 0.09 milligrams per kilogram). The MTD was determined to be 0.06 mg/kg. Five patients achieved stable disease (2.6-10.8 months, including 2 minor responses). One patient had a metabolic response on positron emission tomography (PET) imaging. RT-PCR analysis detected TUSC2 plasmid expression in 7 of 8 post-treatment tumor specimens but not in pretreatment specimens and peripheral blood lymphocyte controls. Proximity ligation assay, performed on paired biopsies from 3 patients, demonstrated low background TUSC2 protein staining in pretreatment tissues compared with intense (10-25 fold increase) TUSC2 protein staining in post-treatment tissues. RT-PCR gene expression profiling analysis of apoptotic pathway genes in two patients with high post-treatment levels of TUSC2 mRNA and protein showed significant post-treatment changes in the intrinsic apoptotic pathway. Twenty-nine genes of the 82 tested in the apoptosis array were identified by Igenuity Pathway Analysis to be significantly altered post-treatment in both patients (Pearson correlation coefficient 0.519; p<0.01). CONCLUSIONS: DOTAP:chol-TUSC2 can be safely administered intravenously in lung cancer patients and results in uptake of the gene by human primary and metastatic tumors, transgene and gene product expression, specific alterations in TUSC2-regulated pathways, and anti-tumor effects (to our knowledge for the first time for systemic DOTAP:cholesterol nanoparticle gene therapy). TRIAL REGISTRATION: ClinicalTrials.gov NCT00059605.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Nanopartículas/uso terapéutico , Proteínas Supresoras de Tumor/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Relación Dosis-Respuesta a Droga , Ácidos Grasos Monoinsaturados/administración & dosificación , Perfilación de la Expresión Génica , Humanos , Dosis Máxima Tolerada , Nanopartículas/administración & dosificación , Plásmidos/administración & dosificación , Tomografía de Emisión de Positrones , Compuestos de Amonio Cuaternario/administración & dosificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/administración & dosificación , Proteínas Supresoras de Tumor/metabolismo
14.
PLoS One ; 5(8): e11994, 2010 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-20700484

RESUMEN

NPRL2, one of the tumor suppressor genes residing in a 120-kb homozygous deletion region of human chromosome band 3p21.3, has a high degree of amino acid sequence homology with the nitrogen permease regulator 2 (NPR2) yeast gene, and mutations of NPRL2 in yeast cells are associated with resistance to cisplatin-mediated cell killing. Previously, we showed that restoration of NPRL2 in NPRL2-negative and cisplatin-resistant cells resensitize lung cancer cells to cisplatin treatment in vitro and in vivo. In this study, we show that sensitization of non-small cell lung cancer (NSCLC) cells to cisplatin by NPRL2 is accomplished through the regulation of key components in the DNA-damage checkpoint pathway. NPRL2 can phosphorylate ataxia telangiectasia mutated (ATM) kinase activated by cisplatin and promote downstream gamma-H2AX formation in vitro and in vivo, which occurs during apoptosis concurrently with the initial appearance of high-molecular-weight DNA fragments. Moreover, this combination treatment results in higher Chk1 and Chk2 kinase activity than does treatment with cisplatin alone and can activate Chk2 in pleural metastases tumor xenograft in mice. Activated Chk1 and Chk2 increase the expression of cell cycle checkpoint proteins, including Cdc25A and Cdc25C, leading to higher levels of G2/M arrest in tumor cells treated with NPRL2 and cisplatin than in tumor cells treated with cisplatin only. Our results therefore suggest that ectopic expression of NPRL2 activates the DNA damage checkpoint pathway in cisplatin-resistant and NPRL2-negative cells; hence, the combination of NPRL2 and cisplatin can resensitize cisplatin nonresponders to cisplatin treatment through the activation of the DNA damage checkpoint pathway, leading to cell arrest in the G2/M phase and induction of apoptosis. The direct implication of this study is that combination treatment with NPRL2 and cisplatin may overcome cisplatin resistance and enhance therapeutic efficacy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Cisplatino/farmacología , Reparación del ADN/efectos de los fármacos , Neoplasias Pulmonares/patología , Transducción de Señal/efectos de los fármacos , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas de la Ataxia Telangiectasia Mutada , Carcinoma de Pulmón de Células no Pequeñas/genética , Caspasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos , Activación Enzimática/efectos de los fármacos , Femenino , Fase G2/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Humanos , Neoplasias Pulmonares/genética , Ratones , Proteínas Nucleares/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
15.
J Biol Chem ; 282(36): 26460-70, 2007 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-17630431

RESUMEN

The up-regulated expression and telomerase activity of human telomerase reverse transcriptase (hTERT) are hallmarks of tumorigenesis. The hTERT promoter has been shown to promote hTERT gene expression selectively in tumor cells but not in normal cells. However, little is known about how tumor cells differentially activate hTERT transcription and induce telomerase activity. In this study, we identified activating enhancer-binding protein-2beta (AP-2beta) as a novel transcription factor that specifically binds to and activates the hTERT promoter in human lung cancer cells. AP-2beta was detected in hTERT promoter DNA-protein complexes formed in nuclear extracts prepared only from lung cancer cells but not from normal cells. We verified the tumor-specific binding activity of AP-2beta for the hTERT promoter in vitro and in vivo and detected high expression levels of AP-2beta in lung cancer cells. We found that ectopic expression of AP-2beta reactivated hTERT promoter-driven reporter green fluorescent protein (GFP) gene and endogenous hTERT gene expression in normal cells, enhanced GFP gene expression in lung cancer cells, and prolonged the life span of primary lung bronchial epithelial cells. Furthermore, we found that inhibition of endogenous AP-2beta expression by AP-2beta gene-specific small interfering RNAs effectively attenuated hTERT promoter-driven GFP expression, suppressed telomerase activity, accelerated telomere shortening, and inhibited tumor cell growth by induction of apoptosis in lung cancer cells. Our results demonstrate the tumor-specific activation of the hTERT promoter by AP-2beta and imply the potential of AP-2beta as a novel tumor marker or a cancer therapeutic target.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/enzimología , Proteínas de Neoplasias/metabolismo , Regiones Promotoras Genéticas , Telomerasa/biosíntesis , Factor de Transcripción AP-2/metabolismo , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Línea Celular Transformada , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Sistema Libre de Células/enzimología , Sistema Libre de Células/patología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Proteínas Fluorescentes Verdes , Humanos , Pulmón/enzimología , Pulmón/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , ARN Interferente Pequeño/farmacología , Mucosa Respiratoria/enzimología , Mucosa Respiratoria/patología , Telomerasa/antagonistas & inhibidores , Telomerasa/genética , Telómero/genética , Telómero/metabolismo , Telómero/patología , Factor de Transcripción AP-2/antagonistas & inhibidores , Factor de Transcripción AP-2/genética , Transcripción Genética/efectos de los fármacos
16.
Cancer Res ; 67(21): 10379-88, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17974981

RESUMEN

The fragile histidine triad (FHIT) gene has been shown to function as a tumor suppressor gene in vitro and in vivo. However, the mechanism of its action is still largely unknown. To elucidate the molecular mechanism and biological pathway in FHIT-mediated tumor suppression, we used a complementary gene and protein expression profiling with DNA microarray and ProteinChip technologies to quantitatively monitor cellular changes in gene and protein expression and discover the molecular targets of FHIT in non-small cell lung carcinoma (NSCLC) cells. The Ras/Rho signaling pathway was identified as one of the unique biological pathways associated with FHIT activity. A significantly down-regulated expression of genes and proteins of multiple key components in the Ras/Rho GTPases molecular switch, including Ran, Rab, Rac, Rap, and Ral, was observed on gene and protein expression profiles and further validated by Western blot analysis. Ectopic activation of FHIT in FHIT-deficient H1299 cells also significantly reduced the invasive potential of tumor cells by down-regulating expression of RhoC, a potential marker of tumor cell invasion and metastases. A simultaneous knockdown of the expression of several key Ras/Rho signaling molecules using gene-specific small interfering RNAs (RHO-siRNA) targeting selected Rab11, Rac1, and Rap1 genes significantly inhibited tumor cell growth and induced apoptosis in NSCLC cells in vitro, and a local injection of RHO-siRNAs complexed with N-[1-(2,3-dioleoyloxyl)propyl]-N,N,N-trimethylammoniummethyl sulfate:cholesterol nanoparticles inhibited tumor growth in A549 tumor xenografts in mice, mimicking the AdFHIT-mediated tumor-suppressing effect. These results suggest a new role of FHIT in down-regulating the Ras/Rho GTPase-associated oncogenic signaling pathway.


Asunto(s)
Ácido Anhídrido Hidrolasas/fisiología , Carcinoma de Pulmón de Células no Pequeñas/prevención & control , Genes ras , Neoplasias Pulmonares/prevención & control , Proteínas de Neoplasias/fisiología , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Animales , Apoptosis , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Pulmonares/patología , Ratones , Invasividad Neoplásica , Proteínas de Neoplasias/análisis , Análisis por Matrices de Proteínas , ARN Interferente Pequeño/farmacología
17.
Genetica ; 119(1): 1-10, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12903741

RESUMEN

Ribosomal protein P0 is one of the highly conserved phosphorylated proteins in the large subunit of eukaryotic ribosomes. P0 has been shown in Drosophila to be a multifunctional protein that associates with elongation factor eEF2 to facilitate translation and also plays a role in DNA repair. In this paper we describe the cloning and characterization of the full-length cDNA encoding P0 from the mosquito, Aedes albopictus. In vitro translation showed that the cDNA encoded a 34kDa protein that corresponded in size to a phosphorylated protein from the large ribosomal subunit after in vivo labeling with [32P]orthophosphate. When cells were exposed to various stresses, expression of P0 appeared to differ from that of rpL34. Preliminary RNAi experiments suggested that downregulation of P0 is correlated with apoptosis in C7-10 mosquito cells.


Asunto(s)
Aedes/genética , ADN Complementario/genética , Expresión Génica/genética , Fosfoproteínas/genética , Proteínas Ribosómicas/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Datos de Secuencia Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA