Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Clin Chem Lab Med ; 57(8): 1142-1152, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31112502

RESUMEN

Background The National Institute of Standards and Technology (NIST) Reference Material RM 8366 was developed to improve the quality of gene copy measurements of EGFR (epidermal growth factor receptor) and MET (proto-oncogene, receptor tyrosine kinase), important targets for cancer diagnostics and treatment. The reference material is composed of genomic DNA prepared from six human cancer cell lines with different levels of amplification of the target genes. Methods The reference values for the ratios of the EGFR and MET gene copy numbers to the copy numbers of reference genes were measured using digital PCR. The digital PCR measurements were confirmed by two additional laboratories. The samples were also characterized using Next Generation Sequencing (NGS) methods including whole genome sequencing (WGS) at three levels of coverage (approximately 1 ×, 5 × and greater than 30 ×), whole exome sequencing (WES), and two different pan-cancer gene panels. The WES data were analyzed using three different bioinformatic algorithms. Results The certified values (digital PCR) for EGFR and MET were in good agreement (within 20%) with the values obtained from the different NGS methods and algorithms for five of the six components; one component had lower NGS values. Conclusions This study shows that NIST RM 8366 is a valuable reference material to evaluate the performance of assays that assess EGFR and MET gene copy number measurements.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/normas , Proteínas Proto-Oncogénicas c-met/genética , ADN de Neoplasias/genética , Receptores ErbB/genética , Receptores ErbB/normas , Dosificación de Gen , Humanos , Reacción en Cadena de la Polimerasa , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-met/normas , Estándares de Referencia , Células Tumorales Cultivadas
2.
Cancer ; 124(5): 1070-1082, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29194591

RESUMEN

BACKGROUND: It is possible that the relative lack of progress in treatment outcomes among adolescent and young adult (AYA) patients with cancer is caused by a difference in disease biology compared with the corresponding diseases in younger and older individuals. There is evidence that colon cancer is more aggressive and has a poorer prognosis in AYA patients than in older adult patients. METHODS: To further understand the molecular basis for this difference, whole-exome sequencing was conducted on a cohort of 30 adult, 30 AYA, and 2 pediatric colon cancers. RESULTS: A statistically significant difference in mutational frequency was observed between AYA and adult samples in 43 genes, including ROBO1, MYC binding protein 2 (MYCBP2), breast cancer 2 (early onset) (BRCA2), MAP3K3, MCPH1, RASGRP3, PTCH1, RAD9B, CTNND1, ATM, NF1; KIT, PTEN, and FBXW7. Many of these mutations were nonsynonymous, missense, stop-gain, or frameshift mutations that were damaging. Next, RNA sequencing was performed on a subset of the samples to confirm the mutations identified by exome sequencing. This confirmation study verified the presence of a significantly greater frequency of damaging mutations in AYA compared with adult colon cancers for 5 of the 43 genes (MYCBP2, BRCA2, PHLPP1, TOPORS, and ATR). CONCLUSIONS: The current results provide the rationale for a more comprehensive study with a larger sample set and experimental validation of the functional impact of the identified variants along with their contribution to the biologic and clinical characteristics of AYA colon cancer. Cancer 2018;124:1070-82. © 2017 American Cancer Society.


Asunto(s)
Colon/metabolismo , Neoplasias del Colon/genética , Secuenciación del Exoma/métodos , Predisposición Genética a la Enfermedad/genética , Mutación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Colon/patología , Neoplasias del Colon/patología , Femenino , Perfilación de la Expresión Génica/métodos , Frecuencia de los Genes , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
3.
J Cell Physiol ; 228(7): 1536-50, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23280476

RESUMEN

Recent studies have suggested that changes in serum phosphate levels influence pathological states associated with aging such as cancer, bone metabolism, and cardiovascular function, even in individuals with normal renal function. The causes are only beginning to be elucidated but are likely a combination of endocrine, paracrine, autocrine, and cell autonomous effects. We have used an integrated quantitative biology approach, combining transcriptomics and proteomics to define a multi-phase, extracellular phosphate-induced, signaling network in pre-osteoblasts as well as primary human and mouse mesenchymal stromal cells. We identified a rapid mitogenic response stimulated by elevated phosphate that results in the induction of immediate early genes including c-fos. The mechanism of activation requires FGF receptor signaling followed by stimulation of N-Ras and activation of AP-1 and serum response elements. A distinct long-term response also requires FGF receptor signaling and results in N-Ras activation and expression of genes and secretion of proteins involved in matrix regulation, calcification, and angiogenesis. The late response is synergistically enhanced by addition of FGF23 peptide. The intermediate phase results in increased oxidative phosphorylation and ATP production and is necessary for the late response providing a functional link between the phases. Collectively, the results define elevated phosphate, as a mitogen and define specific mechanisms by which phosphate stimulates proliferation and matrix regulation. Our approach provides a comprehensive understanding of the cellular response to elevated extracellular phosphate, functionally connecting temporally coordinated signaling, transcriptional, and metabolic events with changes in long-term cell behavior.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Fosfatos/metabolismo , Transducción de Señal/fisiología , Células 3T3 , Adenosina Trifosfato/biosíntesis , Animales , Células Cultivadas , Biología Computacional , Espacio Extracelular/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas de Unión al GTP/metabolismo , Expresión Génica , Genes Inmediatos-Precoces , Genes fos , Genes ras , Humanos , Ratones , Neovascularización Fisiológica , Osteoblastos/metabolismo , Regiones Promotoras Genéticas , Proteínas/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo
4.
Nanomedicine ; 8(6): 793-803, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22100753

RESUMEN

Bone is a dynamic tissue that undergoes renewal throughout life in a process whereby osteoclasts resorb worn bone and osteoblasts synthesize new bone. Imbalances in bone turnover lead to bone loss and development of osteoporosis and ultimately fracture, a debilitating condition with high morbidity and mortality. Silica is a ubiquitous biocontaminant that is considered to have high biocompatibility. The authors report that silica nanoparticles (NPs) mediate potent inhibitory effects on osteoclasts and stimulatory effects on osteoblasts in vitro. The mechanism of bioactivity is a consequence of an intrinsic capacity to antagonize activation of NF-κB, a signal transduction pathway required for osteoclastic bone resorption but inhibitory to osteoblastic bone formation. We further demonstrate that silica NPs promote a significant enhancement of bone mineral density (BMD) in mice in vivo, providing a proof of principle for the potential application of silica NPs as a pharmacological agent to enhance BMD and protect against bone fracture.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Resorción Ósea/prevención & control , Resorción Ósea/fisiopatología , Nanocápsulas/administración & dosificación , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Dióxido de Silicio/administración & dosificación , Células 3T3 , Animales , Resorción Ósea/patología , Sustitutos de Huesos/administración & dosificación , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Osteoblastos/patología , Osteoclastos/patología , Osteogénesis/efectos de los fármacos
5.
Chem Commun (Camb) ; (20): 2881-3, 2009 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19436897

RESUMEN

A new synthetic method has been developed to prepare fluorescent silica nanoparticles without employing isothiocyanated dye molecules and (3-aminopropyl)triethoxysilane (APS) for the thiourea linkage formation; the resulting fluorescent silica nanoparticles show excellent photochemical, thermal and pH stabilities and a good biocompatibility with over 85% viability from various cell types.


Asunto(s)
Nanopartículas/química , Nanopartículas/toxicidad , Nanotecnología/métodos , Dióxido de Silicio/química , Dióxido de Silicio/toxicidad , Células 3T3 , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Fluorescencia , Humanos , Concentración de Iones de Hidrógeno , Ratones , Tamaño de la Partícula , Fotoquímica , Silanos/química , Temperatura
6.
Cancer Res ; 65(14): 6034-41, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16024603

RESUMEN

Programmed cell death 4 (Pdcd4) is a novel repressor of in vitro transformation. Pdcd4 directly inhibits the helicase activity of eukaryotic translation initiation factor 4A, a component of the translation initiation complex. To ascertain whether Pdcd4 suppresses tumor development in vivo, we have generated transgenic mice that overexpress Pdcd4 in the epidermis (K14-Pdcd4). K14-regulated Pdcd4 expression caused a neonatal short-hair phenotype due to early catagen entry compared with matched wild-type siblings. In response to the 7,12-dimethylbenz(a)anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA) mouse skin carcinogenesis protocol, K14-Pdcd4 mice showed significant reductions in papilloma formation, carcinoma incidence, and papilloma-to-carcinoma conversion frequency compared with wild-type mice. The translational efficiency of an mRNA engineered to form a structured 5' untranslated region (UTR) was attenuated in primary keratinocytes when Pdcd4 was overexpressed. Pdcd4 inhibited by 46% TPA-induced activator protein-1 (AP-1)-dependent transcription, an event required for tumorigenesis. CDK4 and ornithine decarboxylase (ODC) are candidates for Pdcd4-regulated translation as their mRNAs contain 5'structured UTRs. In K14-Pdcd4 primary keratinocytes expressing activated Ha-Ras to mimic DMBA-initiated epidermis, ODC and CDK4 protein levels were decreased by 40% and 46%, respectively. Expression of a protein encoded by 5' unstructured mRNA showed no change. These results extend to an in vivo model the observations that Pdcd4 inhibits both translation initiation and AP-1 activation while decreasing benign tumor development and malignant progression. The K14-Pdcd4 mice seem to validate translation initiation as a novel target for cancer prevention.


Asunto(s)
Carcinoma de Células Escamosas/prevención & control , Papiloma/prevención & control , Proteínas de Unión al ARN/fisiología , Neoplasias Cutáneas/prevención & control , Regiones no Traducidas 5' , Animales , Proteínas Reguladoras de la Apoptosis , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Citoplasma/metabolismo , Progresión de la Enfermedad , Epidermis/metabolismo , Epidermis/patología , Femenino , Luciferasas/antagonistas & inhibidores , Luciferasas/biosíntesis , Luciferasas/genética , Ratones , Ratones Transgénicos , Papiloma/genética , Papiloma/metabolismo , Papiloma/patología , Regiones Promotoras Genéticas , Biosíntesis de Proteínas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Factor de Transcripción AP-1/antagonistas & inhibidores , Factor de Transcripción AP-1/fisiología , Activación Transcripcional
7.
Cancer Inform ; 15: 65-71, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27147817

RESUMEN

With rapid advances in DNA sequencing technologies, whole exome sequencing (WES) has become a popular approach for detecting somatic mutations in oncology studies. The initial intent of WES was to characterize single nucleotide variants, but it was observed that the number of sequencing reads that mapped to a genomic region correlated with the DNA copy number variants (CNVs). We propose a method RefCNV that uses a reference set to estimate the distribution of the coverage for each exon. The construction of the reference set includes an evaluation of the sources of variability in the coverage distribution. We observed that the processing steps had an impact on the coverage distribution. For each exon, we compared the observed coverage with the expected normal coverage. Thresholds for determining CNVs were selected to control the false-positive error rate. RefCNV prediction correlated significantly (r = 0.96-0.86) with CNV measured by digital polymerase chain reaction for MET (7q31), EGFR (7p12), or ERBB2 (17q12) in 13 tumor cell lines. The genome-wide CNV analysis showed a good overall correlation (Spearman's coefficient = 0.82) between RefCNV estimation and publicly available CNV data in Cancer Cell Line Encyclopedia. RefCNV also showed better performance than three other CNV estimation methods in genome-wide CNV analysis.

8.
J Mol Diagn ; 18(5): 753-761, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27455875

RESUMEN

The National Institute of Standards and Technology (NIST) Standard Reference Materials 2373 is a set of genomic DNA samples prepared from five breast cancer cell lines with certified values for the ratio of the HER2 gene copy number to the copy numbers of reference genes determined by real-time quantitative PCR and digital PCR. Targeted-amplicon, whole-exome, and whole-genome sequencing measurements were used with the reference material to compare the performance of both the laboratory steps and the bioinformatic approaches of the different methods using a range of amplification ratios. Although good reproducibility was observed in each next-generation sequencing method, slightly different HER2 copy numbers associated with platform-specific biases were obtained. This study clearly demonstrates the value of Standard Reference Materials 2373 as reference material and as a calibrator for evaluating assay performance as well as for increasing confidence in reporting HER2 amplification for clinical applications.


Asunto(s)
Amplificación de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Receptor ErbB-2/genética , Estándares de Referencia , Línea Celular Tumoral , Exoma , Femenino , Dosificación de Gen , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/normas , Humanos , Masculino , Neoplasias/diagnóstico , Neoplasias/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
J Mol Diagn ; 18(3): 336-349, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27105923

RESUMEN

Although next-generation sequencing technologies have been widely adapted for clinical diagnostic applications, an urgent need exists for multianalyte calibrator materials and controls to evaluate the performance of these assays. Control materials will also play a major role in the assessment, development, and selection of appropriate alignment and variant calling pipelines. We report an approach to provide effective multianalyte controls for next-generation sequencing assays, referred to as the control plasmid spiked-in genome (CPSG). Control plasmids that contain approximately 1000 bases of human genomic sequence with a specific mutation of interest positioned near the middle of the insert and a nearby 6-bp molecular barcode were synthesized, linearized, quantitated, and spiked into genomic DNA derived from formalin-fixed, paraffin-embedded-prepared hapmap cell lines at defined copy number ratios. Serial titration experiments demonstrated the CPSGs performed with similar efficiency of variant detection as formalin-fixed, paraffin-embedded cell line genomic DNA. Repetitive analyses of one lot of CPSGs 90 times during 18 months revealed that the reagents were stable with consistent detection of each of the plasmids at similar variant allele frequencies. CPSGs are designed to work across most next-generation sequencing methods, platforms, and data analysis pipelines. CPSGs are robust controls and can be used to evaluate the performance of different next-generation sequencing diagnostic assays, assess data analysis pipelines, and ensure robust assay performance metrics.


Asunto(s)
Pruebas Genéticas/métodos , Pruebas Genéticas/normas , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/normas , Plásmidos/genética , Control de Calidad , Estándares de Referencia , Biología Computacional/métodos , Código de Barras del ADN Taxonómico/métodos , Código de Barras del ADN Taxonómico/normas , Genómica/métodos , Genómica/normas , Humanos , Reproducibilidad de los Resultados , Flujo de Trabajo
10.
Mol Cancer Ther ; 3(2): 103-10, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14985450

RESUMEN

Programmed cell death 4 (Pdcd4), originally identified as an inhibitor of murine cellular transformation, inhibits protein synthesis by directly interacting with eukaryotic initiation factor 4A (eIF4A) of the translation initiation complex. The relevance of Pdcd4 to a broad range of human cancers derived from multiple tissue sites is unknown. Protein expression patterns from the National Cancer Institute drug-screening panel of 60 human cancer cells (NCI60) were analyzed by Western blot methods and revealed frequent reduction of Pdcd4 protein levels in renal-, lung-, and glia-derived tumors. Greater than mean Pdcd4 protein levels correlated with the antitumor activity of geldanamycin and tamoxifen. Stable expression of antisense PDCD4 significantly reduced the sensitivity of MCF-7 breast cancer cells to geldanamycin and to tamoxifen. Sensitivity to geldanamycin significantly increased in UO-31 renal cancer cells expressing sense PDCD4 cDNA. Increased geldanamycin sensitivity was accompanied by enhanced cell cycle arrest and apoptosis. One primary mode of inactivation of Pdcd4 in human cancers appears to involve down-regulated expression, and this down-regulation causes a decreased sensitivity to geldanamycin cytotoxicity. Thus, up-regulating Pdcd4 expression may be promising for geldanamycin-based combination therapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/metabolismo , Proteínas de Unión al ARN/metabolismo , Tacrolimus/análogos & derivados , Proteínas Reguladoras de la Apoptosis , Benzoquinonas , División Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Fase G2/efectos de los fármacos , Humanos , Neoplasias Renales/metabolismo , Lactamas Macrocíclicas , Mitosis/efectos de los fármacos , Pronóstico , Quinonas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteína de Retinoblastoma/metabolismo , Tacrolimus/farmacología , Tamoxifeno/farmacología
11.
Soft Mater ; 11(2): 195-203, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24058289

RESUMEN

Nanomaterials are diverse in size, shape and charge and these differences likely alter their physicochemical properties in biological systems. We have investigated how these properties alter the initial and long-term dynamics of endocytosis, cell viability, cell division, exocytosis, and interaction with a collagen extracellular matrix using silica-based fluorescent nanoparticles and the murine pre-osteoblast cell line, MC3T3-E1. Three surface modified nanoparticles were analyzed: positively charged (PTMA), negatively charged (OH), and neutrally charged polyethylene glycol (PEG). Positively charged PTMA-modified nanoparticles demonstrated the most rapid uptake, within 2 hours, while PEG modified and negatively charged OH nanoparticles demonstrated slower uptake. Cell viability was >80% irrespective of nanoparticle surface charge suggesting a general lack of toxicity. Long-term monitoring of fluorescent intensity revealed that nanoparticles were passed to daughter cells during mitotic cell division with a corresponding decrease in fluorescent intensity. These data suggest that irrespective of surface charge silica nanoparticles have the potential to internalize into osteoblasts, albeit with different kinetics. Furthermore, long lived nanoparticles have the potential to be transferred to daughter cells during mitosis and can be maintained for weeks intracellularly or within a collagen matrix without toxicity and limited exocytosis.

12.
Transl Res ; 161(3): 145-55, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23022285

RESUMEN

Thiazolidinedione (TZD) therapy has been associated with an increased risk of bone fractures. Studies in rodents have led to a model in which decreased bone quality in response to TZDs is due to a competition of lineage commitment between osteoblasts (OBs) and adipocytes (ADs) for a common precursor cell, resulting in decreased OB numbers. Our goal was to investigate the effects of TZD exposure on OB-AD lineage determination from primary human bone marrow stromal cells (hBMSCs) both in vitro and in vivo from nondiabetic subjects and patients with type 2 diabetics. Our experimental design included 2 phases. Phase 1 was an in vitro study of TZD effects on the differentiation of hBMSCs into OBs and ADs in nondiabetic subjects. Phase 2 was a randomized, placebo-controlled trial to determine the effects of 6-month pioglitazone treatment in vivo on hBMSC differentiation using AD/OB colony forming unit assays in patients with type 2 diabetes. In vitro, TZDs (pioglitazone and rosiglitazone) enhanced the adipogenesis of hBMSCs, whereas neither altered OB differentiation or function as measured by alkaline phosphatase activity, gene expression, and mineralization. The ability of TZDs to enhance adipogenesis occurred at a specific time/stage of the differentiation process, and pretreating with TZDs did not further enhance adipogenesis. In vivo, 6-month TZD treatment decreased OB precursors, increased AD precursors, and increased total colony number in patients with type 2 diabetes. Our results indicate that TZD exposure in vitro potently stimulates adipogenesis but does not directly alter OB differentiation/mineralization or lineage commitment from hBMSCs. However, TZD treatment in type 2 diabetic patients results in decreased osteoblastogenesis from hBMSCs compared with placebo, indicating an indirect negative effect on OBs and suggesting an alternative model by which TZDs might negatively regulate bone quality.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/patología , Hipoglucemiantes/efectos adversos , Células Madre Mesenquimatosas/efectos de los fármacos , Tiazolidinedionas/efectos adversos , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/patología , Adipogénesis/efectos de los fármacos , Densidad Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Fracturas Óseas/inducido químicamente , Expresión Génica/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/metabolismo , Células Madre Multipotentes/patología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoblastos/patología , Osteogénesis/efectos de los fármacos , Osteoporosis/inducido químicamente , Pioglitazona , Investigación Biomédica Traslacional
13.
J Bone Miner Res ; 26(1): 209-19, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20683885

RESUMEN

Tumor necrosis factor α (TNF-α) promotes bone loss and inhibits bone formation. Osterix (Osx, SP7) is a transcription factor required for osteoblast (OB) differentiation because deletion results in a cartilaginous skeleton. We previously described a TNF suppressor element in the Osx promoter that was used to isolate nuclear proteins mediating TNF inhibition of OB differentiation. Nuclear extracts from TNF-treated pre-OBs were incubated with the TNF suppressor element for protein pull-down, and tryptic fragments were analyzed by mass spectrometry. Chromatin immunoprecipitation (ChIP) assay confirmed eight bound transcription factors. One protein, the paired related homeobox protein (Prx1), had been shown previously to have a critical role in limb bud formation and skeletal patterning. PCR revealed Prx1 expression in primary stromal cells (MSCs), C3H10T1/2 cells, and MC3T3 preosteoblasts. TNF stimulated a 14-fold increase in mRNA for Prx1, rapid cell accumulation in MC3T3 cells, and expression in periosteal and trabecular lining cells in vivo. Transient expression of Prx inhibited transcription of Osx and RUNX2. Expression of the Prx1b isoform or Prx2 decreased Osx and RUNX2 mRNA and OB differentiation in preosteoblasts. Silencing of Prx1 with siRNA abrogated TNF suppression of Osx mRNA and increased basal Osx expression. Electrophoretic mobility shift revealed Prx1b as the preferred isoform binding the Osx promoter. These results identify the homeobox protein Prx1 as an obligate mediator of TNF inhibition of Osx and differentiation of OB progenitors. Activation of Prx1 by TNF may contribute to reduced bone formation in inflammatory arthritis, menopause, and aging.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Factores de Transcripción/genética , Factor de Necrosis Tumoral alfa/farmacología , Animales , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Ratones , Osteoblastos/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factor de Transcripción Sp7 , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos
14.
Cancer Prev Res (Phila) ; 3(3): 359-70, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20145188

RESUMEN

Recent results suggest a paradigm shift from viewing inorganic phosphate as a passive requirement for basic cell functions to an active regulator of cell behavior. We have previously shown that elevated concentrations of phosphate increased cell proliferation and expression of protumorigenic genes such as Fra-1 and osteopontin in a preosteoblast cell line. Therefore, we hypothesized that elevated phosphate concentrations would promote cell transformation in vitro and tumorigenesis in vivo. Supplementation of medium with phosphate increased anchorage-independent transformation and proliferation of BALB/c mouse JB6 epidermal cells, activation of N-ras, ERK1/2, and activator protein-1, and increased gene expression of Fra-1, COX-2, and osteopontin in a dose-dependent manner. These in vitro results led to the hypothesis that varying the levels of dietary inorganic phosphate would alter tumorigenesis in the mouse model of skin carcinogenesis. Female FVB/N mice were treated with 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphorbol-13-acetate and fed high- or low-phosphate diets (1.2% versus 0.2% of the diet) for 19 weeks. The high-phosphate diet increased skin papilloma number by approximately 50% without changing feed intake and body weights. High dietary phosphate increased serum concentrations of phosphate, parathyroid hormone, and osteopontin and decreased serum concentrations of calcium. Thus, we conclude that elevated phosphate promotes cell transformation and skin tumorigenesis partly by increasing the availability of phosphate for activation of N-ras and its downstream targets, which defines reducing dietary phosphate as a novel target for chemoprevention.


Asunto(s)
Transformación Celular Neoplásica , Genes ras/fisiología , Papiloma/etiología , Fósforo Dietético/administración & dosificación , Neoplasias Cutáneas/etiología , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Northern Blotting , Western Blotting , Carcinógenos/toxicidad , Inmunoprecipitación de Cromatina , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Epidermis/efectos de los fármacos , Femenino , Luciferasas/metabolismo , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Osteopontina/sangre , Osteopontina/genética , Osteopontina/metabolismo , Papiloma/metabolismo , Hormona Paratiroidea/sangre , Fosfatos/sangre , Fosfatos/farmacología , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Neoplasias Cutáneas/metabolismo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Transfección
15.
J Cell Physiol ; 210(2): 325-35, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17096383

RESUMEN

Many key processes central to bone formation and homeostasis require the involvement of osteoblasts, cells responsible for accumulation and mineralization of the extracellular matrix (ECM). During this complex and only partially understood process, osteoblasts generate and secrete matrix vesicles (MVs) into the ECM to initiate mineralization. Although they are considered an important component of mineralization process, MVs still remain a mystery. To better understand their function and biogenesis, a proteomic analysis of MVs has been conducted. MVs were harvested by two sample preparation approaches and mass spectrometry was utilized for protein identification. A total of 133 proteins were identified in common from the two MV preparations, among which were previously known proteins, such as annexins and peptidases, along with many novel proteins including a variety of enzymes, osteoblast-specific factors, ion channels, and signal transduction molecules, such as 14-3-3 family members and Rab-related proteins. To compare the proteome of MV with that of the ECM we conducted a large-scale proteomic analysis of collagenase digested mineralizing osteoblast matrix. This analysis resulted in the identification of 1,327 unique proteins. A comparison of the proteins identified from the two MV preparations with the ECM analysis revealed 83 unique, non-redundant proteins identified in all three samples. This investigation represents the first systematic proteomic analysis of MVs and provides insights into both the function and origin of these important mineralization-regulating vesicles.


Asunto(s)
Matriz Ósea/metabolismo , Calcificación Fisiológica/fisiología , Vesículas Citoplasmáticas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Osteoblastos/metabolismo , Proteoma/metabolismo , Animales , Desarrollo Óseo/fisiología , Matriz Ósea/ultraestructura , Línea Celular , Vesículas Citoplasmáticas/ultraestructura , Proteínas de la Matriz Extracelular/análisis , Técnica del Anticuerpo Fluorescente , Espectrometría de Masas , Ratones , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Osteoblastos/ultraestructura , Osteogénesis/fisiología , Proteómica
16.
J Proteome Res ; 5(8): 1931-9, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16889415

RESUMEN

Current advances in proteomics have allowed for a rapidly expanding integration of associated methodologies with more traditional molecular and biochemical approaches to the study of cell function. Recent studies on the role of inorganic phosphate have suggested this ion is a novel signaling molecule capable of altering the function of numerous cell types. Elevated inorganic phosphate generated in the extracellular microenvironment by differentiating osteoblasts has recently been determined to act through a largely uncharacterized mechanism as an important signaling molecule responsible for altering the transcription of various genes during osteoblast differentiation. The transcription factor, early growth response protein 1 (EGR1), has previously been shown to be involved in the early response of osteoblasts to inorganic phosphate. To elucidate the role of EGR1 as a potential early regulator of transcription in the inorganic phosphate response, an oligoprecipitation procedure was optimized to capture the DNA bound, transcriptionally active form of EGR1. The interacting proteins thusly captured were identified using mass spectrometry (MS). Proteins involved in transcription, RNA processing, and chromatin modification were identified by this approach. The combined oligoprecipitation-MS approach presented here is highly effective for isolating and characterizing entire transcriptional complexes in the DNA bound state and is broadly extendable to the identification of both known and unknown transcription factor protein complexes.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Espectrometría de Masas/métodos , Osteoblastos/fisiología , Regiones Promotoras Genéticas , Proteoma/análisis , Células 3T3 , Animales , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Ratones , Datos de Secuencia Molecular , Osteoblastos/citología , Fosfatos/metabolismo , Transcripción Genética
17.
Mol Cell Proteomics ; 4(9): 1284-96, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15958391

RESUMEN

Inorganic phosphate, which is generated during osteoblast differentiation and mineralization, has recently been identified as an important signaling molecule capable of altering signal transduction pathways and gene expression. A large scale quantitative proteomic investigation of pre-osteoblasts stimulated with inorganic phosphate for 24 h resulted in the identification of 2501 proteins, of which 410 (16%) had an altered abundance ratio of greater than or equal to 1.75-fold, either up or down, revealing both novel and previously defined osteoblast-regulated proteins. A pathway/function analysis of these proteins revealed an increase in cell cycle and proliferation that was subsequently verified by conventional biochemical means. To further analyze the mechanisms by which inorganic phosphate regulates cellular protein levels, we undertook a mRNA microarray analysis of pre-osteoblast cells at 18, 21, and 24 h after inorganic phosphate exposure. Comparison of the mRNA microarray data with the 24-hour quantitative proteomic data resulted in a generally weak overall correlation; the 21-hour RNA sample showed the highest correlation to the proteomic data. However, an analysis of osteoblast relevant proteins revealed a much higher correlation at all time points. A comparison of the microarray and proteomic datasets allowed for the identification of a number of candidate proteins that are post-transcriptionally regulated by elevated inorganic phosphate, including Fra-1, a member of the activator protein-1 family of transcription factors. The analysis of the data presented here not only sheds new light on the important roles of inorganic phosphate in osteoblast function but also begins to address the contribution of post-transcriptional and post-translational regulation to a cell's expressed proteome. The ability to accurately measure changes in both protein abundance and mRNA levels on a system-wide scale represents a novel means to extract data from previously one-dimensional datasets.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Análisis por Micromatrices , Osteoblastos/efectos de los fármacos , Fosfatos/farmacología , Proteoma/análisis , Células 3T3 , Animales , Western Blotting , Isótopos de Carbono , Proliferación Celular , Citometría de Flujo , Marcaje Isotópico , Espectrometría de Masas , Ratones , Osteoblastos/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA