Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Genome Res ; 27(7): 1112-1125, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28411194

RESUMEN

RNA editing, a widespread post-transcriptional mechanism, has emerged as a new player in cancer biology. Recent studies have reported key roles for individual miRNA editing events, but a comprehensive picture of miRNA editing in human cancers remains largely unexplored. Here, we systematically characterized the miRNA editing profiles of 8595 samples across 20 cancer types from miRNA sequencing data of The Cancer Genome Atlas and identified 19 adenosine-to-inosine (A-to-I) RNA editing hotspots. We independently validated 15 of them by perturbation experiments in several cancer cell lines. These miRNA editing events show extensive correlations with key clinical variables (e.g., tumor subtype, disease stage, and patient survival time) and other molecular drivers. Focusing on the RNA editing hotspot in miR-200b, a key tumor metastasis suppressor, we found that the miR-200b editing level correlates with patient prognosis opposite to the pattern observed for the wild-type miR-200b expression. We further experimentally showed that, in contrast to wild-type miRNA, the edited miR-200b can promote cell invasion and migration through its impaired ability to inhibit ZEB1/ZEB2 and acquired concomitant ability to repress new targets, including LIFR, a well-characterized metastasis suppressor. Our study highlights the importance of miRNA editing in gene regulation and suggests its potential as a biomarker for cancer prognosis and therapy.


Asunto(s)
Genes Supresores de Tumor , MicroARNs/metabolismo , Neoplasias/metabolismo , Edición de ARN , ARN Neoplásico/metabolismo , Adenosina/genética , Adenosina/metabolismo , Femenino , Humanos , Inosina/genética , Inosina/metabolismo , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/genética , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Masculino , MicroARNs/genética , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/patología , ARN Neoplásico/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
2.
Mol Cell Proteomics ; 17(6): 1245-1258, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29531020

RESUMEN

Molecular analysis of tumors forms the basis for personalized cancer medicine and increasingly guides patient selection for targeted therapy. Future opportunities for personalized medicine are highlighted by the measurement of protein expression levels via immunohistochemistry, protein arrays, and other approaches; however, sample type, sample quantity, batch effects, and "time to result" are limiting factors for clinical application. Here, we present a development pipeline for a novel multiplexed DNA-labeled antibody platform which digitally quantifies protein expression from lysate samples. We implemented a rigorous validation process for each antibody and show that the platform is amenable to multiple protocols covering nitrocellulose and plate-based methods. Results are highly reproducible across technical and biological replicates, and there are no observed "batch effects" which are common for most multiplex molecular assays. Tests from basal and perturbed cancer cell lines indicate that this platform is comparable to orthogonal proteomic assays such as Reverse-Phase Protein Array, and applicable to measuring the pharmacodynamic effects of clinically-relevant cancer therapeutics. Furthermore, we demonstrate the potential clinical utility of the platform with protein profiling from breast cancer patient samples to identify molecular subtypes. Together, these findings highlight the potential of this platform for enhancing our understanding of cancer biology in a clinical translation setting.


Asunto(s)
Anticuerpos/química , ADN/química , Neoplasias/metabolismo , Proteínas/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Proteómica
3.
Genome Res ; 22(11): 2120-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23028188

RESUMEN

Endometrial cancer is the most common gynecological malignancy, with more than 280,000 cases occurring annually worldwide. Although previous studies have identified important common somatic mutations in endometrial cancer, they have primarily focused on a small set of known cancer genes and have thus provided a limited view of the molecular basis underlying this disease. Here we have developed an integrated systems-biology approach to identifying novel cancer genes contributing to endometrial tumorigenesis. We first performed whole-exome sequencing on 13 endometrial cancers and matched normal samples, systematically identifying somatic alterations with high precision and sensitivity. We then combined bioinformatics prioritization with high-throughput screening (including both shRNA-mediated knockdown and expression of wild-type and mutant constructs) in a highly sensitive cell viability assay. Our results revealed 12 potential driver cancer genes including 10 tumor-suppressor candidates (ARID1A, INHBA, KMO, TTLL5, GRM8, IGFBP3, AKTIP, PHKA2, TRPS1, and WNT11) and two oncogene candidates (ERBB3 and RPS6KC1). The results in the "sensor" cell line were recapitulated by siRNA-mediated knockdown in endometrial cancer cell lines. Focusing on ARID1A, we integrated mutation profiles with functional proteomics in 222 endometrial cancer samples, demonstrating that ARID1A mutations frequently co-occur with mutations in the phosphatidylinositol 3-kinase (PI3K) pathway and are associated with PI3K pathway activation. siRNA knockdown in endometrial cancer cell lines increased AKT phosphorylation supporting ARID1A as a novel regulator of PI3K pathway activity. Our study presents the first unbiased view of somatic coding mutations in endometrial cancer and provides functional evidence for diverse driver genes and mutations in this disease.


Asunto(s)
Neoplasias Endometriales/genética , Exoma , Genes Supresores de Tumor , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Oncogenes , Estudios de Casos y Controles , Transformación Celular Neoplásica/genética , Femenino , Humanos , Mutación , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , ARN Interferente Pequeño , Análisis de Secuencia de ADN , Biología de Sistemas
4.
Cancer Cell ; 9(3): 225-38, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16530706

RESUMEN

S1P has been proposed to contribute to cancer progression by regulating tumor proliferation, invasion, and angiogenesis. We developed a biospecific monoclonal antibody to S1P to investigate its role in tumorigenesis. The anti-S1P mAb substantially reduced tumor progression and in some cases eliminated measurable tumors in murine xenograft and allograft models. Tumor growth inhibition was attributed to antiangiogenic and antitumorigenic effects of the antibody. The anti-S1P mAb blocked EC migration and resulting capillary formation, inhibited blood vessel formation induced by VEGF and bFGF, and arrested tumor-associated angiogenesis. The anti-S1P mAb also neutralized S1P-induced proliferation, release of proangiogenic cytokines, and the ability of S1P to protect tumor cells from apoptosis in several tumor cell lines, validating S1P as a target for therapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Lisofosfolípidos/inmunología , Invasividad Neoplásica/prevención & control , Neoplasias Experimentales/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Esfingosina/análogos & derivados , Animales , Especificidad de Anticuerpos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Ratones , Esfingosina/inmunología
5.
Proc Natl Acad Sci U S A ; 105(13): 5248-53, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18367674

RESUMEN

In primary mammalian cells, expression of oncogenes such as activated Ras induces premature senescence rather than transformation. We show that homozygous deletion of glycogen synthase kinase (GSK) 3beta (GSK3beta-/-) bypasses senescence induced by mutant Ras(V12) allowing primary mouse embryo fibroblasts (MEFs) as well as immortalized MEFs to exhibit a transformed phenotype in vitro and in vivo. Both catalytic activity and Axin-binding of GSK3beta are required to optimally suppress Ras transformation. The expression of Ras(V12) in GSK3beta-/-, but not in GSK3beta+/+ MEFs results in translocation of beta-catenin to the nucleus with concomitant up-regulation of cyclin D1. siRNA-mediated knockdown of beta-catenin decreases both cyclin D1 expression and anchorage-independent growth of transformed cells indicating a causal role for beta-catenin. Thus Ras(V12) and the lack of GSK3beta act in concert to activate the beta-catenin pathway, which may underlie the bypass of senescence and tumorigenic transformation by Ras.


Asunto(s)
Senescencia Celular/fisiología , Eliminación de Gen , Glucógeno Sintasa Quinasa 3/deficiencia , Glucógeno Sintasa Quinasa 3/metabolismo , Homocigoto , Transgenes/genética , Proteínas ras/metabolismo , Transporte Activo de Núcleo Celular , Animales , Catálisis , Células Cultivadas , Ciclina D1/metabolismo , Fibroblastos , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Ratones , Ratones Noqueados , beta Catenina/metabolismo , Proteínas ras/genética
6.
Mol Cancer ; 9: 140, 2010 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-20529378

RESUMEN

BACKGROUND: Although the incidence of melanoma in the U.S. is rising faster than any other cancer, the FDA-approved chemotherapies lack efficacy for advanced disease, which results in poor overall survival. Lysophosphatidic acid (LPA), autotaxin (ATX), the enzyme that produces LPA, and the LPA receptors represent an emerging group of therapeutic targets in cancer, although it is not known which of these is most effective. RESULTS: Herein we demonstrate that thio-ccPA 18:1, a stabilized phosphonothionate analogue of carba cyclic phosphatidic acid, ATX inhibitor and LPA1/3 receptor antagonist, induced a marked reduction in the viability of B16F10 metastatic melanoma cells compared with PBS-treated control by 80-100%. Exogenous LPA 18:1 or D-sn-1-O-oleoyl-2-O-methylglyceryl-3-phosphothioate did not reverse the effect of thio-ccPA 18:1. The reduction in viability mediated by thio-ccPA 18:1 was also observed in A375 and MeWo melanoma cell lines, suggesting that the effects are generalizable. Interestingly, siRNA to LPA3 (siLPA3) but not other LPA receptors recapitulated the effects of thio-ccPA 18:1 on viability, suggesting that inhibition of the LPA3 receptor is an important dualistic function of the compound. In addition, siLPA3 reduced proliferation, plasma membrane integrity and altered morphology of A375 cells. Another experimental compound designed to antagonize the LPA1/3 receptors significantly reduced viability in MeWo cells, which predominantly express the LPA3 receptor. CONCLUSIONS: Thus the ability of thio-ccPA 18:1 to inhibit the LPA3 receptor and ATX are key to its molecular mechanism, particularly in melanoma cells that predominantly express the LPA3 receptor. These observations necessitate further exploration and exploitation of these targets in melanoma.


Asunto(s)
Antineoplásicos/farmacología , Melanoma Experimental/tratamiento farmacológico , Ácidos Fosfatidicos/farmacología , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Expresión Génica , Humanos , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones , Complejos Multienzimáticos/antagonistas & inhibidores , Fosfodiesterasa I/antagonistas & inhibidores , Hidrolasas Diéster Fosfóricas , Pirofosfatasas/antagonistas & inhibidores , ARN Interferente Pequeño , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Clin Invest ; 129(12): 5343-5356, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31682236

RESUMEN

Both miRNAs and A-to-I RNA editing, a widespread nucleotide modification mechanism, have recently emerged as key players in cancer pathophysiology. However, the functional impact of RNA editing of miRNAs in cancer remains largely unexplored. Here, we focused on an ADAR2-catalyzed RNA editing site within the miR-379-5p seed region. This site was under-edited in tumors relative to normal tissues, with a high editing level being correlated with better patient survival times across cancer types. We demonstrated that in contrast to wild-type miRNA, edited miR-379-5p inhibited cell proliferation and promoted apoptosis in diverse tumor contexts in vitro, which was due to the ability of edited but not wild-type miR-379-5p to target CD97. Importantly, through nanoliposomal delivery, edited miR-379-5p mimics significantly inhibited tumor growth and extended survival of mice. Our study indicates a role of RNA editing in diversifying miRNA function during cancer progression and highlights the translational potential of edited miRNAs as a new class of cancer therapeutics.


Asunto(s)
Antígenos CD/fisiología , Apoptosis , MicroARNs/fisiología , Neoplasias/terapia , Edición de ARN , Receptores Acoplados a Proteínas G/fisiología , Animales , Antígenos CD/genética , Línea Celular Tumoral , Proliferación Celular , Femenino , Ratones , Neoplasias/patología , Receptores Acoplados a Proteínas G/genética
8.
Clin Cancer Res ; 13(24): 7421-31, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18094426

RESUMEN

PURPOSE: It is critical to develop methods to quantify the early pharmacodynamic effects of targeted therapeutics in vivo to make drug development more efficient and ensure biologically relevant dosing. Furthermore, an ability to identify patients likely to respond to targeted therapeutics would decrease the size, duration, and cost of clinical trials, resulting in more efficient translation to improved patient outcomes. Recent studies suggest that perifosine inhibits the phosphatidylinositol-3'-kinase (PI3K) pathway by preventing cell membrane recruitment of the AKT pleckstrin homology domain. EXPERIMENTAL DESIGN: A novel functional proteomics technology, reverse phase protein array, was used to establish and quantify pharmacodynamic markers of perifosine efficacy. RESULTS: Perifosine selectively prevents AKT recruitment to the membrane and blocks activation of downstream effectors. Perifosine inhibited breast, ovarian, and prostate cancer models. Growth inhibition was associated with apoptosis. Activation of AKT as a consequence of genomic aberrations predicted perifosine efficacy. In cell lines and xenografts, there was a highly statistically significant correlation between the degree of antitumor efficacy of different perifosine doses and quantified down-regulation of phosphorylation of AKT and of its downstream targets, particularly S6. CONCLUSIONS: Because of a strong correlation between proportional modulation of PI3K pathway biomarkers and quantified perifosine efficacy, it is likely that early measurement of such pharmacodynamic biomarkers with reverse phase protein array will optimize selection of responding patients and guide perifosine dosing. Furthermore, PI3K pathway activation status may allow baseline selection of patients most likely to respond to perifosine alone or in combination with other therapies.


Asunto(s)
Antineoplásicos/farmacocinética , Neoplasias/tratamiento farmacológico , Fosforilcolina/análogos & derivados , Análisis por Matrices de Proteínas/métodos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Immunoblotting , Ratones , Ratones Desnudos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosforilación , Fosforilcolina/farmacocinética , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Res ; 66(5): 2740-8, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16510595

RESUMEN

Growth-regulated oncogene alpha (GROalpha), a member of the chemokine superfamily, is commonly expressed in transformed cells and contributes to angiogenesis and tumorigenesis. Here, we report that increased GROalpha levels are detected in the plasma and ascites of ovarian cancer patients. Ovarian cancer cell lines in culture express and secrete GROalpha. However, when they are starved in serum-free medium, ovarian cancer cells ceased producing GROalpha, suggesting that GROalpha is not constitutively expressed but rather is produced in response to exogenous growth factors in ovarian cancer cells. The prototype peptide growth factors present in serum such as platelet-derived growth factor, insulin-like growth factor I, and insulin do not stimulate GROalpha production by ovarian cancer cells. In contrast, lysophosphatidic acid (LPA), a glycerol backbone phospholipid mediator present in serum and ascites of ovarian cancer patients, is a potent inducer of GROalpha expression in ovarian cancer cell lines. Treatment of ovarian cancer cells with LPA leads to transcriptional activation of the GROalpha gene promoter and robust accumulation of GROalpha protein in culture supernatants. The action of LPA on GROalpha expression is mediated by LPA receptors, particularly the LPA(2) receptor in that ectopic expression of these receptors restores the LPA-dependent GROalpha production in nonresponsive cells. Down-regulation of LPA(2) expression by small interfering RNA (siRNA) in ovarian cancer cells desensitizes GROalpha production in response to LPA. The effect of serum on GROalpha production is also significantly decreased by siRNA inhibition of LPA(2) expression. These studies identify LPA as a primary regulator of GROalpha expression in ovarian cancer.


Asunto(s)
Quimiocinas CXC/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Lisofosfolípidos/farmacología , Neoplasias Ováricas/metabolismo , Ascitis/metabolismo , Línea Celular Tumoral , Quimiocina CXCL1 , Quimiocinas CXC/sangre , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Regulación hacia Abajo , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/sangre , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Isoxazoles/farmacología , Neoplasias Ováricas/sangre , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Toxina del Pertussis/farmacología , Propionatos/farmacología , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Receptores del Ácido Lisofosfatídico/metabolismo , Activación Transcripcional/efectos de los fármacos
10.
Cancer Cell ; 33(5): 817-828.e7, 2018 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-29706454

RESUMEN

Adenosine (A) to inosine (I) RNA editing introduces many nucleotide changes in cancer transcriptomes. However, due to the complexity of post-transcriptional regulation, the contribution of RNA editing to proteomic diversity in human cancers remains unclear. Here, we performed an integrated analysis of TCGA genomic data and CPTAC proteomic data. Despite limited site diversity, we demonstrate that A-to-I RNA editing contributes to proteomic diversity in breast cancer through changes in amino acid sequences. We validate the presence of editing events at both RNA and protein levels. The edited COPA protein increases proliferation, migration, and invasion of cancer cells in vitro. Our study suggests an important contribution of A-to-I RNA editing to protein diversity in cancer and highlights its translational potential.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proteómica/métodos , Edición de ARN , Adenosina/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Bases de Datos Genéticas , Regulación Neoplásica de la Expresión Génica , Humanos , Inosina/genética , Análisis de Secuencia de ARN , Espectrometría de Masas en Tándem
11.
Cancer Cell ; 33(3): 450-462.e10, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29533785

RESUMEN

The functional impact of the vast majority of cancer somatic mutations remains unknown, representing a critical knowledge gap for implementing precision oncology. Here, we report the development of a moderate-throughput functional genomic platform consisting of efficient mutant generation, sensitive viability assays using two growth factor-dependent cell models, and functional proteomic profiling of signaling effects for select aberrations. We apply the platform to annotate >1,000 genomic aberrations, including gene amplifications, point mutations, indels, and gene fusions, potentially doubling the number of driver mutations characterized in clinically actionable genes. Further, the platform is sufficiently sensitive to identify weak drivers. Our data are accessible through a user-friendly, public data portal. Our study will facilitate biomarker discovery, prediction algorithm improvement, and drug development.


Asunto(s)
Biomarcadores de Tumor/genética , Mutación/genética , Neoplasias/diagnóstico , Neoplasias/genética , Algoritmos , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Medicina de Precisión , Proteómica
12.
Mol Cell Biol ; 22(7): 2099-110, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11884598

RESUMEN

Lysophosphatidic acid (LPA) is a natural phospholipid with multiple biological functions. We show here that LPA induces phosphorylation and inactivation of glycogen synthase kinase 3 (GSK-3), a multifunctional serine/threonine kinase. The effect of LPA can be reconstituted by expression of Edg-4 or Edg-7 in cells lacking LPA responses. Compared to insulin, LPA stimulates only modest phosphatidylinositol 3-kinase (PI3K)-dependent activation of protein kinase B (PKB/Akt) that does not correlate with the magnitude of GSK-3 phosphorylation induced by LPA. PI3K inhibitors block insulin- but not LPA-induced GSK-3 phosphorylation. In contrast, the effect of LPA, but not that of insulin or platelet-derived growth factor (PDGF), is sensitive to protein kinase C (PKC) inhibitors. Downregulation of endogenous PKC activity selectively reduces LPA-mediated GSK-3 phosphorylation. Furthermore, several PKC isotypes phosphorylate GSK-3 in vitro and in vivo. To confirm a specific role for PKC in regulation of GSK-3, we further studied signaling properties of PDGF receptor beta subunit (PDGFRbeta) in HEK293 cells lacking endogenous PDGF receptors. In clones expressing a PDGFRbeta mutant wherein the residues that couple to PI3K and other signaling functions are mutated with the link to phospholipase Cgamma (PLCgamma) left intact, PDGF is fully capable of stimulating GSK-3 phosphorylation. The process is sensitive to PKC inhibitors in contrast to the response through the wild-type PDGFRbeta. Therefore, growth factors, such as PDGF, which control GSK-3 mainly through the PI3K-PKB/Akt module, possess the ability to regulate GSK-3 through an alternative, redundant PLCgamma-PKC pathway. LPA and potentially other natural ligands primarily utilize a PKC-dependent pathway to modulate GSK-3.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Lisofosfolípidos/farmacología , Proteína Quinasa C/metabolismo , Proteínas Serina-Treonina Quinasas , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Western Blotting , Línea Celular , Glucógeno Sintasa Quinasa 3 , Glucógeno Sintasa Quinasas , Humanos , Isoenzimas/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfolipasa C gamma , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Células Tumorales Cultivadas , Fosfolipasas de Tipo C/metabolismo
13.
Sci Rep ; 6: 27391, 2016 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-27272709

RESUMEN

Mutation of PPP2R1A has been observed at high frequency in endometrial serous carcinomas but at low frequency in ovarian clear cell carcinoma. However, the biological role of mutation of PPP2R1A in ovarian and endometrial cancer progression remains unclear. In this study, we found that PPP2R1A expression is elevated in high-grade primary tumor patients with papillary serous tumors of the ovary. To determine whether increased levels or mutation of PPP2R1A might contribute to cancer progression, the effects of overexpression or mutation of PPP2R1A on cell proliferation, migration, and PP2A phosphatase activity were investigated using ovarian and endometrial cancer cell lines. Among the mutations, PPP2R1A-W257G enhanced cell migration in vitro through activating SRC-JNK-c-Jun pathway. Overexpression of wild type (WT) PPP2R1A increased its binding ability with B56 regulatory subunits, whereas PPP2R1A-mutations lost the ability to bind to most B56 subunits except B56δ. Total PP2A activity and PPP2R1A-associated PP2Ac activity were significantly increased in cells overexpressing PPP2R1A-WT. In addition, overexpression of PPP2R1A-WT increased cell proliferation in vitro and tumor growth in vivo.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Mutación , Metástasis de la Neoplasia/genética , Proteína Fosfatasa 2/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Proliferación Celular , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Femenino , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Fosforilación
14.
Cell Rep ; 15(7): 1493-1504, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27160903

RESUMEN

3q26.2 amplification in high-grade serous ovarian cancer leads to increased expression of mature microRNA miR551b-3p, which is associated with poor clinical outcome. Importantly, miR551b-3p contributes to resistance to apoptosis and increased survival and proliferation of cancer cells in vitro and in vivo. miR551b-3p upregulates STAT3 protein levels, and STAT3 is required for the effects of miR551b-3p on cell proliferation. Rather than decreasing levels of target mRNA as expected, we demonstrate that miR551b-3p binds a complementary sequence on the STAT3 promoter, recruiting RNA polymerase II and the TWIST1 transcription factor to activate STAT3 transcription, and thus directly upregulates STAT3 expression. Furthermore, anti-miR551b reduced STAT3 expression in ovarian cancer cells in vitro and in vivo and reduced ovarian cancer growth in vivo. Together, our data demonstrate a role for miR551b-3p in transcriptional activation. Thus, miR551b-3p represents a promising candidate biomarker and therapeutic target in ovarian cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Factor de Transcripción STAT3/genética , Regulación hacia Arriba/genética , Animales , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Proliferación Celular/genética , Autorrenovación de las Células , Supervivencia Celular/genética , Regulación hacia Abajo/genética , Femenino , Amplificación de Genes , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Ratones Desnudos , Metástasis de la Neoplasia , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/metabolismo , Factor de Transcripción STAT3/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Transcripción Genética , Resultado del Tratamiento , Carga Tumoral/genética , Proteína 1 Relacionada con Twist/metabolismo
15.
Biochim Biophys Acta ; 1582(1-3): 257-64, 2002 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-12069836

RESUMEN

Lysophosphatidic acid (LPA) is a naturally occurring phospholipid that exhibits pleiotrophic biological activities, ranging from rapid morphological changes to long-term cellular effects such as induction of gene expression and stimulation of cell proliferation and survival on a wide spectrum of cell types. LPA binds and activates distinct members of the Edg/LP subfamily of G protein-coupled receptors that link to multiple G proteins including Gi, Gq and G12/13 to elicit cellular responses. LPA plays a critical role as a general growth, survival and pro-angiogenic factor, in the regulation of physiological and pathophysiological processes in vivo and in vitro. Our previous work indicates that abnormalities in LPA metabolism and function in ovarian cancer patients may contribute to the initiation and progression of the disease. Thus, LPA could be a potential target for cancer therapy. This review summarizes evidence that implicates LPA in the pathophysiology of human ovarian cancer and likely other types of human malignancies.


Asunto(s)
Lisofosfolípidos/fisiología , Neoplasias Ováricas/fisiopatología , Receptores Acoplados a Proteínas G , Femenino , Humanos , Receptores de Superficie Celular/fisiología , Receptores del Ácido Lisofosfatídico , Valores de Referencia
16.
J Med Chem ; 48(9): 3319-27, 2005 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-15857137

RESUMEN

Lysophosphatidic acid (LPA, 1- or 2-acyl-sn-glycerol 3-phosphate) displays an intriguing cell biology that is mediated via interactions with seven-transmembrane G-protein-coupled receptors (GPCRs) and the nuclear hormone receptor PPARgamma. To identify receptor-selective LPA analogues, we describe a series of fluorinated LPA analogues in which either the sn-1 or sn-2 hydroxyl group was replaced by a fluoro or fluoromethyl substituent. We also describe stabilized phosphonate analogues in which the bridging oxygen of the monophosphate was replaced by an alpha-monofluoromethylene (-CHF-) or alpha-difluoromethylene (-CF(2)-) moiety. The sn-2- and sn-1-fluoro-LPA analogues were unable to undergo acyl migration, effectively "freezing" them in the sn-1-O-acyl or sn-2-O-acyl forms, respectively. We first tested these LPA analogues on insect Sf9 cells induced to express human LPA(1), LPA(2), and LPA(3) receptors. While none of the analogues were found to be more potent than 1-oleoyl-LPA at LPA(1) and LPA(2), several LPA analogues were potent LPA(3)-selective agonists. In contrast, 1-oleoyl-LPA had similar activity at all three receptors. The alpha-fluoromethylene phosphonate analogue 15 activated calcium release in LPA(3)-transfected insect Sf9 cells at a concentration 100-fold lower than that of 1-oleoyl-LPA. This activation was enantioselective, with the (2S)-enantiomer showing 1000-fold more activity than the (2R)-enantiomer. Similar results were found for calcium release in HT-29 and OVCAR8 cells. Analogue 15 was also more effective than 1-oleoyl-LPA in activating MAPK and AKT in cells expressing high levels of LPA(3). The alpha-fluoromethylene phosphonate moiety greatly increased the half-life of 15 in cell culture. Thus, alpha-fluoromethylene LPA analogues are unique new phosphatase-resistant ligands that provide enantiospecific and receptor-specific biological readouts.


Asunto(s)
Flúor , Lisofosfolípidos/síntesis química , Receptores del Ácido Lisofosfatídico/agonistas , Animales , Calcio/metabolismo , Línea Celular , Línea Celular Tumoral , Quimiotaxis/efectos de los fármacos , Humanos , Insectos , Ligandos , Lisofosfolípidos/química , Lisofosfolípidos/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Organofosfonatos/química , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Receptores del Ácido Lisofosfatídico/genética , Estereoisomerismo , Relación Estructura-Actividad
17.
Cancer Cell ; 28(4): 515-528, 2015 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-26439496

RESUMEN

Adenosine-to-inosine (A-to-I) RNA editing is a widespread post-transcriptional mechanism, but its genomic landscape and clinical relevance in cancer have not been investigated systematically. We characterized the global A-to-I RNA editing profiles of 6,236 patient samples of 17 cancer types from The Cancer Genome Atlas and revealed a striking diversity of altered RNA-editing patterns in tumors relative to normal tissues. We identified an appreciable number of clinically relevant editing events, many of which are in noncoding regions. We experimentally demonstrated the effects of several cross-tumor nonsynonymous RNA editing events on cell viability and provide the evidence that RNA editing could selectively affect drug sensitivity. These results highlight RNA editing as an exciting theme for investigating cancer mechanisms, biomarkers, and treatments.


Asunto(s)
Adenosina/metabolismo , Inosina/metabolismo , Neoplasias/genética , Edición de ARN , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular , Genoma Humano , Humanos , Neoplasias/patología
18.
Semin Oncol ; 31(1 Suppl 3): 39-53, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15052542

RESUMEN

The ability to analyze the genetic and epigenetic aberrations present in a particular patient's tumor on a global basis is rapidly maturing. The emerging fields of functional genomics and functional proteomics offer the opportunity to translate these advances into a full comprehension of the pathophysiology of cancer. Linking these approaches to chemical genomics and molecular therapeutics should provide an expanding repertoire of targeted therapeutics for clinical evaluation. Novel clinical trial designs that can determine the efficacy of targeted therapeutics in patients selected for aberrations in the target are needed to evaluate the wealth of new drugs becoming available. The promise of these technologies and advances in our understanding of cancer is immense, making it our responsibility to see them to fruition. These technologies should lead to a new era of individualized molecular medicine, wherein we will treat each patient with a "prescription" based on the genetic changes in each patient's tumor and their own genetic make-up, resulting in more effective and less toxic therapy.


Asunto(s)
Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Farmacogenética , Transducción de Señal/efectos de los fármacos , Animales , Perfilación de la Expresión Génica , Inestabilidad Genómica , Humanos , Proteínas de Neoplasias , Neoplasias/metabolismo
19.
Cancer Cell ; 26(4): 479-94, 2014 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-25284480

RESUMEN

PIK3R1 (p85α regulatory subunit of PI3K) is frequently mutated across cancer lineages. Herein, we demonstrate that the most common recurrent PIK3R1 mutation PIK3R1(R348∗) and a nearby mutation PIK3R1(L370fs), in contrast to wild-type and mutations in other regions of PIK3R1, confers an unexpected sensitivity to MEK and JNK inhibitors in vitro and in vivo. Consistent with the response to inhibitors, PIK3R1(R348∗) and PIK3R1(L370fs) unexpectedly increase JNK and ERK phosphorylation. Surprisingly, p85α R348(∗) and L370fs localize to the nucleus where the mutants provide a scaffold for multiple JNK pathway components facilitating nuclear JNK pathway activation. Our findings uncover an unexpected neomorphic role for PIK3R1(R348∗) and neighboring truncation mutations in cellular signaling, providing a rationale for therapeutic targeting of these mutant tumors.


Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mutación , Fosfatidilinositol 3-Quinasas/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Núcleo Celular/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ia , Activación Enzimática , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Transporte de Proteínas
20.
Cancer Cell ; 26(6): 863-879, 2014 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-25490449

RESUMEN

Small noncoding miRNAs represent underexplored targets of genomic aberrations and emerging therapeutic targets. The 3q26.2 amplicon is among the most frequent genomic aberrations in multiple cancer lineages including ovarian and breast cancers. We demonstrate that hsa-miR-569 (hereafter designated as miR569), which is overexpressed in a subset of ovarian and breast cancers, at least in part due to the 3q26.2 amplicon, alters cell survival and proliferation. Downregulation of TP53INP1 expression by miR569 is required for the effects of miR569 on survival and proliferation. Targeting miR569 sensitizes ovarian and breast cancer cells overexpressing miR569 to cisplatin by increasing cell death both in vitro and in vivo. Thus targeting miR569 could potentially benefit patients with the 3q26.2 amplicon and subsequent miR569 elevation.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Portadoras/metabolismo , Proteínas de Choque Térmico/metabolismo , MicroARNs/metabolismo , Neoplasias Glandulares y Epiteliales/genética , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/genética , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cromosomas Humanos Par 3 , Cisplatino/farmacología , Femenino , Amplificación de Genes , Duplicación de Gen , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , MicroARNs/genética , Neoplasias Experimentales , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA