Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Proc Natl Acad Sci U S A ; 115(42): 10666-10671, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30266789

RESUMEN

Scientific progress depends on formulating testable hypotheses informed by the literature. In many domains, however, this model is strained because the number of research papers exceeds human readability. Here, we developed computational assistance to analyze the biomedical literature by reading PubMed abstracts to suggest new hypotheses. The approach was tested experimentally on the tumor suppressor p53 by ranking its most likely kinases, based on all available abstracts. Many of the best-ranked kinases were found to bind and phosphorylate p53 (P value = 0.005), suggesting six likely p53 kinases so far. One of these, NEK2, was studied in detail. A known mitosis promoter, NEK2 was shown to phosphorylate p53 at Ser315 in vitro and in vivo and to functionally inhibit p53. These bona fide validations of text-based predictions of p53 phosphorylation, and the discovery of an inhibitory p53 kinase of pharmaceutical interest, suggest that automated reasoning using a large body of literature can generate valuable molecular hypotheses and has the potential to accelerate scientific discovery.


Asunto(s)
Indización y Redacción de Resúmenes , Quinasas Relacionadas con NIMA/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Células HCT116 , Células HEK293 , Humanos , Quinasas Relacionadas con NIMA/genética , Procesamiento de Lenguaje Natural , Fosforilación , PubMed , Proteína p53 Supresora de Tumor/genética
2.
J Pathol ; 232(5): 522-33, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24374933

RESUMEN

Mutations in the TP53 tumour suppressor gene occur in half of all human cancers, indicating its critical importance in inhibiting cancer development. Despite extensive studies, the mechanisms by which mutant p53 enhances tumour progression remain only partially understood. Here, using data from the Cancer Genome Atlas (TCGA), genomic and transcriptomic analyses were performed on 2256 tumours from 10 human cancer types. We show that tumours with TP53 mutations have altered gene expression profiles compared to tumours retaining two wild-type TP53 alleles. Among 113 known p53-up-regulated target genes identified from cell culture assays, 10 were consistently up-regulated in at least eight of 10 cancer types that retain both copies of wild-type TP53. RPS27L, CDKN1A (p21(CIP1)) and ZMAT3 were significantly up-regulated in all 10 cancer types retaining wild-type TP53. Using this p53-based expression analysis as a discovery tool, we used cell-based assays to identify five novel p53 target genes from genes consistently up-regulated in wild-type p53 cancers. Global gene expression analyses revealed that cell cycle regulatory genes and transcription factors E2F1, MYBL2 and FOXM1 were disproportionately up-regulated in many TP53 mutant cancer types. Finally, > 93% of tumours with a TP53 mutation exhibited greatly reduced wild-type p53 messenger expression, due to loss of heterozygosity or copy neutral loss of heterozygosity, supporting the concept of p53 as a recessive tumour suppressor. The data indicate that tumours with wild-type TP53 retain some aspects of p53-mediated growth inhibitory signalling through activation of p53 target genes and suppression of cell cycle regulatory genes.


Asunto(s)
Biomarcadores de Tumor/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Mutación , Neoplasias/genética , Proteína p53 Supresora de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Pérdida de Heterocigocidad , Neoplasias/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
3.
J Exp Med ; 203(2): 371-81, 2006 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-16446383

RESUMEN

Mutations constitutively activating FLT3 kinase are detected in approximately 30% of acute myelogenous leukemia (AML) patients and affect downstream pathways such as extracellular signal-regulated kinase (ERK)1/2. We found that activation of FLT3 in human AML inhibits CCAAT/enhancer binding protein alpha (C/EBPalpha) function by ERK1/2-mediated phosphorylation, which may explain the differentiation block of leukemic blasts. In MV4;11 cells, pharmacological inhibition of either FLT3 or MEK1 leads to granulocytic differentiation. Differentiation of MV4;11 cells was also observed when C/EBPalpha mutated at serine 21 to alanine (S21A) was stably expressed. In contrast, there was no effect when serine 21 was mutated to aspartate (S21D), which mimics phosphorylation of C/EBPalpha. Thus, our results suggest that therapies targeting the MEK/ERK cascade or development of protein therapies based on transduction of constitutively active C/EBPalpha may prove effective in treatment of FLT3 mutant leukemias resistant to the FLT3 inhibitor therapies.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mutación Puntual , Tirosina Quinasa 3 Similar a fms/genética , Anciano , Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Activación Enzimática/genética , Femenino , Granulocitos/patología , Humanos , Células K562 , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/metabolismo , Masculino , Células Mieloides/patología , Fosforilación , Piperazinas/farmacología , Quinazolinas/farmacología , Serina/metabolismo , Células Tumorales Cultivadas , Células U937 , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/metabolismo
4.
Cell Cycle ; 19(4): 479-491, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31959038

RESUMEN

The phosphoinositide-3-kinase like kinases (PIKK) such as ATM and ATR play a key role in initiating the cellular DNA damage response (DDR). One key ATM target is the cyclin-dependent kinase inhibitor p27Kip1 that promotes G1 arrest. ATM activates p27Kip1-induced arrest in part through phosphorylation of p27Kip1 at Serine 140. Here we show that this site is dephosphorylated by the type 2C serine/threonine phosphatase, WIP1 (Wildtype p53-Induced Phosphatase-1), encoded by the PPM1D gene. WIP1 has been shown to dephosphorylate numerous ATM target sites in DDR proteins, and its overexpression and/or mutation has often been associated with oncogenesis. We demonstrate that wildtype, but not phosphatase-dead WIP1, efficiently dephosphorylates p27Kip1 Ser140 both in vitro and in cells and that this dephosphorylation is sensitive to the WIP1-specific inhibitor GSK 2830371. Increased expression of wildtype WIP1 reduces stability of p27Kip1 while increased expression of similar amounts of phosphatase-dead WIP1 has no effect on p27Kip1 protein stability. Overexpression of wildtype p27Kip1 reduces cell proliferation and colony forming capability relative to the S140A (constitutively non-phosphorylated) form of p27. Thus, WIP1 plays a significant role in homeostatic modulation of p27Kip1 activity following activation by ATM.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteína Fosfatasa 2C/metabolismo , Serina/metabolismo , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Células HEK293 , Humanos , Células MCF-7 , Mutación/genética , Fosfopéptidos/metabolismo , Fosforilación , Estabilidad Proteica , Reproducibilidad de los Resultados , Ensayo de Tumor de Célula Madre
5.
Mol Cell Biol ; 26(3): 1109-23, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16428462

RESUMEN

The leucine zipper family transcription factor CCAAT enhancer binding protein alpha (C/EBPalpha) inhibits proliferation and promotes differentiation in various cell types. In this study, we show, using a lung-specific conditional mouse model of C/EBPalpha deletion, that loss of C/EBPalpha in the respiratory epithelium leads to respiratory failure at birth due to an arrest in the type II alveolar cell differentiation program. This differentiation arrest results in the lack of type I alveolar cells and differentiated surfactant-secreting type II alveolar cells. In addition to showing a block in type II cell differentiation, the neonatal lungs display increased numbers of proliferating cells and decreased numbers of apoptotic cells, leading to epithelial expansion and loss of airspace. Consistent with the phenotype observed, genes associated with alveolar maturation, survival, and proliferation were differentially expressed. Taken together, these results identify C/EBPalpha as a master regulator of airway epithelial maturation and suggest that the loss of C/EBPalpha could also be an important event in the multistep process of lung tumorigenesis. Furthermore, this study indicates that exploring the C/EBPalpha pathway might have therapeutic benefits for patients with respiratory distress syndromes.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/deficiencia , Pulmón/crecimiento & desarrollo , Pulmón/patología , Alveolos Pulmonares/patología , Insuficiencia Respiratoria/genética , Insuficiencia Respiratoria/patología , Animales , Apoptosis , Proteína alfa Potenciadora de Unión a CCAAT/genética , Diferenciación Celular , Proliferación Celular , Epitelio/metabolismo , Epitelio/patología , Eliminación de Gen , Perfilación de la Expresión Génica , Pulmón/metabolismo , Masculino , Ratones , Ratones Mutantes , Alveolos Pulmonares/citología , Alveolos Pulmonares/crecimiento & desarrollo , Eliminación de Secuencia
6.
J Cell Physiol ; 216(2): 309-14, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18366075

RESUMEN

Viruses have been linked to approximately 20% of all human tumors worldwide. These transforming viruses encode viral oncoproteins that interact with cellular proteins to enhance viral replication. The transcriptional and post-transcriptional effects of these viral oncoproteins ultimately result in cellular transformation. Historically, viral research has been vital to the discovery of oncogenes and tumor suppressors with more current research aiding in unraveling some mechanisms of carcinogenesis. Interestingly, since transforming viruses affect some of the same pathways that are dysregulated in human cancers, their study enhances our understanding of the multistep process of tumorigenesis. This review will examine the cellular mechanisms targeted by oncogenic human viruses and the processes by which these effects contribute to transformation. In particular, we will focus on three transforming viruses, human T-cell leukemia virus type-I, hepatitis B virus and human papillomavirus. These viruses all encode specific oncogenes that promote cell cycle progression, inhibit DNA damage checkpoint responses and prevent programmed cell death in an effort to promote viral propagation. While the transforming properties of these viruses are probably unintended consequences of replication strategies, they provide excellent systems in which to study cancer development.


Asunto(s)
Transformación Celular Viral , Neoplasias , Virus Oncogénicos , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Supervivencia Celular , Reparación del ADN , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/virología , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Oncogenes , Virus Oncogénicos/genética , Virus Oncogénicos/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral , Receptor fas/metabolismo
7.
N Engl J Med ; 352(8): 786-92, 2005 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-15728811

RESUMEN

Mutations of the epidermal growth factor receptor (EGFR) gene have been identified in specimens from patients with non-small-cell lung cancer who have a response to anilinoquinazoline EGFR inhibitors. Despite the dramatic responses to such inhibitors, most patients ultimately have a relapse. The mechanism of the drug resistance is unknown. Here we report the case of a patient with EGFR-mutant, gefitinib-responsive, advanced non-small-cell lung cancer who had a relapse after two years of complete remission during treatment with gefitinib. The DNA sequence of the EGFR gene in his tumor biopsy specimen at relapse revealed the presence of a second point mutation, resulting in threonine-to-methionine amino acid change at position 790 of EGFR. Structural modeling and biochemical studies showed that this second mutation led to gefitinib resistance.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Quinazolinas/uso terapéutico , Anciano , Antineoplásicos/efectos adversos , Biopsia , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , ADN de Neoplasias , Receptores ErbB/química , Clorhidrato de Erlotinib , Gefitinib , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Modelos Estructurales , Estructura Molecular , Recurrencia Local de Neoplasia/patología , Mutación Puntual , Quinazolinas/efectos adversos , Quinazolinas/química , Quinazolinas/metabolismo , ARN Neoplásico , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN
8.
J Virol ; 81(23): 13075-81, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17898074

RESUMEN

Serum response factor (SRF) was recently shown to bind and activate the human T-cell leukemia virus type 1 (HTLV-1) promoter at bases -116 to -125 relative to the transcription start site. In addition to the SRF binding site (CArG box), serum response elements (SRE) also typically contain a binding site for a member of the ternary complex factor (TCF) family. Here we demonstrate the presence of two TCF binding sites upstream of the viral CArG box. Binding of the TCF family member Elk-1 to these sites was shown to activate transcription of the promoter. Based on these results, the position of the previously described viral SRE (vSRE) within the HTLV-1 promoter can be extended from -116 to -157 to include the two newly identified TCF sites. Purified Elk-1 bound to a probe containing the vSRE, and this complex formed a ternary complex with SRF. In addition, the complex formed by nuclear extract on this probe contained Elk-1, as shown by electrophoretic mobility shift assay supershift. Both of the predicted TCF sites independently bound Elk-1. Elk-1 activated transcription of the HTLV-1 long terminal repeat (LTR), and mutations within either of the TCF sites or the CArG box reduced responsiveness of the LTR to Elk-1. Chromatin immunoprecipitation demonstrated that Elk-1 associates with the HTLV-1 LTR in vivo. These results identify a functional SRE within the HTLV-1 LTR and suggest that both Elk-1 and SRF play important roles in regulating basal HTLV-1 gene expression.


Asunto(s)
Regulación Viral de la Expresión Génica , Virus Linfotrópico T Tipo 1 Humano/fisiología , ARN Viral/biosíntesis , Secuencias Repetidas Terminales , Transcripción Genética/fisiología , Proteína Elk-1 con Dominio ets/metabolismo , Sitios de Unión/genética , Línea Celular , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Humanos , Mutación , Regiones Promotoras Genéticas , Unión Proteica , ARN Mensajero/biosíntesis
9.
Cell Stem Cell ; 23(5): 700-713.e6, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388424

RESUMEN

Clonal hematopoiesis (CH), in which stem cell clones dominate blood production, becomes increasingly common with age and can presage malignancy development. The conditions that promote ascendancy of particular clones are unclear. We found that mutations in PPM1D (protein phosphatase Mn2+/Mg2+-dependent 1D), a DNA damage response regulator that is frequently mutated in CH, were present in one-fifth of patients with therapy-related acute myeloid leukemia or myelodysplastic syndrome and strongly correlated with cisplatin exposure. Cell lines with hyperactive PPM1D mutations expand to outcompete normal cells after exposure to cytotoxic DNA damaging agents including cisplatin, and this effect was predominantly mediated by increased resistance to apoptosis. Moreover, heterozygous mutant Ppm1d hematopoietic cells outcompeted their wild-type counterparts in vivo after exposure to cisplatin and doxorubicin, but not during recovery from bone marrow transplantation. These findings establish the clinical relevance of PPM1D mutations in CH and the importance of studying mutation-treatment interactions. VIDEO ABSTRACT.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Células Clonales/efectos de los fármacos , Doxorrubicina/farmacología , Hematopoyesis/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Mutación , Proteína Fosfatasa 2C/genética , Anciano , Animales , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Cisplatino/química , Doxorrubicina/química , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Células HEK293 , Hematopoyesis/genética , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Persona de Mediana Edad , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Proteína Fosfatasa 2C/metabolismo
10.
Retrovirology ; 4: 95, 2007 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-18081936

RESUMEN

BACKGROUND: The HTLV-I oncoprotein, Tax, is a pleiotropic protein whose activity is partially regulated by its ability to interact with, and perturb the functions of, numerous cellular proteins. Tax is predominantly a nuclear protein that localizes to nuclear foci known as Tax Speckled Structures (TSS). We recently reported that the localization of Tax and its interactions with cellular proteins are altered in response to various forms of genotoxic and cellular stress. The level of cytoplasmic Tax increases in response to stress and this relocalization depends upon the interaction of Tax with CRM1. Cellular pathways and signals that regulate the subcellular localization of Tax remain to be determined. However, post-translational modifications including sumoylation and ubiquitination are known to influence the subcellular localization of Tax and its interactions with cellular proteins. The sumoylated form of Tax exists predominantly in the nucleus while ubiquitinated Tax exists predominantly in the cytoplasm. Therefore, we hypothesized that post-translational modifications of Tax that occur in response to DNA damage regulate the localization of Tax and its interactions with cellular proteins. RESULTS: We found a significant increase in mono-ubiquitination of Tax in response to UV irradiation. Mutation of specific lysine residues (K280 and K284) within Tax inhibited DNA damage-induced ubiquitination. In contrast to wild-type Tax, which undergoes transient nucleocytoplasmic shuttling in response to DNA damage, the K280 and K284 mutants were retained in nuclear foci following UV irradiation and remained co-localized with the cellular TSS protein, sc35. CONCLUSION: This study demonstrates that the localization of Tax, and its interactions with cellular proteins, are dynamic following DNA damage and depend on the post-translational modification status of Tax. Specifically, DNA damage induces the ubiquitination of Tax at K280 and K284. Ubiquitination of these residues facilitates the dissociation of Tax from sc35-containing nuclear foci, and stimulates nuclear export of Tax through the CRM1 pathway.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Núcleo Celular/metabolismo , Daño del ADN/fisiología , Productos del Gen tax/fisiología , Infecciones por HTLV-I/fisiopatología , Línea Celular Transformada , Productos del Gen tax/metabolismo , Virus Linfotrópico T Tipo 1 Humano/química , Humanos , Procesamiento Proteico-Postraduccional , Ubiquitinación/fisiología
11.
Cancer Res ; 64(12): 4137-47, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15205324

RESUMEN

We showed previously that CCAAT/enhancer binding protein alpha (C/EBP alpha), a tissue-specific transcription factor, is a candidate tumor suppressor in lung cancer. In the present study, we have performed a transcriptional profiling study of C/EBP alpha target genes using an inducible cell line system. This study led to the identification of hepatocyte nuclear factor 3beta (HNF3 beta), a transcription factor known to play a role in airway differentiation, as a downstream target of C/EBP alpha. We found down-regulation of HNF3 beta expression in a large proportion of lung cancer cell lines examined and identified two novel mutants of HNF3 beta, as well as hypermethylation of the HNF3 beta promoter. We also developed a tetracycline-inducible cell line model to study the cellular consequences of HNF3 beta expression. Conditional expression of HNF3 beta led to significant growth reduction, proliferation arrest, apoptosis, and loss of clonogenic ability, suggesting additionally that HNF3 beta is a novel tumor suppressor in lung cancer. This is the first study to show genetic abnormalities of lung-specific differentiation pathways in the development of lung cancer.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas de Unión al ADN/genética , Neoplasias Pulmonares/genética , Proteínas Nucleares/genética , Factores de Transcripción , Apoptosis/genética , División Celular/genética , Metilación de ADN , Proteínas de Unión al ADN/biosíntesis , Regulación hacia Abajo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Factor Nuclear 3-beta del Hepatocito , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Mutación , Proteínas Nucleares/biosíntesis , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Transcripción Genética
12.
PLoS One ; 10(2): e0115635, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25658463

RESUMEN

The wild-type p53-induced phosphatase 1 (WIP1) is a serine/threonine phosphatase that negatively regulates multiple proteins involved in DNA damage response including p53, CHK2, Histone H2AX, and ATM, and it has been shown to be overexpressed or amplified in human cancers including breast and ovarian cancers. We examined WIP1 mRNA levels across multiple tumor types and found the highest levels in breast cancer, leukemia, medulloblastoma and neuroblastoma. Neuroblastoma is an exclusively TP53 wild type tumor at diagnosis and inhibition of p53 is required for tumorigenesis. Neuroblastomas in particular have previously been shown to have 17q amplification, harboring the WIP1 (PPM1D) gene and associated with poor clinical outcome. We therefore sought to determine whether inhibiting WIP1 with a selective antagonist, GSK2830371, can attenuate neuroblastoma cell growth through reactivation of p53 mediated tumor suppression. Neuroblastoma cell lines with wild-type TP53 alleles were highly sensitive to GSK2830371 treatment, while cell lines with mutant TP53 were resistant to GSK2830371. The majority of tested neuroblastoma cell lines with copy number gains of the PPM1D locus were also TP53 wild-type and sensitive to GSK2830371A; in contrast cell lines with no copy gain of PPM1D were mixed in their sensitivity to WIP1 inhibition, with the primary determinant being TP53 mutational status. Since WIP1 is involved in the cellular response to DNA damage and drugs used in neuroblastoma treatment induce apoptosis through DNA damage, we sought to determine whether GSK2830371 could act synergistically with standard of care chemotherapeutics. Treatment of wild-type TP53 neuroblastoma cell lines with both GSK2830371 and either doxorubicin or carboplatin resulted in enhanced cell death, mediated through caspase 3/7 induction, as compared to either agent alone. Our data suggests that WIP1 inhibition represents a novel therapeutic approach to neuroblastoma that could be integrated with current chemotherapeutic approaches.


Asunto(s)
Aminopiridinas/farmacología , Dipéptidos/farmacología , Mutación , Neuroblastoma/tratamiento farmacológico , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Alelos , Línea Celular Tumoral , Femenino , Sitios Genéticos , Humanos , Masculino , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Proteína Fosfatasa 2C , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
13.
PLoS One ; 8(2): e55989, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23405243

RESUMEN

Genomic instability stemming from dysregulation of cell cycle checkpoints and DNA damage response (DDR) is a common feature of many cancers. The cancer adult T cell leukemia (ATL) can occur in individuals infected with human T cell leukemia virus type 1 (HTLV-1), and ATL cells contain extensive chromosomal abnormalities, suggesting that they have defects in the recognition or repair of DNA damage. Since Tax is the transforming protein encoded by HTLV-1, we asked whether Tax can affect cell cycle checkpoints and the DDR. Using a combination of flow cytometry and DNA repair assays we showed that Tax-expressing cells exit G(1) phase and initiate DNA replication prematurely following damage. Reduced phosphorylation of H2AX (γH2AX) and RPA2, phosphoproteins that are essential to properly initiate the DDR, was also observed in Tax-expressing cells. To determine the cause of decreased DDR protein phosphorylation in Tax-expressing cells, we examined the cellular phosphatase, WIP1, which is known to dephosphorylate γH2AX. We found that Tax can interact with Wip1 in vivo and in vitro, and that Tax-expressing cells display elevated levels of Wip1 mRNA. In vitro phosphatase assays showed that Tax can enhance Wip1 activity on a γH2AX peptide target by 2-fold. Thus, loss of γH2AX in vivo could be due, in part, to increased expression and activity of WIP1 in the presence of Tax. siRNA knockdown of WIP1 in Tax-expressing cells rescued γH2AX in response to damage, confirming the role of WIP1 in the DDR. These studies demonstrate that Tax can disengage the G(1)/S checkpoint by enhancing WIP1 activity, resulting in reduced DDR. Premature G(1) exit of Tax-expressing cells in the presence of DNA lesions creates an environment that tolerates incorporation of random mutations into the host genome.


Asunto(s)
Daño del ADN/fisiología , Fase G1/fisiología , Productos del Gen tax/genética , Fragmentos de Péptidos/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Fase S/fisiología , Adulto , Animales , Western Blotting , Células Cultivadas , Daño del ADN/efectos de la radiación , Reparación del ADN/fisiología , Reparación del ADN/efectos de la radiación , Replicación del ADN/efectos de la radiación , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/efectos de la radiación , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Fase G1/efectos de la radiación , Histonas/genética , Histonas/metabolismo , Virus Linfotrópico T Tipo 1 Humano/genética , Humanos , Inmunoprecipitación , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/genética , Fosforilación , Proteína Fosfatasa 2C , Dímeros de Pirimidina , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fase S/efectos de la radiación , Rayos Ultravioleta
14.
Lung Cancer ; 77(1): 31-7, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22341411

RESUMEN

PURPOSE: We sought to determine the mechanisms of downregulation of the airway transcription factor Foxa2 in lung cancer and the expression status of Foxa2 in non-small-cell lung cancer (NSCLC). METHODS: A series of 25 lung cancer cell lines were evaluated for Foxa2 protein expression, FOXA2 mRNA levels, FOXA2 mutations, FOXA2 copy number changes and for evidence of FOXA2 promoter hypermethylation. In addition, 32 NSCLCs were sequenced for FOXA2 mutations and 173 primary NSCLC tumors evaluated for Foxa2 expression using an immunohistochemical assay. RESULTS: Out of the 25 cell lines, 13 (52%) had undetectable FOXA2 mRNA. The expression of FOXA2 mRNA and Foxa2 protein were congruent in 19/22 cells (p = 0.001). FOXA2 mutations were not identified in primary NSCLCs and were infrequent in cell lines. Focal or broad chromosomal deletions involving FOXA2 were not present. The promoter region of FOXA2 had evidence of hypermethylation, with an inverse correlation between FOXA2 mRNA expression and presence of CpG dinucleotide methylation (p < 0.0001). In primary NSCLC tumor specimens, there was a high frequency of either absence (42/173, 24.2%) or no/low expression (96/173, 55.4%) of Foxa2. In 130 patients with stage I NSCLC there was a trend towards decreased survival in tumors with no/low expression of Foxa2 (HR of 1.6, 95%CI 0.9-3.1; p = 0.122). CONCLUSIONS: Loss of expression of Foxa2 is frequent in lung cancer cell lines and NSCLCs. The main mechanism of downregulation of Foxa2 is epigenetic silencing through promoter hypermethylation. Further elucidation of the involvement of Foxa2 and other airway transcription factors in the pathogenesis of lung cancer may identify novel therapeutic targets.


Asunto(s)
Adenocarcinoma/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Regulación hacia Abajo , Epigénesis Genética , Factor Nuclear 3-beta del Hepatocito/genética , Neoplasias Pulmonares/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Metilación de ADN , Análisis Mutacional de ADN , Femenino , Dosificación de Gen , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas
16.
Virology ; 416(1-2): 1-8, 2011 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-21571351

RESUMEN

The HTLV-I oncoprotein Tax interferes with DNA double strand break repair. Since non-homologous end joining (NHEJ) is a major pathway used to repair DNA double strand breaks we examined the effect of Tax on this pathway, with particular interest in the expression and function of Ku80, a critical component of the NHEJ pathway. Tax expression decreased Ku80 mRNA and protein levels, and repressed transcription from the Ku80 promoter. Conversely, Ku80 mRNA increased following siRNA knockdown of Tax in HTLV-I infected cells. Tax expression was associated with an elevated number of micronuclei and nucleoplasmic bridges, hallmarks of improper DNA double strand break repair. Our studies identified Tax as a transcriptional repressor of Ku80 that correlates with decreased DNA repair function. The reduction of Ku80 transcription by Tax may deplete the cell of an essential DNA break binding protein, resulting in reduced repair of DNA double strand breaks and accumulation genomic mutations.


Asunto(s)
Antígenos Nucleares/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Viral de la Expresión Génica/fisiología , Productos del Gen tax/metabolismo , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Antígenos Nucleares/genética , Línea Celular , Roturas del ADN de Doble Cadena , Reparación del ADN , ADN Viral/química , Proteínas de Unión al ADN/genética , Productos del Gen tax/genética , Virus Linfotrópico T Tipo 1 Humano/genética , Humanos , Autoantígeno Ku , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo
17.
Cancer Res ; 69(6): 2559-67, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19276372

RESUMEN

Osteosarcoma is the primary malignant cancer of bone and particularly affects adolescents and young adults, causing debilitation and sometimes death. As a model for human osteosarcoma, we have been studying p53(+/-) mice, which develop osteosarcoma at high frequency. To discover genes that cooperate with p53 deficiency in osteosarcoma formation, we have integrated array comparative genomic hybridization, microarray expression analyses in mouse and human osteosarcomas, and functional assays. In this study, we found seven frequent regions of copy number gain and loss in the mouse p53(+/-) osteosarcomas but have focused on a recurrent amplification event on mouse chromosome 9A1. This amplicon is syntenic with a similar chromosome 11q22 amplicon identified in several human tumor types. Three genes on this amplicon, the matrix metalloproteinase gene MMP13 and the antiapoptotic genes Birc2 (cIAP1) and Birc3 (cIAP2), show elevated expression in mouse and human osteosarcomas. We developed a functional assay using clonal osteosarcoma cell lines transduced with lentiviral short hairpin RNA vectors to show that down-regulation of MMP13, Birc2, or Birc3 resulted in reduced tumor growth when transplanted into immunodeficient recipient mice. These experiments revealed that high MMP13 expression enhances osteosarcoma cell survival and that Birc2 and Birc3 also enhance cell survival but only in osteosarcoma cells with the chromosome 9A1 amplicon. We conclude that the antiapoptotic genes Birc2 and Birc3 are potential oncogenic drivers in the chromosome 9A1 amplicon.


Asunto(s)
Neoplasias Óseas/genética , Proteínas Inhibidoras de la Apoptosis/genética , Metaloproteinasa 13 de la Matriz/genética , Osteosarcoma/genética , Proteína p53 Supresora de Tumor/deficiencia , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteína 3 que Contiene Repeticiones IAP de Baculovirus , Neoplasias Óseas/enzimología , Proteínas de Ciclo Celular , Transformación Celular Neoplásica/genética , Hibridación Genómica Comparativa , Amplificación de Genes , Inestabilidad Genómica , Humanos , Proteínas Inhibidoras de la Apoptosis/deficiencia , Metaloproteinasa 13 de la Matriz/biosíntesis , Ratones , Ratones Endogámicos C57BL , Osteosarcoma/enzimología , Fosfoproteínas/biosíntesis , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas , Proteínas Señalizadoras YAP
18.
Genes Dev ; 22(15): 2085-92, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18676813

RESUMEN

The transcription factor PU.1 is an important regulator of hematopoiesis; precise expression levels are critical for normal hematopoietic development and suppression of leukemia. We show here that noncoding antisense RNAs are important modulators of proper dosages of PU.1. Antisense and sense RNAs are regulated by shared evolutionarily conserved cis-regulatory elements, and we can show that antisense RNAs inhibit PU.1 expression by modulating mRNA translation. We propose that such antisense RNAs will likely be important in the regulation of many genes and may be the reason for the large number of overlapping complementary transcripts with so far unknown function.


Asunto(s)
Expresión Génica , Proteínas Proto-Oncogénicas/genética , ARN sin Sentido/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Transactivadores/genética , Animales , Línea Celular , Electroporación , Granulocitos/citología , Granulocitos/metabolismo , Células HL-60 , Humanos , Separación Inmunomagnética , Células Jurkat , Macrófagos/metabolismo , Ratones , Modelos Genéticos , Biosíntesis de Proteínas , Interferencia de ARN , ARN sin Sentido/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Linfocitos T/metabolismo , Transcripción Genética , Células U937
19.
Nat Genet ; 40(1): 51-60, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17994017

RESUMEN

Both PU.1 (also called SFPI1), an Ets-family transcription factor, and AML1 (also called RUNX1), a DNA-binding subunit of the CBF transcription factor family, are crucial for the generation of all hematopoietic lineages, and both act as tumor suppressors in leukemia. An upstream regulatory element (URE) of PU.1 has both enhancer and repressor activity and tightly regulates PU.1 expression. Here we show that AML1 binds to functionally important sites within the PU.1 upstream regulatory element and regulates PU.1 expression at both embryonic and adult stages of development. Analysis of mice carrying conditional AML1 knockout alleles and knock-in mice carrying mutations in all three AML1 sites of the URE proximal region demonstrated that AML1 regulates PU.1 both positively and negatively in a lineage dependent manner. Dysregulation of PU.1 expression contributed to each of the phenotypes observed in these mice, and restoration of proper PU.1 expression rescued or partially rescued each phenotype. Thus, our data demonstrate that PU.1 is a major downstream target gene of AML1.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Animales , Plaquetas/metabolismo , Ratones , Ratones Noqueados , Elementos Reguladores de la Transcripción , Linfocitos T/metabolismo
20.
Nat Immunol ; 7(7): 732-9, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16751774

RESUMEN

During 'emergency' situations such as infections, host defense requires rapid mobilization of bone marrow granulocyte progenitors. 'Steady-state' granulopoiesis is absolutely dependent on the C/EBPalpha transcription factor, but the transcriptional mechanisms underlying emergency granulopoiesis remain unclear. Here we show that large numbers of granulocytes were generated from C/EBPalpha-deficient progenitors after cytokine stimulation in vivo. Cytokine treatment or fungal infection induced upregulation of C/EBPbeta but not C/EBPalpha or C/EBPepsilon transcripts in granulocyte progenitors, and C/EBPbeta-deficient progenitors showed decreased emergency-induced granulopoiesis in vitro and in vivo. C/EBPbeta inhibited proliferation less severely than did C/EBPalpha. These data suggest a critical function for C/EBPbeta in emergency granulopoiesis, which demands both differentiation and proliferation of granulocyte precursors.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/fisiología , Granulocitos/citología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Infecciones/inmunología , Animales , Animales Congénicos , Proteína alfa Potenciadora de Unión a CCAAT/deficiencia , Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Proteína beta Potenciadora de Unión a CCAAT/deficiencia , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Candidiasis/inmunología , Candidiasis/fisiopatología , Ciclo Celular , Diferenciación Celular , Células Cultivadas/metabolismo , Ensayo de Unidades Formadoras de Colonias , Estradiol/farmacología , Factor Estimulante de Colonias de Granulocitos/genética , Factor Estimulante de Colonias de Granulocitos/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Hematopoyesis/genética , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Interleucina-3/genética , Interleucina-3/fisiología , Células K562/citología , Ratones , Ratones Endogámicos C57BL , Quimera por Radiación , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/genética , Proteínas Recombinantes de Fusión/fisiología , Transducción Genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA