Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Prostate ; 84(1): 87-99, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37812042

RESUMEN

PURPOSE: Despite well-informed work in several malignancies, the phenotypic effects of TP53 mutations in metastatic castration-sensitive prostate cancer (mCSPC) progression and metastasis are not clear. We characterized the structure-function and clinical impact of TP53 mutations in mCSPC. PATIENTS AND METHODS: We performed an international retrospective review of men with mCSPC who underwent next-generation sequencing and were stratified according to TP53 mutational status and metastatic burden. Clinical outcomes included radiographic progression-free survival (rPFS) and overall survival (OS) evaluated with Kaplan-Meier and multivariable Cox regression. We also utilized isogenic cancer cell lines to assess the effect of TP53 mutations and APR-246 treatment on migration, invasion, colony formation in vitro, and tumor growth in vivo. Preclinical experimental observations were compared using t-tests and ANOVA. RESULTS: Dominant-negative (DN) TP53 mutations were enriched in patients with synchronous (vs. metachronous) (20.7% vs. 6.3%, p < 0.01) and polymetastatic (vs. oligometastatic) (14.4% vs. 7.9%, p < 0.01) disease. On multivariable analysis, DN mutations were associated with worse rPFS (hazards ratio [HR] = 1.97, 95% confidence interval [CI]: 1.31-2.98) and overall survival [OS] (HR = 2.05, 95% CI: 1.14-3.68) compared to TP53 wild type (WT). In vitro, 22Rv1 TP53 R175H cells possessed stronger migration, invasion, colony formation ability, and cellular movement pathway enrichment in RNA sequencing analysis compared to 22Rv1 TP53 WT cells. Treatment with APR-246 reversed the effects of TP53 mutations in vitro and inhibited 22Rv1 TP53 R175H tumor growth in vivo in a dosage-dependent manner. CONCLUSIONS: DN TP53 mutations correlated with worse prognosis in prostate cancer patients and higher metastatic potential, which could be counteracted by APR-246 treatment suggesting a potential future therapeutic avenue.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Neoplasias de la Próstata , Masculino , Humanos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Pronóstico , Supervivencia sin Progresión , Mutación , Relación Estructura-Actividad , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Proteína p53 Supresora de Tumor/genética
2.
Int J Cancer ; 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39056587
3.
Int J Hyperthermia ; 36(1): 712-720, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31345068

RESUMEN

Purpose: A proposed mechanism for the enhanced effectiveness of hyperthermia and doxorubicin (Dox) combinations is increased intracellular Dox concentrations resulting from heat-induced cell stress. The purpose of this study was to determine whether specific varied Dox and heat combinations produce measurable effects greater than the additive combination, and whether these effects can be attributed to heat-induced increases in intracellular Dox concentrations. Methods: HCT116, HT29 and CT26 cells were exposed to Dox and water bath heating independently. A clonogenic survival assay was used to determine cell killing and intracellular Dox concentrations were measured in HCT116 cells with mass spectrometry. Cells were exposed to heating at 42 °C (60 min) and 0.5 µg/ml of Dox at varying intervals. Synergy was determined by curve-fitting and isobologram analysis. Results: All cell lines displayed synergistic effects of combined heating and Dox. A maximum synergistic effect was achieved with simultaneous cell exposure to Dox and heat. For exposures at 42 °C, the synergistic effect was most pronounced at Dox concentrations <0.5 µg/ml. Increased intracellular concentrations of Dox in HCT116 cells caused by heat-stress did not generate a concomitant thermal enhancement. Conclusions: Simultaneous exposure of HCT116 cells to heating and Dox is more effective than sequential exposure. Heat-induced cell responses are accompanied by increased intracellular Dox concentrations; however, clonogenic survival data do not support this as the cause for synergistic cytotoxicity.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Calor , Transporte Biológico , Muerte Celular , Línea Celular Tumoral , Humanos
4.
Biochim Biophys Acta ; 1865(2): 220-7, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26951863

RESUMEN

The story of p53 and how we came to understand it is punctuated by fundamental insights into the essence of cancer. In the decades since its discovery, p53 has been shown to be centrally involved in most, if not all, of the cellular processes that maintain tissue homeostasis. Extensive functional analyses of p53 and its tumor-associated mutants have illuminated many of the common defects shared by most cancer cells. As the central character in a tale that continues to unfold, p53 has become increasingly familiar and yet remains surprisingly inscrutable. New relationships periodically come to light, and surprising, novel activities continue to emerge, thereby revealing new dimensions and aspects of its function. What lies at the very core of this complex protagonist? What is its prime motivation? As every avid reader knows, the elements of character are profoundly shaped by adversity--originating from within and without. And so it is with p53. This review will briefly recap the coordinated responses of p53 to viral infection, and outline a hypothetical model that would explain how an abundance of seemingly unrelated phenotypic attributes may in the end reflect a singular function. All stories eventually draw to a conclusion. This epic tale may eventually leave us with the realization that p53, most simply described, is a protein that evolved to mediate immune surveillance.


Asunto(s)
Inmunidad Innata , Proteína p53 Supresora de Tumor/fisiología , Animales , Comunicación Celular , Daño del ADN , Glucosa/metabolismo , Humanos , Neoplasias/etiología
5.
J Biol Chem ; 289(47): 33020-31, 2014 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-25296753

RESUMEN

The p53-mediated responses to DNA damage and the Hedgehog (Hh) signaling pathway are each recurrently dysregulated in many types of human cancer. Here we describe PTCH53, a p53 target gene that is homologous to the tumor suppressor gene PTCH1 and can function as a repressor of Hh pathway activation. PTCH53 (previously designated PTCHD4) was highly responsive to p53 in vitro and was among a small number of genes that were consistently expressed at reduced levels in diverse TP53 mutant cell lines and human tumors. Increased expression of PTCH53 inhibited canonical Hh signaling by the G protein-coupled receptor SMO. PTCH53 thus delineates a novel, inducible pathway by which p53 can repress tumorigenic Hh signals.


Asunto(s)
Proteínas Hedgehog/genética , Proteínas Represoras/genética , Transducción de Señal/genética , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética , Sitios de Unión/genética , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Regulación hacia Abajo , Células HCT116 , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación , Regiones Promotoras Genéticas/genética , Unión Proteica , Interferencia de ARN , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/metabolismo
7.
Dis Res ; 4(1): 31-39, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38962090

RESUMEN

As key modulators of the immune response, interferons play critical roles following infection and during the pathogenesis of cancer. The idea that these cytokines might be developed as new therapies emerged soon after their discovery. While enthusiasm for this approach to cancer therapy has waxed and waned over the ensuing decades, recent advances in cancer immunotherapy and our improved understanding of the tumor immune environment have led to a resurgence of interest in this unique class of biologic drug. Here, we review how interferons influence the growth of colorectal cancers (CRCs) and highlight new insights into how interferons and drugs that modulate interferon expression might be most effectively deployed in the clinic.

8.
PLoS Genet ; 6(2): e1000863, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20195506

RESUMEN

The activation of phase-specific cyclin-dependent kinases (Cdks) is associated with ordered cell cycle transitions. Among the mammalian Cdks, only Cdk1 is essential for somatic cell proliferation. Cdk1 can apparently substitute for Cdk2, Cdk4, and Cdk6, which are individually dispensable in mice. It is unclear if all functions of non-essential Cdks are fully redundant with Cdk1. Using a genetic approach, we show that Cdk2, the S-phase Cdk, uniquely controls the G(2)/M checkpoint that prevents cells with damaged DNA from initiating mitosis. CDK2-nullizygous human cells exposed to ionizing radiation failed to exclude Cdk1 from the nucleus and exhibited a marked defect in G(2)/M arrest that was unmasked by the disruption of P53. The DNA replication licensing protein Cdc6, which is normally stabilized by Cdk2, was physically associated with the checkpoint regulator ATR and was required for efficient ATR-Chk1-Cdc25A signaling. These findings demonstrate that Cdk2 maintains a balance of S-phase regulatory proteins and thereby coordinates subsequent p53-independent G(2)/M checkpoint activation.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/metabolismo , Fase G2 , Mitosis , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa 2 Dependiente de la Ciclina/deficiencia , Citoplasma/metabolismo , Citoplasma/efectos de la radiación , Activación Enzimática/efectos de la radiación , Estabilidad de Enzimas/efectos de la radiación , Fase G2/efectos de la radiación , Células HCT116 , Humanos , Ratones , Mitosis/efectos de la radiación , Modelos Biológicos , Proteínas Nucleares/metabolismo , Fosforilación/efectos de la radiación , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de la radiación , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas/efectos de la radiación , Radiación Ionizante , Fosfatasas cdc25/metabolismo
9.
bioRxiv ; 2023 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-37162859

RESUMEN

Cancers commonly harbor point mutations in TP53 that cause overexpression of functionally inactive p53 proteins. These mutant forms of p53 are immunogenic, and therefore present tantalizing targets for new forms of immunotherapy. Understanding how the immune system recognizes p53 is an important prerequisite for the development of targeted therapeutic strategies designed to exploit this common neoantigen. Monoclonal antibodies have been extensively used to probe the structural conformation of the varied isoforms of p53 and their respective mutants, and are still indispensable tools for studying the complex biological functions of these proteins. In this report, we describe the mapping of a novel epitope on p53 that appears to be shared by heat shock proteins (HSPs), which are typically upregulated in response to a variety of viral infections.

10.
Stem Cells ; 29(3): 418-29, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21308861

RESUMEN

The identification of novel approaches to specifically target the DNA-damage checkpoint response in chemotherapy-resistant cancer stem cells (CSC) of solid tumors has recently attracted great interest. We show here in colon cancer cell lines and primary colon cancer cells that inhibition of checkpoint-modulating phosphoinositide 3-kinase-related (PIK) kinases preferentially depletes the chemoresistant and exclusively tumorigenic CD133(+) cell fraction. We observed a time- and dose-dependent disproportionally pronounced loss of CD133(+) cells and the consecutive lack of in vitro and in vivo tumorigenicity of the remaining cells. Depletion of CD133(+) cells was initiated through apoptosis of cycling CD133(+) cells and further substantiated through subsequent recruitment of quiescent CD133(+) cells into the cell cycle followed by their elimination. Models using specific PIK kinase inhibitors, somatic cell gene targeting, and RNA interference demonstrated that the observed detrimental effects of caffeine on CSC were attributable specifically to the inhibition of the PIK kinase ataxia telangiectasia- and Rad3-related (ATR). Mechanistically, phosphorylation of CHK1 checkpoint homolog (S. pombe; CHK1) was significantly enhanced in CD133(+) as compared with CD133(-) cells on treatment with DNA interstrand-crosslinking (ICL) agents, indicating a preferential activation of the ATR/CHK1-dependent DNA-damage response in tumorigenic CD133(+) cells. Consistently, the chemoresistance of CD133(+) cells toward DNA ICL agents was overcome through inhibition of ATR/CHK1-signaling. In conclusion, our study illustrates a novel target to eliminate the tumorigenic CD133(+) cell population in colon cancer and provides another rationale for the development of specific ATR-inhibitors.


Asunto(s)
Carcinoma/patología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Transformación Celular Neoplásica/genética , Neoplasias del Colon/patología , Células Madre Neoplásicas/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/terapia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Línea Celular Tumoral , Separación Celular/métodos , Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/terapia , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Regulación hacia Abajo/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Terapia Genética , Glicoproteínas/metabolismo , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida/métodos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Péptidos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , ARN Interferente Pequeño/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Proc Natl Acad Sci U S A ; 106(10): 3964-9, 2009 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-19225112

RESUMEN

Through targeted homologous recombination, we developed a panel of matched colorectal cancer cell lines that differ only with respect to their endogenous TP53 status. We then used these lines to define the genes whose expression was altered after DNA damage induced by ionizing radiation. Transcriptome analyses revealed a consistent up-regulation of polo-like kinase 1 (PLK1) as well as other genes controlling the G(2)/M transition in the cells whose TP53 genes were inactivated compared with those with WT TP53 genes. This led to the hypothesis that the viability of stressed cells without WT TP53 depended on PLK1. This hypothesis was validated by demonstrating that stressed cancer cells without WT TP53 alleles were highly sensitive to PLK1 inhibitors, both in vivo and in vitro.


Asunto(s)
Neoplasias/patología , Neoplasias/terapia , Proteína p53 Supresora de Tumor/metabolismo , Alelos , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Fase G1/efectos de los fármacos , Perfilación de la Expresión Génica , Marcación de Gen , Genotipo , Humanos , Imidazoles/metabolismo , Ratones , Piperazinas/metabolismo , Pteridinas/farmacología , Transcripción Genética/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
STAR Protoc ; 3(1): 101123, 2022 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-35098167

RESUMEN

The AdenoBuilder platform enables the in vitro assembly of recombinant vectors from plasmid inserts that span the adenovirus genome. Two advantages of AdenoBuilder are the ease of modifying the genome and the ability to produce multicomponent vectors in a single step, facilitating parallel approaches to vector optimization. This protocol describes how to introduce transgenes in place of the endogenous Human Adenovirus serotype 5 (HAd5) E1 and/or E3 genes and can be applied to other parts of the HAd5 genome. For complete details on the use and execution of this protocol, please refer to Miciak et al. (2018).


Asunto(s)
Adenoviridae/genética , Recombinación Genética , Vectores Genéticos , Plásmidos , Transgenes
13.
Proc Natl Acad Sci U S A ; 105(52): 20752-7, 2008 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-19091954

RESUMEN

Chk1 is widely known as a DNA damage checkpoint signaling protein. Unlike many other checkpoint proteins, Chk1 also plays an essential but poorly defined role in the proliferation of unperturbed cells. Activation of Chk1 after DNA damage is known to require the phosphorylation of several C-terminal residues, including the highly conserved S317 and S345 sites. To evaluate the respective roles of these individual sites and assess their contribution to the functions of Chk1, we used a gene targeting approach to introduce point mutations into the endogenous human CHK1 locus. We report that the essential and nonessential functions of Chk1 are regulated through distinct phosphorylation events and can be genetically uncoupled. The DNA damage response function of Chk1 was nonessential. Targeted mutation of S317 abrogated G(2)/M checkpoint activation, prevented subsequent phosphorylation of Chk1, impaired efficient progression of DNA replication forks, and increased fork stalling, but did not impact viability. Thus, the nonessential DNA damage response function of Chk1 could be unambiguously linked to its role in DNA replication control. In contrast, a CHK1 allele with mutated S345 did not support viability, indicating an essential role for this residue during the unperturbed cell cycle. A distinct, physiologic mode of S345 phosphorylation, initiated at the centrosome during unperturbed mitosis was independent of codon 317 status and mechanistically distinct from the ordered and sequential phosphorylation of serine residues on Chk1 induced by DNA damage. Our findings suggest an essential regulatory role for Chk1 phosphorylation during mitotic progression.


Asunto(s)
Centrosoma/metabolismo , Daño del ADN/fisiología , Replicación del ADN/fisiología , Fase G2/fisiología , Metafase/fisiología , Proteínas Quinasas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/fisiología , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Marcación de Gen/métodos , Humanos , Mutación Missense , Fosforilación/fisiología , Proteínas Quinasas/genética , Sitios de Carácter Cuantitativo/fisiología
14.
Elife ; 102021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34726152

RESUMEN

In pigs, nitrate supplements can protect salivary glands from the damage caused by radiation therapy to the head and neck.


Asunto(s)
Neoplasias de Cabeza y Cuello , Animales , Glándulas Salivales , Porcinos
15.
Nucleic Acids Res ; 36(19): e127, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18784188

RESUMEN

Epitope tagging is a powerful and commonly used approach for studying the physical properties of proteins and their functions and localization in eukaryotic cells. In the case of Saccharomyces cerevisiae, it has been possible to exploit the high efficiency of homologous recombination to tag proteins by modifying their endogenous genes, making it possible to tag virtually every endogenous gene and perform genome-wide proteomics experiments. However, due to the relative inefficiency of homologous recombination in cultured human cells, epitope-tagging approaches have been limited to ectopically expressed transgenes, with the attendant limitations of their nonphysiological transcriptional regulation and levels of expression. To overcome this limitation, a modification and extension of adeno-associated virus-mediated human somatic cell gene targeting technology is described that makes it possible to simply and easily create an endogenous epitope tag in the same way that it is possible to knock out a gene. Using this approach, we have created and validated human cell lines with epitope-tagged alleles of two cancer-related genes in a variety of untransformed and transformed human cell lines. This straightforward approach makes it possible to study the physical and biological properties of endogenous proteins in human cells without the need for specialized antibodies for individual proteins of interest.


Asunto(s)
Línea Celular , Epítopos/análisis , Marcación de Gen/métodos , Péptidos/análisis , Alelos , Línea Celular Transformada , Dependovirus/genética , Genes p53 , Vectores Genéticos , Humanos , Oligopéptidos , Fosfohidrolasa PTEN/genética , Péptidos/inmunología
16.
Cancer Res ; 80(2): 153-155, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31941677

RESUMEN

To what extent does the acquisition of mesenchymal phenotypes by tumor epithelial cells contribute to metastasis? A definitive answer to this question has remained elusive despite much experimentation and debate. Recently, an influential study based on fluorescence-based lineage tracing technology provided evidence that very few of the cells that populate experimental metastases in fact undergo the epithelial-to-mesenchymal transition. Persistent questions regarding the concordance between marker conversion and cell phenotypes prompted Lourenco and colleagues to complement their lineage tracking system with single-cell analysis. This granular approach provides an unprecedented view of the phenotypic transitions that take place during metastasis and their striking heterogeneity.See related article by Lourenco et al., p. 163.


Asunto(s)
Neoplasias de la Mama , Células Epiteliales , Transición Epitelial-Mesenquimal , Humanos , Fenotipo
17.
Cancer Res ; 66(3): 1684-936, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16452228

RESUMEN

Hypoxia-inducible factor-1alpha (HIF-1alpha) is a transcription factor that directly transactivates genes important for the growth and metabolism of solid tumors. HIF-1alpha is overexpressed in cancer, and its level of expression is correlated with patient mortality. Increased synthesis or stability of HIF-1alpha can be induced by hypoxia-dependent or hypoxia-independent factors. Thus, HIF-1alpha is expressed in both nonhypoxic and hypoxic cancer cells. The role of HIF-1alpha in nonhypoxia-mediated cancer cell proliferation remains speculative. We have disrupted HIF-1alpha by targeted homologous recombination in HCT116 and RKO human colon cancer cells. Loss of HIF-1alpha significantly reduced nonhypoxia-mediated cell proliferation in vitro and in vivo. Paradoxically, loss of HIF-1alpha expression did not grossly affect the hypoxic compartments within tumor xenografts in vivo, although HIF-1alpha promoted cell proliferation and survival under hypoxia in vitro. To further test the role of HIF-1alpha within tumor compartments, we generated cells with combined disruptions of both HIF-1alpha and vascular endothelial growth factor (VEGF). In all xenografts, disruption of VEGF led to marked expansion of the hypoxic compartments and growth delay. Nonetheless, the presence or absence of HIF-1alpha did not grossly affect these expanded hypoxic compartments. These data provide compelling evidence that, in a subset of colon cancers, (a) HIF-1alpha is a positive factor for nonhypoxia-mediated cell proliferation in vitro and in vivo and (b) HIF-1alpha is a positive factor for cell proliferation and survival under hypoxic conditions in vitro, but does not grossly contribute to the tumor hypoxic compartments in vivo.


Asunto(s)
Neoplasias del Colon/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Animales , Procesos de Crecimiento Celular/genética , Procesos de Crecimiento Celular/fisiología , Hipoxia de la Célula , Supervivencia Celular/genética , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , ADN de Neoplasias/biosíntesis , Femenino , Glucólisis , Células HCT116 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones Desnudos , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/fisiología
18.
Mol Cancer Ther ; 6(4): 1406-13, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17431119

RESUMEN

The diverse responses of human cells to various forms of DNA damage are controlled by a complex network of signaling proteins. There has been considerable interest in the components of this signaling apparatus as potential targets for new forms of anticancer therapy. In this report, we examine the contributions of an upstream signaling molecule, the ataxia telangiectasia mutated- and Rad3-related (ATR) protein kinase, to the resistance of cancer cells to DNA-damaging agents that are commonly used as anticancer therapeutics. Loss of ATR function in knock-in cancer cells strikingly enhanced the effects of several of the most commonly used therapeutic compounds, impeding the progression of the cell cycle and reducing long-term cancer cell survival. Loss of ATR function potentiated the toxicity of alkylating agents most strikingly, antimetabolites moderately, and double-strand break-inducing agents to a lesser extent. These results suggest that specific inhibition of ATR activity will be a valid strategy to increase the effectiveness of currently used modes of therapy.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Ciclo Celular/metabolismo , Neoplasias/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Fluorouracilo/farmacología , Genotipo , Células HCT116 , Humanos , Neoplasias/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Ensayo de Tumor de Célula Madre
19.
PLoS One ; 13(6): e0199563, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29949649

RESUMEN

The adenoviruses are essential tools for basic research and therapeutic development. Robust methods for the generation of mutant and recombinant viruses are crucial for these diverse applications. Here we describe a simple plasmid-based method that permits highly efficient modification of the adenoviral genome and rapid production of high-titer virus stocks. The 36-kilobase genome of adenovirus serotype 5 was divided into seven tractable blocks that could be individually modified in a single step and reassembled in vitro. Because the system is composed of compact modules, modifications at different loci can be readily recombined. Viral assemblies were delivered to packaging cells by electroporation, a strategy that consistently resulted in the de novo production of 108 infectious units in 3-5 days. In principle, a similar strategy could be applied to any other adenovirus serotype or to other double-strand DNA viruses.


Asunto(s)
Adenovirus Humanos/genética , ADN Recombinante , Técnicas Genéticas , Genoma Viral , Plásmidos , Ensamble de Virus , Animales , Chlorocebus aethiops , Electroporación , Vectores Genéticos , Células HEK293 , Humanos , Células Vero
20.
Nucleic Acids Res ; 33(18): e158, 2005 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-16214806

RESUMEN

Recent studies have demonstrated the utility of recombinant adeno-associated viral (rAAV) vectors in the generation of human knockout cell lines. The efficiency with which such cell lines can be generated using rAAV, in comparison with more extensively described plasmid-based approaches, has not been directly tested. In this report, we demonstrate that targeting constructs delivered by rAAV vectors were nearly 25-fold more efficient than transfected plasmids that target the same exon. In addition, we describe a novel vector configuration which we term the synthetic exon promoter trap (SEPT). This targeting element further improved the efficiency of knockout generation and uniquely facilitated the generation of knockin alterations. An rAAV-based SEPT targeting construct was used to transfer a mutant CTNNB1 allele, encoding an oncogenic form of beta-catenin, from one cell line to another. This versatile method was thus shown to facilitate the efficient integration of small, defined sequence alterations into the chromosomes of cultured human cells.


Asunto(s)
Marcación de Gen/métodos , Mutación , Línea Celular Tumoral , Proteínas del Citoesqueleto/genética , Dependovirus/genética , Exones , Vectores Genéticos , Humanos , Plásmidos , Regiones Promotoras Genéticas , Eliminación de Secuencia , Transactivadores/genética , beta Catenina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA