Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Epigenetics ; 14(1): 96, 2022 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-35902886

RESUMEN

BACKGROUND: Interferon gamma (IFNγ) is a pro-inflammatory cytokine that directly activates the JAK/STAT pathway. However, the temporal dynamics of chromatin remodeling and transcriptional activation initiated by IFNγ have not been systematically profiled in an unbiased manner. Herein, we integrated transcriptomic and epigenomic profiling to characterize the acute epigenetic changes induced by IFNγ stimulation in a murine breast cancer model. RESULTS: We identified de novo activation of cis-regulatory elements bound by Irf1 that were characterized by increased chromatin accessibility, differential usage of pro-inflammatory enhancers, and downstream recruitment of BET proteins and RNA polymerase II. To functionally validate this hierarchical model of IFNγ-driven transcription, we applied selective antagonists of histone acetyltransferases P300/CBP or acetyl-lysine readers of the BET family. This highlighted that histone acetylation is an antecedent event in IFNγ-driven transcription, whereby targeting of P300/CBP acetyltransferase activity but not BET inhibition could curtail the epigenetic remodeling induced by IFNγ through suppression of Irf1 transactivation. CONCLUSIONS: These data highlight the ability for epigenetic therapies to reprogram pro-inflammatory gene expression, which may have therapeutic implications for anti-tumor immunity and inflammatory diseases.


Asunto(s)
Neoplasias de la Mama , Interferón gamma , Acetilación , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Metilación de ADN , Proteína p300 Asociada a E1A , Femenino , Interferón gamma/farmacología , Quinasas Janus , Proteínas de la Membrana , Ratones , Fosfoproteínas , Factores de Transcripción STAT , Transducción de Señal
2.
ACS Med Chem Lett ; 12(5): 726-731, 2021 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-34055218

RESUMEN

Aberrant gene activation driven by the histone acetyltransferases p300 and CREB binding protein (CBP) has been linked to several diseases, including cancers. Because of this, many efforts have been aimed toward the targeting of the closely related paralogues, p300 and CBP, but these endeavors have been exclusively directed toward noncovalent inhibitors. X-ray crystallography of A-485 revealed that both p300 and CBP possess a cysteine (C1450) near the active site, thus rendering covalent inhibition an attractive chemical approach. Herein we report the development of compound 2, an acrylamide-based inhibitor of p300/CBP that forms a covalent adduct with C1450. We demonstrated using mass spectrometry that compound 2 selectively targets C1450, and we also validated covalent binding using kinetics experiments and cellular washout studies. The discovery of covalent inhibitor 2 gives us a unique tool for the study of p300/CBP biology.

3.
Mol Cell ; 81(10): 2183-2200.e13, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34019788

RESUMEN

To separate causal effects of histone acetylation on chromatin accessibility and transcriptional output, we used integrated epigenomic and transcriptomic analyses following acute inhibition of major cellular lysine acetyltransferases P300 and CBP in hematological malignancies. We found that catalytic P300/CBP inhibition dynamically perturbs steady-state acetylation kinetics and suppresses oncogenic transcriptional networks in the absence of changes to chromatin accessibility. CRISPR-Cas9 screening identified NCOR1 and HDAC3 transcriptional co-repressors as the principal antagonists of P300/CBP by counteracting acetylation turnover kinetics. Finally, deacetylation of H3K27 provides nucleation sites for reciprocal methylation switching, a feature that can be exploited therapeutically by concomitant KDM6A and P300/CBP inhibition. Overall, this study indicates that the steady-state histone acetylation-methylation equilibrium functions as a molecular rheostat governing cellular transcription that is amenable to therapeutic exploitation as an anti-cancer regimen.


Asunto(s)
Biocatálisis , Histonas/metabolismo , Oncogenes , Transcripción Genética , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Línea Celular , Cromatina/metabolismo , Proteínas Co-Represoras/metabolismo , Secuencia Conservada , Evolución Molecular , Redes Reguladoras de Genes , Genoma , Histona Desacetilasas/metabolismo , Humanos , Cinética , Metilación , Modelos Biológicos , ARN Polimerasa II/metabolismo
4.
Bioorg Med Chem Lett ; 39: 127854, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33631370

RESUMEN

p300 and CREB-binding protein (CBP) are essential for a multitude of cellular processes. Dysregulation of p300/CBP histone acetyltransferase activity is linked to a broad spectrum of human diseases including cancers. A novel drug-like spirohydantoin (21) has been discovered as a selective orally bioavailable inhibitor of p300/CBP histone acetyltransferase. Lead compound 21 is more potent than the first-in-class lead A-485 in both enzymatic and cellular assays and lacks the off-target inhibition of dopamine and serotonin transporters, that was observed with A-485.


Asunto(s)
Proteína de Unión a CREB/antagonistas & inhibidores , Descubrimiento de Drogas , Proteína p300 Asociada a E1A/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Hidantoínas/farmacología , Compuestos de Espiro/farmacología , Administración Oral , Disponibilidad Biológica , Proteína de Unión a CREB/metabolismo , Relación Dosis-Respuesta a Droga , Proteína p300 Asociada a E1A/metabolismo , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/metabolismo , Humanos , Hidantoínas/administración & dosificación , Hidantoínas/metabolismo , Estructura Molecular , Compuestos de Espiro/administración & dosificación , Compuestos de Espiro/metabolismo , Relación Estructura-Actividad
5.
Nature ; 558(7710): E1, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29769713

RESUMEN

In the originally published version of this Letter, the authors Arthur F. Kluge, Michael A. Patane and Ce Wang were inadvertently omitted from the author list. Their affiliations are: I-to-D, Inc., PO Box 6177, Lincoln, Massachusetts 01773, USA (A.F.K.); Mitobridge, Inc. 1030 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA (M.A.P.); and China Novartis Institutes for BioMedical Research, No. 4218 Jinke Road, Zhangjiang Hi-Tech Park, Pudong District, Shanghai 201203, China (C.W.). These authors contributed to the interpretation of results and design of compounds. In addition, author 'Edward A. Kesicki' was misspelled as 'Ed Kesicki'. These errors have been corrected online.

6.
Cell ; 174(1): 231-244.e12, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29804834

RESUMEN

The acetyltransferases CBP and p300 are multifunctional transcriptional co-activators. Here, we combined quantitative proteomics with CBP/p300-specific catalytic inhibitors, bromodomain inhibitor, and gene knockout to reveal a comprehensive map of regulated acetylation sites and their dynamic turnover rates. CBP/p300 acetylates thousands of sites, including signature histone sites and a multitude of sites on signaling effectors and enhancer-associated transcriptional regulators. Time-resolved acetylome analyses identified a subset of CBP/p300-regulated sites with very rapid (<30 min) acetylation turnover, revealing a dynamic balance between acetylation and deacetylation. Quantification of acetylation, mRNA, and protein abundance after CBP/p300 inhibition reveals a kinetically competent network of gene expression that strictly depends on CBP/p300-catalyzed rapid acetylation. Collectively, our in-depth acetylome analyses reveal systems attributes of CBP/p300 targets, and the resource dataset provides a framework for investigating CBP/p300 functions and for understanding the impact of small-molecule inhibitors targeting its catalytic and bromodomain activities.


Asunto(s)
Acetiltransferasas/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Acetilación/efectos de los fármacos , Acetiltransferasas/antagonistas & inhibidores , Animales , Línea Celular , Técnicas de Inactivación de Genes , Semivida , Compuestos Heterocíclicos de 4 o más Anillos/química , Compuestos Heterocíclicos de 4 o más Anillos/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Histonas/metabolismo , Humanos , Marcaje Isotópico , Cinética , Espectrometría de Masas , Ratones , Péptidos/análisis , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Transcriptoma/efectos de los fármacos , Factores de Transcripción p300-CBP/antagonistas & inhibidores , Factores de Transcripción p300-CBP/genética
7.
ACS Med Chem Lett ; 9(1): 28-33, 2018 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-29348807

RESUMEN

p300 and its paralog CBP can acetylate histones and other proteins and have been implicated in a number of diseases characterized by aberrant gene activation, such as cancer. A novel, highly selective, orally bioavailable histone acetyltransferase (HAT) domain inhibitor has been identified through virtual ligand screening and subsequent optimization of a unique hydantoin screening hit. Conformational restraint in the form of a spirocyclization followed by substitution with a urea led to a significant improvement in potency. Replacement of the hydantoin moiety with an oxazolidinedione followed by fluoro substitution led to A-485, which exhibits potent cell activity, low clearance, and high oral bioavailability.

8.
Nature ; 550(7674): 128-132, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28953875

RESUMEN

The dynamic and reversible acetylation of proteins, catalysed by histone acetyltransferases (HATs) and histone deacetylases (HDACs), is a major epigenetic regulatory mechanism of gene transcription and is associated with multiple diseases. Histone deacetylase inhibitors are currently approved to treat certain cancers, but progress on the development of drug-like histone actyltransferase inhibitors has lagged behind. The histone acetyltransferase paralogues p300 and CREB-binding protein (CBP) are key transcriptional co-activators that are essential for a multitude of cellular processes, and have also been implicated in human pathological conditions (including cancer). Current inhibitors of the p300 and CBP histone acetyltransferase domains, including natural products, bi-substrate analogues and the widely used small molecule C646, lack potency or selectivity. Here, we describe A-485, a potent, selective and drug-like catalytic inhibitor of p300 and CBP. We present a high resolution (1.95 Å) co-crystal structure of a small molecule bound to the catalytic active site of p300 and demonstrate that A-485 competes with acetyl coenzyme A (acetyl-CoA). A-485 selectively inhibited proliferation in lineage-specific tumour types, including several haematological malignancies and androgen receptor-positive prostate cancer. A-485 inhibited the androgen receptor transcriptional program in both androgen-sensitive and castration-resistant prostate cancer and inhibited tumour growth in a castration-resistant xenograft model. These results demonstrate the feasibility of using small molecule inhibitors to selectively target the catalytic activity of histone acetyltransferases, which may provide effective treatments for transcriptional activator-driven malignancies and diseases.


Asunto(s)
Linaje de la Célula , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Histona Acetiltransferasas/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Factores de Transcripción p300-CBP/antagonistas & inhibidores , Acetilcoenzima A/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Unión Competitiva , Biocatálisis/efectos de los fármacos , Dominio Catalítico/efectos de los fármacos , Línea Celular Tumoral , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/enzimología , Neoplasias Hematológicas/patología , Compuestos Heterocíclicos de 4 o más Anillos/química , Histona Acetiltransferasas/química , Histona Acetiltransferasas/metabolismo , Humanos , Masculino , Ratones , Ratones SCID , Modelos Moleculares , Neoplasias/enzimología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/enzimología , Neoplasias de la Próstata Resistentes a la Castración/patología , Conformación Proteica , Receptores Androgénicos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Factores de Transcripción p300-CBP/química , Factores de Transcripción p300-CBP/metabolismo
9.
Nat Chem Biol ; 13(3): 317-324, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28114273

RESUMEN

Protein lysine methyltransferases (PKMTs) regulate diverse physiological processes including transcription and the maintenance of genomic integrity. Genetic studies suggest that the PKMTs SUV420H1 and SUV420H2 facilitate proficient nonhomologous end-joining (NHEJ)-directed DNA repair by catalyzing the di- and trimethylation (me2 and me3, respectively) of lysine 20 on histone 4 (H4K20). Here we report the identification of A-196, a potent and selective inhibitor of SUV420H1 and SUV420H2. Biochemical and co-crystallization analyses demonstrate that A-196 is a substrate-competitive inhibitor of both SUV4-20 enzymes. In cells, A-196 induced a global decrease in H4K20me2 and H4K20me3 and a concomitant increase in H4K20me1. A-196 inhibited 53BP1 foci formation upon ionizing radiation and reduced NHEJ-mediated DNA-break repair but did not affect homology-directed repair. These results demonstrate the role of SUV4-20 enzymatic activity in H4K20 methylation and DNA repair. A-196 represents a first-in-class chemical probe of SUV4-20 to investigate the role of histone methyltransferases in genomic integrity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Inestabilidad Genómica/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Línea Celular Tumoral , Cristalografía por Rayos X , Reparación del ADN/efectos de los fármacos , Inhibidores Enzimáticos/química , Compuestos Heterocíclicos de 4 o más Anillos/química , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Metilación/efectos de los fármacos , Modelos Moleculares , Estructura Molecular
10.
ACS Med Chem Lett ; 6(1): 58-62, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25589931

RESUMEN

Aided by molecular modeling, compounds with a pyrimidine-based tricyclic scaffold were designed and confirmed to inhibit Wee1 kinase. Structure-activity studies identified key pharmacophores at the aminoaryl and halo-benzene regions responsible for binding affinity with sub-nM K i values. The potent inhibitors demonstrated sub-µM activities in both functional and mechanism-based cellular assays and also possessed desirable pharmacokinetic profiles. The lead molecule, 31, showed oral efficacy in potentiating the antiproliferative activity of irinotecan, a cytotoxic agent, in a NCI-H1299 mouse xenograft model.

11.
J Biol Chem ; 285(2): 1358-70, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19861414

RESUMEN

Activation of the G(o/i)-coupled cannabinoid 1 receptor (CB1R) has been shown to induce neurite outgrowth in Neuro2A cells through activation of Src kinase and STAT3 transcription factor. Signaling by the interleukin 6 receptor (IL-6R) also activates STAT3 through Jak kinase. We studied if signals from the two pathways could be integrated in a synergistic manner to trigger neurite outgrowth in Neuro2A cells. At low concentrations, when agonist at either receptor by itself has no effect, we found that CB1R and IL-6R stimulation together induced synergistic neurite outgrowth. Signal integration requires activation of transcription factors by Src, Jak, and mitogen-activated protein kinases. Mitogen-activated protein kinase can be activated by both receptors and shows enhanced early activation in the presence of both ligands. CREB and STAT3 transcription factors are required for synergy and show enhanced DNA-binding activity when both receptors are activated. STAT3 plays a critical role in integration of the signals downstream of the two receptors. When both pathways are activated, STAT3 phosphorylation is sustained for 6 h. This prolonged activation of STAT3 requires deactivation of SHP2 phosphatase. Reduction of SHP2 levels by RNA interference results in greater synergy in neurite outgrowth. Simultaneous knockdown of both SHP2 and STAT3 blocks the synergistic triggering of neurite outgrowth, indicating that STAT3 is downstream of SHP2. CB1R and IL-6R co-stimulation enhanced the differentiation of rat cortical neuron primary cultures. These results provide a mechanism where multiple protein kinases and transcription factors interact to integrate signals from G protein-coupled and cytokine receptor to evoke neurite outgrowth in Neuro2A cells.


Asunto(s)
Diferenciación Celular/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Neuritas/metabolismo , Receptor Cannabinoide CB1/metabolismo , Receptores de Interleucina-6/metabolismo , Animales , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Quinasas Janus/genética , Quinasas Janus/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Ratas , Receptor Cannabinoide CB1/genética , Receptores de Interleucina-6/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Factores de Tiempo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
12.
Science ; 320(5878): 903-9, 2008 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-18487186

RESUMEN

Cannabinoid receptor 1 (CB1R) regulates neuronal differentiation. To understand the logic underlying decision-making in the signaling network controlling CB1R-induced neurite outgrowth, we profiled the activation of several hundred transcription factors after cell stimulation. We assembled an in silico signaling network by connecting CB1R to 23 activated transcription factors. Statistical analyses of this network predicted a role for the breast cancer 1 protein BRCA1 in neuronal differentiation and a new pathway from CB1R through phosphoinositol 3-kinase to the transcription factor paired box 6 (PAX6). Both predictions were experimentally confirmed. Results of transcription factor activation experiments that used pharmacological inhibitors of kinases revealed a network organization of partial OR gates regulating kinases stacked above AND gates that control transcription factors, which together allow for distributed decision-making in CB1R-induced neurite outgrowth.


Asunto(s)
Neuritas/fisiología , Neuronas/citología , Receptor Cannabinoide CB1/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Proteína BRCA1/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Células Cultivadas , Biología Computacional , Simulación por Computador , Proteínas del Ojo/metabolismo , Hipocampo/citología , Proteínas de Homeodominio/metabolismo , Redes y Vías Metabólicas , Ratones , Neuronas/metabolismo , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Mapeo de Interacción de Proteínas , Ratas , Proteínas Represoras/metabolismo , Factores de Transcripción/antagonistas & inhibidores
13.
Front Biosci ; 13: 4544-57, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508528

RESUMEN

Neurogenesis is a long and winding journey. A neural progenitor cell migrates long distances, differentiates by forming a single axon and multiple dendrites, undergoes maturation, and ultimately survives. The initial formation of neurites during neuronal differentiation, commonly referred to as "neurite outgrowth," can be induced by a large repertoire of signals that stimulate an array of receptors and downstream signaling pathways. The G(i/o) family of heterotrimeric G-proteins are abundantly expressed in the brain and enriched at neuronal growth cones. Recent evidence has uncovered several G(i/o)-coupled receptors that induce neurite outgrowth and has begun to elucidate the underlying molecular mechanisms. Emerging data suggests that signals from several G(i/o)-coupled receptors converge at the transcription factor STAT3 to regulate neurite outgrowth and at Rac1 and Cdc42 to regulate cytoskeletal reorganization. Physiologically, signaling through G(i/o)-coupled cannabinoid receptors is critical for pro percentral nervous system development. As the mechanisms by which G(i/o)-coupled receptors regulate neurite outgrowth are clarified, it is becoming evident that modulating signals from G(i/o) and their receptors has great potential for the treatment of neurodegenerative diseases.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Neuritas/fisiología , Animales , Cannabinoides/metabolismo , Diferenciación Celular/fisiología , División Celular , Conos de Crecimiento/fisiología , Degeneración Nerviosa/fisiopatología , Transducción de Señal
14.
PLoS One ; 3(2): e1609, 2008 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-18270596

RESUMEN

Growing evidence supports the importance of ubiquitin ligases in the pathogenesis of muscular disorders, although underlying mechanisms remain largely elusive. Here we show that the expression of RNF5 (aka RMA1), an ER-anchored RING finger E3 ligase implicated in muscle organization and in recognition and processing of malfolded proteins, is elevated and mislocalized to cytoplasmic aggregates in biopsies from patients suffering from sporadic-Inclusion Body Myositis (sIBM). Consistent with these findings, an animal model for hereditary IBM (hIBM), but not their control littermates, revealed deregulated expression of RNF5. Further studies for the role of RNF5 in the pathogenesis of s-IBM and more generally in muscle physiology were performed using RNF5 transgenic and KO animals. Transgenic mice carrying inducible expression of RNF5, under control of beta-actin or muscle specific promoter, exhibit an early onset of muscle wasting, muscle degeneration and extensive fiber regeneration. Prolonged expression of RNF5 in the muscle also results in the formation of fibers containing congophilic material, blue-rimmed vacuoles and inclusion bodies. These phenotypes were associated with altered expression and activity of ER chaperones, characteristic of myodegenerative diseases such as s-IBM. Conversely, muscle regeneration and induction of ER stress markers were delayed in RNF5 KO mice subjected to cardiotoxin treatment. While supporting a role for RNF5 Tg mice as model for s-IBM, our study also establishes the importance of RNF5 in muscle physiology and its deregulation in ER stress associated muscular disorders.


Asunto(s)
Cuerpos de Inclusión/metabolismo , Proteínas de la Membrana/fisiología , Enfermedades Musculares/etiología , Miositis , Ubiquitina-Proteína Ligasas/fisiología , Animales , Proteínas de Unión al ADN , Expresión Génica , Cuerpos de Inclusión/química , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Fenotipo , Ubiquitina-Proteína Ligasas/genética
15.
Cancer Res ; 67(17): 8172-9, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17804730

RESUMEN

The selective ubiquitination of proteins by ubiquitin E3 ligases plays an important regulatory role in control of cell differentiation, growth, and transformation and their dysregulation is often associated with pathologic outcomes, including tumorigenesis. RNF5 is an E3 ubiquitin ligase that has been implicated in motility and endoplasmic reticulum stress response. Here, we show that RNF5 expression is up-regulated in breast cancer tumors and related cell lines. Elevated expression of RNF5 was seen in breast cancer cell lines that became more sensitive to cytochalasin D- and paclitaxel-induced apoptosis following its knockdown with specific short interfering RNA. Inhibition of RNF5 expression markedly decreased cell proliferation and caused a reorganization of the actin cytoskeleton in response to stress in MCF-7 but not in p53 mutant breast cancer cells, suggesting a p53-dependent function. Significantly, high levels of RNF5 were associated with decreased survival in human breast cancer specimens. Similarly, RNF5 levels were higher in metastatic melanoma specimens and in melanoma, leukemia, ovarian, and renal tumor-derived cell lines, suggesting that increased RNF5 expression may be a common event during tumor progression. These results indicate that RNF5 is a novel regulator of breast cancer progression through its effect on actin cytoskeletal alterations, which also affect sensitivity of breast cancer cells to cytoskeletal targeting antineoplastic agents.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Proteínas de Unión al ADN/genética , Ubiquitina-Proteína Ligasas/genética , Actinas/metabolismo , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Proteínas de Unión al ADN/antagonistas & inhibidores , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , ARN Interferente Pequeño/farmacología , Análisis de Supervivencia , Análisis de Matrices Tisulares
16.
Mol Cell Biol ; 25(19): 8619-30, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16166642

RESUMEN

We report the identification and characterization of JAMP (JNK1 [Jun N-terminal kinase 1]-associated membrane protein), a predicted seven-transmembrane protein that is localized primarily within the plasma membrane and associates with JNK1 through its C-terminal domain. JAMP association with JNK1 outcompetes JNK1 association with mitogen-activated protein kinase phosphatase 5, resulting in increased and prolonged JNK1 activity following stress. Elevated expression of JAMP following UV or tunicamycin treatment results in sustained JNK activity and a higher level of JNK-dependent apoptosis. Inhibition of JAMP expression by RNA interference reduces the degree and duration of JNK activation and concomitantly the level of stress-induced apoptosis. Through its regulation of JNK1 activity, JAMP emerges as a membrane-anchored regulator of the duration of JNK1 activity in response to diverse stress stimuli.


Asunto(s)
Proteínas Portadoras/fisiología , Membrana Celular/metabolismo , Regulación Enzimológica de la Expresión Génica , Glicoproteínas de Membrana/fisiología , Células 3T3 , Secuencia de Aminoácidos , Animales , Apoptosis , Proteínas Portadoras/biosíntesis , Línea Celular , Línea Celular Tumoral , Movimiento Celular , ADN/metabolismo , ADN Complementario/metabolismo , Fosfatasas de Especificidad Dual , Glicosilación , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Inmunoprecipitación , Sistema de Señalización de MAP Quinasas , Glicoproteínas de Membrana/biosíntesis , Ratones , Microscopía Confocal , Datos de Secuencia Molecular , Células 3T3 NIH , Fosfoproteínas Fosfatasas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fracciones Subcelulares/metabolismo , Factores de Tiempo , Distribución Tisular , Transfección , Tunicamicina/farmacología , Rayos Ultravioleta
17.
Biochemistry ; 43(42): 13416-23, 2004 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-15491148

RESUMEN

The DNA ligation reaction of topoisomerase II is essential for genomic integrity. However, it has been impossible to examine many fundamental aspects of this reaction because ligation assays historically required the enzyme to cleave a DNA substrate before sealing the nucleic acid break. Recently, a cleavage-independent DNA ligation assay was developed for human topoisomerase IIalpha [Bromberg, K. D., Hendricks, C., Burgin, A. B., and Osheroff, N. (2002) J. Biol. Chem. 277, 31201-31206]. This assay overcomes the requirement for DNA cleavage by monitoring the ability of the enzyme to ligate a nicked oligonucleotide in which the 5'-terminal phosphate at the nick has been activated by covalent attachment to the tyrosine mimic, p-nitrophenol. The cleavage-independent ligation assay was used to more fully characterize the DNA ligation activity of human topoisomerase IIalpha. Results suggest that the active site tyrosine contributes little to the catalysis of DNA ligation beyond its primary role as an activating/leaving group. Although arginine 804 (the residue immediately N-terminal to the active site tyrosine) has been proposed to help anchor the 5'-DNA terminus during cleavage, conversion of this residue to alanine had only a modest effect on DNA ligation. Thus, it appears that arginine 804 does not play an essential role in DNA strand joining. In contrast, disruption of base pairing at the 5'-DNA terminus abrogated DNA ligation in the absence of a covalent enzyme-DNA bond. Therefore, it is proposed that base pairing represents a secondary mechanism for aligning the 5'-DNA termini for ligation. Finally, the human enzyme appears to ligate the two scissile bonds of a cleavage site in a nonconcerted fashion.


Asunto(s)
Reparación del ADN , ADN-Topoisomerasas de Tipo II/metabolismo , Antígenos de Neoplasias , Emparejamiento Base , Catálisis , Daño del ADN , ADN Superhelicoidal/metabolismo , Proteínas de Unión al ADN , Humanos , Hidrólisis , Oligonucleótidos/metabolismo , Plásmidos , Especificidad por Sustrato
18.
Biochemistry ; 43(23): 7563-74, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15182198

RESUMEN

Benzene is a human carcinogen that induces hematopoietic malignancies. It is believed that benzene does not initiate leukemias directly, but rather generates DNA damage through a series of phenolic metabolites, especially 1,4-benzoquinone. The cellular consequences of 1,4-benzoquinone are consistent with those of topoisomerase II-targeted drugs. Therefore, it has been proposed that the compound initiates specific leukemias by acting as a topoisomerase II poison. This hypothesis, however, has not been supported by in vitro studies. While 1,4-benzoquinone has been shown to inhibit topoisomerase II catalysis, increases in enzyme-mediated DNA cleavage have not been reported. Because of the potential involvement of topoisomerase II in benzene-induced leukemias, we re-examined the effects of the compound on DNA cleavage mediated by human topoisomerase IIalpha. In contrast to previous reports, we found that 1,4-benzoquinone was a strong topoisomerase II poison and was more potent in vitro than the anticancer drug etoposide. DNA cleavage enhancement probably was unseen in previous studies due to the presence of reducing agents in reaction buffers and the incubation of 1,4-benzoquinone with the enzyme prior to the addition of DNA. 1,4-Benzoquinone increased topoisomerase II-mediated DNA cleavage primarily by enhancing the forward rate of scission. In vitro, the compound induced cleavage at DNA sites proximal to a defined leukemic chromosomal breakpoint and displayed a sequence specificity that differed from that of etoposide. Finally, 1,4-benzoquinone stimulated DNA cleavage by topoisomerase IIalpha in cultured human cells. The present findings are consistent with the hypothesis that topoisomerase IIalpha plays a role in the initiation of specific leukemias induced by benzene and its metabolites.


Asunto(s)
Benzoquinonas/farmacología , Benzoquinonas/toxicidad , Inhibidores de Topoisomerasa II , Secuencia de Bases , Benzoquinonas/química , Catálisis , Células Cultivadas , ADN-Topoisomerasas de Tipo II/metabolismo , ADN Circular/genética , ADN Circular/metabolismo , Ditiotreitol/farmacología , Glutatión/farmacología , Humanos , Plásmidos/genética , Plásmidos/metabolismo
19.
Biochemistry ; 42(12): 3393-8, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12653542

RESUMEN

Several important antineoplastic drugs kill cells by increasing levels of topoisomerase II-mediated DNA breaks. These compounds act by two distinct mechanisms. Agents such as etoposide inhibit the ability of topoisomerase II to ligate enzyme-linked DNA breaks. Conversely, compounds such as quinolones have little effect on ligation and are believed to stimulate the forward rate of topoisomerase II-mediated DNA cleavage. The fact that there are two scissile bonds per double-stranded DNA break implies that there are two sites for drug action in every enzyme-DNA cleavage complex. However, since agents in the latter group are believed to act by locally perturbing DNA structure, it is possible that quinolone interactions at a single scissile bond are sufficient to distort both strands of the double helix and generate an enzyme-mediated double-stranded DNA break. Therefore, an oligonucleotide system was established to further define the actions of topoisomerase II-targeted drugs that stimulate the forward rate of DNA cleavage. Results indicate that the presence of the quinolone CP-115,953 at one scissile bond increased the extent of enzyme-mediated scission at the opposite scissile bond and was sufficient to stimulate the formation of a double-stranded DNA break by human topoisomerase IIalpha. These findings stand in marked contrast to those for etoposide, which must be present at both scissile bonds to stabilize a double-stranded DNA break [Bromberg, K. D., et al. (2003) J. Biol. Chem. 278, 7406-7412]. Moreover, they underscore important mechanistic differences between drugs that enhance DNA cleavage and those that inhibit ligation.


Asunto(s)
Antiinfecciosos/farmacología , ADN-Topoisomerasas de Tipo II/metabolismo , ADN/metabolismo , Fluoroquinolonas/farmacología , Sustitución de Aminoácidos , Antígenos de Neoplasias , Antineoplásicos/farmacología , Secuencia de Bases , Sitios de Unión , Dominio Catalítico/genética , ADN/química , ADN/genética , Daño del ADN , Reparación del ADN , ADN-Topoisomerasas de Tipo II/genética , Proteínas de Unión al ADN , Activación Enzimática/efectos de los fármacos , Etopósido/farmacología , Humanos , Técnicas In Vitro , Cinética , Modelos Biológicos , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/genética , Oligodesoxirribonucleótidos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
20.
J Biol Chem ; 278(9): 7406-12, 2003 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-12473657

RESUMEN

The widely used anticancer drug etoposide kills cells by increasing levels of topoisomerase II-mediated DNA breaks. While it is known that the drug acts by inhibiting the ability of topoisomerase II to ligate cleaved DNA molecules, the precise mechanism by which it accomplishes this action is not well understood. Because there are two scissile bonds per enzyme-mediated double-stranded DNA break, it has been assumed that there are two sites for etoposide in every cleavage complex. However, it is not known whether the action of etoposide at only one scissile bond is sufficient to stabilize a double-stranded DNA break or whether both drug sites need to be occupied. An oligonucleotide system was utilized to address this important issue. Results of DNA cleavage and ligation assays support a two-drug model for the action of etoposide against human topoisomerase IIalpha. This model postulates that drug interactions at both scissile bonds are required in order to increase enzyme-mediated double-stranded DNA breaks. Etoposide actions at either of the two scissile bonds appear to be independent of one another, with each individual drug molecule stabilizing a strand-specific nick rather than a double-stranded DNA break. This finding suggests (at least in the presence of drug) that there is little or no communication between the two promoter active sites of topoisomerase II. The two-drug model has implications for cancer chemotherapy, the cellular processing of etoposide-stabilized enzyme-DNA cleavage complexes, and the catalytic mechanism of eukaryotic topoisomerase II.


Asunto(s)
ADN/efectos de los fármacos , Etopósido/farmacología , Inhibidores de Topoisomerasa II , Antígenos de Neoplasias , Sitios de Unión , ADN/metabolismo , ADN-Topoisomerasas de Tipo II , Proteínas de Unión al ADN , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Modelos Biológicos , Oligonucleótidos/farmacología , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...