Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Chembiochem ; 21(20): 2916-2920, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32501609

RESUMEN

Novel chemical biology probes linking a serine hydrolase-directed fluorophosphonate warhead and cereblon-binding pomalidomide were assessed for the degradation of serine hydrolases. A quantitative proteomics approach to detect degraded proteins revealed that, despite the engagement of ∼40 serine hydrolases, degradation was achieved for only a single serine hydrolase, lysophospholipase II (LYPLA2).


Asunto(s)
Colorantes Fluorescentes/química , Hidrolasas/análisis , Fosfatos/química , Proteómica , Serina/análisis , Talidomida/análogos & derivados , Colorantes Fluorescentes/metabolismo , Hidrolasas/metabolismo , Estructura Molecular , Fosfatos/metabolismo , Serina/metabolismo , Talidomida/química , Talidomida/metabolismo
2.
Proc Natl Acad Sci U S A ; 111(37): E3929-36, 2014 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-25197063

RESUMEN

The repressor element 1 (RE1) silencing transcription factor (REST) in stem cells represses hundreds of genes essential to neuronal function. During neurogenesis, REST is degraded in neural progenitors to promote subsequent elaboration of a mature neuronal phenotype. Prior studies indicate that part of the degradation mechanism involves phosphorylation of two sites in the C terminus of REST that require activity of beta-transducin repeat containing E3 ubiquitin protein ligase, ßTrCP. We identify a proline-directed phosphorylation motif, at serines 861/864 upstream of these sites, which is a substrate for the peptidylprolyl cis/trans isomerase, Pin1, as well as the ERK1/2 kinases. Mutation at S861/864 stabilizes REST, as does inhibition of Pin1 activity. Interestingly, we find that C-terminal domain small phosphatase 1 (CTDSP1), which is recruited by REST to neuronal genes, is present in REST immunocomplexes, dephosphorylates S861/864, and stabilizes REST. Expression of a REST peptide containing S861/864 in neural progenitors inhibits terminal neuronal differentiation. Together with previous work indicating that both REST and CTDSP1 are expressed to high levels in stem cells and down-regulated during neurogenesis, our results suggest that CTDSP1 activity stabilizes REST in stem cells and that ERK-dependent phosphorylation combined with Pin1 activity promotes REST degradation in neural progenitors.


Asunto(s)
Diferenciación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Represoras/metabolismo , Animales , Pollos , Cromatina/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Mutación/genética , Peptidilprolil Isomerasa de Interacción con NIMA , Células PC12 , Péptidos/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Estabilidad Proteica , Proteolisis , Ratas , Proteínas con Repetición de beta-Transducina/metabolismo , Proteínas ras/metabolismo
3.
Toxicol Sci ; 194(1): 53-69, 2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37228089

RESUMEN

The degradation tag (dTAG) system for target protein degradation can remove proteins from biological systems without the drawbacks of some genetic methods, such as slow kinetics, lack of reversibility, low specificity, and the inability to titrate dosage. These drawbacks can make it difficult to compare toxicity resulting from genetic and pharmacological interventions, especially in vivo. Because the dTAG system has not been studied extensively in vivo, we explored the use of this system to study the physiological sequalae resulting from CDK2 or CDK5 degradation in adult mice. Mice with homozygous knock-in of the dTAG sequence onto CDK2 and CDK5 were born at Mendelian ratios despite decreased CDK2 or CDK5 protein levels in comparison with wild-type mice. In bone marrow cells and duodenum organoids derived from these mice, treatment with the dTAG degrader dTAG-13 resulted in rapid and robust protein degradation but caused no appreciable change in viability or the transcriptome. Repeated delivery of dTAG-13 in vivo for toxicity studies proved challenging; we explored multiple formulations in an effort to maximize degradation while minimizing formulation-related toxicity. Degradation of CDK2 or CDK5 in all organs except the brain, where dTAG-13 likely did not cross the blood brain barrier, only caused microscopic changes in the testis of CDK2dTAG mice. These findings were corroborated with conditional CDK2 knockout in adult mice. Our results suggest that the dTAG system can provide robust protein degradation in vivo and that loss of CDK2 or CDK5 in adult mice causes no previously unknown phenotypes.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina , Proteínas , Masculino , Ratones , Animales , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Proteínas/metabolismo , Proteolisis
4.
Sci Rep ; 12(1): 12501, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35864202

RESUMEN

The synthetic lethal association between BRCA deficiency and poly (ADP-ribose) polymerase (PARP) inhibition supports PARP inhibitor (PARPi) clinical efficacy in BRCA-mutated tumors. PARPis also demonstrate activity in non-BRCA mutated tumors presumably through induction of PARP1-DNA trapping. Despite pronounced clinical response, therapeutic resistance to PARPis inevitably develops. An abundance of knowledge has been built around resistance mechanisms in BRCA-mutated tumors, however, parallel understanding in non-BRCA mutated settings remains insufficient. In this study, we find a strong correlation between the epithelial-mesenchymal transition (EMT) signature and resistance to a clinical PARPi, Talazoparib, in non-BRCA mutated tumor cells. Genetic profiling demonstrates that SNAI2, a master EMT transcription factor, is transcriptionally induced by Talazoparib treatment or PARP1 depletion and this induction is partially responsible for the emerging resistance. Mechanistically, we find that the PARP1 protein directly binds to SNAI2 gene promoter and suppresses its transcription. Talazoparib treatment or PARP1 depletion lifts PARP1-mediated suppression and increases chromatin accessibility around SNAI2 promoters, thus driving SNAI2 transcription and drug resistance. We also find that depletion of the chromatin remodeler CHD1L suppresses SNAI2 expression and reverts acquired resistance to Talazoparib. The PARP1/CHD1L/SNAI2 transcription axis might be therapeutically targeted to re-sensitize Talazoparib in non-BRCA mutated tumors.


Asunto(s)
Antineoplásicos , Neoplasias , Antineoplásicos/farmacología , Cromatina , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Humanos , Neoplasias/genética , Ftalazinas/farmacología , Ftalazinas/uso terapéutico , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Poli(ADP-Ribosa) Polimerasas/genética , Factores de Transcripción de la Familia Snail/genética
5.
Cancer Discov ; 12(1): 186-203, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34417224

RESUMEN

Mutations in epigenetic regulators are common in relapsed pediatric acute lymphoblastic leukemia (ALL). Here, we uncovered the mechanism underlying the relapse of ALL driven by an activating mutation of the NSD2 histone methyltransferase (p.E1099K). Using high-throughput drug screening, we found that NSD2-mutant cells were specifically resistant to glucocorticoids. Correction of this mutation restored glucocorticoid sensitivity. The transcriptional response to glucocorticoids was blocked in NSD2-mutant cells due to depressed glucocorticoid receptor (GR) levels and the failure of glucocorticoids to autoactivate GR expression. Although H3K27me3 was globally decreased by NSD2 p.E1099K, H3K27me3 accumulated at the NR3C1 (GR) promoter. Pretreatment of NSD2 p.E1099K cell lines and patient-derived xenograft samples with PRC2 inhibitors reversed glucocorticoid resistance in vitro and in vivo. PRC2 inhibitors restored NR3C1 autoactivation by glucocorticoids, increasing GR levels and allowing GR binding and activation of proapoptotic genes. These findings suggest a new therapeutic approach to relapsed ALL associated with NSD2 mutation. SIGNIFICANCE: NSD2 histone methyltransferase mutations observed in relapsed pediatric ALL drove glucocorticoid resistance by repression of the GR and abrogation of GR gene autoactivation due to accumulation of K3K27me3 at its promoter. Pretreatment with PRC2 inhibitors reversed resistance, suggesting a new therapeutic approach to these patients with ALL.This article is highlighted in the In This Issue feature, p. 1.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Glucocorticoides/uso terapéutico , Histona Metiltransferasas/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Proteínas Represoras/genética , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular , Niño , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/farmacología , Femenino , Glucocorticoides/farmacología , Humanos , Masculino , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología
6.
Mutat Res ; 706(1-2): 21-7, 2011 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-21035468

RESUMEN

A hallmark of aberrant DNA methylation-associated silencing is reversibility. However, long-term stability of reactivated promoters has not been explored. To examine this issue, spontaneous reactivant clones were isolated from mouse embryonal carcinoma cells bearing aberrantly silenced Aprt alleles and re-silencing frequencies were determined as long as three months after reactivation occurred. Despite continuous selection for expression of the reactivated Aprt alleles, exceptionally high spontaneous re-silencing frequencies were observed. A DNA methylation analysis demonstrated retention of sporadic methylation of CpG sites in a protected region of the Aprt promoter in many reactivant alleles suggesting a role for these methylated sites in the re-silencing process. In contrast, a chromatin immunoprecipitation (ChIP) analysis for methyl-H3K4, acetyl-H3K9, and dimethyl-H3K9 levels failed to reveal a specific histone modification that could explain high frequency re-silencing. These results demonstrate that aberrantly silenced and reactivated promoters retain a persistent memory of having undergone the silencing process and suggest the failure to eliminate all CpG methylation as a potential contributing mechanism.


Asunto(s)
Adenina Fosforribosiltransferasa/genética , Islas de CpG/genética , Metilación de ADN , Regiones Promotoras Genéticas/genética , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Metilasas de Modificación del ADN/antagonistas & inhibidores , Decitabina , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Ácidos Hidroxámicos/farmacología , Lisina/metabolismo , Metilación , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
7.
Oncogene ; 38(5): 671-686, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30171259

RESUMEN

NSD2, a histone methyltransferase specific for methylation of histone 3 lysine 36 (H3K36), exhibits a glutamic acid to lysine mutation at residue 1099 (E1099K) in childhood acute lymphocytic leukemia (ALL), and cells harboring this mutation can become the predominant clone in relapsing disease. We studied the effects of this mutant enzyme in silico, in vitro, and in vivo using gene edited cell lines. The E1099K mutation altered enzyme/substrate binding and enhanced the rate of H3K36 methylation. As a result, cell lines harboring E1099K exhibit increased H3K36 dimethylation and reduced H3K27 trimethylation, particularly on nucleosomes containing histone H3.1. Mutant NSD2 cells exhibit reduced apoptosis and enhanced proliferation, clonogenicity, adhesion, and migration. In mouse xenografts, mutant NSD2 cells are more lethal and brain invasive than wildtype cells. Transcriptional profiling demonstrates that mutant NSD2 aberrantly activates factors commonly associated with neural and stromal lineages in addition to signaling and adhesion genes. Identification of these pathways provides new avenues for therapeutic interventions in NSD2 dysregulated malignancies.


Asunto(s)
Reprogramación Celular , N-Metiltransferasa de Histona-Lisina , Mutación Missense , Proteínas de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras , Proteínas Represoras , Sustitución de Aminoácidos , Células HeLa , Xenoinjertos , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
8.
Cancer Discov ; 9(10): 1438-1451, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31337617

RESUMEN

By examination of the cancer genomics database, we identified a new set of mutations in core histones that frequently recur in cancer patient samples and are predicted to disrupt nucleosome stability. In support of this idea, we characterized a glutamate to lysine mutation of histone H2B at amino acid 76 (H2B-E76K), found particularly in bladder and head and neck cancers, that disrupts the interaction between H2B and H4. Although H2B-E76K forms dimers with H2A, it does not form stable histone octamers with H3 and H4 in vitro, and when reconstituted with DNA forms unstable nucleosomes with increased sensitivity to nuclease. Expression of the equivalent H2B mutant in yeast restricted growth at high temperature and led to defective nucleosome-mediated gene repression. Significantly, H2B-E76K expression in the normal mammary epithelial cell line MCF10A increased cellular proliferation, cooperated with mutant PIK3CA to promote colony formation, and caused a significant drift in gene expression and fundamental changes in chromatin accessibility, particularly at gene regulatory elements. Taken together, these data demonstrate that mutations in the globular domains of core histones may give rise to an oncogenic program due to nucleosome dysfunction and deregulation of gene expression. SIGNIFICANCE: Mutations in the core histones frequently occur in cancer and represent a new mechanism of epigenetic dysfunction that involves destabilization of the nucleosome, deregulation of chromatin accessibility, and alteration of gene expression to drive cellular transformation.See related commentary by Sarthy and Henikoff, p. 1346.This article is highlighted in the In This Issue feature, p. 1325.


Asunto(s)
Histonas/genética , Mutación , Neoplasias/genética , Oncogenes , Alelos , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Histonas/química , Histonas/metabolismo , Humanos , Mutación Missense , Neoplasias/metabolismo , Nucleosomas/metabolismo , Multimerización de Proteína , Levaduras/genética , Levaduras/metabolismo
9.
J Med Chem ; 61(19): 8504-8535, 2018 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-29718665

RESUMEN

Small-molecule (SM) leads in the early drug discovery pipeline are progressed primarily based on potency against the intended target(s) and selectivity against a very narrow slice of the proteome. So, why is there a tendency to wait until SMs are matured before probing for a deeper mechanistic understanding? For one, there is a concern about the interpretation of complex -omic data outputs and the resources needed to test these hypotheses. However, with recent advances in broad endpoint profiling assays that have companion reference databases and refined technology integration strategies, we argue that data complexity can translate into meaningful decision-making. This same strategy can also prioritize phenotypic screening hits to increase the likelihood of accessing unprecedented target space. In this Perspective. we will highlight a cohesive process that supports SM hit prosecution, providing a data-driven rationale and a suite of methods for direct identification of SM targets driving relevant biological end points.


Asunto(s)
Descubrimiento de Drogas , Proteoma/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Ensayos Analíticos de Alto Rendimiento , Humanos
10.
Cell Rep ; 21(3): 628-640, 2017 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-29045832

RESUMEN

Loss or inactivation of the histone H3K27 demethylase UTX occurs in several malignancies, including multiple myeloma (MM). Using an isogenic cell system, we found that loss of UTX leads to deactivation of gene expression ultimately promoting the proliferation, clonogenicity, adhesion, and tumorigenicity of MM cells. Moreover, UTX mutant cells showed increased in vitro and in vivo sensitivity to inhibition of EZH2, a histone methyltransferase that generates H3K27me3. Such sensitivity was related to a decrease in the levels of IRF4 and c-MYC and an activation of repressors of IRF4 characteristic of germinal center B cells such as BCL6 and IRF1. Rebalance of H3K27me3 levels at specific genes through EZH2 inhibitors may be a therapeutic strategy in MM cases harboring UTX mutations.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Histona Demetilasas/deficiencia , Mieloma Múltiple/patología , Proteínas Nucleares/deficiencia , Animales , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Carcinogénesis/patología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Desdiferenciación Celular/efectos de los fármacos , Desdiferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Clonales , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histona Demetilasas/metabolismo , Histonas/metabolismo , Indazoles/farmacología , Factores Reguladores del Interferón/metabolismo , Lisina/metabolismo , Metilación , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mieloma Múltiple/genética , Mutación/genética , Proteínas Nucleares/metabolismo , Fenotipo , Piridonas/farmacología , Transcripción Genética/efectos de los fármacos
11.
Elife ; 3: e04235, 2014 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-25250711

RESUMEN

The bivalent hypothesis posits that genes encoding developmental regulators required for early lineage decisions are poised in stem/progenitor cells by the balance between a repressor histone modification (H3K27me3), mediated by the Polycomb Repressor Complex 2 (PRC2), and an activator modification (H3K4me3). In this study, we test whether this mechanism applies equally to genes that are not required until terminal differentiation. We focus on the RE1 Silencing Transcription Factor (REST) because it is expressed highly in stem cells and is an established global repressor of terminal neuronal genes. Elucidation of the REST complex, and comparison of chromatin marks and gene expression levels in control and REST-deficient stem cells, shows that REST target genes are poised by a mechanism independent of Polycomb, even at promoters which bear the H3K27me3 mark. Specifically, genes under REST control are actively repressed in stem cells by a balance of the H3K4me3 mark and a repressor complex that relies on histone deacetylase activity. Thus, chromatin distinctions between pro-neural and terminal neuronal genes are established at the embryonic stem cell stage by two parallel, but distinct, repressor pathways.


Asunto(s)
Diferenciación Celular , Histona Desacetilasas/metabolismo , Neuronas/citología , Neuronas/enzimología , Complejo Represivo Polycomb 2/metabolismo , Proteínas Represoras/metabolismo , Animales , Cromatina/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Lisina/metabolismo , Metilación , Ratones , Fenotipo , Regiones Promotoras Genéticas
12.
J Am Coll Cardiol ; 60(7): 618-25, 2012 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-22703929

RESUMEN

OBJECTIVES: This study sought to evaluate the contribution of microvascular functional rarefaction and changes in vascular mechanical properties to the development of hypertension and secondary ventricular remodeling that occurs with anti-vascular endothelial growth factor (VEGF) therapy. BACKGROUND: Hypertension is a common side effect of VEGF inhibitors used in cancer medicine. METHODS: Mice were treated for 5 weeks with an anti-murine VEGF-A monoclonal antibody, antibody plus ramipril, or sham treatment. Microvascular blood flow (MBF) and blood volume (MBV) were quantified by contrast-enhanced ultrasound in skeletal muscle, left ventricle (LV), and kidney. Echocardiography and invasive hemodynamics were used to assess ventricular function, dimensions and vascular mechanical properties. RESULTS: Ambulatory blood pressure increased gradually over the first 3 weeks of anti-VEGF therapy. Compared with controls, anti-VEGF-treated mice had similar aortic elastic modulus and histological appearance, but a marked increase in arterial elastance, indicating increased afterload, and elevated plasma angiotensin II. Increased afterload in treated mice led to concentric LV remodeling and reduced stroke volume without impaired LV contractility determined by LV peak change in pressure over time (dp/dt) and the end-systolic dimension-pressure relation. Anti-VEGF therapy did not alter MBF or MBV in skeletal muscle, myocardium, or kidney; but did produce cortical mesangial glomerulosclerosis. Ramipril therapy almost entirely prevented the adverse hemodynamic effects, increased afterload, and LV remodeling in anti-VEGF-treated mice. CONCLUSIONS: Neither reduced functional microvascular density nor major alterations in arterial mechanical properties are primary causes of hypertension during anti-VEGF therapy. Inhibition of VEGF leads to an afterload mismatch state, increased angiotensin II, and LV remodeling, which are all ameliorated by angiotensin-converting enzyme inhibition.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Hipertensión/inducido químicamente , Microcirculación/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Aorta/patología , Ecocardiografía , Hemodinámica/efectos de los fármacos , Hipertensión/diagnóstico por imagen , Hipertensión/patología , Riñón/efectos de los fármacos , Riñón/patología , Ratones , Ratones Endogámicos C57BL , Ramipril/administración & dosificación , Circulación Renal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/inmunología
13.
PLoS One ; 4(3): e4832, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19279688

RESUMEN

BACKGROUND: Aberrant epigenetic silencing plays a major role in cancer formation by inactivating tumor suppressor genes. While the endpoints of aberrant silencing are known, i.e., promoter region DNA methylation and altered histone modifications, the triggers of silencing are not known. We used the tet-off system to test the hypothesis that a transient reduction in gene expression will sensitize a promoter to undergo epigenetic silencing. METHODOLOGY/PRINCIPAL FINDINGS: The tet responsive promoter (P(TRE)) was used to drive expression of the selectable human HPRT cDNA in independent transfectants of an Hprt deficient mouse cell line. In this system, high basal HPRT expression is greatly reduced when doxycycline (Dox) is added to the culture medium. Exposure of the P(TRE)-HPRT transfectants to Dox induced HPRT deficient clones in a time dependent manner. A molecular analysis demonstrated promoter region DNA methylation, loss of histone modifications associated with expression (i.e., H3 lysine 9 and 14 acetylation and lysine 4 methylation), and acquisition of the repressive histone modification H3 lysine 9 methylation. These changes, which are consistent with aberrant epigenetic silencing, were not present in the Dox-treated cultures, with the exception of reduced H3 lysine 14 acetylation. Silenced alleles readily reactivated spontaneously or after treatment of cells with inhibitors of histone deacetylation and/or DNA methylation, but re-silencing of reactivated alleles did not require a new round of Dox exposure. Inhibition of histone deacetylation inhibited both the induction of silencing and re-silencing, whereas inhibition of DNA methylation had no such effect. CONCLUSIONS/SIGNIFICANCE: This study demonstrates that a transient reduction in gene expression triggers a pathway for aberrant silencing in mammalian cells and identifies histone deacetylation as a critical early step in this process. DNA methylation, in contrast, is a secondary step in the silencing pathway under study. A model to explain these observations is offered.


Asunto(s)
Epigénesis Genética , Regulación de la Expresión Génica , Silenciador del Gen , Acetilación , Alelos , Animales , Medios de Cultivo , Cartilla de ADN , Doxiciclina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hipoxantina Fosforribosiltransferasa/genética , Ratones , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA