Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
PLoS One ; 13(3): e0194423, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29554128

RESUMEN

Autophagy is a cellular mechanism that can generate energy for cells or clear misfolded or aggregated proteins, and upregulating this process has been proposed as a therapeutic approach for neurodegenerative diseases. Here we describe a novel set of LC3B-II and p62 time-resolved fluorescence resonance energy transfer (TR-FRET) assays that can detect changes in autophagy in the absence of exogenous labels. Lipidated LC3 is a marker of autophagosomes, while p62 is a substrate of autophagy. These assays can be employed in high-throughput screens to identify novel autophagy upregulators, and can measure autophagy changes in cultured cells or tissues after genetic or pharmacological interventions. We also demonstrate that different cells exhibit varying autophagic responses to pharmacological interventions. Overall, it is clear that a battery of readouts is required to make conclusions about changes in autophagy.


Asunto(s)
Autofagosomas/metabolismo , Autofagia/fisiología , Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Sequestosoma-1/metabolismo , Animales , Células HEK293 , Humanos , Ratas
2.
Proc Natl Acad Sci U S A ; 114(50): E10809-E10818, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29162692

RESUMEN

Posttranslational modifications can have profound effects on the biological and biophysical properties of proteins associated with misfolding and aggregation. However, their detection and quantification in clinical samples and an understanding of the mechanisms underlying the pathological properties of misfolding- and aggregation-prone proteins remain a challenge for diagnostics and therapeutics development. We have applied an ultrasensitive immunoassay platform to develop and validate a quantitative assay for detecting a posttranslational modification (phosphorylation at residue T3) of a protein associated with polyglutamine repeat expansion, namely Huntingtin, and characterized its presence in a variety of preclinical and clinical samples. We find that T3 phosphorylation is greatly reduced in samples from Huntington's disease models and in Huntington's disease patients, and we provide evidence that bona-fide T3 phosphorylation alters Huntingtin exon 1 protein conformation and aggregation properties. These findings have significant implications for both mechanisms of disease pathogenesis and the development of therapeutics and diagnostics for Huntington's disease.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Inmunoensayo/métodos , Animales , Células Cultivadas , Exones , Células HEK293 , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/genética , Ratones , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fosforilación , Conformación Proteica , Procesamiento Proteico-Postraduccional , Sensibilidad y Especificidad
3.
J Huntingtons Dis ; 6(4): 349-361, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29125493

RESUMEN

BACKGROUND: The measurement of disease-relevant biomarkers has become a major component of clinical trial design, but in the absence of rigorous clinical and analytical validation of detection methodology, interpretation of results may be misleading. In Huntington's disease (HD), measurement of the concentration of mutant huntingtin protein (mHTT) in cerebrospinal fluid (CSF) of patients may serve as both a disease progression biomarker and a pharmacodynamic readout for HTT-lowering therapeutic approaches. We recently published the quantification of mHTT levels in HD patient CSF by a novel ultrasensitive immunoassay-based technology and here analytically validate it for use. OBJECTIVE: This work aims to analytically and clinically validate our ultrasensitive assay for mHTT measurement in human HD CSF, for application as a pharmacodynamic biomarker of CNS mHTT lowering in clinical trials. METHODS: The single molecule counting (SMC) assay is an ultrasensitive bead-based immunoassay where upon specific recognition, dye-labeled antibodies are excited by a confocal laser and emit fluorescent light as a readout. The detection of mHTT by this technology was clinically validated following established Food and Drug Administration and European Medicine Agency guidelines. RESULTS: The SMC assay was demonstrated to be accurate, precise, specific, and reproducible. While no matrix influence was detected, a list of interfering substances was compiled as a guideline for proper collection and storage of patient CSF samples. In addition, a set of recommendations on result interpretation is provided. CONCLUSIONS: This SMC assay is a robust and ultrasensitive method for the relative quantification of mHTT in human CSF.


Asunto(s)
Proteína Huntingtina/líquido cefalorraquídeo , Enfermedad de Huntington/líquido cefalorraquídeo , Inmunoensayo/métodos , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Calibración , Fibroblastos/metabolismo , Silenciador del Gen , Humanos , Proteína Huntingtina/sangre , Proteína Huntingtina/genética , Enfermedad de Huntington/sangre , Enfermedad de Huntington/genética , Inmunoensayo/instrumentación , ARN Interferente Pequeño , Proteínas Recombinantes/sangre , Proteínas Recombinantes/líquido cefalorraquídeo , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Expansión de Repetición de Trinucleótido
4.
Eur J Med Chem ; 108: 53-67, 2016 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-26629860

RESUMEN

In the last decades, inhibitors of histone deacetylases (HDAC) have become an important class of anti-cancer agents. In a previous study we described the synthesis of spiro[chromane-2,4'-piperidine]hydroxamic acid derivatives able to inhibit histone deacetylase enzymes. Herein, we present our exploration for new derivatives by replacing the piperidine moiety with various cycloamines. The goal was to obtain highly potent compounds with a good in vitro ADME profile. In addition, molecular modeling studies unravelled the binding mode of these inhibitors.


Asunto(s)
Cromonas/farmacología , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Modelos Moleculares , Compuestos de Espiro/farmacología , Cromonas/síntesis química , Cromonas/química , Relación Dosis-Respuesta a Droga , Inhibidores de Histona Desacetilasas/química , Humanos , Estructura Molecular , Compuestos de Espiro/síntesis química , Compuestos de Espiro/química , Relación Estructura-Actividad
5.
J Clin Invest ; 125(5): 1979-86, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25844897

RESUMEN

BACKGROUND: Quantification of disease-associated proteins in the cerebrospinal fluid (CSF) has been critical for the study and treatment of several neurodegenerative disorders; however, mutant huntingtin protein (mHTT), the cause of Huntington's disease (HD), is at very low levels in CSF and, to our knowledge, has never been measured previously. METHODS: We developed an ultrasensitive single-molecule counting (SMC) mHTT immunoassay that was used to quantify mHTT levels in CSF samples from individuals bearing the HD mutation and from control individuals in 2 independent cohorts. RESULTS: This SMC mHTT immunoassay demonstrated high specificity for mHTT, high sensitivity with a femtomolar detection threshold, and a broad dynamic range. Analysis of the CSF samples showed that mHTT was undetectable in CSF from all controls but quantifiable in nearly all mutation carriers. The mHTT concentration in CSF was approximately 3-fold higher in patients with manifest HD than in premanifest mutation carriers. Moreover, mHTT levels increased as the disease progressed and were associated with 5-year onset probability. The mHTT concentration independently predicted cognitive and motor dysfunction. Furthermore, the level of mHTT was associated with the concentrations of tau and neurofilament light chain in the CSF, suggesting a neuronal origin for the detected mHTT. CONCLUSIONS: We have demonstrated that mHTT can be quantified in CSF from HD patients using the described SMC mHTT immunoassay. Moreover, the level of mHTT detected is associated with proximity to disease onset and diminished cognitive and motor function. The ability to quantify CSF mHTT will facilitate the study of HD, and mHTT quantification could potentially serve as a biomarker for the development and testing of experimental mHTT-lowering therapies for HD. TRIAL REGISTRATION: Not applicable. FUNDING: CHDI Foundation Inc.; Medical Research Council (MRC) UK; National Institutes for Health Research (NIHR); Rosetrees Trust; Swedish Research Council; and Knut and Alice Wallenberg Foundation.


Asunto(s)
Enfermedad de Huntington/líquido cefalorraquídeo , Inmunoensayo/métodos , Proteínas del Tejido Nervioso/líquido cefalorraquídeo , Adulto , Edad de Inicio , Anciano , Colombia Británica , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Tamización de Portadores Genéticos , Humanos , Proteína Huntingtina , Londres , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Proteínas de Neurofilamentos/líquido cefalorraquídeo , Proteínas Recombinantes de Fusión/metabolismo , Sensibilidad y Especificidad , Método Simple Ciego , Expansión de Repetición de Trinucleótido , Proteínas tau/líquido cefalorraquídeo
6.
PLoS One ; 9(12): e112262, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25464275

RESUMEN

BACKGROUND: In Huntington's disease, expansion of a CAG triplet repeat occurs in exon 1 of the huntingtin gene (HTT), resulting in a protein bearing>35 polyglutamine residues whose N-terminal fragments display a high propensity to misfold and aggregate. Recent data demonstrate that polyglutamine expansion results in conformational changes in the huntingtin protein (HTT), which likely influence its biological and biophysical properties. Developing assays to characterize and measure these conformational changes in isolated proteins and biological samples would advance the testing of novel therapeutic approaches aimed at correcting mutant HTT misfolding. Time-resolved Förster energy transfer (TR-FRET)-based assays represent high-throughput, homogeneous, sensitive immunoassays widely employed for the quantification of proteins of interest. TR-FRET is extremely sensitive to small distances and can therefore provide conformational information based on detection of exposure and relative position of epitopes present on the target protein as recognized by selective antibodies. We have previously reported TR-FRET assays to quantify HTT proteins based on the use of antibodies specific for different amino-terminal HTT epitopes. Here, we investigate the possibility of interrogating HTT protein conformation using these assays. METHODOLOGY/PRINCIPAL FINDINGS: By performing TR-FRET measurements on the same samples (purified recombinant proteins or lysates from cells expressing HTT fragments or full length protein) at different temperatures, we have discovered a temperature-dependent, reversible, polyglutamine-dependent conformational change of wild type and expanded mutant HTT proteins. Circular dichroism spectroscopy confirms the temperature and polyglutamine-dependent change in HTT structure, revealing an effect of polyglutamine length and of temperature on the alpha-helical content of the protein. CONCLUSIONS/SIGNIFICANCE: The temperature- and polyglutamine-dependent effects observed with TR-FRET on HTT proteins represent a simple, scalable, quantitative and sensitive assay to identify genetic and pharmacological modulators of mutant HTT conformation, and potentially to assess the relevance of conformational changes during onset and progression of Huntington's disease.


Asunto(s)
Proteínas Mutantes/química , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Péptidos/química , Aminoácidos/química , Dicroismo Circular , Progresión de la Enfermedad , Epítopos/química , Exones , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Proteína Huntingtina , Inmunoensayo , Mutación , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Temperatura , Tiorredoxinas/química
7.
ChemMedChem ; 9(7): 1574-85, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24753447

RESUMEN

Heat-shock protein 90 (Hsp90) is a molecular chaperone involved in the stabilization of key oncogenic signaling proteins, and therefore, inhibition of Hsp90 represents a new strategy in cancer therapy. 2-Amino-7-[4-fluoro-2-(3-pyridyl)phenyl]-4-methyl-7,8-dihydro-6H-quinazolin-5-one oxime is a racemic Hsp90 inhibitor that targets the N-terminal adenosine triphosphatase site. We developed a method to resolve the enantiomers and evaluated their inhibitory activity on Hsp90 and the consequent antitumor effects. The (S) stereoisomer emerged as a potent Hsp90 inhibitor in biochemical and cellular assays. In addition, this enantiomer exhibited high oral bioavailability in mice and excellent antitumor activity in two different human cancer xenograft models.


Asunto(s)
Antineoplásicos/química , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Oximas/química , Quinazolinonas/química , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Sitios de Unión , Línea Celular Tumoral , Femenino , Células HCT116 , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Ratones , Ratones Desnudos , Microsomas/metabolismo , Simulación del Acoplamiento Molecular , Neoplasias/tratamiento farmacológico , Oximas/farmacología , Oximas/uso terapéutico , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Estereoisomerismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Eur J Med Chem ; 64: 273-84, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23644210

RESUMEN

Histone Deacetylases (HDACs) have become important targets for the treatment of cancer and other diseases. In previous studies we described the development of novel spirocyclic HDAC inhibitors based on the combination of privileged structures with hydroxamic acid moieties as zinc binding group. Herein, we report further explorations, which resulted in the discovery of a new class of spiro[2H-(1,3)-benzoxazine-2,4'-piperidine] derivatives. Several compounds showed good potency of around 100 nM and less in the HDAC inhibition assays, submicromolar IC50 values when tested against tumour cell lines and a remarkable stability in human and mouse microsomes. Two representative examples exhibited a good pharmacokinetic profile with an oral bioavailability equal or higher than 35% and one of them studied in an HCT116 murine xenograft model showing a robust tumour growth inhibition. In addition, the two benzoxazines were found to have a minor affinity for the hERG potassium channel compared to their corresponding ketone analogues.


Asunto(s)
Antineoplásicos/farmacología , Benzoxazinas/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Compuestos de Espiro/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Benzoxazinas/síntesis química , Benzoxazinas/química , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HCT116 , Células HeLa , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/química , Humanos , Células K562 , Ratones , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Estructura Molecular , Neoplasias Experimentales/patología , Compuestos de Espiro/síntesis química , Compuestos de Espiro/química , Relación Estructura-Actividad
9.
ChemMedChem ; 8(5): 800-11, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23526814

RESUMEN

Herein we report novel pyrrole- and benzene-based hydroxamates (8, 10) and 2'-aminoanilides (9, 11) bearing the tert-butylcarbamate group at the CAP moiety as histone deacetylase (HDAC) inhibitors. Compounds 8 b and 10 c selectively inhibited HDAC6 at the nanomolar level, whereas the other hydroxamates effected an increase in acetyl-α-tubulin levels in human acute myeloid leukemia U937 cells. In the same cell line, compounds 8 b and 10 c elicited 18.4 and 21.4 % apoptosis, respectively (SAHA: 16.9 %), and the pyrrole anilide 9 c displayed the highest cytodifferentiating effect (90.9 %). In tests against a wide range of various cancer cell lines to determine its antiproliferative effects, compound 10 c exhibited growth inhibition from sub-micromolar (neuroblastoma LAN-5 and SH-SY5Y cells, chronic myeloid leukemia K562 cells) to low-micromolar (lung H1299 and A549, colon HCT116 and HT29 cancer cells) concentrations. In HT29 cells, 10 c increased histone H3 acetylation, and decreased the colony-forming potential of the cancer cells by up to 60 %.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carbamatos/farmacología , Diferenciación Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Neoplasias/patología , Antineoplásicos/síntesis química , Antineoplásicos/química , Carbamatos/síntesis química , Carbamatos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HCT116 , Células HT29 , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 6 , Inhibidores de Histona Desacetilasas/síntesis química , Histona Desacetilasas/metabolismo , Humanos , Células K562 , Células MCF-7 , Modelos Moleculares , Estructura Molecular , Neoplasias/tratamiento farmacológico , Proteínas Recombinantes/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Relación Estructura-Actividad , Células U937
10.
ChemMedChem ; 7(4): 709-21, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22354629

RESUMEN

A series of spiro[chromane-2,4'-piperidine] derivatives based on a previously published lead benzyl spirocycle 1 and bearing various N-aryl and N-alkylaryl substituents on the piperidine ring were prepared as novel histone deacetylase (HDAC) inhibitors. The compounds were evaluated for their abilities to inhibit nuclear HDACs, their in vitro antiproliferative activities, and in vitro ADME profiles. Based on these activities, 4-fluorobenzyl and 2-phenylethyl spirocycles were selected for further characterization. In vivo pharmacokinetic (PK) studies showed that both compounds exhibit an overall lower clearance rate, an increased half-life, and higher AUCs after intravenous and oral administration than spiropiperidine 1 under the conditions used. The improved PK behavior of these two compounds also correlated with superior in vivo antitumor activity in an HCT-116 xenograft model.


Asunto(s)
Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Piperidinas/química , Administración Oral , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Proteínas Sanguíneas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos del Citocromo P-450 , Evaluación Preclínica de Medicamentos , Semivida , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/farmacocinética , Inyecciones Intravenosas , Tasa de Depuración Metabólica , Ratones , Ratones Desnudos , Estructura Molecular , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Med Chem ; 54(8): 3051-64, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21417419

RESUMEN

New spiro[chromane-2,4'-piperidine] and spiro[benzofuran-2,4'-piperidine] hydroxamic acid derivatives as HDAC inhibitors have been identified by combining privileged structures with a hydroxamic acid moiety as zinc binding group. The compounds were evaluated for their ability to inhibit nuclear extract HDACs and for their in vitro antiproliferative activity on different tumor cell lines. This work resulted in the discovery of spirocycle 30d that shows good oral bioavailability and tumor growth inhibition in an HCT-116 murine xenograft model.


Asunto(s)
Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/farmacología , Administración Oral , Animales , Disponibilidad Biológica , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Inhibidores de Histona Desacetilasas/síntesis química , Humanos , Ácidos Hidroxámicos/síntesis química , Espectroscopía de Resonancia Magnética , Ratones , Estructura Molecular , Espectrometría de Masa por Ionización de Electrospray
12.
ChemMedChem ; 5(8): 1359-72, 2010 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-20572281

RESUMEN

A series of amidopropenyl hydroxamic acid derivatives were prepared as novel inhibitors of human histone deacetylases (HDACs). Several compounds showed potency at <100 nM in the HDAC inhibition assays, sub-micromolar IC(50) values in tests against three tumor cell lines, and remarkable stability in human and mouse microsomes was observed. Three representative compounds were selected for further characterization and submitted to a selectivity profile against a series of class I and class II HDACs as well as to preliminary in vivo pharmacokinetic (PK) experiments. Despite their high microsomal stability, the compounds showed medium-to-high clearance rates in in vivo PK studies as well as in rat and human hepatocytes, indicating that a major metabolic pathway is catalyzed by non-microsomal enzymes.


Asunto(s)
Antineoplásicos/síntesis química , Inhibidores de Histona Desacetilasas/síntesis química , Histona Desacetilasas/química , Ácidos Hidroxámicos/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacocinética , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/síntesis química , Ácidos Hidroxámicos/farmacología , Ratones , Microsomas Hepáticos/metabolismo , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/metabolismo , Relación Estructura-Actividad
13.
J Med Chem ; 53(2): 822-39, 2010 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-20017493

RESUMEN

The histone deacetylases (HDACs) are able to regulate gene expression, and histone deacetylase inhibitors (HDACi) emerged as a new class of agents in the treatment of cancer as well as other human disorders such as neurodegenerative diseases. In the present investigation, we report on the synthesis and biological evaluation of compounds derived from the expansion of a HDAC inhibitor scaffold having N-hydroxy-3-phenyl-2-propenamide and N-hydroxy-3-(pyridin-2-yl)-2-propenamide as core structures and containing a phenyloxopropenyl moiety, either unsubstituted or substituted by a 4-methylpiperazin-1-yl or 4-methylpiperazin-1-ylmethyl group. The compounds were evaluated for their ability to inhibit nuclear HDACs, as well as for their in vitro antiproliferative activity. Moreover, their metabolic stability in microsomes and aqueous solubility were studied and selected compounds were further characterized by in vivo pharmacokinetic experiments. These compounds showed a remarkable stability in vivo, compared to hydroxamic acid HDAC inhibitors that have already entered clinical trials. The representative compound 30b showed in vivo antitumor activity in a human colon carcinoma xenograft model.


Asunto(s)
Acrilamidas/síntesis química , Antineoplásicos/síntesis química , Inhibidores de Histona Desacetilasas/síntesis química , Acrilamidas/farmacología , Antineoplásicos/farmacocinética , Derivados del Benceno , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Estabilidad de Medicamentos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/uso terapéutico , Células HeLa , Inhibidores de Histona Desacetilasas/farmacocinética , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Piridinas , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
16.
PLoS One ; 3(7): e2704, 2008 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-18628979

RESUMEN

BACKGROUND: The Polycomb Repressive Complex 2 (PRC2) functions as a transcriptional repressor through a mechanism that involves methylation of Histone H3 at lysine 27. The PRC2 complex activity is essential for cellular proliferation, development, and cell fate decisions. PRC2 target genes include important regulators of development and proliferation as well as tumor suppressor genes. Consistent with this, the activity of several Polycomb group (PcG) proteins is deregulated in human cancer suggesting an important role for PcGs in tumor development. Whereas the downstream functions of PcGs are well characterized, the mechanisms of their recruitment to target genes and the regulation of their activity are not fully understood. PRINCIPAL FINDINGS: Here we show that the two PRC2 components SUZ12 and EZH2 are sumoylated in vitro and in vivo. Among several putative sumoylation sites we have mapped the major site of SUZ12 sumoylation. Furthermore, we show that SUZ12 interacts with the E2-conjugating enzyme UBC9 both in vitro and in vivo and that mutation of the SUZ12 sumoylation site does not abolish this binding. Finally, we provide evidence that the E3-ligase PIASXbeta interacts and enhances the sumoylation of SUZ12 in vivo suggesting that PIASXbeta could function as an E3-ligase for SUZ12. CONCLUSIONS: Taken together, our data identify sumoylation as a novel post-translational modification of components of the PRC2 complex, which could suggest a potential new mechanism to modulate PRC2 repressive activity. Further work aimed to identify the physiological conditions for these modifications will be required to understand the role of SUZ12 and EZH2 sumoylation in PcG-mediated epigenetic regulation of transcription.


Asunto(s)
Regulación de la Expresión Génica , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Proteínas Portadoras/metabolismo , Línea Celular , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Humanos , Datos de Secuencia Molecular , Proteínas de Neoplasias , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Proteínas Inhibidoras de STAT Activados/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/química , Proteína SUMO-1/metabolismo , Homología de Secuencia de Aminoácido , Factores de Transcripción/metabolismo
17.
J Biol Chem ; 282(21): 15376-82, 2007 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-17392274

RESUMEN

SUMO-1 (small ubiquitin-related modifier-1) is a ubiquitin-like family member that is conjugated to its substrates through three discrete enzymatic steps, activation (involving the E1 enzyme (SAE1/SAE2)), conjugation (involving the E2 enzyme), and substrate modification (through the cooperation of the E2 and E3 protein ligases). The adenoviral protein Gam1 inactivates E1, both in vitro and in vivo, followed by SAE1/SAE2 degradation. We have shown here that Gam1 possesses a C-terminal SOCS domain that allows its interaction with two cellular cullin RING (really interesting new gene) ubiquitin ligases. We demonstrate that Gam1 is necessary for the recruitment of SAE1/SAE2 into Cul2/5-EloB/C-Roc1 ubiquitin ligase complexes and for subsequent SAE1 ubiquitylation and degradation. The degradation of SAE2 is not tightly related to Gam1 but is a consequent effect of SAE1 disappearance. These results reveal the mechanism by which a viral protein inactivates and subsequently degrades an essential cellular enzyme, arresting a key regulatory pathway.


Asunto(s)
Adenovirus A Aviar/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Proteína SUMO-1/metabolismo , Enzimas Activadoras de Ubiquitina/metabolismo , Proteínas Virales/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Células HeLa , Humanos , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Estructura Terciaria de Proteína/genética , Proteína SUMO-1/genética , Enzimas Activadoras de Ubiquitina/genética , Proteínas Virales/genética
18.
Curr Opin Microbiol ; 9(4): 430-6, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16815735

RESUMEN

Since its discovery in 1997, SUMO (small ubiquitin-like modifier) has been implicated in a range of activities, indicating that this protein is as important in the cell as ubiquitin is. Although it can function throughout the cell, it appears to be involved more in nuclear functions. The growing list of substrates that are covalently modified by SUMO includes many viral proteins; SUMO appears to facilitate viral infection of cells, making it a possible target for antiviral therapies. It therefore is important to understand how viruses manipulate the cellular sumoylation system and how sumoylation affects viral functions.


Asunto(s)
Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/fisiología , Interferencia Viral , Proteínas Virales/fisiología , Animales , Citomegalovirus/fisiología , Humanos , FN-kappa B/fisiología , Papillomaviridae/fisiología , Simplexvirus/fisiología , Transcripción Genética , Ubiquitina/metabolismo
19.
Cell Cycle ; 4(4): 533-5, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15876861

RESUMEN

Post-translational modifications of proteins have critical roles in many cellular processes because they can cause rapid changes in the functions of preexisting proteins, multiprotein complexes and subcellular structures. Sumoylation, a ubiquitin-like dynamic and reversible post-translational modification system, is an enzymatic cascade leading to the covalent attachment of SUMO to it target proteins. This modification involves three steps and different enzymes: SUMO-activating enzyme E1 (SAE1/SAE2), SUMO-conjugating enzyme E2 (UBC9), SUMO ligases E3s, and SUMO cleaving enzymes. Although the identification of SUMO-modified substrates has progressed rapidly, the biological function of SUMO and regulation of SUMO conjugation are still not well understood. Some viral proteins have been identified as substrates for SUMO modification as well as altering the sumoylation status of host cell proteins. We have been studying an unusual adenoviral protein, Gam1, a strong and global transcriptional activator of both viral and cellular genes that inactivates HDAC1. We have recently expanded the known functions of Gam1 by demonstrating that Gam1 also inhibits the SUMO pathway by interfering with the activity of E1 heterodimer (SAE1/SAE2), leading to the accumulation of SUMO-unmodified substrates. Our data provides a clear example of the effects of a viral infection on host sumoylation and supports the idea that viruses have multifunctional protein that can target essential biochemical pathways.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación Fúngica de la Expresión Génica , Proteína SUMO-1/química , Proteínas de Saccharomyces cerevisiae/fisiología , Factores de Transcripción/fisiología , Adenosina Trifosfatasas , Adenoviridae/genética , Adenoviridae/metabolismo , Secuencias de Aminoácidos , Animales , Ciclo Celular , Dimerización , Células HeLa , Humanos , Modelos Biológicos , Unión Proteica , Procesamiento Proteico-Postraduccional , Activación Transcripcional
20.
Mol Cell ; 16(4): 549-61, 2004 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-15546615

RESUMEN

The SUMO pathway parallels the classical ubiquitinylation pathway with three discrete steps: activation involving the enzyme E1, conjugation involving the E2 enzyme UBC9, and substrate modification through the cooperative association of UBC9 and E3 ligases. We report here that the adenoviral protein Gam1 inhibits the SUMO pathway by interfering with the activity of E1 (SAE1/SAE2). In vivo, Gam1 expression leads to SAE1/SAE2 inactivation, both SAE1/SAE2 and UBC9 disappearance, and overall inhibition of protein sumoylation. This results in transcriptional activation of some promoters and is directly linked to inhibition of sumoylation of the transcriptional activators involved. Our results identify a mechanism for interfering with the SUMO pathway and with transcription that could have an impact in the design of novel pharmaceutical agents. They also point out once again to the extraordinary ability of eukaryotic viruses to interfere with the biology of host cells by targeting fundamental biochemical processes.


Asunto(s)
Ligasas/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinas/metabolismo , Western Blotting , Proteínas de Unión al ADN/metabolismo , Genes Reporteros , Glutatión Transferasa/metabolismo , Células HeLa , Humanos , Luciferasas/metabolismo , Modelos Biológicos , Pruebas de Precipitina , Regiones Promotoras Genéticas , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Transactivadores/antagonistas & inhibidores , Transcripción Genética , Activación Transcripcional , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...