Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Antiviral Res ; 173: 104649, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31711927

RESUMEN

Herpesviral deubiquitinating enzymes (DUBs) were discovered in 2005, are highly conserved across the family, and are proving to be increasingly important players in herpesviral infection. EBV's DUB, BPLF1, is known to regulate both cellular and viral target activities, yet remains largely unstudied. Our work has implicated BPLF1 in a wide range of processes including infectivity, viral DNA replication, and DNA repair. Additionally, knockout of BPLF1 delays and reduces human B-cell immortalization and lymphoma formation in humanized mice. These findings underscore the importance of BPLF1 in viral infectivity and pathogenesis and suggest that inhibition of EBV's DUB activity may offer a new approach to specific therapy for EBV infections. We set out to discover and characterize small molecule inhibitors of BPLF1 deubiquitinating activity through high-throughput screening. An initial small pilot screen resulted in discovery of 10 compounds yielding >80% decrease in BPLF1 DUB activity at a 10 µM concentration. Follow-up dose response curves of top hits identified several compounds with an IC50 in the low micromolar range. Four of these hits were tested for their ability to cleave ubiquitin chains as well as their effects on viral infectivity and cell viability. Further characterization of the top hit, commonly known as suramin was found to not be selective yet decreased viral infectivity by approximately 90% with no apparent effects on cell viability. Due to the conserved nature of Herpesviral deubiquitinating enzymes, identification of an inhibitor of BPLF1 may prove to be an effective and promising new avenue of therapy for EBV and other herpesviral family members.


Asunto(s)
Antivirales/farmacología , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Evaluación Preclínica de Medicamentos , Herpesvirus Humano 4/efectos de los fármacos , Herpesvirus Humano 4/enzimología , Proteínas Reguladoras y Accesorias Virales/antagonistas & inhibidores , Supervivencia Celular , Enzimas Desubicuitinizantes/genética , Enzimas Desubicuitinizantes/metabolismo , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Bibliotecas de Moléculas Pequeñas , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
2.
Sci Rep ; 9(1): 208, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30659232

RESUMEN

Epstein-Barr Virus latent membrane protein-1 (LMP1) interacts with the SUMO-conjugating enzyme Ubc9, which induces protein sumoylation and may contribute to LMP1-mediated oncogenesis. After analyzing human lymphoma tissues and EBV-positive cell lines, we now document a strong correlation between LMP1 and sumo-1/2/3 or SUMO-1/2/3 levels, and show that LMP1-induced sumo expression requires the activation of NF-κB signaling through CTAR1 and CTAR2. Together, these results point to a second mechanism by which LMP1 dysregulates sumoylation processes and adds EBV-associated lymphomas to the list of malignancies associated with increased SUMO expression.


Asunto(s)
Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Proteínas de la Matriz Viral/metabolismo , Línea Celular , Células HEK293 , Herpesvirus Humano 4/metabolismo , Humanos , Linfoma/metabolismo , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Unión Proteica , Proteína SUMO-1/genética , Transducción de Señal , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Sumoilación , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinas/metabolismo , Proteínas de la Matriz Viral/fisiología
3.
Asian Pac J Cancer Prev ; 19(11): 3153-3159, 2018 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-30486603

RESUMEN

The Epstein-Barr virus (EBV) is a herpesvirus infecting more than 90% of the human population. The tropism of EBV for B lymphocytes is evidenced in its association with many lymphoproliferative disorders. Different types of EBV (EBV-1 and EBV-2), classified on the basis of EBV nuclear antigen-2 (EBNA-2) genotyping, have been reported in benign and malignant pathologies, but there is almost no information about their frequency in the Pakistani population. The aim of this study was to determine the frequency and distribution of EBNA-2-based EBV genotypes in lymphoma patients. Genomic DNA was extracted from formalin-fixed paraffin embedded (FFPE) tissue samples obtained from 73 EBV-DNA-positive lymphoma patients. The ß-globin gene was amplified to assess the presence and quality of cellular DNA from all samples. EBER-1 DNA was detected by PCR to confirm EBV presence in tissue samples. EBNA-1 mRNA relative quantification done by quantitative PCR substantiated EBNA-1 mRNA overexpression in 43.8% of EBV-positive cases in comparison to EBV-positive control cell line. EBNA-2 genotyping was done by nested PCR. Among typable samples, EBV-1 was found in 90.7% of samples while EBV-2 was present in 9.3% cases. These results show that EBV-1 was the most prevalent type in the lymphoma population of Pakistan. This epidemiology of EBV in Pakistani lymphoma patients represents an important first step in using EBV for prognosis and monitoring treatment response.


Asunto(s)
Infecciones por Virus de Epstein-Barr/epidemiología , Antígenos Nucleares del Virus de Epstein-Barr/genética , Herpesvirus Humano 4/genética , Linfoma/virología , Proteínas Virales/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Femenino , Estudios de Seguimiento , Genotipo , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 4/aislamiento & purificación , Humanos , Linfoma/patología , Masculino , Persona de Mediana Edad , Pakistán/epidemiología , Prevalencia , Pronóstico , Estudios Retrospectivos
4.
Blood ; 127(12): 1524-5, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-27013211

RESUMEN

In this issue of Blood, Bedekovics et al have demonstrated that a multifunctional molecule of the ubiquitin system ubiquitin C-terminal hydrolase L1 (UCH-L1) is induced in diffuse large B-cell lymphomas (DLBCLs), and that levels of this molecule are higher in germinal center (GC) B-cell DLBCL (GCB-DLBCL) compared with activated B-cell DLBCL (ABC-DLBCL) and predict poor outcomes.


Asunto(s)
Linfocitos B/patología , Regulación Neoplásica de la Expresión Génica , Centro Germinal/patología , Linfoma de Células B Grandes Difuso/genética , Ubiquitina Tiolesterasa/genética , Animales , Humanos
5.
Virology ; 448: 293-302, 2014 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-24314660

RESUMEN

Ubiquitin C-terminal Hydrolase L1 (UCH-L1) has oncogenic properties and is highly expressed during malignancies. We recently documented that Epstein-Barr virus (EBV) infection induces uch-l1 expression. Here we show that Kaposi's Sarcoma-associated herpesvirus (KSHV) infection induced UCH-L1 expression, via cooperation of KSHV Latency-Associated Nuclear Antigen (LANA) and RBP-Jκ and activation of the uch-l1 promoter. UCH-L1 expression was also increased in Primary Effusion Lymphoma (PEL) cells co-infected with KSHV and EBV compared with PEL cells infected only with KSHV, suggesting EBV augments the effect of LANA on uch-l1. EBV latent membrane protein 1 (LMP1) is one of the few EBV products expressed in PEL cells. Results showed that LMP1 was sufficient to induce uch-l1 expression, and co-expression of LMP1 and LANA had an additive effect on uch-l1 expression. These results indicate that viral latency products of both human γ-herpesviruses contribute to uch-l1 expression, which may contribute to the progression of lymphoid malignancies.


Asunto(s)
Antígenos Virales/metabolismo , Transformación Celular Viral , Infecciones por Virus de Epstein-Barr/enzimología , Infecciones por Herpesviridae/enzimología , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina Tiolesterasa/genética , Proteínas de la Matriz Viral/metabolismo , Antígenos Virales/genética , Línea Celular Transformada , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/virología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/virología , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/genética , Humanos , Proteínas Nucleares/genética , Ubiquitina Tiolesterasa/metabolismo , Regulación hacia Arriba , Proteínas de la Matriz Viral/genética
6.
J Gen Virol ; 94(Pt 3): 507-513, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23223620

RESUMEN

A global regulator of chromatin remodelling and gene expression, special AT-rich-binding protein 1 (SATB1) has been implicated in promotion of growth and metastasis of a number of cancers. Here, we demonstrate that the principal oncogene of Epstein-Barr virus (EBV), latent membrane protein 1 (LMP1) upregulates SATB1 RNA and protein expression in human nasopharyngeal cell lines. Silencing of endogenously expressed SATB1 with specific short hairpin RNA decreases cell proliferation and resistance to apoptosis induced by growth factor withdrawal. Additionally, we provide evidence that LMP1-mediated expression of Survivin, a multifunctional protein involved in promoting cell growth and survival, is mediated at least in part by SATB1 in human nasopharyngeal cells. Finally, we show that SATB1 protein levels are elevated in tissue samples from patients with nasopharyngeal carcinoma (NPC), and are directly correlated with the expression of LMP1. Taken together, our results suggest that SATB1 functions as a pro-metastatic effector of LMP1 signalling in EBV-positive NPC.


Asunto(s)
Herpesvirus Humano 4/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Proteínas de la Matriz Viral/metabolismo , Carcinoma , Línea Celular , Proliferación Celular , Regulación de la Expresión Génica/fisiología , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Neoplasias Nasofaríngeas/virología , Nariz/citología , Faringe/citología , ARN Interferente Pequeño , Transducción de Señal , Proteínas de la Matriz Viral/genética
7.
J Virol ; 86(22): 12251-61, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22951831

RESUMEN

Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) induces multiple signal transduction pathways during latent EBV infection via its C-terminal activating region 1 (CTAR1), CTAR2, and the less-studied CTAR3. One mechanism by which LMP1 regulates cellular activation is through the induction of protein posttranslational modifications, including phosphorylation and ubiquitination. We recently documented that LMP1 induces a third major protein modification by physically interacting with the SUMO-conjugating enzyme Ubc9 through CTAR3 and inducing the sumoylation of cellular proteins in latently infected cells. We have now identified a specific target of LMP1-induced sumoylation, interferon regulatory factor 7 (IRF7). We hypothesize that during EBV latency, LMP1 induces the sumoylation of IRF7, limiting its transcriptional activity and modulating the activation of innate immune responses. Our data show that endogenously sumoylated IRF7 is detected in latently infected EBV lymphoblastoid cell lines. LMP1 expression coincided with increased sumoylation of IRF7 in a CTAR3-dependent manner. Additional experiments show that LMP1 CTAR3-induced sumoylation regulates the expression and function of IRF7 by decreasing its turnover, increasing its nuclear retention, decreasing its DNA binding, and limiting its transcriptional activation. Finally, we identified that IRF7 is sumoylated at lysine 452. These data demonstrate that LMP1 CTAR3 does in fact function in intracellular signaling, leading to biologic effects. We propose that CTAR3 is an important signaling region of LMP1 that regulates protein function by sumoylation. We have shown specifically that LMP1 CTAR3, in cooperation with CTAR2, can limit the ability of IRF7 to induce innate immune responses by inducing the sumoylation of IRF7.


Asunto(s)
Regulación Viral de la Expresión Génica , Factor 7 Regulador del Interferón/metabolismo , Proteínas de la Matriz Viral/metabolismo , Núcleo Celular/metabolismo , Cromatina/química , Infecciones por Virus de Epstein-Barr/virología , Células HEK293 , Humanos , Inmunidad Innata , Luciferasas/metabolismo , Lisina/química , Mutagénesis , Unión Proteica , Procesamiento Proteico-Postraduccional , Transducción de Señal , Sumoilación
8.
J Virol ; 86(15): 8097-106, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22623772

RESUMEN

PCNA is monoubiquitinated in response to DNA damage and fork stalling and then initiates recruitment of specialized polymerases in the DNA damage tolerance pathway, translesion synthesis (TLS). Since PCNA is reported to associate with Epstein-Barr virus (EBV) DNA during its replication, we investigated whether the EBV deubiquitinating (DUB) enzyme encoded by BPLF1 targets ubiquitinated PCNA and disrupts TLS. An N-terminal BPLF1 fragment (a BPLF1 construct containing the first 246 amino acids [BPLF1 1-246]) associated with PCNA and attenuated its ubiquitination in response to fork-stalling agents UV and hydroxyurea in cultured cells. Moreover, monoubiquitinated PCNA was deubiquitinated after incubation with purified BPLF1 1-246 in vitro. BPLF1 1-246 dysregulated TLS by reducing recruitment of the specialized repair polymerase polymerase η (Polη) to the detergent-resistant chromatin compartment and virtually abolished localization of Polη to nuclear repair foci, both hallmarks of TLS. Expression of BPLF1 1-246 decreased viability of UV-treated cells and led to cell death, presumably through deubiquitination of PCNA and the inability to repair damaged DNA. Importantly, deubiquitination of PCNA could be detected endogenously in EBV-infected cells in comparison with samples expressing short hairpin RNA (shRNA) against BPLF1. Further, the specificity of the interaction between BPLF1 and PCNA was dependent upon a PCNA-interacting peptide (PIP) domain within the N-terminal region of BPLF1. Both DUB activity and PIP sequence are conserved in the members of the family Herpesviridae. Thus, deubiquitination of PCNA, normally deubiquitinated by cellular USP1, by the viral DUB can disrupt repair of DNA damage by compromising recruitment of TLS polymerase to stalled replication forks. PCNA is the first cellular target identified for BPLF1 and its deubiquitinating activity.


Asunto(s)
Daño del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Infecciones por Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/enzimología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ubiquitinación , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas de Arabidopsis , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , ADN Polimerasa Dirigida por ADN/genética , Endopeptidasas/genética , Endopeptidasas/metabolismo , Infecciones por Virus de Epstein-Barr/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Células HEK293 , Herpesvirus Humano 4/genética , Humanos , Proteínas de Transporte de Membrana , Antígeno Nuclear de Célula en Proliferación/química , Antígeno Nuclear de Célula en Proliferación/genética , Estructura Terciaria de Proteína , Proteasas Ubiquitina-Específicas , Rayos Ultravioleta , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Reguladoras y Accesorias Virales/genética
9.
Biomaterials ; 33(15): 3887-98, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22364702

RESUMEN

One of the major tasks of tissue engineering is to produce tissue grafts for the replacement or regeneration of damaged tissue, and natural and recombinant silk-based polymer scaffolds are promising candidates for such grafts. Here, we compared two porous scaffolds made from different silk proteins, fibroin of Bombyx mori and a recombinant analog of Nephila clavipes spidroin 1 known as rS1/9, and their biocompatibility and degradation behavior in vitro and in vivo. The vascularization and intergrowth of the connective tissue, which was penetrated with nerve fibers, at 8 weeks after subcutaneous implantation in Balb/c mice was more profound using the rS1/9 scaffolds. Implantation of both scaffolds into bone defects in Wistar rats accelerated repair compared to controls with no implanted scaffold at 4 weeks. Based on the number of macrophages and multinuclear giant cells in the subcutaneous area and the number of osteoclasts in the bone, regeneration was determined to be more effective after the rS1/9 scaffolds were implanted. Microscopic examination of the morphology of the matrices revealed differences in their internal microstructures. In contrast to fibroin-based scaffolds, the walls of the rS1/9 scaffolds were visibly thicker and contained specific micropores. We suggest that the porous inner structure of the rS1/9 scaffolds provided a better micro-environment for the regenerating tissue, which makes the matrices derived from the recombinant rS1/9 protein favorable candidates for future in vivo applications.


Asunto(s)
Fibroínas/farmacología , Regeneración Tisular Dirigida/métodos , Proteínas Recombinantes/farmacología , Andamios del Tejido/química , Células 3T3 , Animales , Bombyx , Regeneración Ósea/efectos de los fármacos , Huesos/efectos de los fármacos , Huesos/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Rastreo , Neovascularización Fisiológica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Polímeros/química , Porosidad/efectos de los fármacos , Implantación de Prótesis , Ratas , Tejido Subcutáneo/irrigación sanguínea , Tejido Subcutáneo/efectos de los fármacos , Tejido Subcutáneo/inervación , Tejido Subcutáneo/patología
10.
Leuk Lymphoma ; 52(7): 1336-47, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21504384

RESUMEN

Elevated levels of ubiquitin C-terminal hydrolase L1 (UCH L1) have been detected in a variety of malignancies, and recent studies show the oncogenic capacity of overexpressed UCH L1 in vivo in animal models. Here we demonstrate that expression of endogenous UCH L1 is significantly higher in B-lymphoma cells than in transformed cells of epithelial and fibroblastic origin. The specific hematopoietic transcription factor PU.1 induces UCH L1 expression through direct activation of the uch l1 promoter. Using chromatin immunoprecipitation (ChIP) assays and direct mutagenesis we identified PU.1 binding sites on the uch l1 promoter, at least three of which are involved in this activation. We also show that the viral transcriptional co-activator EBNA2 dramatically increases PU.1-dependent up-regulation of endogenous UCH L1 expression. Finally, inhibition of PU.1 expression with specific shRNA resulted in reduction of UCH L1 mRNA and protein levels in Epstein-Barr virus (EBV)-transformed B-cells. We propose that the ubiquitin-editing enzyme UCH L1 is a multifunctional pro-oncogenic factor involved in development and progression of certain lymphoid malignancies, including EBV-associated lymphomas.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Linfoma de Células B/enzimología , Linfoma de Células B/genética , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Sitios de Unión , Linfoma de Burkitt/enzimología , Linfoma de Burkitt/genética , Línea Celular Tumoral , Endonucleasas , Herpesvirus Humano 4/metabolismo , Humanos , Modelos Biológicos , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Regulación hacia Arriba
11.
Virology ; 402(1): 121-8, 2010 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-20381110

RESUMEN

Activation of interferon regulatory factors (IRFs) 3 and 7 is essential for the induction of Type I interferons (IFN) and innate antiviral responses, and herpesviruses have evolved mechanisms to evade such responses. We previously reported that Epstein-Barr virus BZLF1, an immediate-early (IE) protein, inhibits the function of IRF7, but the role of BRLF1, the other IE transactivator, in IRF regulation has not been examined. We now show that BRLF1 expression decreased induction of IFN-beta, and reduced expression of IRF3 and IRF7; effects were dependent on N- and C-terminal regions of BRLF1 and its nuclear localization signal. Endogenous IRF3 and IRF7 RNA and protein levels were also decreased during cytolytic EBV infection. Finally, production of IFN-beta was decreased during lytic EBV infection and was associated with increased susceptibility to superinfection with Sendai virus. These data suggest a new role for BRLF1 with the ability to evade host innate immune responses.


Asunto(s)
Proteínas Aviares/antagonistas & inhibidores , Herpesvirus Humano 4/patogenicidad , Interacciones Huésped-Patógeno , Proteínas Inmediatas-Precoces/fisiología , Factor 7 Regulador del Interferón/antagonistas & inhibidores , Factores Reguladores del Interferón/antagonistas & inhibidores , Interferón beta/antagonistas & inhibidores , Transactivadores/fisiología , Proteínas Aviares/inmunología , Línea Celular , Herpesvirus Humano 4/inmunología , Humanos , Factor 7 Regulador del Interferón/inmunología , Factores Reguladores del Interferón/inmunología , Interferón beta/inmunología , Señales de Localización Nuclear , Mapeo de Interacción de Proteínas , Virus Sendai/crecimiento & desarrollo , Transcripción Genética
12.
Cell Cycle ; 9(5): 980-94, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20160478

RESUMEN

Microtubules are essential components of the cytoskeleton and are involved in many aspects of cell responses including cell division, migration, and intracellular signal transduction. Among other factors, post-translational modifications play a significant role in the regulation of microtubule dynamics. Here, we demonstrate that the ubiquitin-editing enzyme UCH L1, abundant expression of which is normally restricted to brain tissue, is also a part of the microtubule network in a variety of transformed cells. Moreover, during mitosis, endogenous UCH L1 is expressed and tightly associated with the mitotic spindle through all stages of M phase, suggesting that UCH L1 is involved in regulation of microtubule dynamics. Indeed, addition of recombinant UCH L1 to the reaction of tubulin polymerization in vitro had an inhibitory effect on microtubule formation. Unexpectedly, western blot analysis of tubulin fractions after polymerization revealed the presence of a specific approximately 50 kDa band of UCH L1 (not the normal approximately 25 kDa) in association with microtubules, but not with free tubulin. In addition, we show that along with 25 kDa UCH L1, endogenous high molecular weight UCH L1 complexes exist in cells, and that levels of 50 kDa UCH L1 complexes are increasing in cells during mitosis. Finally, we provide evidence that ubiquitination is involved in tubulin polymerization: the presence of ubiquitin during polymerization in vitro by itself inhibited microtubule formation and enhanced the inhibitory effect of added UCH L1. The inhibitory effects of UCH L1 correlate with an increase in ubiquitination of microtubule components. Since besides being a deubiquitinating enzyme, UCH L1 as a dimer has also been shown to exhibit ubiquitin ligase activity, we discuss the possibility that the approximately 50 kDa UCH L1 observed is a dimer which prevents microtubule formation through ubiquitination of tubulins and/or microtubule-associated proteins.


Asunto(s)
Microtúbulos/metabolismo , Mitosis , Ubiquitina Tiolesterasa/metabolismo , Animales , División Celular , Línea Celular , Dimerización , Fase G2 , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Huso Acromático/fisiología , Tubulina (Proteína)/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitinación
13.
PLoS One ; 4(8): e6764, 2009 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-19707515

RESUMEN

Deubiquitinating enzymes (DUBs) have been increasingly implicated in regulation of cellular processes, but a functional role for Ubiquitin C-terminal Hydrolases (UCHs), which has been largely relegated to processing of small ubiquitinated peptides, remains unexplored. One member of the UCH family, UCH L1, is expressed in a number of malignancies suggesting that this DUB might be involved in oncogenic processes, and increased expression and activity of UCH L1 have been detected in EBV-immortalized cell lines. Here we present an analysis of genes regulated by UCH L1 shown by microarray profiles obtained from cells in which expression of the gene was inhibited by RNAi. Microarray data were verified with subsequent real-time PCR analysis. We found that inhibition of UCH L1 activates genes that control apoptosis, cell cycle arrest and at the same time suppresses expression of genes involved in proliferation and migration pathways. These findings are complemented by biological assays for apoptosis, cell cycle progression and migration that support the data obtained from microarray analysis, and suggest that the multi-functional molecule UCH L1 plays a role in regulating principal pathways involved in oncogenesis.


Asunto(s)
Regulación de la Expresión Génica , Ubiquitina Tiolesterasa/fisiología , Apoptosis , Secuencia de Bases , Ciclo Celular , Línea Celular , Movimiento Celular , Proliferación Celular , Cartilla de ADN , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
PLoS One ; 4(6): e5955, 2009 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-19536331

RESUMEN

Deubiquitinating enzymes (DUBs) are involved in the regulation of distinct critical cellular processes. Ubiquitin C-terminal Hydrolase L1 (UCH L1) has been linked to several neurological diseases as well as human cancer, but the physiological targets and the regulation of UCH L1 expression in vivo have been largely unexplored. Here we demonstrate that UCH L1 up-regulates beta-catenin/TCF signaling: UCH L1 forms endogenous complexes with beta-catenin, stabilizes it and up-regulates beta-catenin/TCF-dependent transcription. We also show that, reciprocally, beta-catenin/TCF signaling up-regulates expression of endogenous UCH L1 mRNA and protein. Moreover, using ChIP assay and direct mutagenesis we identify two TCF4-binding sites on the uch l1 promoter that are involved in this regulation. Since the expression and deubiquitinating activity of UCH L1 are required for its own basic promoter activity, we propose that UCH L1 up-regulates its expression by activation of the oncogenic beta-catenin/TCF signaling in transformed cells.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Factores de Transcripción TCF/química , Ubiquitina Tiolesterasa/química , beta Catenina/química , Animales , Sitios de Unión , Línea Celular Transformada , Humanos , Ratones , Mutagénesis , Mutagénesis Sitio-Dirigida , Células 3T3 NIH , Estructura Terciaria de Proteína , Transducción de Señal , Regulación hacia Arriba
15.
J Virol ; 83(9): 4345-53, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19244336

RESUMEN

A newly discovered virally encoded deubiquitinating enzyme (DUB) is strictly conserved across the Herpesviridae. Epstein-Barr virus (EBV) BPLF1 encodes a tegument protein (3,149 amino acids) that exhibits deubiquitinating (DUB) activity that is lost upon mutation of the active-site cysteine. However, targets for the herpesviral DUBs have remained elusive. To investigate a predicted interaction between EBV BPLF1 and EBV ribonucleotide reductase (RR), a functional clone of the first 246 N-terminal amino acids of BPLF1 (BPLF1 1-246) was constructed. Immunoprecipitation verified an interaction between the small subunit of the viral RR2 and BPLF1 proteins. In addition, the large subunit (RR1) of the RR appeared to be ubiquitinated both in vivo and in vitro; however, ubiquitinated forms of the small subunit, RR2, were not detected. Ubiquitination of RR1 requires the expression of both subunits of the RR complex. Furthermore, coexpression of RR1 and RR2 with BPLF1 1-246 abolishes ubiquitination of RR1. EBV RR1, RR2, and BPLF1 1-246 colocalized to the cytoplasm in HEK 293T cells. Finally, expression of enzymatically active BPLF1 1-246 decreased RR activity, whereas a nonfunctional active-site mutant (BPLF1 C61S) had no effect. These results indicate that the EBV deubiquitinating enzyme interacts with, deubiquitinates, and influences the activity of the EBV RR. This is the first verified protein target of the EBV deubiquitinating enzyme.


Asunto(s)
Herpesvirus Humano 4/enzimología , Ribonucleótido Reductasas/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Línea Celular , Citoplasma/enzimología , Activación Enzimática , Genoma Viral/genética , Herpesvirus Humano 4/genética , Humanos , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Interferencia de ARN , ARN Mensajero/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ribonucleótido Reductasas/genética , Ubiquitinación , Proteínas Reguladoras y Accesorias Virales/genética
16.
BMC Biochem ; 8 Suppl 1: S8, 2007 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-18047745

RESUMEN

Tumor viruses are linked to approximately 20% of human malignancies worldwide. This review focuses on examples of human oncogenic viruses that manipulate the ubiquitin system in a subset of viral malignancies; those associated with AIDS. The viruses include Kaposi's sarcoma herpesvirus, Epstein-Barr virus and human papilloma virus, which are causally linked to Kaposi's sarcoma, certain B-cell lymphomas and cervical cancer, respectively. We discuss the molecular mechanisms by which these viruses subvert the ubiquitin system and potential viral targets for anti-cancer therapy from the perspective of this system. Publication history: Republished from Current BioData's Targeted Proteins database (TPdb; http://www.targetedproteinsdb.com).


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/enzimología , Síndrome de Inmunodeficiencia Adquirida/virología , Neoplasias/enzimología , Neoplasias/virología , Virus Oncogénicos/fisiología , Infecciones Tumorales por Virus/enzimología , Infecciones Tumorales por Virus/virología , Ubiquitina/fisiología , Síndrome de Inmunodeficiencia Adquirida/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Humanos , Neoplasias/tratamiento farmacológico , Virus Oncogénicos/efectos de los fármacos , Infecciones Tumorales por Virus/tratamiento farmacológico
17.
J Virol ; 80(4): 2045-50, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16439560

RESUMEN

Epstein-Barr virus (EBV) nuclear antigen 2 (EBNA2) transactivates EBV genes in latently infected B cells. We have shown that mitotic hyperphosphorylation of EBNA2 suppresses its ability to transactivate the latent membrane protein 1 (LMP1) promoter. In this follow-up study, we identify EBNA2 Ser243 as a phosphorylation site for mitotic cdc2/cyclin B1 kinase. Mutation at Ser243, which mimics constitutive phosphorylation of the protein, decreases endogenous levels of both LMP1 and EBNA2. Moreover, mutation at Ser243 reduces the ability of EBNA2 to transactivate Cp, the promoter for all six EBV EBNA genes. Our data implicate EBNA2 Ser243 as a cdc2/cyclin B1 site of phosphorylation important for EBNA2's cotranscriptional function in mitosis.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Ciclina B/metabolismo , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Mitosis , Serina/metabolismo , Sustitución de Aminoácidos , Ciclina B1 , Antígenos Nucleares del Virus de Epstein-Barr/química , Regulación Viral de la Expresión Génica , Células HeLa , Humanos , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , ARN Mensajero/análisis , ARN Viral/análisis , Alineación de Secuencia , Transcripción Genética , Activación Transcripcional , Proteínas de la Matriz Viral/análisis , Proteínas Virales
18.
Proc Natl Acad Sci U S A ; 102(51): 18431-6, 2005 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-16344472

RESUMEN

The protein levels of beta-catenin are tightly regulated by the ubiquitin/proteasome system. We provide evidence that two distinct ubiquitin-dependent degradation pathways for beta-catenin are active in the same Burkitt's lymphoma cells: Along with the classical glycogen-synthase kinase 3beta-dependent destruction machinery, degradation of beta-catenin through seven in absentia homolog 1 (Siah-1) ubiquitin ligase is functional in these cells. We show that inhibition of endogenous Siah-1 stabilizes and activates beta-catenin in B cells. The principal Epstein-Barr virus oncoprotein, latent membrane protein 1, is involved in beta-catenin up-regulation, and expression of latent membrane protein 1 in B lymphoma cells is associated with decreased Siah-1 RNA and protein levels. Thus, we demonstrate the significance of the endogenous Siah-1-dependent ubiquitin/proteasome pathway for beta-catenin degradation in malignant human cells and its regulation by a viral oncogene.


Asunto(s)
Linfoma de Células B/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Oncogénicas Virales/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Regulación hacia Arriba , Proteínas de la Matriz Viral/metabolismo , beta Catenina/metabolismo , Línea Celular , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Linfoma de Células B/genética , Linfoma de Células B/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas Virales/genética , Unión Proteica , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de la Matriz Viral/genética
19.
Essays Biochem ; 41: 139-56, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16250903

RESUMEN

The ability of viruses to co-opt cell signalling pathways has, over millions of years of co-evolution, come to pervade nearly every facet of cellular functions. Recognition of the extent to which the ubiquitin-proteasome system can be directed or subverted by viruses is relatively recent. Viral products interact with, and adjust, the ubiquitin-proteasome machinery precisely and at many levels, and they do so at distinct stages of viral life-cycles. The implications for both cells and viruses are fundamental, and understanding viral strategies in this context opens up fascinating new areas for research that span from basic cell biology to therapeutic interventions against both viruses and malignancies.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/fisiología , Ubiquitina/fisiología , Fenómenos Fisiológicos de los Virus , Animales , Presentación de Antígeno/inmunología , Humanos , Inmunidad Celular , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal , Ubiquitina/metabolismo , Virión/fisiología , Virosis/inmunología , Virosis/metabolismo , Virosis/virología , Virus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...