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.
Biochem J ; 477(14): 2721-2733, 2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32639530

RESUMEN

Merkel cell carcinoma (MCC) is an aggressive skin cancer with high rates of recurrence and metastasis. Merkel cell polyomavirus (MCPyV) is associated with the majority of MCC cases. MCPyV-induced tumourigenesis is largely dependent on the expression of the small tumour antigen (ST). Recent findings implicate MCPyV ST expression in the highly metastatic nature of MCC by promoting cell motility and migration, through differential expression of cellular proteins that lead to microtubule destabilisation, filopodium formation and breakdown of cell-cell junctions. However, the molecular mechanisms which dysregulate these cellular processes are yet to be fully elucidated. Here, we demonstrate that MCPyV ST expression activates p38 MAPK signalling to drive cell migration and motility. Notably, MCPyV ST-mediated p38 MAPK signalling occurs through MKK4, as opposed to the canonical MKK3/6 signalling pathway. In addition, our results indicate that an interaction between MCPyV ST and the cellular phospatase subunit PP4C is essential for its effect on p38 MAPK signalling. These results provide novel opportunities for the treatment of metastatic MCC given the intense interest in p38 MAPK inhibitors as therapeutic agents.


Asunto(s)
Antígenos Virales de Tumores/metabolismo , Carcinoma de Células de Merkel/virología , Poliomavirus de Células de Merkel/patogenicidad , Neoplasias Cutáneas/virología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Antígenos Virales de Tumores/genética , Carcinoma de Células de Merkel/genética , Carcinoma de Células de Merkel/metabolismo , Carcinoma de Células de Merkel/patología , Movimiento Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Imidazoles/farmacología , MAP Quinasa Quinasa 4/metabolismo , Poliomavirus de Células de Merkel/inmunología , Fosfoproteínas Fosfatasas/metabolismo , Piridinas/farmacología , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/patología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
2.
Chem Commun (Camb) ; 55(66): 9765-9768, 2019 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-31355394

RESUMEN

Bacterial quorum sensing has been implicated in a number of pathogenic bacterial processes, such as biofilm formation, making it a crucial target for developing materials with a novel antibiotic mode of action. This paper describes poly(N-isopropyl acrylamide) that has been covalently linked, at multiple chain ends, to homoserine lactone to give a highly branched polymer functionalized with a key messenger molecule implicated in QS. This novel functional material has shown promising anti-QS activity in a Chromobacterium violaceum assay.


Asunto(s)
Acrilamidas/farmacología , Chromobacterium/efectos de los fármacos , Percepción de Quorum/efectos de los fármacos , Biopelículas/efectos de los fármacos , Chromobacterium/fisiología
3.
PLoS Pathog ; 14(9): e1007276, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30188954

RESUMEN

Merkel cell carcinoma (MCC) is an aggressive skin cancer with a high propensity for recurrence and metastasis. Merkel cell polyomavirus (MCPyV) is recognised as the causative factor in the majority of MCC cases. The MCPyV small tumour antigen (ST) is considered to be the main viral transforming factor, however potential mechanisms linking ST expression to the highly metastatic nature of MCC are yet to be fully elucidated. Metastasis is a complex process, with several discrete steps required for the formation of secondary tumour sites. One essential trait that underpins the ability of cancer cells to metastasise is how they interact with adjoining tumour cells and the surrounding extracellular matrix. Here we demonstrate that MCPyV ST expression disrupts the integrity of cell-cell junctions, thereby enhancing cell dissociation and implicate the cellular sheddases, A disintegrin and metalloproteinase (ADAM) 10 and 17 proteins in this process. Inhibition of ADAM 10 and 17 activity reduced MCPyV ST-induced cell dissociation and motility, attributing their function as critical to the MCPyV-induced metastatic processes. Consistent with these data, we confirm that ADAM 10 and 17 are upregulated in MCPyV-positive primary MCC tumours. These novel findings implicate cellular sheddases as key host cell factors contributing to virus-mediated cellular transformation and metastasis. Notably, ADAM protein expression may be a novel biomarker of MCC prognosis and given the current interest in cellular sheddase inhibitors for cancer therapeutics, it highlights ADAM 10 and 17 activity as a novel opportunity for targeted interventions for disseminated MCC.


Asunto(s)
Antígenos Virales de Tumores/fisiología , Carcinoma de Células de Merkel/etiología , Poliomavirus de Células de Merkel/patogenicidad , Infecciones por Polyomavirus/etiología , Neoplasias Cutáneas/etiología , Infecciones Tumorales por Virus/etiología , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Carcinoma de Células de Merkel/enzimología , Carcinoma de Células de Merkel/secundario , Movimiento Celular , Células HEK293 , Humanos , Uniones Intercelulares/patología , Uniones Intercelulares/fisiología , Proteínas de la Membrana/metabolismo , Poliomavirus de Células de Merkel/inmunología , Poliomavirus de Células de Merkel/fisiología , Invasividad Neoplásica/patología , Invasividad Neoplásica/fisiopatología , Infecciones por Polyomavirus/enzimología , Infecciones por Polyomavirus/patología , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/patología , Infecciones Tumorales por Virus/enzimología , Infecciones Tumorales por Virus/patología
4.
J Invest Dermatol ; 138(11): 2343-2354, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29777657

RESUMEN

Virus-encoded microRNAs are emerging as key regulators of persistent infection and host-cell immune evasion. Merkel cell polyomavirus, the predominant etiological agent of Merkel cell carcinoma, encodes a single microRNA, MCV-miR-M1, which targets the oncogenic Merkel cell polyomavirus large T antigen. MCV-miR-M1 has previously been shown to play an important role in the establishment of long-term infection, however, the underlying mechanism is not fully understood. A key unanswered question is whether, in addition to autoregulating large T antigen, MCV-miR-M1 also targets cellular transcripts to orchestrate an environment conducive to persistent infection. To address this, we adopted an RNA sequencing-based approach to identify cellular targets of MCV-miR-M1. Intriguingly, bioinformatics analysis of transcripts that are differentially expressed in cells expressing MCV-miR-M1 revealed several genes implicated in immune evasion. Subsequent target validation led to the identification of the innate immunity protein, SP100, as a direct target of MCV-miR-M1. Moreover, MCV-miR-M1-mediated modulation of SP100 was associated with a significant decrease in CXCL8 secretion, resulting in the attenuation of neutrophil chemotaxis toward Merkel cells harboring synthetic Merkel cell polyomavirus. Based on these observations, we propose that MCV-miR-M1 targets key immune response regulators to help facilitate persistent infection, which is a prerequisite for cellular transformation in Merkel cell carcinoma.


Asunto(s)
Carcinoma de Células de Merkel/inmunología , Poliomavirus de Células de Merkel/fisiología , MicroARNs/genética , Neutrófilos/inmunología , Infecciones por Polyomavirus/inmunología , ARN Viral/genética , Infecciones Tumorales por Virus/inmunología , Antígenos Nucleares/genética , Antígenos Virales de Tumores/genética , Autoantígenos/genética , Carcinoma de Células de Merkel/genética , Quimiotaxis , Células HEK293 , Humanos , Evasión Inmune , Inmunidad Innata/genética , Interleucina-8/metabolismo , Infecciones por Polyomavirus/genética , Infecciones Tumorales por Virus/genética
5.
J Biol Chem ; 293(12): 4582-4590, 2018 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-29462791

RESUMEN

Ion channels regulate many aspects of cell physiology, including cell proliferation, motility, and migration, and aberrant expression and activity of ion channels is associated with various stages of tumor development, with K+ and Cl- channels now being considered the most active during tumorigenesis. Accordingly, emerging in vitro and preclinical studies have revealed that pharmacological manipulation of ion channel activity offers protection against several cancers. Merkel cell polyomavirus (MCPyV) is a major cause of Merkel cell carcinoma (MCC), primarily because of the expression of two early regulatory proteins termed small and large tumor antigens (ST and LT, respectively). Several molecular mechanisms have been attributed to MCPyV-mediated cancer formation but, thus far, no studies have investigated any potential link to cellular ion channels. Here we demonstrate that Cl- channel modulation can reduce MCPyV ST-induced cell motility and invasiveness. Proteomic analysis revealed that MCPyV ST up-regulates two Cl- channels, CLIC1 and CLIC4, which when silenced, inhibit MCPyV ST-induced motility and invasiveness, implicating their function as critical to MCPyV-induced metastatic processes. Consistent with these data, we confirmed that CLIC1 and CLIC4 are up-regulated in primary MCPyV-positive MCC patient samples. We therefore, for the first time, implicate cellular ion channels as a key host cell factor contributing to virus-mediated cellular transformation. Given the intense interest in ion channel modulating drugs for human disease. This highlights CLIC1 and CLIC4 activity as potential targets for MCPyV-induced MCC.


Asunto(s)
Carcinoma de Células de Merkel/patología , Movimiento Celular , Canales de Cloruro/metabolismo , Poliomavirus de Células de Merkel/fisiología , Infecciones por Polyomavirus/complicaciones , Neoplasias Cutáneas/secundario , Infecciones Tumorales por Virus/complicaciones , Antígenos Virales de Tumores/genética , Antígenos Virales de Tumores/metabolismo , Carcinoma de Células de Merkel/epidemiología , Carcinoma de Células de Merkel/virología , Proliferación Celular , Canales de Cloruro/genética , Cloruros/metabolismo , Células HEK293 , Humanos , Incidencia , Invasividad Neoplásica , Infecciones por Polyomavirus/patología , Infecciones por Polyomavirus/virología , Proteoma/análisis , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/virología , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología
6.
J Virol ; 90(20): 9543-55, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27512077

RESUMEN

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of commonly fatal malignancies of immunocompromised individuals, including primary effusion lymphoma (PEL) and Kaposi's sarcoma (KS). A hallmark of all herpesviruses is their biphasic life cycle-viral latency and the productive lytic cycle-and it is well established that reactivation of the KSHV lytic cycle is associated with KS pathogenesis. Therefore, a thorough appreciation of the mechanisms that govern reactivation is required to better understand disease progression. The viral protein replication and transcription activator (RTA) is the KSHV lytic switch protein due to its ability to drive the expression of various lytic genes, leading to reactivation of the entire lytic cycle. While the mechanisms for activating lytic gene expression have received much attention, how RTA impacts cellular function is less well understood. To address this, we developed a cell line with doxycycline-inducible RTA expression and applied stable isotope labeling of amino acids in cell culture (SILAC)-based quantitative proteomics. Using this methodology, we have identified a novel cellular protein (AT-rich interacting domain containing 3B [ARID3B]) whose expression was enhanced by RTA and that relocalized to replication compartments upon lytic reactivation. We also show that small interfering RNA (siRNA) knockdown or overexpression of ARID3B led to an enhancement or inhibition of lytic reactivation, respectively. Furthermore, DNA affinity and chromatin immunoprecipitation assays demonstrated that ARID3B specifically interacts with A/T-rich elements in the KSHV origin of lytic replication (oriLyt), and this was dependent on lytic cycle reactivation. Therefore, we have identified a novel cellular protein whose expression is enhanced by KSHV RTA with the ability to inhibit KSHV reactivation. IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of fatal malignancies of immunocompromised individuals, including Kaposi's sarcoma (KS). Herpesviruses are able to establish a latent infection, in which they escape immune detection by restricting viral gene expression. Importantly, however, reactivation of productive viral replication (the lytic cycle) is necessary for the pathogenesis of KS. Therefore, it is important that we comprehensively understand the mechanisms that govern lytic reactivation, to better understand disease progression. In this study, we have identified a novel cellular protein (AT-rich interacting domain protein 3B [ARID3B]) that we show is able to temper lytic reactivation. We showed that the master lytic switch protein, RTA, enhanced ARID3B levels, which then interacted with viral DNA in a lytic cycle-dependent manner. Therefore, we have added a new factor to the list of cellular proteins that regulate the KSHV lytic cycle, which has implications for our understanding of KSHV biology.


Asunto(s)
Proteínas de Unión al ADN/genética , Herpesvirus Humano 8/genética , Sarcoma de Kaposi/virología , Proteínas Virales/genética , Línea Celular Tumoral , Inmunoprecipitación de Cromatina/métodos , Replicación del ADN/genética , ADN Viral/genética , Regulación Viral de la Expresión Génica/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Linfoma de Efusión Primaria/genética , Linfoma de Efusión Primaria/virología , ARN Interferente Pequeño/genética , Transactivadores/genética , Activación Viral/genética , Latencia del Virus/genética , Replicación Viral/genética
7.
Int J Oncol ; 45(6): 2181-6, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25269471

RESUMEN

Metastatic melanoma is the leading cause of skin-cancer related deaths and while in recent years some progress has been made with targeted therapies, there remains an urgent unmet need for novel therapeutic treatments and reliable diagnostic, prognostic and predictive biomarkers. The emergence of next generation sequencing (NGS) has seen a growing appreciation for the role played by non-coding genomic transcripts in regulating gene expression and by extension impacting on disease progression. The long non-coding RNAs (lncRNAs) represent the most enigmatic of these new regulatory molecules. Our understanding of how lncRNAs regulate biological functions and their importance to disease aetiology, while still limited, is rapidly improving, in particular with regards to their role in cancer. Herein we review the identification of several lncRNAs shown to impact on melanoma disease progression and discuss how these molecules are operating at the molecular level.


Asunto(s)
Melanoma/genética , Proteínas de Neoplasias/biosíntesis , ARN Largo no Codificante/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Melanoma/etiología , Melanoma/patología , Metástasis de la Neoplasia , Pronóstico
8.
PLoS Pathog ; 7(7): e1002138, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21814512

RESUMEN

The hTREX complex mediates cellular bulk mRNA nuclear export by recruiting the nuclear export factor, TAP, via a direct interaction with the export adaptor, Aly. Intriguingly however, depletion of Aly only leads to a modest reduction in cellular mRNA nuclear export, suggesting the existence of additional mRNA nuclear export adaptor proteins. In order to efficiently export Kaposi's sarcoma-associated herpesvirus (KSHV) intronless mRNAs from the nucleus, the KSHV ORF57 protein recruits hTREX onto viral intronless mRNAs allowing access to the TAP-mediated export pathway. Similarly however, depletion of Aly only leads to a modest reduction in the nuclear export of KSHV intronless mRNAs. Herein, we identify a novel interaction between ORF57 and the cellular protein, UIF. We provide the first evidence that the ORF57-UIF interaction enables the recruitment of hTREX and TAP to KSHV intronless mRNAs in Aly-depleted cells. Strikingly, depletion of both Aly and UIF inhibits the formation of an ORF57-mediated nuclear export competent ribonucleoprotein particle and consequently prevents ORF57-mediated mRNA nuclear export and KSHV protein production. Importantly, these findings highlight that redundancy exists in the eukaryotic system for certain hTREX components involved in the mRNA nuclear export of intronless KSHV mRNAs.


Asunto(s)
Núcleo Celular/metabolismo , Herpesvirus Humano 8/metabolismo , Proteínas Nucleares/metabolismo , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular/genética , Núcleo Celular/genética , Núcleo Celular/virología , Células HEK293 , Herpesvirus Humano 8/genética , Humanos , Proteínas Nucleares/genética , ARN Mensajero/genética , ARN Viral/genética , Proteínas de Unión al ARN/genética , Proteínas Virales/genética
9.
J Virol ; 85(15): 7881-91, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21593148

RESUMEN

The Kaposi's sarcoma-associated herpesvirus (KSHV) ORF57 protein is essential for virus lytic replication. ORF57 regulates virus gene expression at multiple levels, enhancing transcription, stability, nuclear export, and translation of viral transcripts. To enhance the nuclear export of viral intronless transcripts, ORF57 (i) binds viral intronless mRNAs, (ii) shuttles between the nucleus, nucleolus, and the cytoplasm, and (iii) interacts with multiple cellular nuclear export proteins to access the TAP-mediated nuclear export pathway. We investigated the implications on the subcellular trafficking, cellular nuclear export factor recruitment, and ultimately nuclear mRNA export of an ORF57 protein unable to bind RNA. We observed that mutation of a carboxy-terminal RGG motif, which prevents RNA binding, affects the subcellular localization and nuclear trafficking of the ORF57 protein, suggesting that it forms subnuclear aggregates. Further analysis of the mutant shows that although it still retains the ability to interact with cellular nuclear export proteins, it is unable to export viral intronless mRNAs from the nucleus. Moreover, computational molecular modeling and biochemical studies suggest that, unlike the wild-type protein, this mutant is unable to self-associate. Therefore, these results suggest the mutation of a carboxy-terminal RGG motif affects ORF57 RNA binding, nuclear trafficking, and multimerization.


Asunto(s)
Núcleo Celular/metabolismo , Herpesvirus Humano 6/genética , Mutación , Sistemas de Lectura Abierta , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas Virales/genética , Secuencia de Bases , Transporte Biológico , Línea Celular , Cartilla de ADN , Humanos , Intrones , Microscopía Fluorescente , ARN Mensajero/química , ARN Mensajero/genética , ARN Viral/química , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
EMBO J ; 29(11): 1851-64, 2010 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-20436455

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) expresses numerous intronless mRNAs that are unable to access splicing-dependent cellular mRNA nuclear export pathways. To circumvent this problem, KSHV encodes the open reading frame 57 (ORF57) protein, which orchestrates the formation of an export-competent virus ribonucleoprotein particle comprising the nuclear export complex hTREX, but not the exon-junction complex (EJC). Interestingly, EJCs stimulate mRNA translation, which raises the intriguing question of how intronless KSHV transcripts are efficiently translated. Herein, we show that ORF57 associates with components of the 48S pre-initiation complex and co-sediments with the 40S ribosomal subunits. Strikingly, we observed a direct interaction between ORF57 and PYM, a cellular protein that enhances translation by recruiting the 48S pre-initiation complex to newly exported mRNAs, through an interaction with the EJC. Moreover, detailed biochemical analysis suggests that ORF57 recruits PYM to intronless KSHV mRNA and PYM then facilitates the association of ORF57 and the cellular translation machinery. We, therefore, propose a model whereby ORF57 interacts directly with PYM to enhance translation of intronless KSHV transcripts.


Asunto(s)
Proteínas Portadoras/metabolismo , Herpesvirus Humano 8/metabolismo , Sistemas de Lectura Abierta/fisiología , ARN Mensajero/metabolismo , Transporte Activo de Núcleo Celular/genética , Proteínas Portadoras/genética , Citoplasma/genética , Citoplasma/metabolismo , Exones , Herpesvirus Humano 8/genética , Humanos , Empalme del ARN , Transporte de ARN/genética , ARN Mensajero/genética , Ribosomas/genética , Ribosomas/metabolismo , Virión/genética , Virión/metabolismo
12.
FEBS Lett ; 583(22): 3549-56, 2009 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-19850040

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) ORF57 plays a pivotal role in mediating the nuclear export of KSHV intronless transcripts. We demonstrate that ORF57 localises and dynamically trafficks through the nucleolus. To assess if nucleolar shuttling of ORF57 is important for the export of intronless viral mRNA, the ability of ORF57 to export mRNA was assessed in cells where the nucleolus was disrupted using Actinomycin D or DRB. We show that rapid disorganisation of the nucleolus was accompanied by a reduction in intronless virus mRNA export, suggesting that an intact nucleolus is essential for efficient KSHV ORF57-mediated intronless mRNA export.


Asunto(s)
Nucléolo Celular/metabolismo , Herpesvirus Humano 8/metabolismo , ARN Mensajero/metabolismo , Proteínas Virales/metabolismo , Transporte Activo de Núcleo Celular , Western Blotting , Línea Celular , Nucléolo Celular/virología , Recuperación de Fluorescencia tras Fotoblanqueo , Técnica del Anticuerpo Fluorescente Indirecta , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Herpesvirus Humano 8/genética , Humanos , Intrones , Transporte de ARN , ARN Mensajero/genética , ARN Viral/genética , ARN Viral/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Proteínas Virales/genética
13.
J Gen Virol ; 90(Pt 6): 1455-1460, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19264631

RESUMEN

Herpesvirus saimiri (HVS) ORF57 nucleocytoplasmic shuttle protein binds viral RNA and interacts with the cellular nuclear export adaptor protein, Aly, to access the TAP-mediated nuclear export pathway. This enables the efficient nuclear export of HVS intronless mRNAs. Herein, we extend these studies and demonstrate that ORF57 recruits several members of hTREX, namely Aly, UAP56 and hTHO-complex proteins, onto the viral mRNAs to assemble an export-competent ribonucleoprotein particle. Moreover, using a transdominant form of Aly which inhibits UAP56 and hTHO-complex association with viral intronless mRNA, we show that complete hTREX recruitment is required for efficient HVS mRNA nuclear export and replication.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Herpesvirus Saimiriino 2/fisiología , Proteínas Nucleares/metabolismo , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Replicación Viral , Transporte Activo de Núcleo Celular , Línea Celular , Humanos , Unión Proteica
14.
J Gen Virol ; 90(Pt 3): 596-601, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19218204

RESUMEN

The herpesvirus saimiri (HVS) ORF57 protein binds viral RNA, enabling the efficient nuclear export of intronless viral mRNAs. However, it is not known how ORF57 recognizes these viral mRNAs. In this study, a systematic evolution of ligands by exponential enrichment (SELEX) approach was used to select RNA sequences that are preferentially bound by the ORF57 protein. Results identified a recurring motif, GAAGRG, within the majority of selected RNAs, which is also present in many late HVS mRNAs. RNA immunopreciptations demonstrated that disruption of this motif within a viral intronless RNA ablates ORF57 binding. These data suggest that the GAAGRG motif may be required within a HVS intronless mRNA for recognition by the ORF57 protein.


Asunto(s)
Herpesvirus Saimiriino 2/genética , ARN Mensajero/genética , Proteínas Represoras/metabolismo , Elementos de Respuesta/genética , Transactivadores/metabolismo , Animales , Secuencia de Bases , Biología Computacional/métodos , Herpesvirus Saimiriino 2/metabolismo , Datos de Secuencia Molecular , ARN Mensajero/química , ARN Mensajero/metabolismo , Técnica SELEX de Producción de Aptámeros
15.
PLoS Pathog ; 4(10): e1000194, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18974867

RESUMEN

A cellular pre-mRNA undergoes various post-transcriptional processing events, including capping, splicing and polyadenylation prior to nuclear export. Splicing is particularly important for mRNA nuclear export as two distinct multi-protein complexes, known as human TREX (hTREX) and the exon-junction complex (EJC), are recruited to the mRNA in a splicing-dependent manner. In contrast, a number of Kaposi's sarcoma-associated herpesvirus (KSHV) lytic mRNAs lack introns and are exported by the virus-encoded ORF57 protein. Herein we show that ORF57 binds to intronless viral mRNAs and functions to recruit the complete hTREX complex, but not the EJC, in order assemble an export component viral ribonucleoprotein particle (vRNP). The formation of this vRNP is mediated by a direct interaction between ORF57 and the hTREX export adapter protein, Aly. Aly in turn interacts directly with the DEAD-box protein UAP56, which functions as a bridge to recruit the remaining hTREX proteins to the complex. Moreover, we show that a point mutation in ORF57 which disrupts the ORF57-Aly interaction leads to a failure in the ORF57-mediated recruitment of the entire hTREX complex to the intronless viral mRNA and inhibits the mRNAs subsequent nuclear export and virus replication. Furthermore, we have utilised a trans-dominant Aly mutant to prevent the assembly of the complete ORF57-hTREX complex; this results in a vRNP consisting of viral mRNA bound to ORF57, Aly and the nuclear export factor, TAP. Strikingly, although both the export adapter Aly and the export factor TAP were present on the viral mRNP, a dramatic decrease in intronless viral mRNA export and virus replication was observed in the absence of the remaining hTREX components (UAP56 and hTHO-complex). Together, these data provide the first direct evidence that the complete hTREX complex is essential for the export of KSHV intronless mRNAs and infectious virus production.


Asunto(s)
Exodesoxirribonucleasas/metabolismo , Herpesvirus Humano 8/metabolismo , Fosfoproteínas/metabolismo , Transporte de ARN , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , ARN Helicasas DEAD-box/metabolismo , Herpesvirus Humano 8/fisiología , Humanos , Complejos Multiproteicos/metabolismo , Complejo Proteico Nuclear de Unión a la Caperuza/metabolismo , Señales de Exportación Nuclear/fisiología , Proteínas Nucleares/metabolismo , Procesamiento Postranscripcional del ARN/fisiología , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo
16.
Virology ; 380(2): 191-202, 2008 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-18775548

RESUMEN

Localisation of both viral and cellular proteins to the nucleolus is determined by a variety of factors including nucleolar localisation signals (NoLSs), but how these signals operate is not clearly understood. The nucleolar trafficking of wild type viral proteins and chimeric proteins, which contain altered NoLSs, were compared to investigate the role of NoLSs in dynamic nucleolar trafficking. Three viral proteins from diverse viruses were selected which localised to the nucleolus; the coronavirus infectious bronchitis virus nucleocapsid (N) protein, the herpesvirus saimiri ORF57 protein and the HIV-1 Rev protein. The chimeric proteins were N protein and ORF57 protein which had their own NoLS replaced with those from ORF57 and Rev proteins, respectively. By analysing the sub-cellular localisation and trafficking of these viral proteins and their chimeras within and between nucleoli using confocal microscopy and photo-bleaching we show that NoLSs are responsible for different nucleolar localisations and trafficking rates.


Asunto(s)
Nucléolo Celular/virología , Señales de Clasificación de Proteína , Proteínas Virales/metabolismo , Animales , Fusión Artificial Génica , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Microscopía Confocal , Transporte de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Productos del Gen rev del Virus de la Inmunodeficiencia Humana/metabolismo
17.
Front Biosci ; 13: 2928-38, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17981766

RESUMEN

Herpesvirus saimiri (HVS) is the prototype gamma-2 herpesvirus and is a useful model to study the basic mechanisms of lytic replication in this herpesvirus subfamily. This review focuses upon the role of an essential lytic protein, ORF57, which is functionally conserved in all classes of herpesviruses. ORF57 is a multidomain, multifunctional protein responsible for both activation and repression of viral gene expression at a post-transcriptional level. ORF57-mediated repression of gene expression is determined by mRNA processing signals, in particular the presence of an intron within the target gene. This may also be linked to the ability of ORF57 to redistribute SC-35 and U2 splicing factors into specific nuclear domains. ORF57 also plays a pivotal role in transactivating viral gene expression by specifically mediating the nuclear export of HVS intronless transcripts. ORF57 has the ability to shuttle between the nucleus and the cytoplasm, bind viral RNA and recruit cellular nuclear export proteins, such as hTREX components and TAP, onto the viral mRNA. This enables the efficient nuclear export and cytoplasmic accumulation of virus intronless mRNA.


Asunto(s)
Núcleo Celular/virología , Regulación Viral de la Expresión Génica , Herpesvirus Saimiriino 2/metabolismo , Proteínas Represoras/fisiología , Transactivadores/fisiología , Proteínas Virales/fisiología , Transporte Activo de Núcleo Celular , Nucléolo Celular/metabolismo , Perfilación de la Expresión Génica , Modelos Biológicos , Unión Proteica , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas Represoras/genética , Transactivadores/genética , Transcripción Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
18.
Eur J Cell Biol ; 85(11): 1201-15, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17005296

RESUMEN

In budding yeast the final stages of the cell division cycle, cytokinesis and cell separation, are distinct events that require to be coupled, both together and with mitotic exit. Here we demonstrate that mutations in genes of the mitotic exit network (MEN) prevent cell separation and are synthetically lethal in combination with both cytokinesis and septation defective mutations. Analysis of the synthetic lethal phenotypes reveals that Iqg1p functions in combination with the MEN components, Tem1p, Cdc15p Dbf20p and Dbf2p to govern the re-polarization of the actin cytoskeleton to either side of the bud neck. In addition phosphorylation of the conserved PCH protein, Hof1p, is dependent upon these activities and requires actin ring assembly. Recruitment of Dbf2p to the bud neck is dependent upon actin ring assembly and correlates with Hof1p phosphorylation. Failure to phosphorylate Hof1p results in the increased stability of the protein and its persistence at the bud neck. These data establish a mechanistic dependency of cell separation upon an intermediate step requiring actomyosin ring assembly.


Asunto(s)
Citoesqueleto de Actina/fisiología , Citocinesis/fisiología , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Citoesqueleto de Actina/ultraestructura , Actinas/análisis , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis/fisiología , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Mutación , Fenotipo , Fosforilación , Proteínas Quinasas/análisis , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/análisis , Proteínas de Saccharomyces cerevisiae/genética , Proteínas Activadoras de ras GTPasa/genética
19.
Proc Natl Acad Sci U S A ; 103(41): 15190-5, 2006 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-17005724

RESUMEN

The nucleolus is the largest subnuclear structure and is plurifunctional in nature. Here, we demonstrate that nucleolar localization of a key herpesvirus regulatory protein is essential for its role in virus mRNA nuclear export. The herpesvirus saimiri ORF57 protein is a nucleocytoplasmic shuttle protein that is conserved in all herpesviruses and orchestrates the nuclear export of viral intronless mRNAs. We demonstrate that expression of the ORF57 protein induces nucleolar redistribution of human TREX (transcription/export) proteins that are involved in mRNA nuclear export. Moreover, we describe a previously unidentified nucleolar localization signal within ORF57 that is composed of two distinct nuclear localization signals. Intriguingly, point mutations that ablate ORF57 nucleolar localization lead to a failure of ORF57-mediated viral mRNA nuclear export. Furthermore, nucleolar retargeting of the ORF57 mutant was achieved by the incorporation of the HIV-1 Rev nucleolar localization signal, and analysis demonstrated that this modification was sufficient to restore viral mRNA nuclear export. This finding represents a unique and fundamental role for the nucleolus in nuclear export of viral mRNA.


Asunto(s)
Nucléolo Celular/metabolismo , Herpesvirus Saimiriino 2/genética , Intrones , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Transporte Activo de Núcleo Celular/genética , Animales , Aotidae , Línea Celular , Células HeLa , Herpesvirus Saimiriino 2/metabolismo , Humanos , Intrones/genética , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transactivadores/biosíntesis , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Proteínas Virales/metabolismo
20.
Curr Gene Ther ; 6(1): 1-15, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16475942

RESUMEN

Herpesviruses possess a number of characteristics which make them promising gene delivery vectors. These include their capacity to package large amounts of heterologous DNA and an ability to establish persistent, lifelong infections, where the viral genome remains as a circular non-integrated episome. Herpesvirus saimiri (HVS) is the prototype gamma-2 herpesvirus and is currently being developed as a potential gene delivery vector. In addition to the above properties, HVS-based vectors have the ability to infect a wide range of human cell lines and primary cultures with high efficiencies. Moreover, upon infection the viral genome persists as high copy number, circular, non-integrated episomes which segregate to progeny cells upon division. This allows the HVS-based vector to stably transduce a dividing cell population and provide sustained heterologous gene expression. As such, it offers the characteristics of an artificial chromosome combined with a highly efficient delivery system. This review aims to describe the assessment of HVS-based vectors in both in vitro and in vivo studies, highlighting new developments and possible applications for the treatment of genetic diseases.


Asunto(s)
Vectores Genéticos , Herpesvirus Saimiriino 2/genética , Transgenes , Animales , Fibroblastos/fisiología , Humanos , Ratas , Transformación Genética , Vesículas Transportadoras/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...