Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Biochem J ; 477(14): 2721-2733, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32639530

RESUMO

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.


Assuntos
Antígenos Virais de Tumores/metabolismo , Carcinoma de Célula de Merkel/virologia , Poliomavírus das Células de Merkel/patogenicidade , Neoplasias Cutâneas/virologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Antígenos Virais de Tumores/genética , Carcinoma de Célula de Merkel/genética , Carcinoma de Célula de Merkel/metabolismo , Carcinoma de Célula de Merkel/patologia , Movimento Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Imidazóis/farmacologia , MAP Quinase Quinase 4/metabolismo , Poliomavírus das Células de Merkel/imunologia , Fosfoproteínas Fosfatases/metabolismo , Piridinas/farmacologia , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
2.
PLoS Pathog ; 14(9): e1007276, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30188954

RESUMO

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.


Assuntos
Antígenos Virais de Tumores/fisiologia , Carcinoma de Célula de Merkel/etiologia , Poliomavírus das Células de Merkel/patogenicidade , Infecções por Polyomavirus/etiologia , Neoplasias Cutâneas/etiologia , Infecções Tumorais por Vírus/etiologia , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Carcinoma de Célula de Merkel/enzimologia , Carcinoma de Célula de Merkel/secundário , Movimento Celular , Células HEK293 , Humanos , Junções Intercelulares/patologia , Junções Intercelulares/fisiologia , Proteínas de Membrana/metabolismo , Poliomavírus das Células de Merkel/imunologia , Poliomavírus das Células de Merkel/fisiologia , Invasividade Neoplásica/patologia , Invasividade Neoplásica/fisiopatologia , Infecções por Polyomavirus/enzimologia , Infecções por Polyomavirus/patologia , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/patologia , Infecções Tumorais por Vírus/enzimologia , Infecções Tumorais por Vírus/patologia
3.
J Biol Chem ; 293(12): 4582-4590, 2018 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-29462791

RESUMO

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.


Assuntos
Carcinoma de Célula de Merkel/patologia , Movimento Celular , Canais de Cloreto/metabolismo , Poliomavírus das Células de Merkel/fisiologia , Infecções por Polyomavirus/complicações , Neoplasias Cutâneas/secundário , Infecções Tumorais por Vírus/complicações , Antígenos Virais de Tumores/genética , Antígenos Virais de Tumores/metabolismo , Carcinoma de Célula de Merkel/epidemiologia , Carcinoma de Célula de Merkel/virologia , Proliferação de Células , Canais de Cloreto/genética , Cloretos/metabolismo , Células HEK293 , Humanos , Incidência , Invasividade Neoplásica , Infecções por Polyomavirus/patologia , Infecções por Polyomavirus/virologia , Proteoma/análise , Neoplasias Cutâneas/epidemiologia , Neoplasias Cutâneas/virologia , Infecções Tumorais por Vírus/patologia , Infecções Tumorais por Vírus/virologia
4.
J Virol ; 90(20): 9543-55, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27512077

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/genética , Herpesvirus Humano 8/genética , Sarcoma de Kaposi/virologia , Proteínas Virais/genética , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina/métodos , Replicação do DNA/genética , DNA Viral/genética , Regulação Viral da Expressão Gênica/genética , Humanos , Proteínas Imediatamente Precoces/genética , Linfoma de Efusão Primária/genética , Linfoma de Efusão Primária/virologia , RNA Interferente Pequeno/genética , Transativadores/genética , Ativação Viral/genética , Latência Viral/genética , Replicação Viral/genética
5.
EMBO J ; 29(11): 1851-64, 2010 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-20436455

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Herpesvirus Humano 8/metabolismo , Fases de Leitura Aberta/fisiologia , RNA Mensageiro/metabolismo , Transporte Ativo do Núcleo Celular/genética , Proteínas de Transporte/genética , Citoplasma/genética , Citoplasma/metabolismo , Éxons , Herpesvirus Humano 8/genética , Humanos , Splicing de RNA , Transporte de RNA/genética , RNA Mensageiro/genética , Ribossomos/genética , Ribossomos/metabolismo , Vírion/genética , Vírion/metabolismo
6.
PLoS One ; 19(6): e0304870, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38900754

RESUMO

The underlying causes of breast cancer are diverse, however, there is a striking association between type 2 diabetes and poor patient outcomes. Platelet activation is a common feature of both type 2 diabetes and breast cancer and has been implicated in tumourigenesis through a multitude of pathways. Here transcriptomic analysis of type 2 diabetes patient-derived platelet microvesicles revealed an altered miRNA signature compared with normoglycaemic control patients. Interestingly, interrogation of these data identifies a shift towards an oncogenic signature in type 2 diabetes-derived platelet microvesicles, with increased levels of miRNAs implicated in breast cancer progression and poor prognosis. Functional studies demonstrate that platelet microvesicles isolated from type 2 diabetes patient blood are internalised by triple-negative breast cancer cells in vitro, and that co-incubation with type 2 diabetes patient-derived platelet microvesicles led to significantly increased expression of epithelial to mesenchymal transition markers and triple-negative breast cancer cell invasion compared with platelet microvesicles from healthy volunteers. Together, these data suggest that circulating PMVs in type 2 diabetes patients may contribute to the progression of triple-negative breast cancer.


Assuntos
Plaquetas , Micropartículas Derivadas de Células , Diabetes Mellitus Tipo 2 , MicroRNAs , Invasividade Neoplásica , Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Feminino , Plaquetas/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Micropartículas Derivadas de Células/metabolismo , Linhagem Celular Tumoral , Pessoa de Meia-Idade , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica
7.
PLoS Pathog ; 7(7): e1002138, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21814512

RESUMO

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.


Assuntos
Núcleo Celular/metabolismo , Herpesvirus Humano 8/metabolismo , Proteínas Nucleares/metabolismo , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Virais/metabolismo , Transporte Ativo do Núcleo Celular/genética , Núcleo Celular/genética , Núcleo Celular/virologia , Células HEK293 , Herpesvirus Humano 8/genética , Humanos , Proteínas Nucleares/genética , RNA Mensageiro/genética , RNA Viral/genética , Proteínas de Ligação a RNA/genética , Proteínas Virais/genética
8.
J Virol ; 85(15): 7881-91, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21593148

RESUMO

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.


Assuntos
Núcleo Celular/metabolismo , Herpesvirus Humano 6/genética , Mutação , Fases de Leitura Aberta , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Proteínas Virais/genética , Sequência de Bases , Transporte Biológico , Linhagem Celular , Primers do DNA , Humanos , Íntrons , Microscopia de Fluorescência , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Viral/química , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
PLoS Pathog ; 4(10): e1000194, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18974867

RESUMO

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.


Assuntos
Exodesoxirribonucleases/metabolismo , Herpesvirus Humano 8/metabolismo , Fosfoproteínas/metabolismo , Transporte de RNA , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , RNA Helicases DEAD-box/metabolismo , Herpesvirus Humano 8/fisiologia , Humanos , Complexos Multiproteicos/metabolismo , Complexo Proteico Nuclear de Ligação ao Cap/metabolismo , Sinais de Exportação Nuclear/fisiologia , Proteínas Nucleares/metabolismo , Processamento Pós-Transcricional do RNA/fisiologia , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo
10.
Chem Commun (Camb) ; 55(66): 9765-9768, 2019 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-31355394

RESUMO

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.


Assuntos
Acrilamidas/farmacologia , Chromobacterium/efeitos dos fármacos , Percepção de Quorum/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Chromobacterium/fisiologia
11.
Front Biosci ; 13: 2928-38, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17981766

RESUMO

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.


Assuntos
Núcleo Celular/virologia , Regulação Viral da Expressão Gênica , Herpesvirus Saimiriíneo 2/metabolismo , Proteínas Repressoras/fisiologia , Transativadores/fisiologia , Proteínas Virais/fisiologia , Transporte Ativo do Núcleo Celular , Nucléolo Celular/metabolismo , Perfilação da Expressão Gênica , Modelos Biológicos , Ligação Proteica , Processamento Pós-Transcricional do RNA , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Proteínas Repressoras/genética , Transativadores/genética , Transcrição Gênica , Proteínas Virais/genética , Proteínas Virais/metabolismo
12.
J Invest Dermatol ; 138(11): 2343-2354, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29777657

RESUMO

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.


Assuntos
Carcinoma de Célula de Merkel/imunologia , Poliomavírus das Células de Merkel/fisiologia , MicroRNAs/genética , Neutrófilos/imunologia , Infecções por Polyomavirus/imunologia , RNA Viral/genética , Infecções Tumorais por Vírus/imunologia , Antígenos Nucleares/genética , Antígenos Virais de Tumores/genética , Autoantígenos/genética , Carcinoma de Célula de Merkel/genética , Quimiotaxia , Células HEK293 , Humanos , Evasão da Resposta Imune , Imunidade Inata/genética , Interleucina-8/metabolismo , Infecções por Polyomavirus/genética , Infecções Tumorais por Vírus/genética
13.
Eur J Cell Biol ; 85(11): 1201-15, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17005296

RESUMO

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.


Assuntos
Citoesqueleto de Actina/fisiologia , Citocinese/fisiologia , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Citoesqueleto de Actina/ultraestrutura , Actinas/análise , Proteínas de Ciclo Celular/análise , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose/fisiologia , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Mutação , Fenótipo , Fosforilação , Proteínas Quinases/análise , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/ultraestrutura , Proteínas de Saccharomyces cerevisiae/análise , Proteínas de Saccharomyces cerevisiae/genética , Proteínas Ativadoras de ras GTPase/genética
14.
Curr Gene Ther ; 6(1): 1-15, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16475942

RESUMO

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.


Assuntos
Vetores Genéticos , Herpesvirus Saimiriíneo 2/genética , Transgenes , Animais , Fibroblastos/fisiologia , Humanos , Ratos , Transformação Genética , Vesículas Transportadoras/fisiologia
15.
Biochem J ; 387(Pt 2): 295-308, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15537388

RESUMO

HVS (herpesvirus saimiri) is the prototype gamma-2 herpesvirus. This is a subfamily of herpesviruses gaining importance since the identification of the first human gamma-2 herpesvirus, Kaposi's sarcoma-associated herpesvirus. The HVS ORF 57 (open reading frame 57) protein is a multifunctional transregulatory protein homologous with genes identified in all classes of herpesviruses. Recent work has demonstrated that ORF 57 has the ability to bind viral RNA, shuttles between the nucleus and cytoplasm and promotes the nuclear export of viral transcripts. In the present study, we show that ORF 57 shuttles between the nucleus and cytoplasm in a CRM-1 (chromosomal region maintenance 1)-independent manner. ORF 57 interacts with the mRNA export factor REF (RNA export factor) and two other components of the exon junction complex, Y14 and Magoh. The association of ORF 57 with REF stimulates recruitment of the cellular mRNA export factor TAP (Tip-associated protein), and HVS infection triggers the relocalization of REF and TAP from the nuclear speckles to several large clumps within the cell. Using a dominant-negative form of TAP and RNA interference to deplete TAP, we show that it is essential for bulk mRNA export in mammalian cells and is required for ORF 57-mediated viral RNA export. Furthermore, we show that the disruption of TAP reduces viral replication. These results indicate that HVS utilizes ORF 57 to recruit components of the exon junction complex and subsequently TAP to promote viral RNA export through the cellular mRNA export pathway.


Assuntos
Herpesvirus Saimiriíneo 2/fisiologia , Proteínas de Transporte Nucleocitoplasmático/fisiologia , Transporte de RNA/fisiologia , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Proteínas Repressoras/fisiologia , Transativadores/fisiologia , Proteínas Virais/fisiologia , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Células COS , Núcleo Celular/metabolismo , Chlorocebus aethiops , Carioferinas/metabolismo , Membrana Nuclear/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Proteína Exportina 1
16.
Int J Oncol ; 45(6): 2181-6, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25269471

RESUMO

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.


Assuntos
Melanoma/genética , Proteínas de Neoplasias/biossíntese , RNA Longo não Codificante/genética , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Melanoma/etiologia , Melanoma/patologia , Metástase Neoplásica , Prognóstico
17.
FEBS Lett ; 583(22): 3549-56, 2009 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-19850040

RESUMO

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.


Assuntos
Nucléolo Celular/metabolismo , Herpesvirus Humano 8/metabolismo , RNA Mensageiro/metabolismo , Proteínas Virais/metabolismo , Transporte Ativo do Núcleo Celular , Western Blotting , Linhagem Celular , Nucléolo Celular/virologia , Recuperação de Fluorescência Após Fotodegradação , Técnica Indireta de Fluorescência para Anticorpo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Herpesvirus Humano 8/genética , Humanos , Íntrons , Transporte de RNA , RNA Mensageiro/genética , RNA Viral/genética , RNA Viral/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Proteínas Virais/genética
18.
J Gen Virol ; 90(Pt 6): 1455-1460, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19264631

RESUMO

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.


Assuntos
RNA Helicases DEAD-box/metabolismo , Herpesvirus Saimiriíneo 2/fisiologia , Proteínas Nucleares/metabolismo , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Replicação Viral , Transporte Ativo do Núcleo Celular , Linhagem Celular , Humanos , Ligação Proteica
19.
J Gen Virol ; 90(Pt 3): 596-601, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19218204

RESUMO

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.


Assuntos
Herpesvirus Saimiriíneo 2/genética , RNA Mensageiro/genética , Proteínas Repressoras/metabolismo , Elementos de Resposta/genética , Transativadores/metabolismo , Animais , Sequência de Bases , Biologia Computacional/métodos , Herpesvirus Saimiriíneo 2/metabolismo , Dados de Sequência Molecular , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Técnica de Seleção de Aptâmeros
20.
Virology ; 380(2): 191-202, 2008 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-18775548

RESUMO

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.


Assuntos
Nucléolo Celular/virologia , Sinais Direcionadores de Proteínas , Proteínas Virais/metabolismo , Animais , Fusão Gênica Artificial , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Microscopia Confocal , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transativadores/genética , Transativadores/metabolismo , Produtos do Gene rev do Vírus da Imunodeficiência Humana/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA